Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 180(5): 822-824, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32142673

RESUMEN

Monoclonal antibodies (mAbs) targeting antigens expressed at the surface of tumor cells are widely used for cancer control in clinics, but these treatments need to be improved. Chew et al. show how an old drug, prochlorperazine, could be repurposed to enhance the efficacy of anti-tumor mAbs by increasing the cell-surface expression of tumor antigens.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Neoplasias , Anticuerpos Monoclonales , Antígenos de Neoplasias , Endocitosis , Humanos
2.
Cell ; 177(7): 1701-1713.e16, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31155232

RESUMEN

Over the last decade, various new therapies have been developed to promote anti-tumor immunity. Despite interesting clinical results in hematological malignancies, the development of bispecific killer-cell-engager antibody formats directed against tumor cells and stimulating anti-tumor T cell immunity has proved challenging, mostly due to toxicity problems. We report here the generation of trifunctional natural killer (NK) cell engagers (NKCEs), targeting two activating receptors, NKp46 and CD16, on NK cells and a tumor antigen on cancer cells. Trifunctional NKCEs were more potent in vitro than clinical therapeutic antibodies targeting the same tumor antigen. They had similar in vivo pharmacokinetics to full IgG antibodies and no off-target effects and efficiently controlled tumor growth in mouse models of solid and invasive tumors. Trifunctional NKCEs thus constitute a new generation of molecules for fighting cancer. VIDEO ABSTRACT.


Asunto(s)
Anticuerpos Biespecíficos , Antígenos Ly/inmunología , Antineoplásicos Inmunológicos , Citotoxicidad Inmunológica/efectos de los fármacos , Células Asesinas Naturales/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología , Neoplasias Experimentales , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Células Asesinas Naturales/patología , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia
3.
Cell ; 175(7): 1731-1743.e13, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30503213

RESUMEN

Checkpoint inhibitors have revolutionized cancer treatment. However, only a minority of patients respond to these immunotherapies. Here, we report that blocking the inhibitory NKG2A receptor enhances tumor immunity by promoting both natural killer (NK) and CD8+ T cell effector functions in mice and humans. Monalizumab, a humanized anti-NKG2A antibody, enhanced NK cell activity against various tumor cells and rescued CD8+ T cell function in combination with PD-x axis blockade. Monalizumab also stimulated NK cell activity against antibody-coated target cells. Interim results of a phase II trial of monalizumab plus cetuximab in previously treated squamous cell carcinoma of the head and neck showed a 31% objective response rate. Most common adverse events were fatigue (17%), pyrexia (13%), and headache (10%). NKG2A targeting with monalizumab is thus a novel checkpoint inhibitory mechanism promoting anti-tumor immunity by enhancing the activity of both T and NK cells, which may complement first-generation immunotherapies against cancer.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma de Células Escamosas , Cetuximab/uso terapéutico , Inmunidad Celular/efectos de los fármacos , Inmunoterapia , Células Asesinas Naturales/inmunología , Subfamília C de Receptores Similares a Lectina de Células NK , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Ensayos Clínicos Fase II como Asunto , Humanos , Células Asesinas Naturales/patología , Ratones , Subfamília C de Receptores Similares a Lectina de Células NK/antagonistas & inhibidores , Subfamília C de Receptores Similares a Lectina de Células NK/inmunología
4.
Nucleic Acids Res ; 49(17): 9886-9905, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34469544

RESUMEN

Telomere maintenance is essential to preserve genomic stability and involves telomere-specific proteins, DNA replication and repair proteins. Lamins are key components of the nuclear envelope and play numerous roles, including maintenance of the nuclear integrity, regulation of transcription, and DNA replication. Elevated levels of lamin B1, one of the major lamins, have been observed in some human pathologies and several cancers. Yet, the effect of lamin B1 dysregulation on telomere maintenance remains unknown. Here, we unveil that lamin B1 overexpression drives telomere instability through the disruption of the shelterin complex. Indeed, lamin B1 dysregulation leads to an increase in telomere dysfunction-induced foci, telomeric fusions and telomere losses in human cells. Telomere aberrations were preceded by mislocalizations of TRF2 and its binding partner RAP1. Interestingly, we identified new interactions between lamin B1 and these shelterin proteins, which are strongly enhanced at the nuclear periphery upon lamin B1 overexpression. Importantly, chromosomal fusions induced by lamin B1 in excess were rescued by TRF2 overexpression. These data indicated that lamin B1 overexpression triggers telomere instability through a mislocalization of TRF2. Altogether our results point to lamin B1 as a new interacting partner of TRF2, that is involved in telomere stability.


Asunto(s)
Lamina Tipo B/metabolismo , Complejo Shelterina/metabolismo , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Células Cultivadas , Humanos , Lamina Tipo B/química , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/química
5.
J Invertebr Pathol ; 196: 107867, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36503887

RESUMEN

European foulbrood (EFB) is a honey bee brood disease caused by the bacterium Melissococcus plutonius. Large-scale EFB outbreaks have been reported in several countries in recent decades, which entail costly sanitation measures of affected apiaries to restrict the spread of this contagious pathogen. To mitigate its impact, a better understanding of the population dynamics of the etiological agent is required. We here used multi-locus sequence typing (MLST) to infer the genetic diversity and geographical distribution of 160 M. plutonius isolates collected from EFB symptomatic honey bee colonies seven years apart. Isolates belonged to three clonal complexes (CCs) known worldwide and to 12 sequence types (STs), of which five were novel. Phylogenetic and clustering analyses showed that some of these novel sequence types have likely evolved locally during a period of outbreak, but most disappeared again. We further screened the isolates for melissotoxin A (mtxA), a putative virulence gene. The prevalence of STs in which mtxA was frequent increased over time, suggesting that this gene promotes spread. Despite the increased frequency of this gene in the population, the total number of cases decreased, which could be due to stricter control measures implemented before the second sampling period. Our results provide a better understanding of M. plutonius population dynamics and help identify knowledge gaps that limit efficient control of this emerging disease.


Asunto(s)
Genética de Población , Abejas , Animales , Larva/microbiología , Tipificación de Secuencias Multilocus , Prevalencia , Filogenia
6.
Eur J Immunol ; 51(8): 1934-1942, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34145579

RESUMEN

Immuno-oncology is revolutionizing the treatment of cancers, by inducing the recognition and elimination of tumor cells by the immune system. Recent advances have focused on generating or unleashing tumor antigen-specific T-cell responses, leading to alternative treatment paradigms for many cancers. Despite these successes, the clinical benefit has been limited to a subset of patients and certain tumor types, highlighting the need for alternative strategies. One innovative approach is to broaden and amplify antitumoral immune responses by targeting innate immunity. Particularly, the aim has been to develop new antibody formats capable of stimulating the antitumor activity of innate immune cells, boosting not only their direct role in tumor elimination, but also their function in eliciting multicellular immune responses ultimately resulting in long-lasting tumor control by adaptive immunity. This review covers the development of a new class of synthetic molecules, natural killer cell engagers (NKCEs), which are built from fragments of monoclonal antibodies (mAbs) and are designed to harness the immune functions of NK cells in cancer. As currently shown in preclinical studies and clinical trials, NKCEs are promising candidates for the next generation of tumor immunotherapies.


Asunto(s)
Inmunoterapia/métodos , Inmunoterapia/tendencias , Células Asesinas Naturales/inmunología , Oncología Médica/tendencias , Neoplasias/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico
7.
Methods ; 180: 35-44, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32156657

RESUMEN

Producing intact recombinant membrane proteins for structural studies is an inherently challenging task due to their requirement for a cell-lipid environment. Most of the procedures developed involve isolating the protein by solubilization with detergent and further reconstitutions into artificial membranes. These procedures are highly time consuming and suffer from further drawbacks, including low yields and high cost. We describe here an alternative method for rapidly obtaining recombinant cell-surface membrane proteins displayed on extracellular vesicles (EVs) derived from cells in culture. Interaction between these membrane proteins and ligands can be analyzed directly on EVs. Moreover, EVs can also be used for protein structure determination or immunization purposes.


Asunto(s)
Vesículas Extracelulares/metabolismo , Proteínas de la Membrana/aislamiento & purificación , Proteínas Recombinantes/aislamiento & purificación , 5'-Nucleotidasa/inmunología , Clonación Molecular , Microscopía por Crioelectrón , Detergentes/química , Dispersión Dinámica de Luz , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/ultraestructura , Proteínas Ligadas a GPI/inmunología , Células HEK293 , Humanos , Ligandos , Espectrometría de Masas , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Electrónica , Plásmidos/genética
9.
J Immunol ; 194(4): 1591-601, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25582852

RESUMEN

We proposed that the killer cell Ig-like receptor KIR3DL2 binding more strongly to HLA-B27 (B27) ß2-microglobulin free H chain (FHC) dimers than other HLA-class I molecules regulates lymphocyte function in arthritis and infection. We compared the function of B27 FHC dimers with other class I H chains and identified contact residues in KIR3DL2. B27 FHC dimers interacted functionally with KIR3DL2 on NK and reporter cells more strongly than did other class I FHCs. Mutagenesis identified key residues in the D0 and other Ig-like domains that were shared and distinct from KIR3DL1 for KIR3DL2 binding to B27 and other class I FHCs. We modeled B27 dimer binding to KIR3DL2 and compared experimental mutagenesis data with computational "hot spot" predictions. Modeling predicts that the stronger binding of B27 dimers to KIR3DL2 is mediated by nonsymmetrical complementary contacts of the D0 and D1 domains with the α1, α2, and α3 domains of both B27 H chains. In contrast, the D2 domain primarily contacts residues in the α2 domain of one B27 H chain. These findings provide novel insights about the molecular basis of KIR3DL2 binding to B27 and other ligands and suggest an important role for KIR3DL2-B27 interactions in controlling the function of NK cells in B27(+) individuals.


Asunto(s)
Antígeno HLA-B27/inmunología , Modelos Moleculares , Multimerización de Proteína , Receptores KIR3DL2/metabolismo , Línea Celular , Citometría de Flujo , Antígeno HLA-B27/química , Humanos , Inmunoprecipitación , Células Asesinas Naturales/inmunología , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Receptores KIR3DL2/química
10.
Proc Biol Sci ; 283(1835)2016 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-27466446

RESUMEN

There is clear evidence for sublethal effects of neonicotinoid insecticides on non-target ecosystem service-providing insects. However, their possible impact on male insect reproduction is currently unknown, despite the key role of sex. Here, we show that two neonicotinoids (4.5 ppb thiamethoxam and 1.5 ppb clothianidin) significantly reduce the reproductive capacity of male honeybees (drones), Apis mellifera Drones were obtained from colonies exposed to the neonicotinoid insecticides or controls, and subsequently maintained in laboratory cages until they reached sexual maturity. While no significant effects were observed for male teneral (newly emerged adult) body mass and sperm quantity, the data clearly showed reduced drone lifespan, as well as reduced sperm viability (percentage living versus dead) and living sperm quantity by 39%. Our results demonstrate for the first time that neonicotinoid insecticides can negatively affect male insect reproductive capacity, and provide a possible mechanistic explanation for managed honeybee queen failure and wild insect pollinator decline. The widespread prophylactic use of neonicotinoids may have previously overlooked inadvertent contraceptive effects on non-target insects, thereby limiting conservation efforts.


Asunto(s)
Abejas/efectos de los fármacos , Anticonceptivos/farmacología , Guanidinas/farmacología , Insecticidas/farmacología , Neonicotinoides/farmacología , Tiazoles/farmacología , Animales , Supervivencia Celular , Masculino , Nitrocompuestos , Oxazinas , Reproducción , Espermatozoides/citología , Tiametoxam
11.
Nucleic Acids Res ; 42(9): 5616-32, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24598253

RESUMEN

The repair of toxic double-strand breaks (DSB) is critical for the maintenance of genome integrity. The major mechanisms that cope with DSB are: homologous recombination (HR) and classical or alternative nonhomologous end joining (C-NHEJ versus A-EJ). Because these pathways compete for the repair of DSB, the choice of the appropriate repair pathway is pivotal. Among the mechanisms that influence this choice, deoxyribonucleic acid (DNA) end resection plays a critical role by driving cells to HR, while accurate C-NHEJ is suppressed. Furthermore, end resection promotes error-prone A-EJ. Increasing evidence define Poly(ADP-ribose) polymerase 3 (PARP3, also known as ARTD3) as an important player in cellular response to DSB. In this work, we reveal a specific feature of PARP3 that together with Ku80 limits DNA end resection and thereby helps in making the choice between HR and NHEJ pathways. PARP3 interacts with and PARylates Ku70/Ku80. The depletion of PARP3 impairs the recruitment of YFP-Ku80 to laser-induced DNA damage sites and induces an imbalance between BRCA1 and 53BP1. Both events result in compromised accurate C-NHEJ and a concomitant increase in DNA end resection. Nevertheless, HR is significantly reduced upon PARP3 silencing while the enhanced end resection causes mutagenic deletions during A-EJ. As a result, the absence of PARP3 confers hypersensitivity to anti-tumoral drugs generating DSB.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Reparación del ADN por Unión de Extremidades , Poli(ADP-Ribosa) Polimerasas/fisiología , Reparación del ADN por Recombinación , Antígenos Nucleares/metabolismo , Antineoplásicos/farmacología , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Etopósido/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Autoantígeno Ku , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteína de Replicación A/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
12.
Int J Cancer ; 136(4): 982-8, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24975135

RESUMEN

The hematopoietic growth factor granulocyte colony-stimulating factor (G-CSF) has a role in proliferation, differentiation and migration of the myeloid lineage and in mobilizing hematopoietic stem and progenitor cells into the bloodstream. However, G-CSF has been newly characterized as a neurotrophic factor in the brain. We recently uncovered that autonomic nerve development in the tumor microenvironment participates actively in prostate tumorigenesis and metastasis. Here, we found that G-CSF constrains cancer to grow and progress by, respectively, supporting the survival of sympathetic nerve fibers in 6-hydroxydopamine-sympathectomized mice and also, promoting the aberrant outgrowth of parasympathetic nerves in transgenic or xenogeneic prostate tumor models. This provides insight into how neurotrophic growth factors may control tumor neurogenesis and may lead to new antineurogenic therapies for prostate cancer.


Asunto(s)
Axones/fisiología , Carcinogénesis/metabolismo , Factor Estimulante de Colonias de Granulocitos/fisiología , Neoplasias de la Próstata/metabolismo , Fibras Adrenérgicas/patología , Fibras Adrenérgicas/fisiología , Animales , Axones/patología , Supervivencia Celular , Células HL-60 , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Factores de Crecimiento Nervioso/fisiología , Próstata/inervación , Neoplasias de la Próstata/patología
13.
Stem Cells ; 32(12): 3257-65, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25098224

RESUMEN

Neurogenesis decreases during aging causing a progressive cognitive decline but it is still controversial whether proliferation defects in neurogenic niches result from a loss of neural stem cells or from an impairment of their progression through the cell cycle. Using an accurate fluorescence-activated cell sorting technique, we show that the pool of neural stem cells is maintained in the subventricular zone of middle-aged mice while they have a reduced proliferative potential eventually leading to the subsequent decrease of their progeny. In addition, we demonstrate that the G1 phase is lengthened during aging specifically in activated stem cells, but not in transit-amplifying cells, and directly impacts on neurogenesis. Finally, we report that inhibition of TGFß signaling restores cell cycle progression defects in stem cells. Our data highlight the significance of cell cycle dysregulation in stem cells in the aged brain and provide an attractive foundation for the development of anti-TGFß regenerative therapies based on stimulating endogenous neural stem cells.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/citología , Diferenciación Celular/fisiología , Fase G1 , Neurogénesis/fisiología , Células Madre/citología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proliferación Celular/fisiología , Fase G1/genética , Ratones Endogámicos C57BL , Nicho de Células Madre/fisiología
14.
J Biol Chem ; 288(4): 2571-9, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23223580

RESUMEN

CD146 is a highly glycosylated junctional adhesion molecule, expressed on human vascular endothelial cells and involved in the control of vessel integrity. Galectin-1 is a lectin produced by vascular cells that can binds N- and O-linked oligosaccharides of cell membrane glycoproteins. Because both CD146 and Galectin-1 are involved in modulation of cell apoptosis, we hypothesized that Galectin-1 could interact with CD146, leading to functional consequences in endothelial cell apoptosis. We first characterized CD146 glycosylations and showed that it is mainly composed of N-glycans able to establish interactions with Galectin-1. We demonstrated a sugar-dependent binding of recombinant CD146 to Galectin-1 using both ELISA and Biacore assays. This interaction is direct, with a K(D) of 3.10(-7) M, and specific as CD146 binds to Galectin-1 and not to Galectin-2. Moreover, co-immunoprecipitation experiments showed that Galectin-1 interacts with endogenous CD146 that is highly expressed by HUVEC. We observed a Galectin-1-induced HUVEC apoptosis in a dose-dependent manner as demonstrated by Annexin-V/7AAD staining. Interestingly, both down-regulation of CD146 cell surface expression using siRNA and antibody-mediated blockade of CD146 increase this apoptosis. Altogether, our results identify Galectin-1 as a novel ligand for CD146 and this interaction protects, in vitro, endothelial cells against apoptosis induced by Galectin-1.


Asunto(s)
Apoptosis , Antígeno CD146/química , Células Endoteliales/citología , Galectina 1/metabolismo , Regulación de la Expresión Génica , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Fibroblastos/metabolismo , Galectina 2/metabolismo , Glicosilación , Células Endoteliales de la Vena Umbilical Humana , Humanos , Cinética , Ligandos , Polisacáridos/química , Unión Proteica , ARN Interferente Pequeño/metabolismo , Resonancia por Plasmón de Superficie , Transfección
15.
Hum Mol Genet ; 21(1): 121-35, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21968513

RESUMEN

Fanconi anemia (FA) is a human rare genetic disorder characterized by congenital defects, bone marrow (BM) failure and predisposition to leukemia. The progressive aplastic anemia suggests a defect in the ability of hematopoietic stem cells (HSC) to sustain hematopoieis. We have examined the role of the nuclear FA core complex gene Fancg in the functionality of HSC. In Fancg-/- mice, we observed a decay of long-term HSC and multipotent progenitors that account for the reduction in the LSK compartment containing primitive hematopoietic cells. Fancg-/- lymphoid and myeloid progenitor cells were also affected, and myeloid progenitors show compromised in vitro functionality. HSC from Fancg-/- mice failed to engraft and to reconstitute at short and long term the hematopoiesis in a competitive transplantation assay. Fancg-/- LSK cells showed a loss of quiescence, an impaired migration in vitro in response to the chemokine CXCL12 and a defective homing to the BM after transplantation. Finally, the expression of several key genes involved in self-renewal, quiescence and migration of HSC was dysregulated in Fancg-deficient LSK subset. Collectively, our data reveal that Fancg should play a role in the regulation of physiological functions of HSC.


Asunto(s)
Proteína del Grupo de Complementación G de la Anemia de Fanconi/deficiencia , Anemia de Fanconi/fisiopatología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Médula Ósea/metabolismo , Movimiento Celular , Quimiocina CXCL12/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Femenino , Hematopoyesis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
16.
Bioconjug Chem ; 25(3): 569-78, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24483299

RESUMEN

Most chemical techniques used to produce antibody-drug conjugates (ADCs) result in a heterogeneous mixture of species with variable drug-to-antibody ratios (DAR) which will potentially display different pharmacokinetics, stability, and safety profiles. Here we investigated two strategies to obtain homogeneous ADCs based on site-specific modification of deglycosylated antibodies by microbial transglutaminase (MTGase), which forms isopeptidic bonds between Gln and Lys residues. We have previously shown that MTGase solely recognizes Gln295 within the heavy chain of IgGs as a substrate and can therefore be exploited to generate ADCs with an exact DAR of 2. The first strategy included the direct, one-step attachment of the antimitotic toxin monomethyl auristatin E (MMAE) to the antibody via different spacer entities with a primary amine functionality that is recognized as a substrate by MTGase. The second strategy was a chemo-enzymatic, two-step approach whereby a reactive spacer entity comprising a bio-orthogonal thiol or azide function was attached to the antibody by MTGase and subsequently reacted with a suitable MMAE-derivative. To this aim, we investigated two different chemical approaches, namely, thiol-maleimide and strain-promoted azide-alkyne cycloaddition (SPAAC). Direct enzymatic attachment of MMAE-spacer derivatives at an 80 molar excess of drug yielded heterogeneous ADCs with a DAR of between 1.0 to 1.6. In contrast to this, the chemo-enzymatic approach only required a 2.5 molar excess of toxin to yield homogeneous ADCs with a DAR of 2.0 in the case of SPAAC and 1.8 for the thiol-maleimide approach. As a proof-of-concept, trastuzumab (Herceptin) was armed with the MMAE via the chemo-enzymatic approach using SPAAC and tested in vitro. Trastuzumab-MMAE efficiently killed BT-474 and SK-BR-3 cells with an IC50 of 89.0 pM and 21.7 pM, respectively. Thus, the chemo-enzymatic approach using MTGase is an elegant strategy to form ADCs with a defined DAR of 2. Furthermore, the approach is directly applicable to a broad variety of antibodies as it does not require prior genetic modifications of the antibody sequence.


Asunto(s)
Anticuerpos Monoclonales Humanizados/química , Anticuerpos/química , Antineoplásicos/farmacología , Oligopéptidos/química , Transglutaminasas/química , Alquinos/química , Alquinos/metabolismo , Anticuerpos/metabolismo , Anticuerpos Monoclonales Humanizados/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Azidas/química , Azidas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Maleimidas/química , Maleimidas/metabolismo , Estructura Molecular , Oligopéptidos/metabolismo , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/metabolismo , Transglutaminasas/metabolismo , Trastuzumab
17.
Proc Natl Acad Sci U S A ; 108(7): 2783-8, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21270334

RESUMEN

The ADP ribosyl transferase [poly(ADP-ribose) polymerase] ARTD3(PARP3) is a newly characterized member of the ARTD(PARP) family that catalyzes the reaction of ADP ribosylation, a key posttranslational modification of proteins involved in different signaling pathways from DNA damage to energy metabolism and organismal memory. This enzyme shares high structural similarities with the DNA repair enzymes PARP1 and PARP2 and accordingly has been found to catalyse poly(ADP ribose) synthesis. However, relatively little is known about its in vivo cellular properties. By combining biochemical studies with the generation and characterization of loss-of-function human and mouse models, we describe PARP3 as a newcomer in genome integrity and mitotic progression. We report a particular role of PARP3 in cellular response to double-strand breaks, most likely in concert with PARP1. We identify PARP3 as a critical player in the stabilization of the mitotic spindle and in telomere integrity notably by associating and regulating the mitotic components NuMA and tankyrase 1. Both functions open stimulating prospects for specifically targeting PARP3 in cancer therapy.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Roturas del ADN de Doble Cadena , Inestabilidad Genómica/genética , Mitosis/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Huso Acromático/fisiología , Adenosina Difosfato/metabolismo , Animales , Antígenos Nucleares/metabolismo , Western Blotting , Línea Celular Tumoral , Ensayo de Unidades Formadoras de Colonias , Ensayo Cometa , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente Indirecta , Inestabilidad Genómica/fisiología , Humanos , Inmunoprecipitación , Hibridación Fluorescente in Situ , Espectrometría de Masas , Ratones , Ratones Noqueados , Microscopía por Video , Mitosis/fisiología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Poli(ADP-Ribosa) Polimerasas/deficiencia , Tanquirasas/metabolismo
18.
Nat Rev Immunol ; 24(7): 471-486, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38273127

RESUMEN

There have been major advances in the immunotherapy of cancer in recent years, including the development of T cell engagers - antibodies engineered to redirect T cells to recognize and kill cancer cells - for the treatment of haematological malignancies. However, the field still faces several challenges to develop agents that are consistently effective in a majority of patients and cancer types, such as optimizing drug dose, overcoming treatment resistance and improving efficacy in solid tumours. A new generation of T cell-targeted molecules was developed to tackle these issues that are potentially more effective and safer. In addition, agents designed to engage the antitumour activities of other immune cells, including natural killer cells and myeloid cells, are showing promise and have the potential to treat a broader range of cancers.


Asunto(s)
Inmunoterapia , Células Asesinas Naturales , Neoplasias , Linfocitos T , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Linfocitos T/inmunología , Animales , Células Mieloides/inmunología
19.
J Biol Chem ; 287(53): 44703-13, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23124203

RESUMEN

During B cell differentiation in the bone marrow, the expression and activation of the pre-B cell receptor (pre-BCR) constitute crucial checkpoints for B cell development. Both constitutive and ligand-dependent pre-BCR activation modes have been described. The pre-BCR constitutes an immunoglobulin heavy chain (Igµ) and a surrogate light chain composed of the invariant λ5 and VpreB proteins. We previously showed that galectin-1 (GAL1), produced by bone marrow stromal cells, is a pre-BCR ligand that induces receptor clustering, leading to efficient pre-BII cell proliferation and differentiation. GAL1 interacts with the pre-BCR via the unique region of λ5 (λ5-UR). Here, we investigated the solution structure of a minimal λ5-UR motif that interacts with GAL1. This motif adopts a stable helical conformation that docks onto a GAL1 hydrophobic surface adjacent to its carbohydrate binding site. We identified key hydrophobic residues from the λ5-UR as crucial for the interaction with GAL1 and for pre-BCR clustering. These residues involved in GAL1-induced pre-BCR activation are different from those essential for autonomous receptor activation. Overall, our results indicate that constitutive and ligand-induced pre-BCR activation could occur in a complementary manner.


Asunto(s)
Galectina 1/química , Galectina 1/metabolismo , Receptores de Células Precursoras de Linfocitos B/química , Receptores de Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Sitios de Unión , Diferenciación Celular , Línea Celular , Proliferación Celular , Cristalografía por Rayos X , Galectina 1/genética , Humanos , Ligandos , Modelos Moleculares , Receptores de Células Precursoras de Linfocitos B/genética , Células Precursoras de Linfocitos B/química , Células Precursoras de Linfocitos B/citología , Células del Estroma/citología , Células del Estroma/metabolismo
20.
Cell Mol Life Sci ; 69(4): 629-40, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21773671

RESUMEN

Functional telomeres are protected from non-homologous end-joining (NHEJ) and homologous recombination (HR) DNA repair pathways. Replication is a critical period for telomeres because of the requirement for reconstitution of functional protected telomere conformations, a process that involves DNA repair proteins. Using knockdown of DNA-PKcs and Rad51 expression in three different cell lines, we demonstrate the respective involvement of NHEJ and HR in the formation of telomere aberrations induced by the G-quadruplex ligand 360A during or after replication. HR contributed to specific chromatid-type aberrations (telomere losses and doublets) affecting the lagging strand telomeres, whereas DNA-PKcs-dependent NHEJ was responsible for sister telomere fusions as a direct consequence of G-quadruplex formation and/or stabilization induced by 360A on parental telomere G strands. NHEJ and HR activation at telomeres altered mitotic progression in treated cells. In particular, NHEJ-mediated sister telomere fusions were associated with altered metaphase-anaphase transition and anaphase bridges and resulted in cell death during mitosis or early G1. Collectively, these data elucidate specific molecular and cellular mechanisms triggered by telomere targeting by the G-quadruplex ligand 360A, leading to cancer cell death.


Asunto(s)
Apoptosis , Proteína Quinasa Activada por ADN/metabolismo , G-Cuádruplex , Mitosis/genética , Proteínas Nucleares/metabolismo , Piridinas/farmacología , Quinolinas/farmacología , Recombinasa Rad51/metabolismo , Telómero , Anafase , Línea Celular , Reparación del ADN por Unión de Extremidades , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/genética , Recombinación Homóloga , Humanos , Ligandos , Metafase , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Recombinasa Rad51/antagonistas & inhibidores , Recombinasa Rad51/genética , Telómero/metabolismo , Telómero/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA