Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Soc Trans ; 49(5): 2021-2035, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34623375

RESUMEN

The RAS superfamily of small GTPases regulates major physiological cellular processes. Mutation or deregulation of these small GTPases, their regulators and/or their effectors are associated with many diseases including cancer. Hence, targeting these classes of proteins is an important therapeutic strategy in cancer. This has been recently achieved with the approval of the first KRASG12C covalent inhibitors for the clinic. However, many other mutants and small GTPases are still considered as 'undruggable' with small molecule inhibitors because of a lack of well-defined pocket(s) at their surface. Therefore, alternative therapeutic strategies have been developed to target these proteins. In this review, we discuss the use of intracellular antibodies and derivatives - reagents that bind their antigen inside the cells - for the discovery of novel inhibitory mechanisms, targetable features and therapeutic strategies to inhibit small GTPases and their downstream pathways. These reagents are also versatile tools used to better understand the biological mechanisms regulated by small GTPases and to accelerate the drug discovery process.


Asunto(s)
Anticuerpos/metabolismo , Proteínas de Repetición de Anquirina Diseñadas/metabolismo , Descubrimiento de Drogas/métodos , Proteínas de Unión al GTP Monoméricas/metabolismo , Neoplasias/enzimología , Transducción de Señal/efectos de los fármacos , Anticuerpos de Dominio Único/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Proteínas de Repetición de Anquirina Diseñadas/farmacología , Humanos , Terapia Molecular Dirigida/métodos , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/inmunología , Neoplasias/tratamiento farmacológico , Unión Proteica , Proteolisis/efectos de los fármacos , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología
2.
Front Immunol ; 12: 704408, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489954

RESUMEN

On murine T cells, mono-ADP ribosyltransferase ARTC2.2 catalyzes ADP-ribosylation of various surface proteins when nicotinamide adenine dinucleotide (NAD+) is released into the extracellular compartment. Covalent ADP-ribosylation of the P2X7 receptor by ARTC2.2 thereby represents an additional mechanism of activation, complementary to its triggering by extracellular ATP. P2X7 is a multifaceted receptor that may represents a potential target in inflammatory, and neurodegenerative diseases, as well as in cancer. We present herein an experimental approach using intramuscular injection of recombinant AAV vectors (rAAV) encoding nanobody-based biologics targeting ARTC2.2 or P2X7. We demonstrate the ability of these in vivo generated biologics to potently and durably block P2X7 or ARTC2.2 activities in vivo, or in contrast, to potentiate NAD+- or ATP-induced activation of P2X7. We additionally demonstrate the ability of rAAV-encoded functional heavy chain antibodies to elicit long-term depletion of T cells expressing high levels of ARTC2.2 or P2X7. Our approach of using rAAV to generate functional nanobody-based biologics in vivo appears promising to evaluate the role of ARTC2.2 and P2X7 in murine acute as well as chronic disease models.


Asunto(s)
ADP Ribosa Transferasas , Productos Biológicos/inmunología , Dependovirus , Vectores Genéticos , Depleción Linfocítica , Receptores Purinérgicos P2X7/inmunología , Anticuerpos de Dominio Único , ADP Ribosa Transferasas/antagonistas & inhibidores , ADP Ribosa Transferasas/inmunología , Animales , Ratones , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología
3.
Recent Pat Anticancer Drug Discov ; 16(4): 552-562, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34365930

RESUMEN

OBJECTIVES: The aim of this study was to formulate fluorescent-labeled targeted immunoliposome to visualize the delivery and distribution of drugs in real-time. METHODS: In this study, fluorescent-labeled liposomes were decorated with anti-HER2 VHH or Herceptin to improve the monitoring of intracellular drug delivery and tumor cell tracking with minimal side effects. The conjugation efficiency of antibodies was analyzed by SDS-PAGE silver staining. In addition, the physicochemical characterization of liposomes was performed using DLS and TEM. Finally, confocal microscopy visualized nanoparticles in the target cells. RESULTS: Quantitative and qualitative methods characterized the intracellular uptake of 110±10 nm particles with near 70% conjugation efficiency. In addition, live-cell trafficking during hours of incubation was monitored by wide-field microscopy imaging. The results show that the fluorescent- labeled nanoparticles can specifically bind to HER2-positive breast cancer with minimal off-target delivery. CONCLUSION: These nanoparticles can have several applications in personalized medicine, especially drug delivery and real-time visualization of cancer therapy. Moreover, this method also can be applied in the targeted delivery of contrast agents in imaging and thermotherapy.


Asunto(s)
Neoplasias de la Mama/terapia , Nanopartículas , Receptor ErbB-2/inmunología , Anticuerpos de Dominio Único/inmunología , Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Medios de Contraste/administración & dosificación , Sistemas de Liberación de Medicamentos , Femenino , Fluorescencia , Humanos , Liposomas , Microscopía Confocal , Imagen Óptica/métodos , Medicina de Precisión/métodos , Trastuzumab/farmacología
4.
Biochem Biophys Res Commun ; 566: 45-52, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34116356

RESUMEN

A newly-emergent beta-coronavirus, SARS-CoV-2, rapidly has become a pandemic since 2020. It is a serious respiratory disease and caused more than 100 million of deaths in the world. WHO named it COVIA-19 and there is no effective targeted drug for it. The main treatment strategies include chemical medicine, traditional Chinese medicine (TCM) and biologics. Due to SARS-CoV-2 uses the spike proteins (S proteins) on its envelope to infect human cells, monoclonal antibodies that neutralize the S protein have become one of the hot research areas in the current research and treatment of SARS-CoV-2. In this study, we reviewed the antibodies that have been reported to have neutralizing activity against the SARS-CoV-2 infection. According to their different binding epitope regions in RBD or NTD, they are classified, and the mechanism of the representative antibodies in each category is discussed in depth, which provides potential foundation for future antibody and vaccine therapy and the development of antibody cocktails against SARS-CoV-2 mutants.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , COVID-19/virología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Enzima Convertidora de Angiotensina 2/química , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/química , Anticuerpos Antivirales/uso terapéutico , COVID-19/terapia , Vacunas contra la COVID-19/inmunología , Epítopos/inmunología , Humanos , Modelos Moleculares , Pruebas de Neutralización , Pandemias , Dominios y Motivos de Interacción de Proteínas , Receptores Virales/química , SARS-CoV-2/genética , Anticuerpos de Dominio Único/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética
5.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33903242

RESUMEN

Infection with obligatory intracellular bacteria is difficult to treat, as intracellular targets and delivery methods of therapeutics are not well known. Ehrlichia translocated factor-1 (Etf-1), a type IV secretion system (T4SS) effector, is a primary virulence factor for an obligatory intracellular bacterium, Ehrlichia chaffeensis In this study, we developed Etf-1-specific nanobodies (Nbs) by immunizing a llama to determine if intracellular Nbs block Etf-1 functions and Ehrlichia infection. Of 24 distinct anti-Etf-1 Nbs, NbD7 blocked mitochondrial localization of Etf-1-GFP in cotransfected cells. NbD7 and control Nb (NbD3) bound to different regions of Etf-1. Size-exclusion chromatography showed that the NbD7 and Etf-1 complex was more stable than the NbD3 and Etf-1 complex. Intracellular expression of NbD7 inhibited three activities of Etf-1 and E. chaffeensis: up-regulation of mitochondrial manganese superoxide dismutase, reduction of intracellular reactive oxygen species, and inhibition of cellular apoptosis. Consequently, intracellular NbD7 inhibited Ehrlichia infection, whereas NbD3 did not. To safely and effectively deliver Nbs into the host cell cytoplasm, NbD7 was conjugated to cyclized cell-permeable peptide 12 (CPP12-NbD7). CPP12-NbD7 effectively entered mammalian cells and abrogated the blockade of cellular apoptosis caused by E. chaffeensis and inhibited infection by E. chaffeensis in cell culture and in a severe combined-immunodeficiency mouse model. Our results demonstrate the development of an Nb that interferes with T4SS effector functions and intracellular pathogen infection, along with an intracellular delivery method for this Nb. This strategy should overcome current barriers to advance mechanistic research and develop therapies complementary or alternative to the current broad-spectrum antibiotic.


Asunto(s)
Ehrlichia chaffeensis/efectos de los fármacos , Ehrlichiosis/tratamiento farmacológico , Anticuerpos de Dominio Único/farmacología , Sistemas de Secreción Tipo IV/genética , Animales , Apoptosis/genética , Subgrupos de Linfocitos B/inmunología , Ehrlichia chaffeensis/genética , Ehrlichia chaffeensis/inmunología , Ehrlichia chaffeensis/patogenicidad , Ehrlichiosis/genética , Ehrlichiosis/inmunología , Ehrlichiosis/patología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Especies Reactivas de Oxígeno/metabolismo , Anticuerpos de Dominio Único/inmunología , Sistemas de Secreción Tipo IV/antagonistas & inhibidores , Sistemas de Secreción Tipo IV/inmunología , Factores de Virulencia
6.
Inflammation ; 44(4): 1620-1628, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33751358

RESUMEN

The currently used anti-cytokine therapeutic antibodies cannot selectively neutralize pathogenic cytokine signalling that cause collateral damage to protective signalling cascades. The single domain chain firstly discovered in Camelidae displays fully functional ability in antigen-binding against variable targets, which has been seemed as attractive candidates for the next-generation biologic drug study. In this study, we established a simple prokaryotic expression system for a dual target-directed single domain-based fusion protein against the interleukin-6 receptor and human serum, albumin, the recombinant anti-IL-6R fusion protein (VHH-0031). VHH-0031 exhibited potent anti-inflammatory effects produced by LPS on cell RAW264.7, where the major cytokines and NO production were downregulated after 24 h incubation with VHH-0031 in a dose-dependent manner. In vivo, VHH-0031 presented significant effects on the degree reduction of joint swelling in the adjuvant-induced arthritis (AIA) rat, having a healthier appearance compared with the dexamethasone. The expression level of JNK protein in the VHH-0031 group was significantly decreased, demonstrating that VHH-0031 provides a low-cost and desirable effect in the treatment of more widely patients.


Asunto(s)
Antiinflamatorios/inmunología , Artritis Experimental/tratamiento farmacológico , Interleucina-6/antagonistas & inhibidores , Albúmina Sérica Humana/antagonistas & inhibidores , Anticuerpos de Dominio Único/inmunología , Animales , Antiinflamatorios/uso terapéutico , Especificidad de Anticuerpos , Artritis Experimental/inmunología , Citocinas/metabolismo , ADN Complementario/genética , Dexametasona/uso terapéutico , Evaluación Preclínica de Medicamentos , Inducción Enzimática/efectos de los fármacos , Humanos , Interleucina-6/inmunología , Lipopolisacáridos/toxicidad , MAP Quinasa Quinasa 4/biosíntesis , MAP Quinasa Quinasa 4/genética , Ratones , Modelos Moleculares , Terapia Molecular Dirigida , Óxido Nítrico/metabolismo , Conformación Proteica , Células RAW 264.7 , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Albúmina Sérica Humana/inmunología , Anticuerpos de Dominio Único/genética
7.
Nature ; 587(7832): 18, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33097846
8.
Anal Chem ; 92(1): 983-990, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31742384

RESUMEN

Due to exceptional electron-accepting ability, light-absorption, and a delocalized conjugated structure, buckminsterfullerene (C60) has attracted fascinating interest in the field of organic solar cells. However, poor delocalization and accumulation of electrons for pristine C60 in physiological aqueous solution and difficulties in conjugation with biomolecules limit its extended photovoltaic applications in bioassay. Herein, we reported the noncovalent coupling of C60 to an electronically complementary porphyrin-derived metal-organic framework (PCN-224) with carboxyl-group terminals. Such assembly not only offered a friendly interface for bioconjugation but also resulted in a long-range ordering C60@PCN-224 donor-acceptor system that demonstrated an unprecedented photocurrent enhancement up to 10 times with respect to each component. As an example, by further cooperating with Nanobodies, the as-prepared C60@PCN-224 was applied to a photoelectrochemical (PEC) immunosensor for S100 calcium-binding protein B with by far the most promising detection activities. This work may open a new venue to unlock the great potential of C60 in PEC biosensing with excellent performances.


Asunto(s)
Técnicas Electroquímicas/métodos , Fulerenos/química , Inmunoensayo/métodos , Estructuras Metalorgánicas/química , Biomarcadores/sangre , Técnicas Biosensibles/métodos , Fulerenos/efectos de la radiación , Humanos , Luz , Límite de Detección , Estructuras Metalorgánicas/efectos de la radiación , Porfirinas/química , Porfirinas/efectos de la radiación , Prueba de Estudio Conceptual , Reproducibilidad de los Resultados , Subunidad beta de la Proteína de Unión al Calcio S100/sangre , Subunidad beta de la Proteína de Unión al Calcio S100/inmunología , Anticuerpos de Dominio Único/inmunología
9.
Talanta ; 195: 55-61, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30625581

RESUMEN

Mimotopes could replace mycotoxins and their conjugates to develop immunoassay methods. The mimotopes obtained by phage display technology were mainly using monoclonal antibodies or polyclonal antibodies as targets. However, the mimotope of recombinant antibody has not been selected and applied to immunoassay for mycotoxin. The purpose of this study was to prove that an immunoassay for mycotoxin could be developed based on both recombinant antibody and its mimotope. Using aflatoxin B1 (AFB1) as a model system, mimotopes of an aflatoxin nanobody Nb28 were screened by phage display. A rapid magnetic beads-based directed competitive ELISA (MB-dcELISA) was developed utilizing Nb28 and its mimotope ME17. The 50% inhibitory concentration and the detection limit of the MB-dcELISA were 0.75 and 0.13 ng/mL, respectively, with a linear range of 0.24-2.21 ng/mL. Further validation study indicated good recovery (84.2-116.2%) with low coefficient of variable (2.2%-15.9%) in spiked corn, rice, peanut, feedstuff, corn germ oil and peanut oil samples. The developed immunoassay based on nanobody and mimotope provides a new strategy for the monitoring of AFB1 and other toxic small molecular weight compounds.


Asunto(s)
Aflatoxina B1/análisis , Contaminación de Alimentos/análisis , Anticuerpos de Dominio Único/inmunología , Aflatoxina B1/inmunología , Alimentación Animal/análisis , Arachis , Biotinilación , Ensayo de Inmunoadsorción Enzimática/métodos , Oryza , Biblioteca de Péptidos , Aceites de Plantas/análisis , Zea mays
10.
Toxins (Basel) ; 10(4)2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29596324

RESUMEN

Toxic effects triggered by crotalic envenoming are mainly related to crotoxin (CTX), composed of a phospholipase A2 (CB) and a subunit with no toxic activity (CA). Camelids produce immunoglobulins G devoid of light chains, in which the antigen recognition domain is called VHH. Given their unique characteristics, VHHs were selected using Phage Display against CTX from Crotalus durissus terrificus. After three rounds of biopanning, four sequence profiles for CB (KF498602, KF498603, KF498604, and KF498605) and one for CA (KF498606) were revealed. All clones presented the VHH hallmark in FR2 and a long CDR3, with the exception of KF498606. After expressing pET22b-VHHs in E. coli, approximately 2 to 6 mg of protein per liter of culture were obtained. When tested for cross-reactivity, VHHs presented specificity for the Crotalus genus and were capable of recognizing CB through Western blot. KF498602 and KF498604 showed thermostability, and displayed affinity constants for CTX in the micro or nanomolar range. They inhibited in vitro CTX PLA2 activity, and CB cytotoxicity. Furthermore, KF498604 inhibited the CTX-induced myotoxicity in mice by 78.8%. Molecular docking revealed that KF498604 interacts with the CA–CB interface of CTX, seeming to block substrate access. Selected VHHs may be alternatives for the crotalic envenoming treatment.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Crotoxina/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Crotoxina/toxicidad , Escherichia coli/genética , Masculino , Ratones , Simulación del Acoplamiento Molecular , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/tratamiento farmacológico , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/uso terapéutico , Mordeduras de Serpientes/diagnóstico , Mordeduras de Serpientes/terapia
11.
Sci Rep ; 7(1): 14289, 2017 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-29084989

RESUMEN

The cell surface ecto-enzyme CD38 is a promising target antigen for the treatment of hematological malignancies, as illustrated by the recent approval of daratumumab for the treatment of multiple myeloma. Our aim was to evaluate the potential of CD38-specific nanobodies as novel diagnostics for hematological malignancies. We successfully identified 22 CD38-specific nanobody families using phage display technology from immunized llamas. Crossblockade analyses and in-tandem epitope binning revealed that the nanobodies recognize three different non-overlapping epitopes, with four nanobody families binding complementary to daratumumab. Three nanobody families inhibit the enzymatic activity of CD38 in vitro, while two others were found to act as enhancers. In vivo, fluorochrome-conjugated CD38 nanobodies efficiently reach CD38 expressing tumors in a rodent model within 2 hours after intravenous injection, thereby allowing for convenient same day in vivo tumor imaging. These nanobodies represent highly specific tools for modulating the enzymatic activity of CD38 and for diagnostic monitoring CD38-expressing tumors.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Glicoproteínas de Membrana/metabolismo , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/tratamiento farmacológico , Anticuerpos de Dominio Único/inmunología , ADP-Ribosil Ciclasa 1/inmunología , Animales , Camélidos del Nuevo Mundo , Línea Celular Tumoral , Técnicas de Visualización de Superficie Celular , Modelos Animales de Enfermedad , Epítopos/inmunología , Colorantes Fluorescentes , Humanos , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Desnudos , Mieloma Múltiple/patología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Bioorg Med Chem Lett ; 27(21): 4844-4848, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28974337

RESUMEN

Survivin, an inhibitor of the apoptosis protein family, is a potent tumor marker for diagnosis and prognosis. The enzyme-linked immunosorbent assay (ELISA) is one of the methods that has been used for detection of survivin. However, ELISA has several disadvantages caused by the use of conventional antibodies, and we have therefore been trying to develop a novel ELISA system using camelid single-domain antibodies (VHHs) as advantageous replacements. Here we report a supplemental approach to improve the VHH-polyclonal antibody sandwich ELISA for survivin detection. Iodoacetyl-functionalized pullulan was synthesized, and its thiol reactivity was characterized by a model reaction with l-cysteine. The thiophilic pullulan was applied to an immunoassay asan additive upon coating of standard assay plates with an anti-survivin VHH fusion protein with C-terminal cysteine. The results showed that the mole ratio of the additive to VHH had a significant effect on the consequent response. Mole ratios of 0.07, 0.7, and 7 led to 90% lower, 15% higher, and 69% lower responses, respectively, than the response of a positive control in which no additive was used. The background levels observed in any additive conditions were as low as that of a negative control lacking both VHH and the additive. These results indicate the applicability of the thiol-reactive pullulan as a response enhancer to VHH-based ELISA.


Asunto(s)
Anticuerpos/inmunología , Ensayo de Inmunoadsorción Enzimática , Glucanos/química , Proteínas Inhibidoras de la Apoptosis/análisis , Anticuerpos de Dominio Único/inmunología , Animales , Anticuerpos/química , Cisteína/química , Humanos , Ácido Yodoacético/química , Anticuerpos de Dominio Único/química , Survivin
13.
Proc Natl Acad Sci U S A ; 114(38): 10184-10189, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28874561

RESUMEN

CD47 is an antiphagocytic ligand broadly expressed on normal and malignant tissues that delivers an inhibitory signal through the receptor signal regulatory protein alpha (SIRPα). Inhibitors of the CD47-SIRPα interaction improve antitumor antibody responses by enhancing antibody-dependent cellular phagocytosis (ADCP) in xenograft models. Endogenous expression of CD47 on a variety of cell types, including erythrocytes, creates a formidable antigen sink that may limit the efficacy of CD47-targeting therapies. We generated a nanobody, A4, that blocks the CD47-SIRPα interaction. A4 synergizes with anti-PD-L1, but not anti-CTLA4, therapy in the syngeneic B16F10 melanoma model. Neither increased dosing nor half-life extension by fusion of A4 to IgG2a Fc (A4Fc) overcame the issue of an antigen sink or, in the case of A4Fc, systemic toxicity. Generation of a B16F10 cell line that secretes the A4 nanobody showed that an enhanced response to several immune therapies requires near-complete blockade of CD47 in the tumor microenvironment. Thus, strategies to localize CD47 blockade to tumors may be particularly valuable for immune therapy.


Asunto(s)
Antígeno CD47/antagonistas & inhibidores , Inmunoterapia/métodos , Melanoma Experimental/terapia , Anticuerpos de Dominio Único/uso terapéutico , Anemia/inducido químicamente , Animales , Antígeno CD47/inmunología , Evaluación Preclínica de Medicamentos , Ratones Endogámicos C57BL , Fagocitosis , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología , Microambiente Tumoral
14.
Sci Rep ; 6: 32256, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27558409

RESUMEN

Candida albicans (C. albicans) is an important human commensal and opportunistic fungal pathogen. Secreted aspartyl proteinases (Saps) are a major virulence trait of C. albicans, and among these proteases Sap2 has the highest expression levels. It is possible that antibodies against Sap2 could provide an antifungal effect. In this study, two phages displaying anti-rSap2 single chain variable fragments (scFvs) were screened from human single fold scFv libraries, and their potential therapeutic roles were evaluated using a murine model infected by C. albicans. The in vivo efficacies were assessed by mortality rates, fungal burden and histological examination. Overall survival rates were significantly increased while the colony counts and infectious foci were significantly decreased after treatment with the scFv-phages relative to the control groups. In order to investigate the immune response provoked by scFv-phages, three kinds of cytokines (Th1, Th2 and Th17 types) were measured and a clear immune response was observed. These findings suggest that anti-rSap2 scFv-phages have potential in the therapy of systemic infection caused by C. albicans.


Asunto(s)
Anticuerpos Antifúngicos/farmacología , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Candida albicans/inmunología , Candidiasis/tratamiento farmacológico , Proteínas Fúngicas/antagonistas & inhibidores , Anticuerpos de Dominio Único/farmacología , Animales , Anticuerpos Antifúngicos/química , Anticuerpos Antifúngicos/genética , Anticuerpos Antifúngicos/inmunología , Ácido Aspártico Endopeptidasas/inmunología , Bacteriófago M13 , Candidiasis/genética , Candidiasis/inmunología , Candidiasis/patología , Modelos Animales de Enfermedad , Femenino , Proteínas Fúngicas/inmunología , Humanos , Ratones Endogámicos BALB C , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , omegacloroacetofenona
15.
Clin Immunol ; 169: 128-138, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27373969

RESUMEN

Vγ9Vδ2-T cells constitute the predominant subset of γδ-T cells in human peripheral blood and have been shown to play an important role in antimicrobial and antitumor immune responses. Several efforts have been initiated to exploit these cells for cancer immunotherapy, e.g. by using phosphoantigens, adoptive cell transfer, and by a bispecific monoclonal antibody based approach. Here, we report the generation of a novel set of Vγ9Vδ2-T cell specific VHH (or nanobody). VHH have several advantages compared to conventional antibodies related to their small size, stability, ease of generating multispecific molecules and low immunogenicity. With high specificity and affinity, the anti-Vγ9Vδ2-T cell receptor VHHs are shown to be useful for FACS, MACS and immunocytochemistry. In addition, some VHH were found to specifically activate Vγ9Vδ2-T cells. Besides being of possible immunotherapeutic value, these single domain antibodies will be of great value in the further study of this important immune effector cell subset.


Asunto(s)
Afinidad de Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Anticuerpos de Dominio Único/inmunología , Linfocitos T/inmunología , Animales , Camélidos del Nuevo Mundo/inmunología , Células Cultivadas , Citometría de Flujo/métodos , Humanos , Inmunohistoquímica/métodos , Separación Inmunomagnética/métodos , Células Jurkat , Microscopía Fluorescente , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Reproducibilidad de los Resultados , Linfocitos T/metabolismo
16.
Biochimie ; 111: 82-106, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25656912

RESUMEN

The deposition of misfolded peptides and proteins in the form of amyloid fibrils is the hallmark of nearly fifty medical disorders, including Alzheimer's disease, Parkinson's disease, prion diseases and type II diabetes. These disorders, referred to as amyloidoses, generally become apparent late in life. Their psycho-sociological and economic incidence in western societies will be therefore considerable in the coming decades due to the ageing of the population. Neither preventing nor curative treatments are available yet. These disorders constitute therefore a medical challenge of great importance. Thus, an extensive research is being carried out to understand, at the molecular level, (i) how amyloidogenic proteins misfold and convert from their soluble form into amyloid fibrils, and (ii) how these aggregates or some of their oligomeric precursor species are toxic. The formation of amyloid fibrils proceeds through a complex nucleation/polymerisation mechanism with the formation of various species, including small oligomers. In this review, we focus on how VHHs or nanobodies, the antigen-binding domains of camelid heavy-chain antibodies, are being increasingly used to characterise each of the species formed on the pathway of fibril formation in terms of structure, stability, kinetics of formation and toxicity. We first introduce the characteristic features of nanobodies compared to those of conventional antibody fragments. Thereafter, we discuss how nanobodies, due to their unique properties, are used as probes to dissect the molecular mechanisms of misfolding and aggregation of six proteins associated with diseases, i.e. human lysozyme, ß2-microglobulin, α-synuclein, prion, polyadenylate binding protein nuclear 1 and amyloid ß-peptide. A brief general presentation of each disease and the associated peptide/protein is also provided. In addition, we discuss how nanobodies could be used as early diagnostic tools and as novel strategies to treat diseases associated with protein misfolding and aggregation.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Cadenas Pesadas de Inmunoglobulina/uso terapéutico , Agregación Patológica de Proteínas/tratamiento farmacológico , Deficiencias en la Proteostasis/tratamiento farmacológico , Anticuerpos de Dominio Único/uso terapéutico , Animales , Camélidos del Nuevo Mundo/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Agregación Patológica de Proteínas/inmunología , Deficiencias en la Proteostasis/inmunología , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología
17.
J Virol ; 88(15): 8278-96, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829341

RESUMEN

UNLABELLED: Influenza virus neuraminidase (NA) is an interesting target of small-molecule antiviral drugs. We isolated a set of H5N1 NA-specific single-domain antibodies (N1-VHHm) and evaluated their in vitro and in vivo antiviral potential. Two of them inhibited the NA activity and in vitro replication of clade 1 and 2 H5N1 viruses. We then generated bivalent derivatives of N1-VHHm by two methods. First, we made N1-VHHb by genetically joining two N1-VHHm moieties with a flexible linker. Second, bivalent N1-VHH-Fc proteins were obtained by genetic fusion of the N1-VHHm moiety with the crystallizable region of mouse IgG2a (Fc). The in vitro antiviral potency against H5N1 of both bivalent N1-VHHb formats was 30- to 240-fold higher than that of their monovalent counterparts, with 50% inhibitory concentrations in the low nanomolar range. Moreover, single-dose prophylactic treatment with bivalent N1-VHHb or N1-VHH-Fc protected BALB/c mice against a lethal challenge with H5N1 virus, including an oseltamivir-resistant H5N1 variant. Surprisingly, an N1-VHH-Fc fusion without in vitro NA-inhibitory or antiviral activity also protected mice against an H5N1 challenge. Virus escape selection experiments indicated that one amino acid residue close to the catalytic site is required for N1-VHHm binding. We conclude that single-domain antibodies directed against influenza virus NA protect against H5N1 virus infection, and when engineered with a conventional Fc domain, they can do so in the absence of detectable NA-inhibitory activity. IMPORTANCE: Highly pathogenic H5N1 viruses are a zoonotic threat. Outbreaks of avian influenza caused by these viruses occur in many parts of the world and are associated with tremendous economic loss, and these viruses can cause very severe disease in humans. In such cases, small-molecule inhibitors of the viral NA are among the few treatment options for patients. However, treatment with such drugs often results in the emergence of resistant viruses. Here we show that single-domain antibody fragments that are specific for NA can bind and inhibit H5N1 viruses in vitro and can protect laboratory mice against a challenge with an H5N1 virus, including an oseltamivir-resistant virus. In addition, plant-produced VHH fused to a conventional Fc domain can protect in vivo even in the absence of NA-inhibitory activity. Thus, NA of influenza virus can be effectively targeted by single-domain antibody fragments, which are amenable to further engineering.


Asunto(s)
Antivirales/uso terapéutico , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Neuraminidasa/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/prevención & control , Anticuerpos de Dominio Único/uso terapéutico , Animales , Antivirales/inmunología , Modelos Animales de Enfermedad , Femenino , Subtipo H5N1 del Virus de la Influenza A/inmunología , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Anticuerpos de Dominio Único/inmunología , Resultado del Tratamiento
18.
Ther Deliv ; 4(10): 1321-36, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24116915

RESUMEN

Nanobodies (Nbs) are small antibody fragments derived from camelid heavy chain antibodies through recombinant gene technology. Their exceptional physicochemical properties, possibility of humanization and unique antigen recognition properties make them excellent candidates for targeted delivery of biologically active components. Several different therapeutic approaches based on the novel camelid Nbs have been developed to treat a wide range of diseases ranging from immune, bone, blood and neurological disorders; infectious diseases and cancer. This review provides a comprehensive overview of the current state of the use of camelid-derived Nbs as novel therapeutic agents against multiple diseases.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Camelus/inmunología , Inmunoterapia/métodos , Inmunotoxinas/uso terapéutico , Nanomedicina/métodos , Anticuerpos de Dominio Único/uso terapéutico , Animales , Especificidad de Anticuerpos , Química Farmacéutica , Portadores de Fármacos , Humanos , Inmunotoxinas/química , Inmunotoxinas/inmunología , Conformación Proteica , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Relación Estructura-Actividad
19.
Curr Opin Investig Drugs ; 10(11): 1212-24, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19876789

RESUMEN

Evolution has been continuously honing the design of antibodies to function as specific molecular markers that are able to alert the immune system to the presence of pathogenic antigens, and to recruit complement- and Fc receptor-bearing effector cells. During the past 25 years, the versatility of antibodies has been applied to several therapeutic applications. The development of new technologies, combined with data obtained using a new generation of antibody reagents, have allowed the adaptation of the design of antibodies to better match drug development requirements. Nanobodies are therapeutic proteins derived from the heavy-chain variable (VHH) domains that occur naturally in heavy-chain-only Ig molecules in camelidae. These VHH domains are the smallest known antigen-binding antibody fragments. Nanobodies can be easily produced in prokaryotic or eukaryotic host organisms, and their unique biophysical and pharmacological characteristics render these molecules ideal candidates for drug development. This review describes the structural properties of nanobodies and focuses on their unique features, which distinguishes these molecules from other antibody formats and small-molecule drugs. Possible therapeutic applications of nanobodies are discussed and data from phase I clinical trials of the novel 'first-in-class' anti-thrombotic agent ALX-0081 (Ablynx NV) are presented.


Asunto(s)
Diseño de Fármacos , Fragmentos de Inmunoglobulinas/uso terapéutico , Cadenas Pesadas de Inmunoglobulina/inmunología , Anticuerpos de Dominio Único/uso terapéutico , Animales , Anticuerpos/inmunología , Camélidos del Nuevo Mundo , Ensayos Clínicos Fase I como Asunto , Fibrinolíticos/inmunología , Fibrinolíticos/farmacología , Humanos , Fragmentos de Inmunoglobulinas/inmunología , Anticuerpos de Dominio Único/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA