Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 1779, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38413606

RESUMO

Human African trypanosomiasis or sleeping sickness, caused by the protozoan parasite Trypanosoma brucei, is characterized by the manipulation of the host's immune response to ensure parasite invasion and persistence. Uncovering key molecules that support parasite establishment is a prerequisite to interfere with this process. We identified Q586B2 as a T. brucei protein that induces IL-10 in myeloid cells, which promotes parasite infection invasiveness. Q586B2 is expressed during all T. brucei life stages and is conserved in all Trypanosomatidae. Deleting the Q586B2-encoding Tb927.6.4140 gene in T. brucei results in a decreased peak parasitemia and prolonged survival, without affecting parasite fitness in vitro, yet promoting short stumpy differentiation in vivo. Accordingly, neutralization of Q586B2 with newly generated nanobodies could hamper myeloid-derived IL-10 production and reduce parasitemia. In addition, immunization with Q586B2 delays mortality upon a challenge with various trypanosomes, including Trypanosoma cruzi. Collectively, we uncovered a conserved protein playing an important regulatory role in Trypanosomatid infection establishment.


Assuntos
Trypanosoma brucei brucei , Trypanosoma cruzi , Tripanossomíase Africana , Animais , Humanos , Trypanosoma brucei brucei/genética , Interleucina-10/genética , Fatores de Virulência , Parasitemia/parasitologia , Tripanossomíase Africana/parasitologia
2.
Proc Natl Acad Sci U S A ; 119(38): e2210769119, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36095215

RESUMO

Nanobodies and chemical cross-linking were used to gain information on the identity and positions of flexible domains of PI3Kα. The application of chemical cross-linking mass spectrometry (CXMS) facilitated the identification of the p85 domains BH, cSH2, and SH3 as well as their docking positions on the PI3Kα catalytic core. Binding of individual nanobodies to PI3Kα induced activation or inhibition of enzyme activity and caused conformational changes that could be correlated with enzyme function. Binding of nanobody Nb3-126 to the BH domain of p85α substantially improved resolution for parts of the PI3Kα complex, and binding of nanobody Nb3-159 induced a conformation of PI3Kα that is distinct from known PI3Kα structures. The analysis of CXMS data also provided mechanistic insights into the molecular underpinning of the flexibility of PI3Kα.


Assuntos
Domínio Catalítico , Classe I de Fosfatidilinositol 3-Quinases , Classe Ia de Fosfatidilinositol 3-Quinase , Classe I de Fosfatidilinositol 3-Quinases/química , Classe Ia de Fosfatidilinositol 3-Quinase/química , Humanos , Espectrometria de Massas/métodos , Anticorpos de Domínio Único
3.
Nanomedicine ; 24: 102103, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31669421

RESUMO

Matrix metalloproteinase-2 (MMP-2) is an endopeptidase involved in cardiovascular disease and cancer. To date, no highly selective MMP-2 inhibitors have been identified for potential use in humans. Aim of our work was to apply the nanobody technology to the generation of highly selective inhibitors of human MMP-2 and to assess their effects on platelet function and their applicability as conjugated nanobodies. We constructed a nanobody library after immunising an alpaca with human active MMP-2 and identified, after phage display and screening, one MMP-2 inhibitory nanobody (VHH-29), able to hinder the effects of MMP-2 on platelet activation, and one nanobody not inhibiting MMP-2 activity (VHH-136) which, chemically conjugated to a fluorescent probe, allowed the detection of human MMP-2 by flow-cytometry and immune-cytochemistry. In conclusion, we have generated and characterized two new nanotechnological molecular tools for human MMP-2 which represent promising agents for the study of MMP-2 in cardiovascular pathophysiology.


Assuntos
Citometria de Fluxo , Metaloproteinase 2 da Matriz/imunologia , Biblioteca de Peptídeos , Anticorpos de Domínio Único , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/imunologia
4.
Int J Parasitol ; 49(8): 635-645, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31150611

RESUMO

Human toxocariasis is a zoonosis resulting from the migration of larval stages of the dog parasite Toxocara canis into the human paratenic host. Despite its well-known limitations, serology remains the most important tool to diagnose the disease. Our objective was to employ camelid single domain antibody fragments also known as nanobodies (Nbs) for a specific and sensitive detection of Toxocara canis excretory/secretory (TES) antigens. From an alpaca immune Nb library, we retrieved different Nbs with specificity for TES antigens. Based on ELISA experiments, these Nbs did not show any cross-reactivity with Ascaris lumbricoides, Ascaris suum, Pseudoterranova decipiens, Anisakis simplex and Angiostrongylus cantonensis larval antigens. Western blot and immunocapturing revealed that Nbs 1TCE39, 1TCE52 and 2TCE49 recognise shared epitopes on different components of TES antigen. The presence of disulphide bonds in the target antigen seems to be essential for recognition of the epitopes by these three Nbs. Three separate sandwich ELISA formats, using monovalent and bivalent Nbs, were assessed to maximise the detection of TES antigens in solution. The combination of biotinylated, bivalent Nb 2TCE49 on a streptavidin pre-coated plate to capture TES antigens, and Nb 1TCE39 chemically coupled to horseradish peroxidase for detection of the captured TES antigens, yielded the most sensitive ELISA with a limit of detection of 0.650 ng/ml of TES antigen, spiked in serum. Moreover, the assay was able to detect TES antigens in sera from mice, taken 3 days after the animals were experimentally infected with T. canis. The specific characteristics of Nbs make this ELISA not only a promising tool for the detection of TES antigens in clinical samples, but also for a detailed structural and functional study of TES antigens.


Assuntos
Antígenos de Helmintos/análise , Anticorpos de Domínio Único/imunologia , Toxocara canis/imunologia , Animais , Anticorpos Anti-Helmínticos/imunologia , Western Blotting , Camelídeos Americanos , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Camundongos , Microesferas , Plasmídeos , Reação em Cadeia da Polimerase
5.
PLoS One ; 13(9): e0204222, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30260981

RESUMO

Campylobacteriosis is a widespread infectious disease, leading to a major health and economic burden. Chickens are considered as the most common infection source for humans. Campylobacter mainly multiplies in the mucus layer of their caeca. No effective control measures are currently available, but passive immunisation of chickens with pathogen-specific maternal IgY antibodies, present in egg yolk of immunised chickens, reduces Campylobacter colonisation. To explore this strategy further, anti-Campylobacter nanobodies, directed against the flagella and major outer membrane proteins, were fused to the constant domains of chicken IgA and IgY, combining the benefits of nanobodies and the effector functions of the Fc-domains. The designer chimeric antibodies were effectively produced in leaves of Nicotiana benthamiana and seeds of Arabidopsis thaliana. Stable expression of the chimeric antibodies in seeds resulted in production levels between 1% and 8% of the total soluble protein. These in planta produced antibodies do not only bind to their purified antigens but also to Campylobacter bacterial cells. In addition, the anti-flagellin chimeric antibodies are reducing the motility of Campylobacter bacteria. These antibody-containing Arabidopsis seeds can be tested for oral passive immunisation of chickens and, if effective, the chimeric antibodies can be produced in crop seeds.


Assuntos
Anticorpos Antibacterianos/metabolismo , Campylobacter/imunologia , Plantas Geneticamente Modificadas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Domínio Único/metabolismo , Animais , Anticorpos Antibacterianos/imunologia , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Campylobacter/fisiologia , Infecções por Campylobacter/imunologia , Infecções por Campylobacter/prevenção & controle , Infecções por Campylobacter/veterinária , Galinhas , Flagelos/genética , Flagelos/imunologia , Flagelina/imunologia , Imunidade Materno-Adquirida , Imunoglobulina A/genética , Imunoglobulina A/metabolismo , Imunoglobulinas/genética , Imunoglobulinas/metabolismo , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/prevenção & controle , Proteínas Recombinantes de Fusão/imunologia , Anticorpos de Domínio Único/imunologia , Nicotiana/genética , Nicotiana/metabolismo
6.
Vet Res ; 48(1): 86, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29216932

RESUMO

Campylobacter infections are among the most prevalent foodborne infections in humans, resulting in a massive disease burden worldwide. Broilers have been identified as the major source of campylobacteriosis and reducing Campylobacter loads in the broiler caeca has been proposed as an effective measure to decrease the number of infections in humans. Failure of current methods to control Campylobacter in broilers stresses the urgency to develop novel mitigation measures. We obtained six nanobodies with a broad specificity, that recognize strains belonging to the two most relevant species, Campylobacter jejuni and Campylobacter coli. The target of the nanobodies was identified as the major outer membrane protein, a porin that contributes to bacterial virulence and viability. Multimerization of the nanobodies led to agglutination of C. jejuni cells, which may affect colonization in the chicken gut. These Campylobacter-specific nanobodies may be useful to develop a strategy for preserving chickens from Campylobacter colonization.


Assuntos
Anticorpos Antibacterianos/imunologia , Infecções por Campylobacter/veterinária , Campylobacter coli/imunologia , Campylobacter jejuni/imunologia , Galinhas , Doenças das Aves Domésticas/prevenção & controle , Anticorpos de Domínio Único/imunologia , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Bactérias/imunologia , Infecções por Campylobacter/imunologia , Infecções por Campylobacter/microbiologia , Infecções por Campylobacter/prevenção & controle , Epitopos/imunologia , Porinas/imunologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia
7.
Sci Rep ; 7(1): 3385, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28611361

RESUMO

Although trypsin-like serine proteases have flexible surface-exposed loops and are known to adopt higher and lower activity conformations, structural determinants for the different conformations have remained largely obscure. The trypsin-like serine protease, urokinase-type plasminogen activator (uPA), is central in tissue remodeling processes and also strongly implicated in tumor metastasis. We solved five X-ray crystal structures of murine uPA (muPA) in the absence and presence of allosteric molecules and/or substrate-like molecules. The structure of unbound muPA revealed an unsuspected non-chymotrypsin-like protease conformation in which two ß-strands in the core of the protease domain undergoes a major antiparallel-to-parallel conformational transition. We next isolated two anti-muPA nanobodies; an active-site binding nanobody and an allosteric nanobody. Crystal structures of the muPA:nanobody complexes and hydrogen-deuterium exchange mass spectrometry revealed molecular insights about molecular factors controlling the antiparallel-to-parallel equilibrium in muPA. Together with muPA activity assays, the data provide valuable insights into regulatory mechanisms and conformational flexibility of uPA and trypsin-like serine proteases in general.


Assuntos
Conformação Proteica , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Especificidade de Anticorpos , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Camundongos , Modelos Moleculares
8.
J Biol Chem ; 292(27): 11452-11465, 2017 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-28526745

RESUMO

The ephrin receptor A4 (EphA4) is one of the receptors in the ephrin system that plays a pivotal role in a variety of cell-cell interactions, mostly studied during development. In addition, EphA4 has been found to play a role in cancer biology as well as in the pathogenesis of several neurological disorders such as stroke, spinal cord injury, multiple sclerosis, amyotrophic lateral sclerosis (ALS), and Alzheimer's disease. Pharmacological blocking of EphA4 has been suggested to be a therapeutic strategy for these disorders. Therefore, the aim of our study was to generate potent and selective Nanobodies against the ligand-binding domain of the human EphA4 receptor. We identified two Nanobodies, Nb 39 and Nb 53, that bind EphA4 with affinities in the nanomolar range. These Nanobodies were most selective for EphA4, with residual binding to EphA7 only. Using Alphascreen technology, we found that both Nanobodies displaced all known EphA4-binding ephrins from the receptor. Furthermore, Nb 39 and Nb 53 inhibited ephrin-induced phosphorylation of the EphA4 protein in a cell-based assay. Finally, in a cortical neuron primary culture, both Nanobodies were able to inhibit endogenous EphA4-mediated growth-cone collapse induced by ephrin-B3. Our results demonstrate the potential of Nanobodies to target the ligand-binding domain of EphA4. These Nanobodies may deserve further evaluation as potential therapeutics in disorders in which EphA4-mediated signaling plays a role.


Assuntos
Afinidade de Anticorpos , Receptor EphA4/imunologia , Anticorpos de Domínio Único/imunologia , Animais , Linhagem Celular , Humanos , Camundongos , Domínios Proteicos , Receptor EphA4/química , Anticorpos de Domínio Único/química
9.
Thromb Haemost ; 116(6): 1032-1040, 2016 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-27604413

RESUMO

Plasminogen activator inhibitor 1 (PAI-1) is the principal physiological inhibitor of tissue-type plasminogen activator (t-PA) and has been identified as a risk factor in cardiovascular diseases. In order to generate nanobodies against PAI-1 to interfere with its functional properties, we constructed three nanobody libraries upon immunisation of three alpacas with three different PAI-1 variants. Three panels of nanobodies were selected against these PAI-1 variants. Evaluation of the amino acid sequence identity of the complementarity determining region-3 (CDR3) reveals 34 clusters in total. Five nanobodies (VHH-s-a98, VHH-2w-64, VHH-s-a27, VHH-s-a93 and VHH-2g-42) representing five clusters exhibit inhibition towards PAI-1 activity. VHH-s-a98 and VHH-2w-64 inhibit both glycosylated and non-glycosylated PAI-1 variants through a substrate-inducing mechanism, and bind to two different regions close to αhC and the hinge region of αhF; the profibrinolytic effect of both nanobodies was confirmed using an in vitro clot lysis assay. VHH-s-a93 may inhibit PAI-1 activity by preventing the formation of the initial PAI-1t-PA complex formation and binds to the hinge region of the reactive centre loop. Epitopes of VHH-s-a27 and VHH-2g-42 could not be deduced yet. These five nanobodies interfere with PAI-1 activity through different mechanisms and merit further evaluation for the development of future profibrinolytic therapeutics.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/imunologia , Anticorpos de Domínio Único/imunologia , Sequência de Aminoácidos , Mapeamento de Epitopos , Fibrinólise , Humanos , Estrutura Terciária de Proteína , Ativador de Plasminogênio Tecidual
10.
Mol Immunol ; 78: 183-192, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27648860

RESUMO

Placental growth factor (PlGF), a member of vascular endothelial growth factors (VEGF) family, is considered as an important antigen associated with pathological conditions such as cancer cell growth, and metastasis. PlGF-targeting via nanobody (Nb) therefore could be beneficial to modulate these pathologies. In this work, phage-display and computational approach was employed to develop a high affinity PlGF-specific Nb. An Nb library was constructed against human recombinant PlGF (rPlGF). After panning on immobilized rPlGF the periplasmic-extract (PE) of individual colonies were screened by ELISA (PE-ELISA). The 3D structures of selected Nbs were then homology modeled and energy minimized using the AMBER force field. Binding score calculations were also assessed to reveal possible Nb-PlGF interactions. Via ELISA-based affinity/specificity determinations, the best-qualified Nb was further evaluated by proliferation, migration, 3D capillary formation, invasion assays and on Chick chorioallantoic membrane (CAM) model. An immune library of 1.5×107 individual Nb clones was constructed. By PE-ELISA 12 clones with strong signals were selected. Three out of 12 sequenced Nbs (Nb-C13, Nb-C18 and Nb-C62) showed high binding scores ranging between -378.7 and -461kcal/mol. Compared to a control Nb, Nb-C18 significantly inhibited proliferation, migration and the 3D-capillary formation of HUVEC cells (p<0.05) with an EC50 of 35nM, 42nM and 24nM and invasion of MDA-MB231was significantly suppressed (p<0.05) with an EC50 of57nM. The result of the CAM assay shows that Nb-C18 could inhibit the vascular formation in the chicken chorioallantoic membrane. This Nb can be used as anti-angiogenesis agent in future.


Assuntos
Inibidores da Angiogênese/farmacologia , Neovascularização Patológica/prevenção & controle , Fator de Crescimento Placentário/antagonistas & inibidores , Anticorpos de Domínio Único/imunologia , Inibidores da Angiogênese/imunologia , Animais , Afinidade de Anticorpos , Especificidade de Anticorpos , Embrião de Galinha , Biologia Computacional , Ensaio de Imunoadsorção Enzimática , Humanos , Modelos Moleculares , Biblioteca de Peptídeos , Anticorpos de Domínio Único/isolamento & purificação , Anticorpos de Domínio Único/farmacologia
11.
Nucleic Acids Res ; 42(20): 12928-38, 2014 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-25324313

RESUMO

The p53 transcription factor plays an important role in genome integrity. To perform this task, p53 regulates the transcription of genes promoting various cellular outcomes including cell cycle arrest, apoptosis or senescence. The precise regulation of this activity remains elusive as numerous mechanisms, e.g. posttranslational modifications of p53 and (non-)covalent p53 binding partners, influence the p53 transcriptional program. We developed a novel, non-invasive tool to manipulate endogenous p53. Nanobodies (Nb), raised against the DNA-binding domain of p53, allow us to distinctively target both wild type and mutant p53 with great specificity. Nb3 preferentially binds 'structural' mutant p53, i.e. R175H and R282W, while a second but distinct nanobody, Nb139, binds both mutant and wild type p53. The co-crystal structure of the p53 DNA-binding domain in complex with Nb139 (1.9 Å resolution) reveals that Nb139 binds opposite the DNA-binding surface. Furthermore, we demonstrate that Nb139 does not disturb the functional architecture of the p53 DNA-binding domain using conformation-specific p53 antibody immunoprecipitations, glutaraldehyde crosslinking assays and chromatin immunoprecipitation. Functionally, the binding of Nb139 to p53 allows us to perturb the transactivation of p53 target genes. We propose that reduced recruitment of transcriptional co-activators or modulation of selected post-transcriptional modifications account for these observations.


Assuntos
Anticorpos de Domínio Único/farmacologia , Ativação Transcricional/efeitos dos fármacos , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/imunologia , Linhagem Celular , Humanos , Modelos Moleculares , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Anticorpos de Domínio Único/imunologia , Proteína Supressora de Tumor p53/antagonistas & inibidores
12.
J Biol Chem ; 289(47): 32682-93, 2014 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-25253690

RESUMO

The biologically and clinically important membrane transporters are challenging proteins to study because of their low level of expression, multidomain structure, and complex molecular dynamics that underlies their activity. ATP7B is a copper transporter that traffics between the intracellular compartments in response to copper elevation. The N-terminal domain of ATP7B (N-ATP7B) is involved in binding copper, but the role of this domain in trafficking is controversial. To clarify the role of N-ATP7B, we generated nanobodies that interact with ATP7B in vitro and in cells. In solution NMR studies, nanobodies revealed the spatial organization of N-ATP7B by detecting transient functionally relevant interactions between metal-binding domains 1-3. Modulation of these interactions by nanobodies in cells enhanced relocalization of the endogenous ATP7B toward the plasma membrane linking molecular and cellular dynamics of the transporter. Stimulation of ATP7B trafficking by nanobodies in the absence of elevated copper provides direct evidence for the important role of N-ATP7B structural dynamics in regulation of ATP7B localization in a cell.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Anticorpos de Domínio Único/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Western Blotting , Camelídeos Americanos , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Membrana Celular/metabolismo , Cobre/química , ATPases Transportadoras de Cobre , Células HEK293 , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Espectroscopia de Ressonância Magnética , Microscopia Confocal , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Homologia de Sequência de Aminoácidos , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/genética
13.
FASEB J ; 28(9): 4004-14, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24891523

RESUMO

Hemiscorpius lepturus scorpionism poses one of the most dangerous health problems in many parts of the world. The common therapy consists of using antivenom antibody fragments derived from a polyclonal immune response raised in horses. However, this immunotherapy creates serious side effects, including anaphylactic shock sometimes even leading to death. Thus, many efforts have been made to introduce new replacement therapeutics that cause less adverse reactions. One of the most attractive approaches to replacing the available therapy is offered by single-domain antibody fragments, or nanobodies (Nbs). We immunized dromedaries with H. lepturus toxin and identified a functional recombinant Nb (referred to as F7Nb) against heminecrolysin (HNc), the major known hemolytic and dermonecrotic fraction of H. lepturus venom. This Nb was retrieved from the immune library by phage display selection. The in vitro neutralization tests indicated that 17.5 nmol of the F7Nb can inhibit 45% of the hemolytic activity of 1 EC100 (7.5 µg/ml) of HNc. The in vivo neutralization tests demonstrated that F7Nb had good antihemolytic and antidermonecrotic effects against HNc in all tested mice. Surprisingly, F7Nb (8.75 nmol) neutralized 1 LD100 of HNc (10 µg) via an intracerebroventricular route or 1 LD100 (80 µg) via a subcutaneous route. All of the control mice died. Hence, this Nb is a potential leading novel candidate for treating H. lepturus scorpionism in the near future.


Assuntos
Anticorpos Neutralizantes/imunologia , Antivenenos/uso terapêutico , Camelus/imunologia , Picadas de Escorpião/terapia , Venenos de Escorpião/química , Escorpiões/metabolismo , Anticorpos de Domínio Único/uso terapêutico , Sequência de Aminoácidos , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Eritrócitos/efeitos dos fármacos , Feminino , Hemólise/efeitos dos fármacos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Necrose , Picadas de Escorpião/imunologia , Picadas de Escorpião/parasitologia , Dermatopatias/patologia , Dermatopatias/prevenção & controle
14.
Thromb Haemost ; 111(5): 824-32, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24402608

RESUMO

One of the main disadvantages of current t-PA thrombolytic treatment is the increased bleeding risk. Upon activation, thrombin activatable fibrinolysis inhibitor (TAFI) is a very powerful antifibrinolytic enzyme. Therefore, co-administration of a TAFI inhibitor during thrombolysis could reduce the required t-PA dose without compromising the thrombolytic efficacy. In this study we generated and characterised a nanobody that is inhibitory towards rat TAFI and evaluated its profibrinolytic property in vitro and in vivo. Nanobody VHH-rTAFI-i81 inhibits (at a 16-fold molar ratio nanobody over TAFI) the thrombin/thrombomodulin (T/TM)-mediated activation of rat TAFI (rTAFI) by 83 ± 1.8% with an IC50 of 0.46 (molar ratio nanobody over TAFI). The affinity (KA) of VHH-rTAFI-i81 for rTAFI, as determined by surface plasmon resonance (Biacore®), is 2.5 ± 0.2 x 10(10) M(-1) and illustrates a very strong binding. In an in vitro clot lysis assay, administration of VHH-rTAFI-i81 strongly enhances the degree of lysis and reduces time to reach full lysis of t-PA-mediated clot lysis. Epitope mapping discloses that Lys392 is of primary importance for the nanobody/rTAFI interaction besides minor contributions of Tyr175 and Glu183. In vivo application of VHH-rTAFI-i81 in a tissue factor-induced mouse thromboembolism model significantly decreases fibrin deposition in the lungs in the absence of exogenous administered t-PA. Nanobody VHH-rTAFI-i81 is a very potent inhibitor of T/TM-mediated TAFI activation. Co-administration of this nanobody and t-PA enhances the fibrinolytic efficacy. In an in vivo mouse thromboembolism model, VHH-rTAFI-i81 reduces fibrin deposition in the lungs.


Assuntos
Anticorpos Bloqueadores/metabolismo , Fibrinolíticos/uso terapêutico , Pulmão/efeitos dos fármacos , Anticorpos de Domínio Único/administração & dosagem , Tromboembolia/tratamento farmacológico , Animais , Anticorpos Bloqueadores/genética , Carboxipeptidase B2/metabolismo , Modelos Animais de Doenças , Quimioterapia Combinada , Mapeamento de Epitopos , Feminino , Fibrina/metabolismo , Humanos , Técnicas In Vitro , Pulmão/metabolismo , Camundongos , Ligação Proteica , Ratos , Anticorpos de Domínio Único/efeitos adversos , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/metabolismo , Ressonância de Plasmônio de Superfície , Trombina/metabolismo , Trombomodulina/metabolismo , Ativador de Plasminogênio Tecidual/uso terapêutico
15.
FASEB J ; 28(4): 1805-18, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24414419

RESUMO

Invadopodia are actin-rich protrusions arising through the orchestrated regulation of precursor assembly, stabilization, and maturation, endowing cancer cells with invasive properties. Using nanobodies (antigen-binding domains of Camelid heavy-chain antibodies) as perturbators of intracellular functions and/or protein domains at the level of the endogenous protein, we examined the specific contribution of fascin and cortactin during invadopodium formation in MDA-MB-231 breast and PC-3 prostate cancer cells. A nanobody (K(d)~35 nM, 1:1 stoichiometry) that disrupts fascin F-actin bundling emphasizes the importance of stable actin bundles in invadopodium array organization and turnover, matrix degradation, and cancer cell invasion. Cortactin-SH3 dependent WIP recruitment toward the plasma membrane was specifically inhibited by a cortactin nanobody (K(d)~75 nM, 1:1 stoichiometry). This functional domain is shown to be important for formation of properly organized invadopodia, MMP-9 secretion, matrix degradation, and cancer cell invasion. Notably, using a subcellular delocalization strategy to trigger protein loss of function, we uncovered a fascin-bundling-independent role in MMP-9 secretion. Hence, we demonstrate that nanobodies enable high resolution protein function mapping in cells.


Assuntos
Proteínas de Transporte/metabolismo , Extensões da Superfície Celular/metabolismo , Cortactina/metabolismo , Proteínas dos Microfilamentos/metabolismo , Anticorpos de Domínio Único/metabolismo , Actinas/metabolismo , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Movimento Celular , Extensões da Superfície Celular/ultraestrutura , Cortactina/genética , Cortactina/imunologia , Proteínas do Citoesqueleto/metabolismo , Epitopos/genética , Epitopos/imunologia , Epitopos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/imunologia , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Pseudópodes/metabolismo , Pseudópodes/ultraestrutura , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/imunologia , Termodinâmica , Domínios de Homologia de src
16.
Breast Cancer Res ; 15(6): R116, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24330716

RESUMO

INTRODUCTION: Aberrant turnover of the actin cytoskeleton is intimately associated with cancer cell migration and invasion. Frequently however, evidence is circumstantial, and a reliable assessment of the therapeutic significance of a gene product is offset by lack of inhibitors that target biologic properties of a protein, as most conventional drugs do, instead of the corresponding gene. Proteomic studies have demonstrated overexpression of CapG, a constituent of the actin cytoskeleton, in breast cancer. Indirect evidence suggests that CapG is involved in tumor cell dissemination and metastasis. In this study, we used llama-derived CapG single-domain antibodies or nanobodies in a breast cancer metastasis model to address whether inhibition of CapG activity holds therapeutic merit. METHODS: We raised single-domain antibodies (nanobodies) against human CapG and used these as intrabodies (immunomodulation) after lentiviral transduction of breast cancer cells. Functional characterization of nanobodies was performed to identify which biochemical properties of CapG are perturbed. Orthotopic and tail vein in vivo models of metastasis in nude mice were used to assess cancer cell spreading. RESULTS: With G-actin and F-actin binding assays, we identified a CapG nanobody that binds with nanomolar affinity to the first CapG domain. Consequently, CapG interaction with actin monomers or actin filaments is blocked. Intracellular delocalization experiments demonstrated that the nanobody interacts with CapG in the cytoplasmic environment. Expression of the nanobody in breast cancer cells restrained cell migration and Matrigel invasion. Notably, the nanobody prevented formation of lung metastatic lesions in orthotopic xenograft and tail-vein models of metastasis in immunodeficient mice. We showed that CapG nanobodies can be delivered into cancer cells by using bacteria harboring a type III protein secretion system (T3SS). CONCLUSIONS: CapG inhibition strongly reduces breast cancer metastasis. A nanobody-based approach offers a fast track for gauging the therapeutic merit of drug targets. Mapping of the nanobody-CapG interface may provide a platform for rational design of pharmacologic compounds.


Assuntos
Actinas/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proteínas dos Microfilamentos/imunologia , Terapia de Alvo Molecular/métodos , Proteínas Nucleares/imunologia , Anticorpos de Domínio Único/farmacologia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Escherichia coli/genética , Feminino , Humanos , Camundongos SCID , Proteínas dos Microfilamentos/genética , Proteínas Nucleares/genética , Estrutura Terciária de Proteína
17.
BMC Genomics ; 14: 828, 2013 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-24274039

RESUMO

BACKGROUND: Gene regulatory processes are largely resulting from binding of transcription factors to specific genomic targets. Leucine-responsive Regulatory Protein (Lrp) is a prevalent transcription factor family in prokaryotes, however, little information is available on biological functions of these proteins in archaea. Here, we study genome-wide binding of the Lrp-like transcription factor Ss-LrpB from Sulfolobus solfataricus. RESULTS: Chromatin immunoprecipitation in combination with DNA microarray analysis (ChIP-chip) has revealed that Ss-LrpB interacts with 36 additional loci besides the four previously identified local targets. Only a subset of the newly identified binding targets, concentrated in a highly variable IS-dense genomic region, is also bound in vitro by pure Ss-LrpB. There is no clear relationship between the in vitro measured DNA-binding specificity of Ss-LrpB and the in vivo association suggesting a limited permissivity of the crenarchaeal chromatin for transcription factor binding. Of 37 identified binding regions, 29 are co-bound by LysM, another Lrp-like transcription factor in S. solfataricus. Comparative gene expression analysis in an Ss-lrpB mutant strain shows no significant Ss-LrpB-mediated regulation for most targeted genes, with exception of the CRISPR B cluster, which is activated by Ss-LrpB through binding to a specific motif in the leader region. CONCLUSIONS: The genome-wide binding profile presented here implies that Ss-LrpB is associated at additional genomic binding sites besides the local gene targets, but acts as a specific transcription regulator in the tested growth conditions. Moreover, we have provided evidence that two Lrp-like transcription factors in S. solfataricus, Ss-LrpB and LysM, interact in vivo.


Assuntos
Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Regulação da Expressão Gênica em Archaea , Sulfolobus solfataricus/genética , Sulfolobus solfataricus/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Sítios de Ligação , Imunoprecipitação da Cromatina , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Sequenciamento de Nucleotídeos em Larga Escala , Motivos de Nucleotídeos , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
18.
Nanomedicine (Lond) ; 8(6): 1013-26, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23730699

RESUMO

Nanobodies are recombinant, antigen-specific, single-domain, variable fragments of camelid heavy chain-only antibodies. The innate supremacy of nanobodies as a renewable source of affinity reagents, together with their high production yield in a broad variety of expression systems, minimal size, great stability, reversible refolding and outstanding solubility in aqueous solutions, and ability to specifically recognize unique epitopes with subnanomolar affinity, have combined to make them a useful class of biomolecules for research and various medical diagnostic and therapeutic applications. This article speculates on a number of technological innovations that might be introduced in the nanobody identification platform to streamline the generation of more potent nanobodies and to expand their application range.


Assuntos
Anticorpos de Domínio Único/uso terapêutico , Animais , Técnicas Biossensoriais/métodos , Diagnóstico por Imagem/métodos , Humanos , Nanomedicina/métodos , Anticorpos de Domínio Único/análise , Anticorpos de Domínio Único/imunologia
19.
Anal Biochem ; 438(1): 82-9, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23541519

RESUMO

The preparation of a V(H)H (nanobody) named IH4 that recognizes human glycophorin A (GPA) is described. IH4 was isolated by screening a library prepared from the lymphocytes of a dromedary immunized by human blood transfusion. Phage display and panning against GPA as the immobilized antigen allowed isolating this V(H)H. IH4, representing 67% of the retrieved V(H)H sequences, was expressed as a soluble correctly folded protein in SHuffle Escherichia coli cells, routinely yielding approximately 100 mg/L fermentation medium. Because IH4 recognizes GPA independently of the blood group antigens, it recognizes red cells of all humans with the possible exception of those with some extremely rare genetic background. The targeted linear epitope comprises the GPA Y52PPE55 sequence. Based on surface plasmon resonance results, the dissociation constant of the IH4-GPA equilibrium is 33 nM. IH4 is a stable protein with a transition melting temperature of 75.8 °C (measured by differential scanning calorimetry). As proof of concept, we fused HIV p24 to IH4 and used the purified construct expressed in E. coli to show that IH4 was amenable to the preparation of autologous erythrocyte agglutination reagents: reconstituted blood prepared with serum from an HIV-positive patient was readily agglutinated by the addition of the bifunctional reagent.


Assuntos
Agregação Eritrocítica , Glicoforinas/imunologia , Proteínas Recombinantes de Fusão/imunologia , Anticorpos de Domínio Único/imunologia , Sequência de Aminoácidos , Infecções por HIV/sangue , Humanos , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/isolamento & purificação , Anticorpos de Domínio Único/metabolismo
20.
Nucleic Acids Res ; 41(5): e59, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23275538

RESUMO

Nanobodies® are single-domain antibody fragments derived from camelid heavy-chain antibodies. Because of their small size, straightforward production in Escherichia coli, easy tailoring, high affinity, specificity, stability and solubility, nanobodies® have been exploited in various biotechnological applications. A major challenge in the post-genomics and post-proteomics era is the identification of regulatory networks involving nucleic acid-protein and protein-protein interactions. Here, we apply a nanobody® in chromatin immunoprecipitation followed by DNA microarray hybridization (ChIP-chip) for genome-wide identification of DNA-protein interactions. The Lrp-like regulator Ss-LrpB, arguably one of the best-studied specific transcription factors of the hyperthermophilic archaeon Sulfolobus solfataricus, was chosen for this proof-of-principle nanobody®-assisted ChIP. Three distinct Ss-LrpB-specific nanobodies®, each interacting with a different epitope, were generated for ChIP. Genome-wide ChIP-chip with one of these nanobodies® identified the well-established Ss-LrpB binding sites and revealed several unknown target sequences. Furthermore, these ChIP-chip profiles revealed auxiliary operator sites in the open reading frame of Ss-lrpB. Our work introduces nanobodies® as a novel class of affinity reagents for ChIP. Taking into account the unique characteristics of nanobodies®, in particular, their short generation time, nanobody®-based ChIP is expected to further streamline ChIP-chip and ChIP-Seq experiments, especially in organisms with no (or limited) possibility of genetic manipulation.


Assuntos
Proteínas Arqueais/metabolismo , Imunoprecipitação da Cromatina , Análise de Sequência com Séries de Oligonucleotídeos , Anticorpos de Domínio Único/química , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Proteínas Arqueais/química , Proteínas Arqueais/imunologia , Sítios de Ligação , Camelídeos Americanos , DNA Arqueal/genética , DNA Arqueal/metabolismo , Mapeamento de Epitopos , Genoma Arqueal , Proteínas Imobilizadas/química , Proteínas Imobilizadas/imunologia , Proteínas Imobilizadas/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Análise de Sequência de DNA , Sulfolobus solfataricus/genética , Sulfolobus solfataricus/metabolismo , Ressonância de Plasmônio de Superfície , Fatores de Transcrição/química , Fatores de Transcrição/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA