Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chem Sci ; 13(18): 5390-5397, 2022 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-35655564

RESUMEN

Labelling of tyrosine residues in peptides and proteins has been reported to selectively occur via a 'tyrosine-click' reaction with triazolinedione reagents (TAD). However, we here demonstrate that TAD reagents are actually not selective for tyrosine and that tryptophan residues are in fact also labelled with these reagents. This off-target labelling remained under the radar as it is challenging to detect these physiologically stable but thermally labile modifications with the commonly used HCD and CID MS/MS techniques. We show that selectivity of tryptophan over tyrosine can be achieved by lowering the pH of the aqueous buffer to effect selective Trp-labelling. Given the low relative abundance of tryptophan compared to tyrosine in natural proteins, this results in a new site-selective bioconjugation method that does not rely on enzymes nor unnatural amino acids and is demonstrated for peptides and recombinant proteins.

2.
Biomed Pharmacother ; 133: 111055, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33378961

RESUMEN

The AKT kinase family is a high-profile target for cancer therapy. Despite their high degree of homology the three AKT isoforms (AKT1, AKT2 and AKT3) are non-redundant and can even have opposing functions. Small-molecule AKT inhibitors affect all three isoforms which severely limits their usefulness as research tool or therapeutic. Using AKT2-specific nanobodies we examined the function of endogenous AKT2 in breast cancer cells. Two AKT2 nanobodies (Nb8 and Nb9) modulate AKT2 and reduce MDA-MB-231 cell viability/proliferation. Nb8 binds the AKT2 hydrophobic motif and reduces IGF-1-induced phosphorylation of this site. This nanobody also affects the phosphorylation and/or expression levels of a wide range of proteins downstream of AKT, resulting in a G0/G1 cell cycle arrest, the induction of autophagy, a reduction in focal adhesion count and loss of stress fibers. While cell cycle progression is likely to be regulated by more than one isoform, our results indicate that both the effects on autophagy and the cytoskeleton are specific to AKT2. By using an isoform-specific nanobody we were able to map a part of the AKT2 pathway. Our results confirm AKT2 and the hydrophobic motif as targets for cancer therapy. Nb8 can be used as a research tool to study AKT2 signalling events and aid in the design of an AKT2-specific inhibitor.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Autofagia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Adhesiones Focales/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Anticuerpos de Dominio Único/farmacología , Secuencias de Aminoácidos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Adhesiones Focales/enzimología , Adhesiones Focales/inmunología , Adhesiones Focales/patología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Factor I del Crecimiento Similar a la Insulina/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
3.
PLoS One ; 15(10): e0240554, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33045011

RESUMEN

The serine/threonine protein kinase AKT is frequently over-activated in cancer and is associated with poor prognosis. As a central node in the PI3K/AKT/mTOR pathway, which regulates various processes considered to be hallmarks of cancer, this kinase has become a prime target for cancer therapy. However, AKT has proven to be a highly complex target as it comes in three isoforms (AKT1, AKT2 and AKT3) which are highly homologous, yet non-redundant. The isoform-specific functions of the AKT kinases can be dependent on context (i.e. different types of cancer) and even opposed to one another. To date, there is no isoform-specific inhibitor available and no alternative to genetic approaches to study the function of a single AKT isoform. We have developed and characterized nanobodies that specifically interact with the AKT1 or AKT2 isoforms. These new tools should enable future studies of AKT1 and AKT2 isoform-specific functions. Furthermore, for both isoforms we obtained a nanobody that interferes with the AKT-PIP3-interaction, an essential step in the activation of the kinase. The nanobodies characterized in this study are a new stepping stone towards unravelling AKT isoform-specific signalling.


Asunto(s)
Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Anticuerpos de Dominio Único/metabolismo , Activación Enzimática , Humanos , Unión Proteica , Dominios Proteicos
4.
N Biotechnol ; 59: 33-43, 2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-32659511

RESUMEN

While conventional antibodies have been an instrument of choice in immunocytochemistry for some time, their small counterparts known as nanobodies have been much less frequently used for this purpose. In this study we took advantage of the availability of nanobody cDNAs to site-specifically introduce a non-standard amino acid carrying an azide/alkyne moiety, allowing subsequent Cu(I)-catalyzed Azide-Alkyne Click Chemistry (CuAAC). This generated a fluorescently labelled nanobody that can be used in single step immunocytochemistry as compared to conventional two step immunocytochemistry. Two strategies were explored to label nanobodies with Alexa Fluor 488. The first involved enzymatic addition of an alkyne-containing peptide to nanobodies using sortase A, while the second consisted of incorporating para-azido phenylalanine at the nanobody C-terminus. Through these approaches, the fluorophore was covalently and site-specifically attached. It was demonstrated that cortactin and ß-catenin, cytoskeletal and adherens junction proteins respectively, can be imaged in cells in this manner through single step immunocytochemistry. However, fixation and permeabilization of cells can alter native protein structure and form a dense cross-linked protein network, encumbering antibody binding. It was shown that photoporation prior to fixation not only allowed delivery of nanobodies into living cells, but also facilitated ß-catenin nanobody Nb86 imaging of its target, which was not possible in fixed cells. Pharmacological inhibitors are lacking for many non-enzymatic proteins, and it is therefore expected that new biological information will be obtained through photoporation of fluorescent nanobodies, which allows the study of short term effects, independent of gene-dependent (intrabody) expression.


Asunto(s)
ADN Complementario/química , Sistemas de Liberación de Medicamentos , Colorantes Fluorescentes/química , Imagen Óptica , Anticuerpos de Dominio Único/química , Coloración y Etiquetado , Biotecnología , Química Clic , Portadores de Fármacos/química , Fluorescencia , Células HeLa , Humanos , Inmunohistoquímica , Estructura Molecular , Procesos Fotoquímicos , Células Tumorales Cultivadas
5.
Sci Rep ; 9(1): 12680, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31481667

RESUMEN

Cervical cancer is caused by a persistent infection of the mucosal epithelia with high-risk human papilloma viruses (HPVs). The viral oncoprotein E6 is responsible for the inactivation of the tumour suppressor p53 and thus plays a crucial role in HPV-induced tumorigenesis. The viral E6 protein forms a trimeric complex with the endogenous E3 ubiquitine ligase E6AP and the DNA-binding domain (DBD) of p53, which results in the polyubiquitination and proteasomal degradation of p53. We have developed nanobodies (Nbs) against the DBD of p53, which substantially stabilise p53 in HeLa cells. The observed effect is specific for HPV-infected cells, since similar effects were not seen for U2OS cells. Despite the fact that the stabilised p53 was strongly nuclear enriched, its tumour suppressive functions were hampered. We argue that the absence of a tumour suppressive effect is caused by inhibition of p53 transactivation in both HPV-infected and HPV-negative cells. The inactivation of the transcriptional activity of p53 was associated with an increased cellular proliferation and viability of HeLa cells. In conclusion, we demonstrate that p53 DBD Nbs positively affect protein stability whilst adversely affecting protein function, attesting to their ability to modulate protein properties in a very subtle manner.


Asunto(s)
Papillomaviridae/fisiología , Anticuerpos de Dominio Único/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Células HeLa , Humanos , Dominios Proteicos/inmunología , Estabilidad Proteica/efectos de los fármacos , Anticuerpos de Dominio Único/farmacología , Activación Transcripcional/efectos de los fármacos , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/inmunología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Vorinostat/farmacología
6.
Eur J Nucl Med Mol Imaging ; 46(9): 1940-1951, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31161257

RESUMEN

INTRODUCTION: We have recently shown that intracerebral delivery of an anti-VEGF monoclonal antibody bevacizumab using an intra-arterial (IA) infusion is more effective than intravenous administration. While antibodies are quickly emerging as therapeutics, their disadvantages such as large size, production logistics and immunogenicity motivate search for alternatives. Thus we have studied brain uptake of nanobodies and polyamidoamine (PAMAM) dendrimers. METHODS: Nanobodies were conjugated with deferoxamine (DFO) to generate NB(DFO)2. Generation-4 PAMAM dendrimers were conjugated with DFO, and subsequently primary amines were capped with butane-1,2-diol functionalities to generate G4(DFO)3(Bdiol)110. Resulting conjugates were radiolabeled with zirconium-89. Brain uptake of 89ZrNB(DFO)2 and 89ZrG4(DFO)3(Bdiol)110 upon carotid artery vs tail vein infusions with intact BBB or osmotic blood-brain barrier opening (OBBBO) with mannitol in mice was monitored by dynamic positron emission tomography (PET) over 30 min to assess brain uptake and clearance, followed by whole-body PET-CT (computed tomography) imaging at 1 h and 24 h post-infusion (pi). Imaging results were subsequently validated by ex-vivo biodistribution. RESULTS: Intravenous administration of 89ZrNB(DFO)2 and 89ZrG4(DFO)3(Bdiol)110 resulted in their negligible brain accumulation regardless of BBB status and timing of OBBBO. Intra-arterial (IA) administration of 89ZrNB(DFO)2 dramatically increased its brain uptake, which was further potentiated with prior OBBBO. Half of the initial brain uptake was retained after 24 h. In contrast, IA infusion of 89ZrG4(DFO)3(Bdiol)110 resulted in poor initial accumulation in the brain, with complete clearance within 1 h of administration. Ex-vivo biodistribution results reflected those on PET-CT. CONCLUSIONS: IA delivery of nanobodies might be an attractive therapeutic platform for CNS disorders where prolonged intracranial retention is necessary.


Asunto(s)
Arterias , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Dendrímeros/metabolismo , Nylons/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Anticuerpos de Dominio Único/metabolismo , Animales , Dendrímeros/química , Procesamiento de Imagen Asistido por Computador , Ratones , Nylons/química , Transporte de Proteínas , Radioisótopos , Distribución Tisular , Circonio
7.
MAbs ; 10(7): 1045-1059, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30111239

RESUMEN

The tumor suppressor p53 is of crucial importance in the prevention of cellular transformation. In the presence of cellular stress signals, the negative feedback loop between p53 and Mdm2, its main negative regulator, is disrupted, which results in the activation and stabilization of p53. Via a complex interplay between both transcription-dependent and - independent functions of p53, the cell will go through transient cell cycle arrest, cellular senescence or apoptosis. However, it remains difficult to completely fathom the mechanisms behind p53 regulation and its responses, considering the presence of multiple layers involved in fine-tuning them. In order to take the next step forward, novel research tools are urgently needed. We have developed single-domain antibodies, also known as nanobodies, that specifically bind with the N-terminal transactivation domain of wild type p53, but that leave the function of p53 as a transcriptional transactivator intact. When the nanobodies are equipped with a mitochondrial-outer-membrane (MOM)-tag, we can capture p53 at the mitochondria. This nanobody-induced mitochondrial delocalization of p53 is, in specific cases, associated with a decrease in cell viability and with morphological changes in the mitochondria. These findings underpin the potential of nanobodies as bona fide research tools to explore protein function and to unravel their biochemical pathways.


Asunto(s)
Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Neoplasias/inmunología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Anticuerpos de Dominio Único/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Ciclo Celular , Senescencia Celular , Humanos , Espacio Intracelular , Dominios Proteicos/genética , Señales de Clasificación de Proteína , Transporte de Proteínas , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/inmunología
8.
Biomed Pharmacother ; 102: 230-241, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29567535

RESUMEN

Cortactin is a multidomain actin binding protein that activates Arp2/3 mediated branched actin polymerization. This is essential for the formation of protrusive structures during cancer cell invasion. Invadopodia are cancer cell-specific membrane protrusions, specialized at extracellular matrix degradation and essential for invasion and tumor metastasis. Given the unequivocal role of cortactin at every stage of invadopodium formation, it is considered an invadopodium marker and potential drug target. We used cortactin nanobodies to examine the role of cortactin domain-specific function at endogenous protein level. Two cortactin nanobodies target the central region of cortactin with high specificity. One nanobody interacts with the actin binding repeats whereas the other targets the proline rich region and was found to reduce EGF-induced cortactin phosphorylation. After intracellular expression as an intrabody, they are both capable of tracing their target in the complex environment of the cytoplasm, and disturb cortactin functions during invadopodia formation and extracellular matrix degradation. These data illustrate the use of nanobodies as a research tool to dissect the role of cortactin in cancer cell motility. This information can contribute to the development of novel therapeutics for tumor cell migration and metastasis.


Asunto(s)
Actinas/metabolismo , Extensiones de la Superficie Celular/metabolismo , Cortactina/metabolismo , Matriz Extracelular/patología , Invasividad Neoplásica , Prolina/metabolismo , Anticuerpos de Dominio Único/fisiología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Proteínas de Microfilamentos/metabolismo , Neoplasias de Células Escamosas/metabolismo , Neoplasias de Células Escamosas/patología , Unión Proteica , Dominios Proteicos
9.
PLoS One ; 12(9): e0185076, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28938008

RESUMEN

Invasive cancer cells develop small actin-based protrusions called invadopodia, which perform a primordial role in metastasis and extracellular matrix remodelling. Neural Wiskott-Aldrich syndrome protein (N-WASp) is a scaffold protein which can directly bind to actin monomers and Arp2/3 and is a crucial player in the formation of an invadopodium precursor. Expression modulation has pointed to an important role for N-WASp in invadopodium formation but the role of its C-terminal VCA domain in this process remains unknown. In this study, we generated alpaca nanobodies against the N-WASp VCA domain and investigated if these nanobodies affect invadopodium formation. By using this approach, we were able to study functions of a selected functional/structural N-WASp protein domain in living cells, without requiring overexpression, dominant negative mutants or siRNAs which target the gene, and hence the entire protein. When expressed as intrabodies, the VCA nanobodies significantly reduced invadopodium formation in both MDA-MB-231 breast cancer and HNSCC61 head and neck squamous cancer cells. Furthermore, expression of distinct VCA Nbs (VCA Nb7 and VCA Nb14) in PC-3 prostate cancer cells resulted in reduced overall matrix degradation without affecting MMP9 secretion/activation or MT1-MMP localisation at invadopodial membranes. From these results, we conclude that we have generated nanobodies targeting N-WASp which reduce invadopodium formation and functioning, most likely via regulation of N-WASp-Arp2/3 complex interaction, indicating that this region of N-WASp plays an important role in these processes.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Podosomas/metabolismo , Anticuerpos de Dominio Único/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/inmunología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/metabolismo , Afinidad de Anticuerpos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Células HEK293 , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Espacio Intracelular/metabolismo , Masculino , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Neoplasias de Células Escamosas/metabolismo , Neoplasias de Células Escamosas/patología , Podosomas/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Unión Proteica , Dominios Proteicos , Anticuerpos de Dominio Único/inmunología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/química
11.
Hum Mol Genet ; 26(7): 1353-1364, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334940

RESUMEN

Gelsolin amyloidosis is a dominantly inherited, incurable type of amyloidosis. A single point mutation in the gelsolin gene (G654A is most common) results in the loss of a Ca2+ binding site in the second gelsolin domain. Consequently, this domain partly unfolds and exposes an otherwise buried furin cleavage site at the surface. During secretion of mutant plasma gelsolin consecutive cleavage by furin and MT1-MMP results in the production of 8 and 5 kDa amyloidogenic peptides. Nanobodies that are able to (partly) inhibit furin or MT1-MMP proteolysis have previously been reported. In this study, the nanobodies have been combined into a single bispecific format able to simultaneously shield mutant plasma gelsolin from intracellular furin and extracellular MT1-MMP activity. We report the successful in vivo expression of this bispecific nanobody following adeno-associated virus serotype 9 gene therapy in gelsolin amyloidosis mice. Using SPECT/CT and immunohistochemistry, a reduction in gelsolin amyloid burden was detected which translated into improved muscle contractile properties. We conclude that a nanobody-based gene therapy using adeno-associated viruses shows great potential as a novel strategy in gelsolin amyloidosis and potentially other amyloid diseases.


Asunto(s)
Amiloidosis/genética , Amiloidosis/terapia , Gelsolina/genética , Terapia Genética , Amiloidosis/patología , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Dependovirus/genética , Dependovirus/inmunología , Modelos Animales de Enfermedad , Furina/inmunología , Furina/uso terapéutico , Gelsolina/inmunología , Humanos , Metaloproteinasa 14 de la Matriz/inmunología , Metaloproteinasa 14 de la Matriz/uso terapéutico , Ratones , Mutación Puntual/genética , Anticuerpos de Dominio Único/administración & dosificación , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología
12.
FASEB J ; 31(6): 2460-2476, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28235780

RESUMEN

Cancer cells exploit different strategies to escape from the primary tumor, gain access to the circulation, disseminate throughout the body, and form metastases, the leading cause of death by cancer. Invadopodia, proteolytically active plasma membrane extensions, are essential in this escape mechanism. Cortactin is involved in every phase of invadopodia formation, and its overexpression is associated with increased invadopodia formation, extracellular matrix degradation, and cancer cell invasion. To analyze endogenous cortactin domain function in these processes, we characterized the effects of nanobodies that are specific for the N-terminal acidic domain of cortactin and expected to target small epitopes within this domain. These nanobodies inhibit cortactin-mediated actin-related protein (Arp)2/3 activation, and, after their intracellular expression in cancer cells, decrease invadopodia formation, extracellular matrix degradation, and cancer cell invasion. In addition, one of the nanobodies affects Arp2/3 interaction and invadopodium stability, and a nanobody targeting the Src homology 3 domain of cortactin enabled comparison of 2 functional regions in invadopodium formation or stability. Given their common and distinct effects, we validate cortactin nanobodies as an instrument to selectively block and study distinct domains within a protein with unprecedented precision, aiding rational future generation of protein domain-selective therapeutic compounds.-Bertier, L., Boucherie, C., Zwaenepoel, O., Vanloo, B., Van Troys, M., Van Audenhove, I., Gettemans, J. Inhibitory cortactin nanobodies delineate the role of NTA- and SH3-domain-specific functions during invadopodium formation and cancer cell invasion.


Asunto(s)
Cortactina/química , Invasividad Neoplásica , Podosomas/fisiología , Anticuerpos de Dominio Único/fisiología , Línea Celular Tumoral , Clonación Molecular , Cortactina/metabolismo , Epítopos , Regulación Neoplásica de la Expresión Génica , Humanos , Indoles/farmacología , Dominios Proteicos
13.
Sci Rep ; 6: 31177, 2016 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-27514728

RESUMEN

Survivin, the smallest member of the inhibitor of apoptosis protein family, plays a central role during mitosis and exerts a cytoprotective function. Survivin is highly expressed in most cancer types and contributes to multiple facets of carcinogenesis. The molecular mechanisms underlying its highly diverse functions need to be extensively explored, which is crucial for rational design of future personalized therapeutics. In this study, we have generated an alpaca survivin nanobody (SVVNb8) that binds with low nanomolar affinity to its target. When expressed as an intrabody in HeLa cells, SVVNb8 faithfully tracks survivin during different phases of mitosis without interfering with survivin function. Furthermore, coupling SVVNb8 with a subcellular delocalization tag efficiently redirects endogenous survivin towards the nucleus, the cytoplasm, peroxisomes and even to the intermembrane space of mitochondria where it presumably interacts with resident mitochondrial survivin. Based on our findings, we believe that SVVNb8 is an excellent instrument to further elucidate survivin biology and topography, and can serve as a model system to investigate mitochondrial and peroxisomal (survivin) protein import.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/metabolismo , Orgánulos/metabolismo , Fracciones Subcelulares/metabolismo , Línea Celular , Humanos , Proteínas Inhibidoras de la Apoptosis/inmunología , Anticuerpos de Dominio Único/inmunología , Survivin
14.
Mol Imaging Biol ; 18(6): 887-897, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27130233

RESUMEN

PURPOSE: Gelsolin amyloidosis (AGel), also known as familial amyloidosis, Finnish type (FAF), is an autosomal, dominant, incurable disease caused by a point mutation (G654A/T) in the gelsolin (GSN) gene. The mutation results in loss of a Ca2+-binding site in the second gelsolin domain. Subsequent incorrect folding exposes a cryptic furin cleavage site, leading to the formation of a 68-kDa C-terminal cleavage product (C68) in the trans-Golgi network. This C68 fragment is cleaved by membrane type 1-matrix metalloproteinase (MT1-MMP) during secretion into the extracellular environment, releasing 8- and 5-kDa amyloidogenic peptides. These peptides aggregate and cause disease-associated symptoms. We set out to investigate whether AGel-specific nanobodies could be used to monitor amyloidogenic gelsolin buildup. PROCEDURES: Three nanobodies (FAF Nb1-3) raised against the 8-kDa fragment were screened as AGel amyloid imaging agents in WT and AGel mice using 99mTc-based single-photon emission computed tomography (SPECT)/X-ray tomography (CT), biodistribution analysis, and immunofluorescence (IF). The quantitative characteristics were analyzed in a follow-up study with a Nb11-expressing mouse model. RESULTS: All three nanobodies possess the characteristics desired for a 99mTc-based SPECT/CT imaging agent, high specificity and a low background signal. FAF Nb1 was identified as the most potent, based on its superior signal-to-noise ratio and signal specificity. As a proof of concept, we implemented 99mTc-FAF Nb1 in a follow-up study of the Nb11-expressing AGel mouse model. Using biodistribution analysis and immunofluorescence, we demonstrated the validity of the data acquired via 99mTc-FAF Nb1 SPECT/CT. CONCLUSION: These findings demonstrate the potential of this nanobody as a non-invasive tool to image amyloidogenic gelsolin deposition and assess the therapeutic capacity of AGel therapeutics currently under development. We propose that this approach can be extended to other amyloid diseases, thereby contributing to the development of specific therapies.


Asunto(s)
Amiloide/metabolismo , Amiloidosis Familiar/diagnóstico por imagen , Anticuerpos de Dominio Único/química , Tecnecio/química , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Animales , Especificidad de Anticuerpos/inmunología , Sitios de Unión , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Gelsolina/química , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miocardio/metabolismo , Miocardio/patología , Relación Señal-Ruido , Coloración y Etiquetado , Distribución Tisular
15.
Hum Mol Genet ; 24(9): 2492-507, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25601851

RESUMEN

Hereditary gelsolin amyloidosis is an autosomal dominantly inherited amyloid disorder. A point mutation in the GSN gene (G654A being the most common one) results in disturbed calcium binding by the second gelsolin domain (G2). As a result, the folding of G2 is hampered, rendering the mutant plasma gelsolin susceptible to a proteolytic cascade. Consecutive cleavage by furin and MT1-MMP-like proteases generates 8 and 5 kDa amyloidogenic peptides that cause neurological, ophthalmological and dermatological findings. To this day, no specific treatment is available to counter the pathogenesis. Using GSN nanobody 11 as a molecular chaperone, we aimed to protect mutant plasma gelsolin from furin proteolysis in the trans-Golgi network. We report a transgenic, GSN nanobody 11 secreting mouse that was used for crossbreeding with gelsolin amyloidosis mice. Insertion of the therapeutic nanobody gene into the gelsolin amyloidosis mouse genome resulted in improved muscle contractility. X-ray crystal structure determination of the gelsolin G2:Nb11 complex revealed that Nb11 does not directly block the furin cleavage site. We conclude that nanobodies can be used to shield substrates from aberrant proteolysis and this approach might establish a novel therapeutic strategy in amyloid diseases.


Asunto(s)
Amiloide/metabolismo , Amiloidosis Familiar/metabolismo , Retículo Endoplásmico/metabolismo , Gelsolina/metabolismo , Anticuerpos de Dominio Único/farmacología , Amiloidosis Familiar/genética , Amiloidosis Familiar/fisiopatología , Animales , Modelos Animales de Enfermedad , Furina/metabolismo , Gelsolina/antagonistas & inhibidores , Gelsolina/química , Gelsolina/genética , Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Contracción Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Mutación , Unión Proteica , Conformación Proteica , Proteolisis/efectos de los fármacos , Anticuerpos de Dominio Único/química , Red trans-Golgi/metabolismo
16.
Nucleic Acids Res ; 42(20): 12928-38, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-25324313

RESUMEN

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.


Asunto(s)
Anticuerpos de Dominio Único/farmacología , Activación Transcripcional/efectos de los fármacos , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/inmunología , Línea Celular , Humanos , Modelos Moleculares , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Anticuerpos de Dominio Único/inmunología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
17.
Mol Ther ; 22(10): 1768-78, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25023329

RESUMEN

Gelsolin amyloidosis is an autosomal dominant incurable disease caused by a point mutation in the GSN gene (G654A/T), specifically affecting secreted plasma gelsolin. Incorrect folding of the mutant (D187N/Y) second gelsolin domain leads to a pathological proteolytic cascade. D187N/Y gelsolin is first cleaved by furin in the trans-Golgi network, generating a 68 kDa fragment (C68). Upon secretion, C68 is cleaved by MT1-MMP-like proteases in the extracellular matrix, releasing 8 kDa and 5 kDa amyloidogenic peptides which aggregate in multiple tissues and cause disease-associated symptoms. We developed nanobodies that recognize the C68 fragment, but not native wild type gelsolin, and used these as molecular chaperones to mitigate gelsolin amyloid buildup in a mouse model that recapitulates the proteolytic cascade. We identified gelsolin nanobodies that potently reduce C68 proteolysis by MT1-MMP in vitro. Converting these nanobodies into an albumin-binding format drastically increased their serum half-life in mice, rendering them suitable for intraperitoneal injection. A 12-week treatment schedule of heterozygote D187N gelsolin transgenic mice with recombinant bispecific gelsolin-albumin nanobody significantly decreased gelsolin buildup in the endomysium and concomitantly improved muscle contractile properties. These findings demonstrate that nanobodies may be of considerable value in the treatment of gelsolin amyloidosis and related diseases.


Asunto(s)
Amiloide/metabolismo , Amiloidosis/metabolismo , Gelsolina/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Chaperonas Moleculares/metabolismo , Anticuerpos de Dominio Único/metabolismo , Amiloidosis Familiar/metabolismo , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/metabolismo , Especificidad de Anticuerpos/inmunología , Modelos Animales de Enfermedad , Gelsolina/química , Gelsolina/inmunología , Humanos , Ratones , Chaperonas Moleculares/química , Chaperonas Moleculares/inmunología , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Péptidos/inmunología , Péptidos/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Anticuerpos de Dominio Único/inmunología
18.
FASEB J ; 28(4): 1805-18, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24414419

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Extensiones de la Superficie Celular/metabolismo , Cortactina/metabolismo , Proteínas de Microfilamentos/metabolismo , Anticuerpos de Dominio Único/metabolismo , Actinas/metabolismo , Western Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Movimiento Celular , Extensiones de la Superficie Celular/ultraestructura , Cortactina/genética , Cortactina/inmunología , Proteínas del Citoesqueleto/metabolismo , Epítopos/genética , Epítopos/inmunología , Epítopos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/inmunología , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Seudópodos/metabolismo , Seudópodos/ultraestructura , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Termodinámica , Dominios Homologos src
19.
Breast Cancer Res ; 15(6): R116, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24330716

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Proteínas de Microfilamentos/inmunología , Terapia Molecular Dirigida/métodos , Proteínas Nucleares/inmunología , Anticuerpos de Dominio Único/farmacología , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Escherichia coli/genética , Femenino , Humanos , Ratones SCID , Proteínas de Microfilamentos/genética , Proteínas Nucleares/genética , Estructura Terciaria de Proteína
20.
Cell Mol Life Sci ; 70(5): 909-22, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23001012

RESUMEN

The T cell integrin receptor LFA-1 orchestrates adhesion between T cells and antigen-presenting cells (APCs), resulting in formation of a contact zone known as the immune synapse (IS) which is supported by the cytoskeleton. L-plastin is a leukocyte-specific actin bundling protein that rapidly redistributes to the immune synapse following T cell-APC engagement. We used single domain antibodies (nanobodies, derived from camelid heavy-chain only antibodies) directed against functional and structural modules of L-plastin to investigate its contribution to formation of an immune synapse between Raji cells and human peripheral blood mononuclear cells or Jurkat T cells. Nanobodies that interact either with the EF hands or the actin binding domains of L-plastin both trapped L-plastin in an inactive conformation, causing perturbation of IS formation, MTOC docking towards the plasma membrane, T cell proliferation and IL-2 secretion. Both nanobodies delayed Ser(5) phosphorylation of L-plastin which is required for enhanced bundling activity. Moreover, one nanobody delayed LFA-1 phosphorylation, reduced the association between LFA-1 and L-plastin and prevented LFA-1 enrichment at the IS. Our findings reveal subtle mechanistic details that are difficult to attain by conventional means and show that L-plastin contributes to immune synapse formation at distinct echelons.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Leucocitos Mononucleares/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas de Microfilamentos/inmunología , Centro Organizador de los Microtúbulos/inmunología , Anticuerpos de Dominio Único/inmunología , Linfocitos T/inmunología , Actinas/metabolismo , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/metabolismo , Calmodulina/inmunología , Calmodulina/metabolismo , Comunicación Celular , Línea Celular , Células Cultivadas , Motivos EF Hand , Humanos , Interleucina-2/inmunología , Células Jurkat , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Centro Organizador de los Microtúbulos/metabolismo , Centro Organizador de los Microtúbulos/ultraestructura , Modelos Moleculares , Fosforilación , Mapeo de Interacción de Proteínas , Linfocitos T/citología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...