Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 13: 1029236, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36299894

RESUMO

The P2X7 ion channel is a key sensor for extracellular ATP and a key trigger of sterile inflammation. Intravenous injection of nanobodies that block P2X7 has shown to be beneficial in mouse models of systemic inflammation. P2X7 has also emerged as an attractive therapeutic target for inflammatory brain diseases. However, little is known about the ability of nanobodies to cross the BBB. Here we evaluated the ability of P2X7-specific nanobodies to reach and to block P2X7 on microglia following intravenous or intracerebral administration. For this study, we reformatted and sequence-optimized P2X7 nanobodies for higher stability and elevated isoelectric point. Following injection of nanobodies or nanobody-encoding adeno-associated viral vectors (AAV), we monitored the occupancy and blockade of microglial P2X7 in vivo using ex vivo flow cytometry. Our results show that P2X7 on microglia was within minutes completely occupied and blocked by intracerebroventricularly injected nanobodies, even at low doses. In contrast, very high doses were required to achieve similar effects when injected intravenously. The endogenous production of P2X7-antagonistic nanobodies following intracerebral or intramuscular injection of nanobody-encoding AAVs resulted in a long-term occupancy and blockade of P2X7 on microglia. Our results provide new insights into the conditions for the delivery of nanobodies to microglial P2X7 and point to AAV-mediated delivery of P2X7 nanobodies as a promising strategy for the treatment of sterile brain inflammation.

2.
ACS Chem Neurosci ; 10(1): 704-715, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30346709

RESUMO

Transthyretin (TTR) has intrinsic neurotrophic physiological activities independent from its thyroxine ligands, which involve activation of signaling pathways through interaction with megalin. Still, the megalin binding motif on TTR is unknown. Nanobodies (Nb) have the ability to bind "hard to reach" epitopes being useful tools for protein/structure function. In this work, we characterize two anti-TTR Nanobodies, with similar mouse TTR binding affinities, although only one is able to block its neuritogenic activity (169F7_Nb). Through epitope mapping, we identified amino acids 14-18, at the entrance of the TTR central channel, to be important for interaction with megalin, and a stable TTR K15N mutant in that region was constructed. The TTR K15N mutant lacks neuritogenic activity, indicating that K15 is critical for TTR neuritogenic activity. Thus, we identify the putative binding site for megalin and describe two Nanobodies that will allow research and clarification of TTR physiological properties, regarding its neurotrophic effects.


Assuntos
Sítios de Ligação/efeitos dos fármacos , Epitopos/efeitos dos fármacos , Pré-Albumina/farmacologia , Anticorpos de Domínio Único/farmacologia , Animais , Humanos , Ligantes , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/efeitos dos fármacos , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos
3.
Front Immunol ; 9: 2553, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524421

RESUMO

CD38 is overexpressed by multiple myeloma cells and has emerged as a target for therapeutic antibodies. Nanobodies are soluble single domain antibody fragments derived from the VHH variable domain of heavy chain antibodies naturally occurring in camelids. We previously identified distinct llama nanobodies that recognize three non-overlapping epitopes of the extracellular domain of CD38. Here, we fused these VHH domains to the hinge, CH2, and CH3 domains of human IgG1, yielding highly soluble chimeric llama/human heavy chain antibodies (hcAbs). We analyzed the capacity of these hcAbs to mediate complement-dependent cytotoxicity (CDC) to CD38-expressing human multiple myeloma and Burkitt lymphoma cell lines. Combinations of two hcAbs that recognize distinct, non-overlapping epitopes of CD38 mediated potent CDC, in contrast to the hcAb monotherapy with only weak CDC capacity. Similarly, combining daratumumab with a hcAb that recognizes a non-overlapping epitope resulted in dramatically enhanced CDC. Further, introducing the E345R HexaBody mutation into the CH3 domain strongly enhanced the CDC potency of hcAbs to CD38-expressing cells. Exploiting their high solubility, we genetically fused two distinct nanobodies into heteromeric dimers via a flexible peptide linker and then fused these nanobody dimers to the hinge, CH2 and CH3 domains of human IgG1, yielding highly soluble, biparatopic hcAbs. These biparatopic hcAbs elicited CDC toward CD38-expressing myeloma cells more effectively than daratumumab. Our results underscore the advantage of nanobodies vs. pairs of VH and VL domains for constructing bispecific antibodies. Moreover, the CD38-specific biparatopic heavy chain antibodies described here represent potential new powerful therapeutics for treatment of multiple myeloma.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Anticorpos Biespecíficos/farmacologia , Antígenos de Neoplasias/imunologia , Antineoplásicos/farmacologia , Epitopos de Linfócito B/imunologia , Imunoterapia/métodos , Mieloma Múltiplo/imunologia , Animais , Anticorpos Biespecíficos/genética , Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos , Camelídeos Americanos , Linhagem Celular Tumoral , Proteínas do Sistema Complemento/metabolismo , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Mieloma Múltiplo/terapia , Proteínas Recombinantes de Fusão/genética , Anticorpos de Domínio Único/genética
4.
Curr Opin Immunol ; 52: 18-26, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29579624

RESUMO

Immune cells express various voltage-gated and ligand-gated ion channels that mediate the influx and efflux of charged ions across the plasma membrane, thereby controlling the membrane potential and mediating intracellular signal transduction pathways. These channels thus present potential targets for experimental modulation of immune responses and for therapeutic interventions in immune disease. Small molecule drugs and natural toxins acting on ion channels have illustrated the potential therapeutic benefit of targeting ion channels on immune cells. Unwanted side effects and immunogenicity have however hampered the application of these molecules. Owing to their high specificity, low immunogenicity and beneficial pharmacodynamics, antibodies targeting membrane and secretory proteins have emerged as potent therapeutics in oncology and inflammation. Nanobodies-single domain fragments derived from heavy chain antibodies naturally occurring in camelids-offer additional benefits versus antibodies, including protrusion into cryptic epitopes and easy formatting of multi-specific reagents. Here we review recent progress in the development and application of antibodies and Nanobodies targeting ion channels on immune cells.


Assuntos
Anticorpos/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/metabolismo , Anticorpos de Domínio Único/farmacologia , Animais , Anticorpos/metabolismo , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Imunomodulação/efeitos dos fármacos , Canais Iônicos/agonistas , Canais Iônicos/antagonistas & inibidores , Transdução de Sinais , Anticorpos de Domínio Único/metabolismo
5.
J Neurochem ; 145(5): 393-408, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29527688

RESUMO

Transthyretin (TTR) is a transport protein of retinol and thyroxine in serum and CSF, which is mainly secreted by liver and choroid plexus, and in smaller amounts in other cells throughout the body. The exact role of TTR and its specific expression in Central Nervous System (CNS) remains understudied. We investigated TTR expression and metabolism in CNS, through the intranasal and intracerebroventricular delivery of a specific anti-TTR Nanobody to the brain, unveiling Nanobody pharmacokinetics to the CNS. In TTR deficient mice, we observed that anti-TTR Nanobody was successfully distributed throughout all brain areas, and also reaching the spinal cord. In wild-type mice, a similar distribution pattern was observed. However, in areas known to be rich in TTR, reduced levels of Nanobody were found, suggesting potential target-mediated effects. Indeed, in wild-type mice, the anti-TTR Nanobody was specifically internalized in a receptor-mediated process, by neuronal-like cells, which were identified as motor neurons. Whereas in KO TTR mice Nanobody was internalized by all cells, for late lysosomal degradation. Moreover, we demonstrate that in vivo motor neurons also actively synthesize TTR. Finally, in vitro cultured primary motor neurons were also found to synthesize and secrete TTR into culture media. Thus, through a novel intranasal CNS distribution study with an anti-TTR Nanobody, we disclose a new cell type capable of synthesizing TTR, which might be important for the understanding of the physiological role of TTR, as well as in pathological conditions where TTR levels are altered in CSF, such as amyotrophic lateral sclerosis.


Assuntos
Encéfalo/metabolismo , Neurônios Motores/metabolismo , Pré-Albumina/metabolismo , Medula Espinal/metabolismo , Administração Intranasal , Animais , Camundongos , Camundongos Knockout , Anticorpos de Domínio Único/administração & dosagem
6.
Sci Rep ; 7(1): 14289, 2017 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-29084989

RESUMO

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.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Glicoproteínas de Membrana/metabolismo , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/tratamento farmacológico , Anticorpos de Domínio Único/imunologia , ADP-Ribosil Ciclase 1/imunologia , Animais , Camelídeos Americanos , Linhagem Celular Tumoral , Técnicas de Visualização da Superfície Celular , Modelos Animais de Doenças , Epitopos/imunologia , Corantes Fluorescentes , Humanos , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Nus , Mieloma Múltiplo/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Front Immunol ; 8: 420, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28443097

RESUMO

Next-generation sequencing (NGS) has been applied successfully to the field of therapeutic antibody discovery, often outperforming conventional screening campaigns which tend to identify only the more abundant selective antibody sequences. We used NGS to mine the functional nanobody repertoire from a phage-displayed camelid immune library directed to the recepteur d'origine nantais (RON) receptor kinase. Challenges to this application of NGS include accurate removal of read errors, correct identification of related sequences, and establishing meaningful inclusion criteria for sequences-of-interest. To this end, a sequence identity threshold was defined to separate unrelated full-length sequence clusters by exploring a large diverse set of publicly available nanobody sequences. When combined with majority-rule consensus building, applying this elegant clustering approach to the NGS data set revealed a wealth of >5,000-enriched candidate RON binders. The huge binding potential predicted by the NGS approach was explored through a set of randomly selected candidates: 90% were confirmed as RON binders, 50% of which functionally blocked RON in an ERK phosphorylation assay. Additional validation came from the correct prediction of all 35 RON binding nanobodies which were identified by a conventional screening campaign of the same immune library. More detailed characterization of a subset of RON binders revealed excellent functional potencies and a promising epitope diversity. In summary, our approach exposes the functional diversity and quality of the outbred camelid heavy chain-only immune response and confirms the power of NGS to identify large numbers of promising nanobodies.

8.
Sci Transl Med ; 8(366): 366ra162, 2016 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-27881823

RESUMO

Ion channels are desirable therapeutic targets, yet ion channel-directed drugs with high selectivity and few side effects are still needed. Unlike small-molecule inhibitors, antibodies are highly selective for target antigens but mostly fail to antagonize ion channel functions. Nanobodies-small, single-domain antibody fragments-may overcome these problems. P2X7 is a ligand-gated ion channel that, upon sensing adenosine 5'-triphosphate released by damaged cells, initiates a proinflammatory signaling cascade, including release of cytokines, such as interleukin-1ß (IL-1ß). To further explore its function, we generated and characterized nanobodies against mouse P2X7 that effectively blocked (13A7) or potentiated (14D5) gating of the channel. Systemic injection of nanobody 13A7 in mice blocked P2X7 on T cells and macrophages in vivo and ameliorated experimental glomerulonephritis and allergic contact dermatitis. We also generated nanobody Dano1, which specifically inhibited human P2X7. In endotoxin-treated human blood, Dano1 was 1000 times more potent in preventing IL-1ß release than small-molecule P2X7 antagonists currently in clinical development. Our results show that nanobody technology can generate potent, specific therapeutics against ion channels, confirm P2X7 as a therapeutic target for inflammatory disorders, and characterize a potent new drug candidate that targets P2X7.


Assuntos
Trifosfato de Adenosina/química , Inflamação/imunologia , Antagonistas do Receptor Purinérgico P2X/química , Receptores Purinérgicos P2X7/química , Anticorpos de Domínio Único/química , Animais , Anticorpos Monoclonais/química , Morte Celular , Linhagem Celular , Proliferação de Células , Dermatite Alérgica de Contato/terapia , Feminino , Glomerulonefrite/terapia , Células HEK293 , Humanos , Interleucina-1beta/química , Ligantes , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Linfócitos T/citologia
9.
PLoS Negl Trop Dis ; 10(8): e0004902, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27483431

RESUMO

Post-exposure prophylaxis (PEP) against rabies infection consists of a combination of passive immunisation with plasma-derived human or equine immune globulins and active immunisation with vaccine delivered shortly after exposure. Since anti-rabies immune globulins are expensive and scarce, there is a need for cheaper alternatives that can be produced more consistently. Previously, we generated potent virus-neutralising VHH, also called Nanobodies, against the rabies glycoprotein that are effectively preventing lethal disease in an in vivo mouse model. The VHH domain is the smallest antigen-binding functional fragment of camelid heavy chain-only antibodies that can be manufactured in microbial expression systems. In the current study we evaluated the efficacy of half-life extended anti-rabies VHH in combination with vaccine for PEP in an intranasal rabies infection model in mice. The PEP combination therapy of systemic anti-rabies VHH and intramuscular vaccine significantly delayed the onset of disease compared to treatment with anti-rabies VHH alone, prolonged median survival time (35 versus 14 days) and decreased mortality (60% versus 19% survival rate), when treated 24 hours after rabies virus challenge. Vaccine alone was unable to rescue mice from lethal disease. As reported also for immune globulins, some interference of anti-rabies VHH with the antigenicity of the vaccine was observed, but this did not impede the synergistic effect. Post exposure treatment with vaccine and human anti-rabies immune globulins was unable to protect mice from lethal challenge. Anti-rabies VHH and vaccine act synergistically to protect mice after rabies virus exposure, which further validates the possible use of anti-rabies VHH for rabies PEP.


Assuntos
Profilaxia Pós-Exposição , Vacina Antirrábica/administração & dosagem , Raiva/mortalidade , Raiva/prevenção & controle , Anticorpos de Domínio Único/administração & dosagem , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Linhagem Celular , Modelos Animais de Doenças , Feminino , Meia-Vida , Humanos , Camundongos , Testes de Neutralização , Vírus da Raiva , Vacinação , Carga Viral
10.
Antimicrob Agents Chemother ; 60(11): 6498-6509, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27550346

RESUMO

ALX-0171 is a trivalent Nanobody derived from monovalent Nb017 that binds to antigenic site II of the human respiratory syncytial virus (hRSV) fusion (F) glycoprotein. ALX-0171 is about 6,000 to 10,000 times more potent than Nb017 in neutralization tests with strains of hRSV antigenic groups A and B. To explore the effect of this enhanced neutralization on escape mutant selection, viruses resistant to either ALX-0171 or Nb017 were isolated after serial passage of the hRSV Long strain in the presence of suboptimal concentrations of the respective Nanobodies. Resistant viruses emerged notably faster with Nb017 than with ALX-0171 and in both cases contained amino acid changes in antigenic site II of hRSV F. Detailed binding and neutralization analyses of these escape mutants as well as previously described mutants resistant to certain monoclonal antibodies (MAbs) offered a comprehensive description of site II mutations which are relevant for neutralization by MAbs and Nanobodies. Notably, ALX-0171 showed a sizeable neutralization potency with most escape mutants, even with some of those selected with the Nanobody, and these findings make ALX-0171 an attractive antiviral for treatment of hRSV infections.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antivirais/farmacologia , Antígenos Virais/imunologia , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Anticorpos de Domínio Único/farmacologia , Proteínas Virais de Fusão/antagonistas & inibidores , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/química , Anticorpos Antivirais/isolamento & purificação , Antígenos Virais/química , Antígenos Virais/genética , Camelídeos Americanos , Linhagem Celular Tumoral , Células Epiteliais/virologia , Epitopos/química , Epitopos/imunologia , Humanos , Evasão da Resposta Imune/genética , Soros Imunes/química , Modelos Moleculares , Mutação , Testes de Neutralização , Ligação Proteica , Estrutura Secundária de Proteína , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/imunologia , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/isolamento & purificação , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
11.
Antimicrob Agents Chemother ; 60(1): 6-13, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26438495

RESUMO

Respiratory syncytial virus (RSV) is an important causative agent of lower respiratory tract infections in infants and elderly individuals. Its fusion (F) protein is critical for virus infection. It is targeted by several investigational antivirals and by palivizumab, a humanized monoclonal antibody used prophylactically in infants considered at high risk of severe RSV disease. ALX-0171 is a trimeric Nanobody that binds the antigenic site II of RSV F protein with subnanomolar affinity. ALX-0171 demonstrated in vitro neutralization superior to that of palivizumab against prototypic RSV subtype A and B strains. Moreover, ALX-0171 completely blocked replication to below the limit of detection for 87% of the viruses tested, whereas palivizumab did so for 18% of the viruses tested at a fixed concentration. Importantly, ALX-0171 was highly effective in reducing both nasal and lung RSV titers when delivered prophylactically or therapeutically directly to the lungs of cotton rats. ALX-0171 represents a potent novel antiviral compound with significant potential to treat RSV-mediated disease.


Assuntos
Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/farmacologia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Anticorpos de Domínio Único/farmacologia , Proteínas Virais de Fusão/antagonistas & inibidores , Administração por Inalação , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Antivirais/imunologia , Antivirais/metabolismo , Antivirais/farmacologia , Feminino , Expressão Gênica , Humanos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/virologia , Masculino , Modelos Moleculares , Cavidade Nasal/efeitos dos fármacos , Cavidade Nasal/imunologia , Cavidade Nasal/virologia , Testes de Neutralização , Palivizumab/biossíntese , Palivizumab/imunologia , Palivizumab/farmacologia , Pichia/genética , Pichia/metabolismo , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/patogenicidade , Sigmodontinae , Anticorpos de Domínio Único/biossíntese , Anticorpos de Domínio Único/imunologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
12.
PLoS One ; 9(10): e109367, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25347556

RESUMO

Rabies virus causes lethal brain infection in about 61000 people per year. Each year, tens of thousands of people receive anti-rabies prophylaxis with plasma-derived immunoglobulins and vaccine soon after exposure. Anti-rabies immunoglobulins are however expensive and have limited availability. VHH are the smallest antigen-binding functional fragments of camelid heavy chain antibodies, also called Nanobodies. The therapeutic potential of anti-rabies VHH was examined in a mouse model using intranasal challenge with a lethal dose of rabies virus. Anti-rabies VHH were administered directly into the brain or systemically, by intraperitoneal injection, 24 hours after virus challenge. Anti-rabies VHH were able to significantly prolong survival or even completely rescue mice from disease. The therapeutic effect depended on the dose, affinity and brain and plasma half-life of the VHH construct. Increasing the affinity by combining two VHH with a glycine-serine linker into bivalent or biparatopic constructs, increased the neutralizing potency to the picomolar range. Upon direct intracerebral administration, a dose as low as 33 µg of the biparatopic Rab-E8/H7 was still able to establish an anti-rabies effect. The effect of systemic treatment was significantly improved by increasing the half-life of Rab-E8/H7 through linkage with a third VHH targeted against albumin. Intraperitoneal treatment with 1.5 mg (2505 IU, 1 ml) of anti-albumin Rab-E8/H7 prolonged the median survival time from 9 to 15 days and completely rescued 43% of mice. For comparison, intraperitoneal treatment with the highest available dose of human anti-rabies immunoglobulins (65 mg, 111 IU, 1 ml) only prolonged survival by 2 days, without rescue. Overall, the therapeutic benefit seemed well correlated with the time of brain exposure and the plasma half-life of the used VHH construct. These results, together with the ease-of-production and superior thermal stability, render anti-rabies VHH into valuable candidates for development of alternative post exposure treatment drugs against rabies.


Assuntos
Vírus da Raiva/imunologia , Raiva/imunologia , Anticorpos de Domínio Único/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linhagem Celular , Modelos Animais de Doenças , Feminino , Meia-Vida , Cadeias Pesadas de Imunoglobulinas/genética , Camundongos , Raiva/prevenção & controle , Raiva/virologia , Vacina Antirrábica/imunologia , Vírus da Raiva/genética , Anticorpos de Domínio Único/administração & dosagem , Anticorpos de Domínio Único/genética , Distribuição Tecidual , Carga Viral
13.
J Biol Chem ; 288(41): 29562-72, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23979133

RESUMO

The chemokine receptor CXCR7, belonging to the membrane-bound G protein-coupled receptor superfamily, is expressed in several tumor types. Inhibition of CXCR7 with either small molecules or small interference (si)RNA has shown promising therapeutic benefits in several tumor models. With the increased interest and effectiveness of biologicals inhibiting membrane-bound receptors we made use of the "Nanobody platform" to target CXCR7. Previously we showed that Nanobodies, i.e. immunoglobulin single variable domains derived from naturally occurring heavy chain-only camelids antibodies, represent new biological tools to efficiently tackle difficult drug targets such as G protein-coupled receptors. In this study we developed and characterized highly selective and potent Nanobodies against CXCR7. Interestingly, the CXCR7-targeting Nanobodies displayed antagonistic properties in contrast with previously reported CXCR7-targeting agents. Several high affinity CXCR7-specific Nanobodies potently inhibited CXCL12-induced ß-arrestin2 recruitment in vitro. A wide variety of tumor biopsies was profiled, showing for the first time high expression of CXCR7 in head and neck cancer. Using a patient-derived CXCR7-expressing head and neck cancer xenograft model in nude mice, tumor growth was inhibited by CXCR7-targeting Nanobody therapy. Mechanistically, CXCR7-targeting Nanobodies did not inhibit cell cycle progression but instead reduced secretion of the angiogenic chemokine CXCL1 from head and neck cancer cells in vitro, thus acting here as inverse agonists, and subsequent angiogenesis in vivo. Hence, with this novel class of CXCR7 inhibitors, we further substantiate the therapeutic relevance of targeting CXCR7 in head and neck cancer.


Assuntos
Neoplasias de Cabeça e Pescoço/imunologia , Receptores CXCR/imunologia , Anticorpos de Domínio Único/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Arrestinas/imunologia , Arrestinas/metabolismo , Ligação Competitiva/imunologia , Camelídeos Americanos/imunologia , Linhagem Celular Tumoral , Quimiocina CXCL12/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/prevenção & controle , Humanos , Camundongos , Camundongos Nus , Células NIH 3T3 , Ensaio Radioligante , Receptores CXCR/genética , Receptores CXCR/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Anticorpos de Domínio Único/farmacologia , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/imunologia , beta-Arrestinas
14.
Antiviral Res ; 92(3): 389-407, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21939690

RESUMO

In 1989, a new type of antibody was identified, first in the sera of dromedaries and later also in all other species of the Camelidae family. These antibodies do not contain a light chain and also lack the first constant heavy domain. Today it is still unclear what the evolutionary advantage of such heavy chain-only antibodies could be. In sharp contrast, the broad applicability of the isolated variable antigen-binding domains (VHH) was rapidly recognized, especially for the development of therapeutic proteins, called Nanobodies(®). Here we summarize first some of the unique characteristics and features of VHHs. These will next be described in the context of different experimental therapeutic applications of Nanobodies against different viruses: HIV, Hepatitis B virus, influenza virus, Respiratory Syncytial virus, Rabies virus, FMDV, Poliovirus, Rotavirus, and PERVs. Next, the diagnostic application of VHHs (Vaccinia virus, Marburg virus and plant Tulip virus X), as well as an industrial application (lytic lactococcal 936 phage) will be described. In addition, the described data show that monovalent Nanobodies can possess unique characteristics not observed with conventional antibodies. The straightforward formatting into bivalent, multivalent, and/or multispecific Nanobodies allowed tailoring molecules for potency and cross-reactivity against viral targets with high sequence diversity.


Assuntos
Anticorpos Antivirais/uso terapêutico , Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/uso terapêutico , Viroses/diagnóstico , Viroses/tratamento farmacológico , Vírus/imunologia , Animais , Anticorpos Antivirais/imunologia , Camelídeos Americanos/imunologia , Humanos , Cadeias Pesadas de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Viroses/imunologia
15.
BMC Genomics ; 9: 387, 2008 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-18702810

RESUMO

BACKGROUND: Lysophosphatidic acid (LPA) is a lipid mediator that acts through specific G protein-coupled receptors to stimulate the proliferation, migration and survival of many cell types. LPA signaling has been implicated in development, wound healing and cancer. While LPA signaling pathways have been studied extensively, it remains unknown how LPA affects global gene expression in its target cells. RESULTS: We have examined the temporal program of global gene expression in quiescent mouse embryonic fibroblasts stimulated with LPA using 32 k oligonucleotide microarrays. In addition to genes involved in growth stimulation and cytoskeletal reorganization, LPA induced many genes that encode secreted factors, including chemokines, growth factors, cytokines, pro-angiogenic and pro-fibrotic factors, components of the plasminogen activator system and metalloproteases. Strikingly, epidermal growth factor induced a broadly overlapping expression pattern, but some 7% of the genes (105 out of 1508 transcripts) showed differential regulation by LPA. The subset of LPA-specific genes was enriched for those associated with cytoskeletal remodeling, in keeping with LPA's ability to regulate cell shape and motility. CONCLUSION: This study highlights the importance of LPA in programming fibroblasts not only to proliferate and migrate but also to produce many paracrine mediators of tissue remodeling, angiogenesis, inflammation and tumor progression. Furthermore, our results show that G protein-coupled receptors and receptor tyrosine kinases can signal independently to regulate broadly overlapping sets of genes in the same cell type.


Assuntos
Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Citoesqueleto/genética , Embrião de Mamíferos , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , RNA/genética , Receptores de Ácidos Lisofosfatídicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
17.
J Biol Chem ; 281(52): 40033-40, 2006 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-17032651

RESUMO

Epidermal growth factor (EGF)-like growth factors bind their ErbB receptors in a highly selective manner, but the molecular basis for this specificity is poorly understood. We have previously shown that certain residues in human EGF (Ser(2)-Asp(3)) and TGFalpha (Glu(26)) are not essential for their binding to ErbB1 but prevent binding to ErbB3 and ErbB4. In the present study, we have used a phage display approach to affinity-optimize the C-terminal linear region of EGF-like growth factors for binding to each ErbB receptor and thereby shown that Arg(45) in EGF impairs binding to both ErbB3 and ErbB4. By omitting all these so-called negative constraints from EGF, we designed a ligand designated panerbin that binds ErbB1, ErbB3, and ErbB4 with similarly high affinity as their wild-type ligands. Homology models, based on the known crystal structure of TGFalpha-bound ErbB1, showed that panerbin is able to bind ErbB1, ErbB3, and ErbB4 in a highly similar manner with respect to position and number of interaction sites. Upon in silico introduction of the experimentally known negative constraints into panerbin, we found that Arg(45) induced local charge repulsion and Glu(26) induced steric hindrance in a receptor-specific manner, whereas Ser(2)-Asp(3) impaired binding due to a disordered conformation. Furthermore, radiolabeled panerbin was used to quantify the level of all three receptors on human breast cancer cells in a single radioreceptor assay. It is concluded that the ErbB specificity of EGF-like growth factors primarily results from the presence of a limited number of residues that impair the unintended interaction with other ErbB receptors.


Assuntos
Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/química , Receptores ErbB/metabolismo , Proteínas Oncogênicas v-erbB/química , Proteínas Oncogênicas v-erbB/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/genética , Receptores ErbB/genética , Humanos , Ligantes , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Oncogênicas v-erbB/genética , Ligação Proteica/genética , Receptor ErbB-3/química , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Receptor ErbB-4 , Homologia de Sequência de Aminoácidos
18.
Mol Cell Biol ; 26(13): 5015-22, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16782887

RESUMO

Autotaxin (ATX), or nucleotide pyrophosphatase-phosphodiesterase 2, is a secreted lysophospholipase D that promotes cell migration, metastasis, and angiogenesis. ATX generates lysophosphatidic acid (LPA), a lipid mitogen and motility factor that acts on several G protein-coupled receptors. Here we report that ATX-deficient mice die at embryonic day 9.5 (E9.5) with profound vascular defects in yolk sac and embryo resembling the Galpha13 knockout phenotype. Furthermore, at E8.5, ATX-deficient embryos showed allantois malformation, neural tube defects, and asymmetric headfolds. The onset of these abnormalities coincided with increased expression of ATX and LPA receptors in normal embryos. ATX heterozygous mice appear healthy but show half-normal ATX activity and plasma LPA levels. Our results reveal a critical role for ATX in vascular development, indicate that ATX is the major LPA-producing enzyme in vivo, and suggest that the vascular defects in ATX-deficient embryos may be explained by loss of LPA signaling through Galpha13.


Assuntos
Vasos Sanguíneos/anormalidades , Embrião de Mamíferos/anormalidades , Genes Letais , Lisofosfolipídeos/metabolismo , Complexos Multienzimáticos/metabolismo , Fosfodiesterase I/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/metabolismo , Animais , Vasos Sanguíneos/enzimologia , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/enzimologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Lisofosfolipídeos/sangue , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/genética , Fosfodiesterase I/genética , Diester Fosfórico Hidrolases/genética , Pirofosfatases/genética , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo
19.
Biochemistry ; 45(14): 4703-10, 2006 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-16584205

RESUMO

The family of epidermal growth factor (EGF)-like ligands binds to ErbB receptors in a highly selective manner. Previous studies indicated that both linear regions of the ligand play a major role in determining receptor selectivity, and phage display studies showed that each region could be optimized independently for enhanced affinity. In this study, we broadened the ErbB binding specificity of EGF by introducing the optimal sequence requirements for ErbB3 binding in both the N- and C-terminal linear regions. One such EGF mutant, designated WVR/EGF/IADIQ, gained high affinity for ErbB3 and showed concomitant ErbB3 activation through ErbB2.ErbB3 heterodimers similar to the natural ErbB3 ligand NRG1beta, while the capacity to bind and activate ErbB1 was fully maintained. Despite its high affinity for ErbB1 and ErbB3, this mutant was unable to activate ErbB1.ErbB3 heterodimers, as shown by the cell survival and receptor phosphorylation analysis. We concluded that despite the fact that no naturally occurring ligand exists with this dual-specificity, high-affinity binding to both ErbB1 and ErbB3 is not mutually exclusive. This mutant can be useful in a direct structural comparison of the ligand-binding characteristics of ErbB1 and ErbB3.


Assuntos
Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Receptor ErbB-3/metabolismo , Animais , Ligação Competitiva , Linhagem Celular , Dimerização , Humanos , Ligantes , Camundongos , Estrutura Quaternária de Proteína , Receptor ErbB-2/metabolismo
20.
J Biol Chem ; 278(40): 39114-23, 2003 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-12869572

RESUMO

Various chimeras of the ErbB1-specific ligands epidermal growth factor (EGF) and transforming growth factor-alpha (TGFalpha) display an enlarged repertoire as activators of ErbB2.ErbB3 heterodimers. Mutational analysis indicated that particularly residues in the N terminus and B-loop region of these ligands are involved in the broadened receptor specificity. In order to understand the receptor specificity of T1E, a chimeric ligand constructed by the introduction of the linear N-terminal region of TGFalpha into EGF, we determined in this study the solution structure and dynamics of T1E by multidimensional NMR analysis. Subsequently, we studied the structural characteristics of T1E binding to both ErbB1 and ErbB3 by superposition modeling of its structure on the known crystal structures of ErbB3 and liganded ErbB1 complexes. The results show that the overall structure of T1E in solution is very similar to that of native EGF and TGFalpha but that its N terminus shows an extended structure that is appropriately positioned to form a triple beta-sheet with the large antiparallel beta-sheet in the B-loop region. This conformational effect of the N terminus together with the large overall flexibility of T1E, as determined by 15N NMR relaxation analysis, may be a facilitative property for its broad receptor specificity. The structural superposition models indicate that hydrophobic and electrostatic interactions of the N terminus and B-loop of T1E are particularly important for its binding to ErbB3.


Assuntos
Fator de Crescimento Epidérmico/química , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Fator de Crescimento Transformador alfa/química , Sequência de Aminoácidos , Análise Mutacional de DNA , Fator de Crescimento Epidérmico/metabolismo , Vetores Genéticos , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Pichia/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Fator de Crescimento Transformador alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...