Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Acta Neuropathol Commun ; 10(1): 82, 2022 06 03.
Article de Anglais | MEDLINE | ID: mdl-35659116

RÉSUMÉ

Based on immunostainings and biochemical analyses, certain post-translationally modified alpha-synuclein (aSyn) variants, including C-terminally truncated (CTT) and Serine-129 phosphorylated (pSer129) aSyn, are proposed to be involved in the pathogenesis of synucleinopathies such as Parkinson's disease with (PDD) and without dementia (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). However, quantitative information about aSyn proteoforms in the human brain in physiological and different pathological conditions is still limited. To address this, we generated sequential biochemical extracts of the substantia nigra, putamen and hippocampus from 28 donors diagnosed and neuropathologically-confirmed with different synucleinopathies (PD/PDD/DLB/MSA), as well as Alzheimer's disease, progressive supranuclear palsy, and aged normal subjects. The tissue extracts were used to build a reverse phase array including 65 aSyn antibodies for detection. In this multiplex approach, we observed increased immunoreactivity in donors with synucleinopathies compared to controls in detergent-insoluble fractions, mainly for antibodies against CT aSyn and pSer129 aSyn. In addition, despite of the restricted sample size, clustering analysis suggested disease-specific immunoreactivity signatures in patient groups with different synucleinopathies. We aimed to validate and quantify these findings using newly developed immunoassays towards total, 119 and 122 CTT, and pSer129 aSyn. In line with previous studies, we found that synucleinopathies shared an enrichment of post-translationally modified aSyn in detergent-insoluble fractions compared to the other analyzed groups. Our measurements allowed for a quantitative separation of PDD/DLB patients from other synucleinopathies based on higher detergent-insoluble pSer129 aSyn concentrations in the hippocampus. In addition, we found that MSA stood out due to enrichment of CTT and pSer129 aSyn also in the detergent-soluble fraction of the SN and putamen. Together, our results achieved by multiplexed and quantitative immunoassay-based approaches in human brain extracts of a limited sample set point to disease-specific biochemical aSyn proteoform profiles in distinct neurodegenerative disorders.


Sujet(s)
Maladie à corps de Lewy , Atrophie multisystématisée , Synucléinopathies , Sujet âgé , Encéphale/anatomopathologie , Détergents , Humains , Maladie à corps de Lewy/anatomopathologie , Atrophie multisystématisée/anatomopathologie , alpha-Synucléine/métabolisme
2.
Acta Neuropathol ; 142(3): 423-448, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34115198

RÉSUMÉ

Various post-translationally modified (PTM) proteoforms of alpha-synuclein (aSyn)-including C-terminally truncated (CTT) and Serine 129 phosphorylated (Ser129-p) aSyn-accumulate in Lewy bodies (LBs) in different regions of the Parkinson's disease (PD) brain. Insight into the distribution of these proteoforms within LBs and subcellular compartments may aid in understanding the orchestration of Lewy pathology in PD. We applied epitope-specific antibodies against CTT and Ser129-p aSyn proteoforms and different aSyn domains in immunohistochemical multiple labelings on post-mortem brain tissue from PD patients and non-neurological, aged controls, which were scanned using high-resolution 3D multicolor confocal and stimulated emission depletion (STED) microscopy. Our multiple labeling setup highlighted a consistent onion skin-type 3D architecture in mature nigral LBs in which an intricate and structured-appearing framework of Ser129-p aSyn and cytoskeletal elements encapsulates a core enriched in CTT aSyn species. By label-free CARS microscopy we found that enrichments of proteins and lipids were mainly localized to the central portion of nigral aSyn-immunopositive (aSyn+) inclusions. Outside LBs, we observed that 122CTT aSyn+ punctae localized at mitochondrial membranes in the cytoplasm of neurons in PD and control brains, suggesting a physiological role for 122CTT aSyn outside of LBs. In contrast, very limited to no Ser129-p aSyn immunoreactivity was observed in brains of non-neurological controls, while the alignment of Ser129-p aSyn in a neuronal cytoplasmic network was characteristic for brains with (incidental) LB disease. Interestingly, Ser129-p aSyn+ network profiles were not only observed in neurons containing LBs but also in neurons without LBs particularly in donors at early disease stage, pointing towards a possible subcellular pathological phenotype preceding LB formation. Together, our high-resolution and 3D multicolor microscopy observations in the post-mortem human brain provide insights into potential mechanisms underlying a regulated LB morphogenesis.


Sujet(s)
Chimie du cerveau , Maladie de Parkinson/métabolisme , Fractions subcellulaires/métabolisme , alpha-Synucléine/métabolisme , Sujet âgé , Biobanques , Cytoplasme/anatomopathologie , Cytoplasme/ultrastructure , Cytosquelette/métabolisme , Cytosquelette/ultrastructure , Humains , Corps d'inclusion/anatomopathologie , Corps d'inclusion/ultrastructure , Corps de Lewy/métabolisme , Mâle , Microscopie confocale , Adulte d'âge moyen , Neurones/anatomopathologie , Neurones/ultrastructure , Maturation post-traductionnelle des protéines , alpha-Synucléine/génétique
3.
Front Immunol ; 10: 1962, 2019.
Article de Anglais | MEDLINE | ID: mdl-31555260

RÉSUMÉ

Anti-tumor immunity is limited by a number of factors including the lack of fully activated T-cells, insufficient antigenic stimulation and the immune-suppressive tumor microenvironment. We addressed these hurdles by developing a novel class of immunoconjugates, Antibody-Targeted Pathogen-derived Peptides (ATPPs), which were designed to efficiently deliver viral T-cell epitopes to tumors with the aim of redirecting virus-specific memory T-cells against the tumor. ATPPs were generated through covalent binding of mature MHC class I peptides to antibodies specific for cell surface-expressed tumor antigens that mediate immunoconjugate internalization. By means of a cleavable linker, the peptides are released in the endosomal compartment, from which they are loaded into MHC class I without the need for further processing. Pulsing of tumor cells with ATPPs was found to sensitize these for recognition by virus-specific CD8+ T-cells with much greater efficiency than exogenous loading with free peptides. Systemic injection of ATPPs into tumor-bearing mice enhanced the recruitment of virus-specific T-cells into the tumor and, when combined with immune checkpoint blockade, suppressed tumor growth. Our data thereby demonstrate the potential of ATPPs as a means of kick-starting the immune response against "cold" tumors and increasing the efficacy of checkpoint inhibitors.


Sujet(s)
Lymphocytes T CD8+/immunologie , Déterminants antigéniques des lymphocytes T/administration et posologie , Immunoconjugués/administration et posologie , Tumeurs/thérapie , Peptides/administration et posologie , Animaux , Lignée cellulaire tumorale , Femelle , Herpèsvirus humain de type 4 , Humains , Immunothérapie , Souris
4.
Nucleic Acids Res ; 47(10): e55, 2019 06 04.
Article de Anglais | MEDLINE | ID: mdl-30809660

RÉSUMÉ

We report a novel system for efficient and specific targeted delivery of large nucleic acids to and into cells. Plasmid DNA and core histones were assembled to chromatin by salt gradient dialysis and subsequently connected to bispecific antibody derivatives (bsAbs) via a nucleic acid binding peptide bridge. The resulting reconstituted vehicles termed 'plasmid-chromatin' deliver packaged nucleic acids to and into cells expressing antigens that are recognized by the bsAb, enabling intracellular functionality without detectable cytotoxicity. High efficiency of intracellular nucleic acid delivery is revealed by intracellular expression of plasmid encoded genes in most (∼90%) target cells to which the vehicles were applied under normal growth/medium conditions in nanomolar concentrations. Specific targeting, uptake and transgene expression depends on antibody-mediated cell surface binding: plasmid chromatin of identical composition but with non-targeting bsAbs or without bsAbs is ineffective. Examples that demonstrate applicability, specificity and efficacy of antibody-targeted plasmid chromatin include reporter gene constructs as well as plasmids that enable CRISPR/Cas9 mediated genome editing of target cells.


Sujet(s)
Anticorps bispécifiques/métabolisme , Systèmes CRISPR-Cas , Chromatine/métabolisme , Édition de gène/méthodes , Plasmides/génétique , Anticorps bispécifiques/immunologie , Chromatine/génétique , ADN/génétique , ADN/métabolisme , Humains , Cellules MCF-7 , Microscopie confocale , Transgènes/génétique
5.
Redox Biol ; 20: 146-156, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30312900

RÉSUMÉ

The diphthamide modification of translation elongation factor 2 is highly conserved in eukaryotes and archaebacteria. Nevertheless, cells lacking diphthamide can carry out protein synthesis and are viable. We have analyzed the phenotypes of diphthamide deficient cells and found that diphthamide deficiency reduces selenocysteine incorporation into selenoproteins. Additional phenotypes resulting from diphthamide deficiency include altered tRNA-synthetase and selenoprotein transcript levels, hypersensitivity to oxidative stress and increased selenite tolerance. Diphthamide-eEF2 occupies the aminoacyl-tRNA translocation site at which UGA either stalls translation or decodes selenocysteine. Its position is in close proximity and mutually exclusive to the ribosomal binding site of release/recycling factor ABCE1, which harbors a redox-sensitive Fe-S cluster and, like diphthamide, is present in eukaryotes and archaea but not in eubacteria. Involvement of diphthamide in UGA-SECIS decoding may explain deregulated selenoprotein expression and as a consequence oxidative stress, NFkB activation and selenite tolerance in diphthamide deficient cells.


Sujet(s)
Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Histidine/analogues et dérivés , Sélénoprotéines/génétique , Amino acyl-tRNA synthetases/génétique , Amino acyl-tRNA synthetases/métabolisme , Lignée cellulaire tumorale , Histidine/pharmacologie , Humains , Facteur de transcription NF-kappa B/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Acide sélénieux/pharmacologie , Sélénocystéine/métabolisme , Sélénoprotéines/métabolisme
6.
Nucleic Acids Res ; 47(2): 953-969, 2019 01 25.
Article de Anglais | MEDLINE | ID: mdl-30462278

RÉSUMÉ

Locked nucleic acid based antisense oligonucleotides (LNA-ASOs) can reach their intracellular RNA targets without delivery modules. Functional cellular uptake involves vesicular accumulation followed by translocation to the cytosol and nucleus. However, it is yet unknown how many LNA-ASO molecules need to be delivered to achieve target knock down. Here we show by quantitative fluorescence imaging combined with LNA-ASO microinjection into the cytosol or unassisted uptake that ∼105 molecules produce >50% knock down of their targets, indicating that a substantial amount of LNA-ASO escapes from endosomes. Microinjected LNA-ASOs redistributed within minutes from the cytosol to the nucleus and remained bound to nuclear components. Together with the fact that RNA levels for a given target are several orders of magnitude lower than the amounts of LNA-ASO, our data indicate that only a minor fraction is available for RNase H1 mediated reduction of target RNA. When non-specific binding sites were blocked by co-administration of non-related LNA-ASOs, the amount of target LNA-ASO required was reduced by an order of magnitude. Therefore, dynamic processes within the nucleus appear to influence the distribution and activity of LNA-ASOs and may represent important parameters for improving their efficacy and potency.


Sujet(s)
Techniques de knock-down de gènes , Oligonucléotides/analyse , Noyau de la cellule/génétique , Redistribution de fluorescence après photoblanchiment , Humains , Cellules MCF-7 , Microinjections , Microscopie de fluorescence , Oligonucléotides/administration et posologie , Oligonucléotides antisens/administration et posologie , Oligonucléotides antisens/analyse
7.
Biol Chem ; 399(7): 711-721, 2018 06 27.
Article de Anglais | MEDLINE | ID: mdl-29466231

RÉSUMÉ

A transcellular shuttle system was generated for the delivery of non-covalently linked payloads across blood-brain barrier (BBB) endothelial cells. Transcytosis-enabling shuttles are composed of bispecific antibodies (bsAbs) that simultaneously bind transferrin receptor (TfR) and haptens such as digoxigenin or biocytinamide. Haptenylated payloads are attached to these vehicles via non-covalent hapten-antibody complexation. This enables targeting to and internalization into human BBB-derived microvascular endothelial hCMEC/D3 cells. In contrast to other shuttles, this system does not require special affinities or formats of their TfR-binding moieties for transcytosis and subsequent release. Non-covalent payload complexation to bsAb is flexible and robust, works for a multitude of payloads and enables separation of payloads from shuttles during transcytosis. Released payloads can enter the brain without connected bsAb entities, minimizing potential interference with distribution or functionality. Intracellular separation of shuttle and payload and recycling to cell surfaces may also enable recharging of the cell-bound BBB shuttle with payload for subsequent (merry-go-round) transport cycles.


Sujet(s)
Anticorps bispécifiques/métabolisme , Barrière hémato-encéphalique/métabolisme , Cellules endothéliales/métabolisme , Transcytose , Lignée cellulaire , Cellules HEK293 , Humains
8.
Proc Natl Acad Sci U S A ; 112(19): 5997-6002, 2015 May 12.
Article de Anglais | MEDLINE | ID: mdl-25918417

RÉSUMÉ

Here, we investigated the influence of the variable fragment (Fv) of IgG antibodies on the binding to the neonatal Fc receptor (FcRn) as well as on FcRn-dependent pharmacokinetics (PK). FcRn plays a key role in IgG homeostasis, and specific manipulation in the crystallizable fragment (Fc) is known to affect FcRn-dependent PK. Although the influence of the antigen-binding fragment (Fab) on FcRn interactions has been reported, the underlying mechanism is hitherto only poorly understood. Therefore, we analyzed the two IgG1 antibodies, briakinumab and ustekinumab, that have similar Fc parts but different terminal half-lives in human and systematically engineered variants of them with cross-over exchanges and varied charge distribution. Using FcRn affinity chromatography, molecular dynamics simulation, and in vivo PK studies in human FcRn transgenic mice, we provide evidence that the charge distribution on the Fv domain is involved in excessive FcRn binding. This excessive binding prevents efficient FcRn-IgG dissociation at physiological pH, thereby reducing FcRn-dependent terminal half-lives. Furthermore, we observed a linear correlation between FcRn column retention times of the antibody variants and the terminal half-lives in vivo. Taken together, our study contributes to a better understanding of the FcRn-IgG interaction, and it could also provide profound potential in FcRn-dependent antibody engineering of the variable Fab region.


Sujet(s)
Antigènes d'histocompatibilité de classe I/composition chimique , Récepteur Fc/composition chimique , Animaux , Anticorps/composition chimique , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux humanisés/composition chimique , Réaction antigène-anticorps , Chromatographie d'affinité , Femelle , Cellules endothéliales de la veine ombilicale humaine , Humains , Concentration en ions d'hydrogène , Fragments Fc des immunoglobulines/composition chimique , Immunoglobuline G/composition chimique , Souris , Souris transgéniques , Microscopie confocale , Simulation de dynamique moléculaire , Liaison aux protéines , Conformation des protéines , Ingénierie des protéines , Multimérisation de protéines , Électricité statique , Résonance plasmonique de surface , Ustékinumab , bêta-2-Microglobuline/composition chimique
9.
FASEB J ; 29(5): 1763-79, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25670234

RÉSUMÉ

Humanized hapten-binding IgGs were designed with an accessible cysteine close to their binding pockets, for specific covalent payload attachment. Individual analyses of known structures of digoxigenin (Dig)- and fluorescein (Fluo) binding antibodies and a new structure of a biotin (Biot)-binder, revealed a "universal" coupling position (52(+2)) in proximity to binding pockets but without contributing to hapten interactions. Payloads that carry a free thiol are positioned on the antibody and covalently linked to it via disulfides. Covalent coupling is achieved and driven toward complete (95-100%) payload occupancy by spontaneous redox shuffling between antibody and payload. Attachment at the universal position works with different haptens, antibodies, and payloads. Examples are the haptens Fluo, Dig, and Biot combined with various fluorescent or peptidic payloads. Disulfide-bonded covalent antibody-payload complexes do not dissociate in vitro and in vivo. Coupling requires the designed cysteine and matching payload thiol because payload or antibody without the Cys/thiol are not linked (<5% nonspecific coupling). Hapten-mediated positioning is necessary as hapten-thiol-payload is only coupled to antibodies that bind matching haptens. Covalent complexes are more stable in vivo than noncovalent counterparts because digoxigeninylated or biotinylated fluorescent payloads without disulfide-linkage are cleared more rapidly in mice (approximately 50% reduced 48 hour serum levels) compared with their covalently linked counterparts. The coupling technology is applicable to many haptens and hapten binding antibodies (confirmed by automated analyses of the structures of 140 additional hapten binding antibodies) and can be applied to modulate the pharmacokinetics of small compounds or peptides. It is also suitable to link payloads in a reduction-releasable manner to tumor- or tissue-targeting delivery vehicles.


Sujet(s)
Anticorps/immunologie , Disulfures/immunologie , Haptènes/immunologie , Fragments peptidiques/immunologie , Animaux , Anticorps/composition chimique , Anticorps/métabolisme , Disulfures/composition chimique , Disulfures/métabolisme , Haptènes/composition chimique , Haptènes/métabolisme , Souris , Fragments peptidiques/composition chimique , Fragments peptidiques/métabolisme , Liaison aux protéines , Thiols/composition chimique , Thiols/immunologie , Thiols/métabolisme
10.
Cancer Immunol Res ; 3(7): 764-76, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-25691327

RÉSUMÉ

Tumor cells escape immune eradication through multiple mechanisms, including loss of antigenicity and local suppression of effector lymphocytes. To counteract these obstacles, we aimed to direct the unique cytomegalovirus (CMV)-specific immune surveillance against tumor cells. We developed a novel generation of fusion proteins composed of a tumor antigen-specific full immunoglobulin connected to a single major histocompatibility class I complex bearing a covalently linked virus-derived peptide (pMHCI-IgG). Here, we show that tumor antigen-expressing cancer cells, which are decorated with pMHCI-IgGs containing a HLA-A*0201 molecule associated with a CMV-derived peptide, are specifically eliminated through engagement of antigen-specific CD8(+) T cells isolated from peripheral blood mononuclear cell preparations of CMV-infected humans. These CD8(+) T cells act without additional expansion, preactivation, or provision of costimulatory signals. Elimination of tumor cells is induced at similar concentrations and with similar time kinetics as those seen with bispecific T-cell engagers (BiTE). However, while BiTE-like reagents indiscriminately activate T cells through binding to the T-cell receptor complex, pMHCI-IgGs selectively engage antigen-specific, constantly renewable, differentiated effector cytotoxic T lymphocytes to tumor cells, thereby representing a novel class of anticancer immunotherapeutics with potentially improved safety and efficacy profiles.


Sujet(s)
Cytomegalovirus/immunologie , Antigène HLA-A2/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Agranulocytes/immunologie , Lymphocytes T cytotoxiques/immunologie , Animaux , Anticorps/immunologie , Lignée cellulaire tumorale , Humains , Activation des lymphocytes/immunologie , Souris , Peptides/immunologie
11.
Mol Cancer Ther ; 12(10): 2031-42, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-23873847

RÉSUMÉ

We report the first preclinical in vitro and in vivo comparison of GA101 (obinutuzumab), a novel glycoengineered type II CD20 monoclonal antibody, with rituximab and ofatumumab, the two currently approved type I CD20 antibodies. The three antibodies were compared in assays measuring direct cell death (AnnexinV/PI staining and time-lapse microscopy), complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), and internalization. The models used for the comparison of their activity in vivo were SU-DHL4 and RL xenografts. GA101 was found to be superior to rituximab and ofatumumab in the induction of direct cell death (independent of mechanical manipulation required for cell aggregate disruption formed by antibody treatment), whereas it was 10 to 1,000 times less potent in mediating CDC. GA101 showed superior activity to rituximab and ofatumumab in ADCC and whole-blood B-cell depletion assays, and was comparable with these two in ADCP. GA101 also showed slower internalization rate upon binding to CD20 than rituximab and ofatumumab. In vivo, GA101 induced a strong antitumor effect, including complete tumor remission in the SU-DHL4 model and overall superior efficacy compared with both rituximab and ofatumumab. When rituximab-pretreated animals were used, second-line treatment with GA101 was still able to control tumor progression, whereas tumors escaped rituximab treatment. Taken together, the preclinical data show that the glyoengineered type II CD20 antibody GA101 is differentiated from the two approved type I CD20 antibodies rituximab and ofatumumab by its overall preclinical activity, further supporting its clinical investigation.


Sujet(s)
Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux d'origine murine/administration et posologie , Anticorps monoclonaux/administration et posologie , Tumeurs/traitement médicamenteux , Animaux , Anticorps monoclonaux humanisés/effets indésirables , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cytotoxicité immunologique/effets des médicaments et des substances chimiques , Humains , Souris , Tumeurs/immunologie , Tumeurs/anatomopathologie , Phagocytose/effets des médicaments et des substances chimiques , Phagocytose/immunologie , Rituximab , Tests d'activité antitumorale sur modèle de xénogreffe
12.
J Nucl Med ; 53(2): 304-11, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22228796

RÉSUMÉ

UNLABELLED: The putative presence of the erythropoietin receptor (EpoR) on human cancer cells has given rise to controversial discussion about the use of recombinant human erythropoietin (rhuEpo) for treatment of patients with chemotherapy-induced anemia. In vivo analysis of the EpoR status in tumors could help in elucidating the role of erythropoietin in cancer. Thus, the aim of this study was to develop a targeted EpoR probe for the investigation of EpoR expression in human lung cancer xenografts by fluorescence-mediated tomography. METHODS: Epo-Cy5.5 was generated by coupling Cy5.5 to rhuEpo. In vitro binding assays were performed using the EpoR-positive non-small cell lung cancer (NSCLC) cell lines A549 (lower EpoR expression) and H838 (higher EpoR expression), the EpoR-negative cell line H2030, and EpoR/EGFP-overexpressing HeLa cells. In vivo specificity of Epo-Cy5.5 was confirmed by competition analyses using micro-CT/fluorescence-mediated tomography fusion imaging. Biodistribution was analyzed over 50 h after injection. Binding of Epo-Cy5.5 was validated on tumor cryosections. RESULTS: After intravenous injection, the probe was rapidly cleared from the circulation. An accumulation was observed in liver and kidneys, with a maximum at 7 h after injection followed by a decline, indicating renal excretion. Almost constant accumulation of Epo-Cy5.5 was found in bone marrow and tumors, indicating specific receptor binding. The probe allowed the discrimination between H838 with higher EpoR expression (89.54 ± 15.91 nM at 25 h) and A549 tumors with lower EpoR expression (60.45 ± 14.59 nM at 25 h, P < 0.05). Tumor accumulation of Epo-Cy5.5 could be significantly reduced by adding unlabeled rhuEpo (P < 0.05 at 4, 7, and 24 h). In vitro validation confirmed specific binding of Epo-Cy5.5 to the tumor cells, and this binding correlated with the EpoR expression level. Binding was also observed on endothelial cells. Vessel density and Epo-Cy5.5 binding on endothelial cells were comparable. CONCLUSION: Epo-Cy5.5 allows the longitudinal analysis of EpoR expression in tumors and thereby can investigate the influence of erythropoietin on EpoR expression, tumor growth, and angiogenesis.


Sujet(s)
Transformation cellulaire néoplasique , Rayons infrarouges , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Imagerie moléculaire/méthodes , Sondes moléculaires/métabolisme , Récepteur érythropoïétine/métabolisme , Animaux , Moelle osseuse/métabolisme , Carbocyanines/métabolisme , Lignée cellulaire tumorale , Cellules endothéliales/métabolisme , Érythropoïétine/métabolisme , Érythropoïétine/pharmacocinétique , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Souris , Sondes moléculaires/pharmacocinétique , Spécificité du substrat
13.
Mol Ther Nucleic Acids ; 1: e46, 2012 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-23344238

RÉSUMÉ

Bispecific antibodies (bsAbs) that bind to cell surface antigens and to digoxigenin (Dig) were used for targeted small interfering RNA (siRNA) delivery. They are derivatives of immunoglobulins G (IgGs) that bind tumor antigens, such as Her2, IGF1-R, CD22, and LeY, with stabilized Dig-binding variable domains fused to the C-terminal ends of the heavy chains. siRNA that was digoxigeninylated at its 3'end was bound in a 2:1 ratio to the bsAbs. These bsAb-siRNA complexes delivered siRNAs specifically to cells that express the corresponding antigen as demonstrated by flow cytometry and confocal microscopy. The complexes internalized into endosomes and Dig-siRNAs separated from bsAbs, but Dig-siRNA was not released into the cytoplasm; bsAb-targeting alone was thus not sufficient for effective mRNA knockdown. This limitation was overcome by formulating the Dig-siRNA into nanoparticles consisting of dynamic polyconjugates (DPCs) or into lipid-based nanoparticles (LNPs). The resulting complexes enabled bsAb-targeted siRNA-specific messenger RNA (mRNA) knockdown with IC(50) siRNA values in the low nanomolar range for a variety of bsAbs, siRNAs, and target cells. Furthermore, pilot studies in mice bearing tumor xenografts indicated mRNA knockdown in endothelial cells following systemic co-administration of bsAbs and siRNA formulated in LNPs that were targeted to the tumor vasculature.Molecular Therapy - Nucleic Acids (2012) 1, e45; doi:10.1038/mtna.2012.39; published online 18 September 2012.

14.
Biochem J ; 442(3): 483-94, 2012 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-22132769

RÉSUMÉ

Hepsin is a type II transmembrane serine protease that is expressed in several human tissues. Overexpression of hepsin has been found to correlate with tumour progression and metastasis, which is so far best studied for prostate cancer, where more than 90% of such tumours show this characteristic. To enable improved future patient treatment, we have developed a monoclonal humanized antibody that selectively inhibits human hepsin and does not inhibit other related proteases. We found that our antibody, hH35, potently inhibits hepsin enzymatic activity at nanomolar concentrations. Kinetic characterization revealed non-linear, slow, tight-binding inhibition. This correlates with the crystal structure we obtained for the human hepsin-hH35 antibody Fab fragment complex, which showed that the antibody binds hepsin around α3-helix, located far from the active centre. The unique allosteric mode of inhibition of hH35 is distinct from the recently described HGFA (hepatocyte growth factor activator) allosteric antibody inhibition. We further explain how a small change in the antibody design induces dramatic structural rearrangements in the hepsin antigen upon binding, leading to complete enzyme inactivation.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Serine endopeptidases/métabolisme , Inhibiteurs de la sérine protéinase/pharmacologie , Régulation allostérique , Animaux , Anticorps monoclonaux/composition chimique , Cristallographie aux rayons X , Humains , Souris , Souris de lignée BALB C , Modèles moléculaires , Conformation des protéines , Inhibiteurs de la sérine protéinase/composition chimique , Transfection
15.
Proc Natl Acad Sci U S A ; 108(20): 8194-9, 2011 May 17.
Article de Anglais | MEDLINE | ID: mdl-21536919

RÉSUMÉ

Bispecific antibodies that bind cell-surface targets as well as digoxigenin (Dig) were generated for targeted payload delivery. Targeting moieties are IgGs that bind the tumor antigens Her2, IGF1R, CD22, or LeY. A Dig-binding single-chain Fv was attached in disulfide-stabilized form to C termini of CH3 domains of targeting antibodies. Bispecific molecules were expressed in mammalian cells and purified in the same manner as unmodified IgGs. They are stable without aggregation propensity and retain binding specificity/affinity to cell-surface antigens and Dig. Digoxigeninylated payloads were generated that retain full functionality and can be complexed to bispecific antibodies in a defined 21 ratio. Payloads include small compounds (Dig-Cy5, Dig-Doxorubicin) and proteins (Dig-GFP). Complexed payloads are targeted by the bispecifics to cancer cells and because these complexes are stable in serum, they can be applied for targeted delivery. Because Dig bispecifics also effectively capture digoxigeninylated compounds under physiological conditions, separate administration of uncharged Dig bispecifics followed by application of Dig payload is sufficient to achieve antibody-mediated targeting in vitro and in vivo.


Sujet(s)
Anticorps bispécifiques/usage thérapeutique , Antinéoplasiques/administration et posologie , Digoxigénine/immunologie , Systèmes de délivrance de médicaments/méthodes , Anticorps bispécifiques/immunologie , Antigènes néoplasiques/immunologie , Carbocyanines/administration et posologie , Lignée cellulaire tumorale , Doxorubicine/administration et posologie , Protéines à fluorescence verte/administration et posologie , Humains , Méthodes , Anticorps à chaîne unique
16.
Blood ; 118(2): 358-67, 2011 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-21444918

RÉSUMÉ

CD20 is a cell-surface marker of normal and malignant B cells. Rituximab, a monoclonal antibody targeting CD20, has improved the treatment of malignant lymphomas. Therapeutic CD20 antibodies are classified as either type I or II based on different mechanisms of killing malignant B cells. To reveal the molecular basis of this distinction, we fine-mapped the epitopes recognized by both types. We also determined the first X-ray structure of a type II antibody by crystallizing the obinutuzumab (GA101) Fab fragment alone and in complex with a CD20 cyclopeptide. Despite recognizing an overlapping epitope, GA101 binds CD20 in a completely different orientation than type I antibodies. Moreover, the elbow angle of GA101 is almost 30° wider than in type I antibodies, potentially resulting in different spatial arrangements of 2 CD20 molecules bound to a single GA101 or rituximab molecule. Using protein tomography, different CD20 complexes were found to be associated with the 2 antibodies, and confocal microscopy showed different membrane compartmentalization of these subpopulations of the cellular CD20 pool. Our findings offer a possible molecular explanation for the different cellular responses elicited by type I and II antibodies.


Sujet(s)
Anticorps monoclonaux/classification , Antigènes CD20/composition chimique , Antigènes CD20/immunologie , Épitopes/composition chimique , Séquence d'acides aminés , Anticorps monoclonaux/analyse , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux humanisés , Anticorps monoclonaux d'origine murine/composition chimique , Spécificité des anticorps , Antigènes CD20/génétique , Lignée cellulaire , Cristallographie aux rayons X , Cartographie épitopique/méthodes , Épitopes/analyse , Humains , Modèles moléculaires , Mutagenèse dirigée , Structure quaternaire des protéines , Structure secondaire des protéines , Rituximab
17.
J Cell Physiol ; 191(1): 60-8, 2002 Apr.
Article de Anglais | MEDLINE | ID: mdl-11920682

RÉSUMÉ

Claudins and occludin constitute the major transmembrane proteins of tight junctions (TJs). We have previously identified the human homologue of the murine Cldn1, CLDN1 (SEMP1) that is expressed in normal, mammary gland-derived epithelial cells but is absent in most human breast cancer cell lines. To investigate the potential functions of CLDN1 protein in tumor and normal epithelial cells, we developed an I-NGFR retroviral vector and monoclonal anti-CLDN1 antibody. In subconfluent and confluent breast cancer cells, MDA-MB-435 and MDA-MB-361, endogenous CLDN1 expression was not detected by an anti-CLDN1 monoclonal antibody by Western blot analysis or quantitative RT-PCR. When CLDN1-negative breast cancer cell lines were transduced with a CLDN1 retrovirus the cells express CLDN1 mRNA constitutively as shown by quantitative RT-PCR. Immunofluorescence analyses of the CLDN1 retroviral transduced breast tumor cells using monoclonal antibodies against CLDN1 reveals a subcellular distribution at cell-cell contact sites similar to the CLDN1 homing pattern in T47-D cells, which express endogenous CLDN1. This cell-cell contact co-localization of CLDN1 was evident in CLDN1-transduced breast tumor cells which fail to express occludin protein (MDA-MB-361 and MDA-MB-435) and express relatively little ZO-1 protein (MDA-MB-435), suggesting that other proteins may be responsible for targeting of CLDN1 to cell-cell contact sites. The re-expression of CLDN1 decreases the paracellular flux of 3 and 40 kDa dextran despite the absence of occludin in the MDA-MB-361 tumor cells. Our findings indicate that in CLDN1-negative breast tumor cells, the basal protein partner requirements for physiological homing of the CLDN1 protein are intact, and that CLDN1 gene transfer and protein expression itself might be sufficient to exert a TJ-mediate gate function in metastatic tumor cells even in the absence of other TJ-associated proteins, such as occludin.


Sujet(s)
Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Protéines membranaires/déficit , Protéines membranaires/génétique , Animaux , Anticorps monoclonaux , Tumeurs du sein/physiopathologie , Communication cellulaire/physiologie , Lignée cellulaire , Claudine-1 , Cellules épithéliales/métabolisme , Femelle , Ciblage de gène , Techniques de transfert de gènes , Vecteurs génétiques , Humains , Insectes , Protéines membranaires/immunologie , Protéines membranaires/métabolisme , Souris , Souris de lignée BALB C , Occludine , Phosphoprotéines/métabolisme , Retroviridae/génétique , Transduction génétique , Protéine-1 de la zonula occludens
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...