Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
Br J Dermatol ; 191(1): 92-106, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38375775

RÉSUMÉ

BACKGROUND: Extracellular matrices play a critical role in tissue structure and function and aberrant remodelling of these matrices is a hallmark of many age-related diseases. In skin, loss of dermal collagens and disorganization of elastic fibre components are key features of photoageing. Although the application of some small matrix-derived peptides to aged skin has been shown to beneficially affect in vitro cell behaviour and, in vivo, molecular architecture and clinical appearance, the discovery of new peptides has lacked a guiding hypothesis. OBJECTIVES: To identify, using protease cleavage site prediction, novel putative matrikines with beneficial activities for skin composition and structure. METHODS: Here, we present an in silico (peptide cleavage prediction) to in vitro (proteomic and transcriptomic activity testing in cultured human dermal fibroblasts) to in vivo (short-term patch test and longer-term split-face clinical study) discovery pipeline, which enables the identification and characterization of peptides with differential activities. RESULTS: Using this pipeline we showed that cultured fibroblasts were responsive to all applied peptides, but their associated bioactivity was sequence-dependent. Based on bioactivity, toxicity and protein source, we further characterized a combination of two novel peptides, GPKG (glycine-proline-lysine-glycine) and LSVD (leucine-serine-valine-aspartate), that acted in vitro to enhance the transcription of matrix -organization and cell proliferation genes and in vivo (in a short-term patch test) to promote processes associated with epithelial and dermal maintenance and remodelling. Prolonged use of a formulation containing these peptides in a split-face clinical study led to significantly improved measures of crow's feet and firmness in a mixed population. CONCLUSIONS: This approach to peptide discovery and testing can identify new synthetic matrikines, providing insights into biological mechanisms of tissue homeostasis and repair and new pathways to clinical intervention.


Like other organs and tissues, the skin is composed of both cells and a complex network of molecules and proteins called an extracellular matrix. This matrix contains proteins such as collagen and elastin and undergoes many changes when the skin is damaged by the sun. We know from previous studies that small parts of matrix proteins (called peptide 'matrikines') can help to treat the signs of sun-related skin ageing. In this UK study, we show that new beneficial peptides (with matrikine activity) can be identified using machine learning (artificial intelligence) techniques that predict where common matrix proteins might be 'cut' by skin enzymes. Candidate peptides were first made in the laboratory and then applied to skin cells in culture. These cell culture screens demonstrated that, while all the peptides showed some matrikine activity, two were particularly promising. These two peptides were then tested in a short-term study on the forearm skin of volunteers and, in a longer-term study, on the face. We found that the combination of these two peptides can prompt forearm skin cells to express genes that are involved in many different aspect of skin health and, over the longer 6-month period, produce visible benefits in the appearance of fine lines and wrinkles and firmness on the face. Our findings suggest that this approach may be able to identify beneficial peptide treatments for not only skin ageing and diseases, but also unwanted changes in the extracellular matrix of other tissues and organs.


Sujet(s)
Fibroblastes , Oligopeptides , Rajeunissement , Vieillissement de la peau , Humains , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Vieillissement de la peau/effets des médicaments et des substances chimiques , Oligopeptides/pharmacologie , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Peau/métabolisme , Cellules cultivées , Femelle , Adulte d'âge moyen , Prolifération cellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/métabolisme , Mâle , Protéines de la matrice extracellulaire/métabolisme , Adulte , Sujet âgé , Protéomique/méthodes
2.
J Am Heart Assoc ; 10(19): e016287, 2021 10 05.
Article de Anglais | MEDLINE | ID: mdl-34569248

RÉSUMÉ

Background Atherosclerosis is a complex pathology in which dysfunctional endothelium, activated leucocytes, macrophages, and lipid-laden foam cells are implicated, and in which plaque disruption is driven by many putative actors. This study aimed to identify accurate targetable biomarkers using new in vivo approaches to propose tools for improved diagnosis and treatment. Methods and Results Human scFv (single-chain fragment variable) selected by in vivo phage display in a rabbit model of atherosclerosis was reformatted as scFv fused to the scFv-Fc (single-chain fragment variable fused to the crystallizable fragment of immunoglobulin G format) antibodies. Their reactivity was tested using flow cytometry and immunoassays, and aorta sections from animal models and human carotid and coronary artery specimens. A pool of atherosclerotic proteins from human endarterectomies was co-immunoprecipitated with the selected scFv-Fc followed by mass spectrometry for target identification. Near-infrared fluorescence imaging was performed in Apoe-/- mice after injection of an Alexa Fluor 647-labeled scFv-Fc-2c antibody produced in a baculovirus system with 2 additional cysteine residues (ie, 2c) for future coupling to nano-objects for theranostic applications. One scFv-Fc clone (P3) displayed the highest cross-reactivity against atherosclerotic lesion sections (rabbit, mouse, and human) and was chosen for translational development. Mass spectrometry identified galectin-3, a ß-galactoside-binding lectin, as the leader target. ELISA and immunofluorescence assays with a commercial anti-galectin-3 antibody confirmed this specificity. P3 scFv-Fc-2c specifically targeted atherosclerotic plaques in the Apoe-/- mouse model. Conclusions These results provide evidence that the P3 antibody holds great promise for molecular imaging of atherosclerosis and other inflammatory pathologies involving macrophages. Recently, galectin-3 was proposed as a high-value biomarker for the assessment of coronary and carotid atherosclerosis.


Sujet(s)
Athérosclérose , Bactériophages , Plaque d'athérosclérose , Anticorps à chaîne unique , Animaux , Apolipoprotéines E , Athérosclérose/diagnostic , Athérosclérose/génétique , Marqueurs biologiques , Galectine -3/génétique , Humains , Souris , Lapins , Anticorps à chaîne unique/génétique
3.
Front Immunol ; 12: 728322, 2021.
Article de Anglais | MEDLINE | ID: mdl-34512662

RÉSUMÉ

Novel molecules that directly target the neonatal Fc receptor (FcRn) and/or Fc gamma receptors (FcγRs) are emerging as promising treatments for immunoglobulin G (IgG)-dependent autoimmune pathologies. Mutated Fc regions and monoclonal antibodies that target FcRn are currently in clinical development and hold promise for reducing the levels of circulating IgG. Additionally, engineered structures containing multimeric Fc regions allow the dual targeting of FcRn and FcγRs; however, their tolerance needs to first be validated in phase I clinical studies. Here, for the first time, we have developed a modified monomeric recombinant Fc optimized for binding to all FcRns and FcγRs without the drawback of possible tolerance associated with FcγR cross-linking. A rational approach using Fc engineering allowed the selection of LFBD192, an Fc with a combination of six mutations that exhibits improved binding to human FcRn and FcγR as well as mouse FcRn and FcγRIV. The potency of LFBD192 was compared with that of intravenous immunoglobulin (IVIg), an FcRn blocker (Fc-MST-HN), and a trimeric Fc that blocks FcRn and/or immune complex-mediated cell activation through FcγR without triggering an immune reaction in several in vitro tests and validated in three mouse models of autoimmune disease.


Sujet(s)
Antirhumatismaux/pharmacologie , Arthrite expérimentale/prévention et contrôle , Auto-immunité/effets des médicaments et des substances chimiques , Fragments Fc des immunoglobulines/pharmacologie , Récepteur Fc/antagonistes et inhibiteurs , Récepteurs du fragment Fc des IgG/antagonistes et inhibiteurs , Animaux , Antirhumatismaux/métabolisme , Arthrite expérimentale/génétique , Arthrite expérimentale/immunologie , Arthrite expérimentale/métabolisme , Fixation compétitive , Complément C5a/métabolisme , Femelle , Antigènes d'histocompatibilité de classe I/génétique , Antigènes d'histocompatibilité de classe I/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Humains , Fragments Fc des immunoglobulines/génétique , Fragments Fc des immunoglobulines/immunologie , Fragments Fc des immunoglobulines/métabolisme , Interleukine-2/métabolisme , Cellules Jurkat , Cinétique , Souris de lignée C57BL , Souris transgéniques , Mutation , Phagocytose/effets des médicaments et des substances chimiques , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Liaison aux protéines , Ingénierie des protéines , Récepteur Fc/génétique , Récepteur Fc/immunologie , Récepteur Fc/métabolisme , Récepteurs du fragment Fc des IgG/génétique , Récepteurs du fragment Fc des IgG/immunologie , Récepteurs du fragment Fc des IgG/métabolisme , Voie de sécrétion , Transduction du signal , Cellules THP-1
4.
Front Immunol ; 12: 683902, 2021.
Article de Anglais | MEDLINE | ID: mdl-34163482

RÉSUMÉ

Respiratory syncytial virus (RSV) is a public health concern that causes acute lower respiratory tract infection. So far, no vaccine candidate under development has reached the market and the only licensed product to prevent RSV infection in at-risk infants and young children is a monoclonal antibody (Synagis®). Polyclonal human anti-RSV hyper-immune immunoglobulins (Igs) have also been used but were superseded by Synagis® owing to their low titer and large infused volume. Here we report a new drug class of immunoglobulins, derived from human non hyper-immune plasma that was generated by an innovative bioprocess, called Ig cracking, combining expertises in plasma-derived products and affinity chromatography. By using the RSV fusion protein (F protein) as ligand, the Ig cracking process provided a purified and concentrated product, designated hyper-enriched anti-RSV IgG, composed of at least 15-20% target-specific-antibodies from normal plasma. These anti-RSV Ig displayed a strong in vitro neutralization effect on RSV replication. Moreover, we described a novel prophylactic strategy based on local nasal administration of this unique hyper-enriched anti-RSV IgG solution using a mouse model of infection with bioluminescent RSV. Our results demonstrated that very low doses of hyper-enriched anti-RSV IgG can be administered locally to ensure rapid and efficient inhibition of virus infection. Thus, the general hyper-enriched Ig concept appeared a promising approach and might provide solutions to prevent and treat other infectious diseases. IMPORTANCE: Respiratory Syncytial Virus (RSV) is the major cause of acute lower respiratory infections in children, and is also recognized as a cause of morbidity in the elderly. There are still no vaccines and no efficient antiviral therapy against this virus. Here, we described an approach of passive immunization with a new class of hyper-enriched anti-RSV immunoglobulins (Ig) manufactured from human normal plasma. This new class of immunoglobulin plasma derived product is generated by an innovative bioprocess, called Ig cracking, which requires a combination of expertise in both plasma derived products and affinity chromatography. The strong efficacy in a small volume of these hyper-enriched anti-RSV IgG to inhibit the viral infection was demonstrated using a mouse model. This new class of immunoglobulin plasma-derived products could be applied to other pathogens to address specific therapeutic needs in the field of infectious diseases or even pandemics, such as COVID-19.


Sujet(s)
Anticorps antiviraux/administration et posologie , Immunisation passive , Immunoglobuline G/administration et posologie , Infections à virus respiratoire syncytial/prévention et contrôle , Virus respiratoire syncytial humain/immunologie , Administration par voie nasale , Animaux , Anticorps antiviraux/immunologie , Anticorps antiviraux/isolement et purification , Modèles animaux de maladie humaine , Humains , Immunoglobuline G/immunologie , Immunoglobuline G/isolement et purification , Poumon/effets des médicaments et des substances chimiques , Poumon/virologie , Tests de neutralisation , Infections à virus respiratoire syncytial/virologie , Cornets/effets des médicaments et des substances chimiques , Cornets/virologie , Protéines de fusion virale/immunologie , Réplication virale/effets des médicaments et des substances chimiques
5.
Blood Adv ; 3(7): 1129-1144, 2019 04 09.
Article de Anglais | MEDLINE | ID: mdl-30952679

RÉSUMÉ

Infectious mononucleosis, caused by infection with the human gamma-herpesvirus Epstein-Barr virus (EBV), manifests with one of the strongest CD8+ T-cell responses described in humans. The resulting T-cell memory response controls EBV infection asymptomatically in the vast majority of persistently infected individuals. Whether and how dendritic cells (DCs) contribute to the priming of this near-perfect immune control remains unclear. Here we show that of all the human DC subsets, plasmacytoid DCs (pDCs) play a central role in the detection of EBV infection in vitro and in mice with reconstituted human immune system components. pDCs respond to EBV by producing the interferon (IFN) subtypes α1, α2, α5, α7, α14, and α17. However, the virus curtails this type I IFN production with its latent EBV gene products EBNA3A and EBNA3C. The induced type I IFNs inhibit EBV entry and the proliferation of latently EBV-transformed B cells but do not influence lytic reactivation of the virus in vitro. In vivo, exogenous IFN-α14 and IFN-α17, as well as pDC expansion, delay EBV infection and the resulting CD8+ T-cell expansion, but pDC depletion does not significantly influence EBV infection. Thus, consistent with the observation that primary immunodeficiencies compromising type I IFN responses affect only alpha- and beta-herpesvirus infections, we found that EBV elicits pDC responses that transiently suppress viral replication and attenuate CD8+ T-cell expansion but are not required to control primary infection.


Sujet(s)
Cellules dendritiques/immunologie , Infections à virus Epstein-Barr/immunologie , Interféron de type I/biosynthèse , Animaux , Lymphocytes T CD8+/anatomopathologie , Prolifération cellulaire , Humains , Interféron de type I/pharmacologie , Souris , Pénétration virale/effets des médicaments et des substances chimiques , Réplication virale
6.
J Immunol ; 202(5): 1582-1594, 2019 03 01.
Article de Anglais | MEDLINE | ID: mdl-30683704

RÉSUMÉ

The long serum t 1/2 of IgGs is ensured by their interaction with the neonatal Fc receptor (FcRn), which salvages IgG from intracellular degradation. Fc glycosylation is thought not to influence FcRn binding and IgG longevity in vivo. In this article, we demonstrate that hypersialylation of asparagine 297 (N297) enhances IgG serum persistence. This polarized glycosylation is achieved using a novel Fc mutation, a glutamate residue deletion at position 294 (Del) that endows IgGs with an up to 9-fold increase in serum lifespan. The strongest impact was observed when the Del was combined with Fc mutations improving FcRn binding (Del-FcRn+). Enzymatic desialylation of a Del-FcRn+ mutant or its production in a cell line unable to hypersialylate reduced the in vivo serum t 1/2 of the desialylated mutants to that of native FcRn+ mutants. Consequently, our study proves that sialylation of the N297 sugar moiety has a direct impact on human IgG serum persistence.


Sujet(s)
Anticorps/sang , Anticorps/usage thérapeutique , Fragments Fc des immunoglobulines/sang , Fragments Fc des immunoglobulines/composition chimique , Immunoglobuline G/sang , Immunoglobuline G/usage thérapeutique , Animaux , Anticorps/composition chimique , Cellules HEK293 , Période , Humains , Immunoglobuline G/composition chimique , Souris , Souris knockout
7.
Sci Rep ; 8(1): 15016, 2018 10 09.
Article de Anglais | MEDLINE | ID: mdl-30302027

RÉSUMÉ

Atherosclerosis is a chronic, progressive inflammatory disease that may develop into vulnerable lesions leading to thrombosis. This pathology is characterized by the deposition of lipids within the arterial wall and infiltration of immune cells leading to amplification of inflammation. Nowadays there is a rising interest to assess directly the molecular and cellular components that underlie the clinical condition of stroke and myocardial infarction. Single chain fragment variable (scFv)-phages issuing from a human combinatorial library were selected on the lesions induced in a rabbit model of atherosclerosis after three rounds of in vivo phage display. We further implemented a high-throughput flow cytometry method on rabbit protein extracts to individually test one thousand of scFv-phages. Two hundred and nine clones were retrieved on the basis of their specificity for atherosclerotic proteins. Immunohistochemistry assays confirmed the robustness of the designed cytometry protocol. Sequencing of candidates demonstrated their high diversity in VH and VL germline usage. The large number of candidates and their diversity open the way in the discovery of new biomarkers. Here, we successfully showed the capacity of combining in vivo phage display and high-throughput cytometry strategies to give new insights in in vivo targetable up-regulated biomarkers in atherosclerosis.


Sujet(s)
Athérosclérose/immunologie , Techniques d'exposition à la surface cellulaire , Cytométrie en flux , Anticorps à chaîne unique/isolement et purification , Animaux , Anticorps monoclonaux/génétique , Anticorps monoclonaux/immunologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Modèles animaux de maladie humaine , Humains , Immunohistochimie/méthodes , Lapins , Anticorps à chaîne unique/immunologie
8.
Mol Cancer Ther ; 16(7): 1312-1323, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28507002

RÉSUMÉ

Exploratory clinical trials using therapeutic anti-HER3 antibodies strongly suggest that neuregulin (NRG1; HER3 ligand) expression at tumor sites is a predictive biomarker of anti-HER3 antibody efficacy in cancer. We hypothesized that in NRG1-expressing tumors, where the ligand is present before antibody treatment, anti-HER3 antibodies that do not compete with NRG1 for receptor binding have a higher receptor-neutralizing action than antibodies competing with the ligand for binding to HER3. Using time-resolved-fluorescence energy transfer (TR-FRET), we demonstrated that in the presence of recombinant NRG1, binding of 9F7-F11 (a nonligand-competing anti-HER3 antibody) to HER3 is increased, whereas that of ligand-competing anti-HER3 antibodies (H4B-121, U3-1287, Ab#6, Mab205.10.2, and MOR09825) is decreased. Moreover, 9F7-F11 showed higher efficacy than antibodies that compete with the ligand for binding to HER3. Specifically, 9F7-F11 inhibition of cell proliferation and of HER3/AKT/ERK1/2 phosphorylation as well as 9F7-F11-dependent cell-mediated cytotoxicity were higher in cancer cells preincubated with recombinant NRG1 compared with cells directly exposed to the anti-HER3 antibody. This translated in vivo into enhanced growth inhibition of NRG1-expressing BxPC3 pancreatic, A549 lung, and HCC-1806 breast cell tumor xenografts in mice treated with 9F7-F11 compared with H4B-121. Conversely, both antibodies had similar antitumor effect in NRG1-negative HPAC pancreatic carcinoma cells. In conclusion, the allosteric modulator 9F7-F11 shows increased anticancer effectiveness in the presence of NRG1 and thus represents a novel treatment strategy for NRG1-addicted tumors. Mol Cancer Ther; 16(7); 1312-23. ©2017 AACR.


Sujet(s)
Anticorps monoclonaux d'origine murine/administration et posologie , Marqueurs biologiques tumoraux/immunologie , Tumeurs/traitement médicamenteux , Neuréguline-1/génétique , Récepteur ErbB-3/immunologie , Cellules A549 , Animaux , Anticorps anti-idiotypiques/administration et posologie , Anticorps anti-idiotypiques/immunologie , Anticorps monoclonaux d'origine murine/immunologie , Marqueurs biologiques tumoraux/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Transfert d'énergie par résonance de fluorescence , Régulation de l'expression des gènes tumoraux , Humains , Souris , Tumeurs/génétique , Tumeurs/immunologie , Tumeurs/anatomopathologie , Neuréguline-1/immunologie , Phosphorylation , Liaison aux protéines , Récepteur ErbB-3/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
9.
J Cosmet Sci ; 68(2): 183-194, 2017.
Article de Anglais | MEDLINE | ID: mdl-29619942

RÉSUMÉ

Skin produces sebum through sebocytes. Hyper-seborrhea creates conditions for the development of inflamed cutaneous alterations through bacteria colonization triggering dead cell accumulation and pro-inflammatory mediator release. Study of sebum production, its modulation, and its consequences requires complementary in vitro models in order to evaluate the effect of molecules on cell metabolisms. Clinical studies need to be performed to confirm in vitro results. Effects of phenylpropanoids, obtained by elicitation and purification from plant cell culture of Syringa vulgaris (CCSV), were studied on sebocytes, keratinocytes, and explants, all derived from normal human skins. Normal human sebocytes (NHSs) expressed markers such as cytokeratin-7, cytokeratin-4, and perilipin-2 (PLIN-2) (1); the latter being colocalized with lipid droplets. Lipid droplets clearly appeared and their size increased rapidly when lipogenic agents were used. NHS, normal human keratinocytes (NHK), and explants reacted to presence of bacterial fragments which trigger pre-inflammatory mediator release. CCSV reduced lipid storage and release of pre-inflammatory mediators in NHS, NHK and explants. CCSV also reduced P. acnes growth and triggered beta-defensin-2 and cathelicidin synthesis by NHS, two natural antimicrobial peptides. On volunteers, sebum production, inflamed blemishes, and retentional lesions were significantly reduced after 1 month treatment with CCSV.


Sujet(s)
Dermite séborrhéique/traitement médicamenteux , Kératinocytes/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Peau/cytologie , Peau/effets des médicaments et des substances chimiques , Syringa/composition chimique , Acné juvénile/traitement médicamenteux , Adulte , Cellules cultivées , Humains , Kératinocytes/physiologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Macrophages/effets des médicaments et des substances chimiques , Extraits de plantes/composition chimique , Extraits de plantes/usage thérapeutique , Sébum/effets des médicaments et des substances chimiques , Techniques de culture de tissus , Jeune adulte
10.
Front Immunol ; 8: 1796, 2017.
Article de Anglais | MEDLINE | ID: mdl-29326697

RÉSUMÉ

Phage-display selection of immunoglobulin (IG) or antibody single chain Fragment variable (scFv) from combinatorial libraries is widely used for identifying new antibodies for novel targets. Next-generation sequencing (NGS) has recently emerged as a new method for the high throughput characterization of IG and T cell receptor (TR) immune repertoires both in vivo and in vitro. However, challenges remain for the NGS sequencing of scFv from combinatorial libraries owing to the scFv length (>800 bp) and the presence of two variable domains [variable heavy (VH) and variable light (VL) for IG] associated by a peptide linker in a single chain. Here, we show that single-molecule real-time (SMRT) sequencing with the Pacific Biosciences RS II platform allows for the generation of full-length scFv reads obtained from an in vivo selection of scFv-phages in an animal model of atherosclerosis. We first amplified the DNA of the phagemid inserts from scFv-phages eluted from an aortic section at the third round of the in vivo selection. From this amplified DNA, 450,558 reads were obtained from 15 SMRT cells. Highly accurate circular consensus sequences from these reads were generated, filtered by quality and then analyzed by IMGT/HighV-QUEST with the functionality for scFv. Full-length scFv were identified and characterized in 348,659 reads. Full-length scFv sequencing is an absolute requirement for analyzing the associated VH and VL domains enriched during the in vivo panning rounds. In order to further validate the ability of SMRT sequencing to provide high quality, full-length scFv sequences, we tracked the reads of an scFv-phage clone P3 previously identified by biological assays and Sanger sequencing. Sixty P3 reads showed 100% identity with the full-length scFv of 767 bp, 53 of them covering the whole insert of 977 bp, which encompassed the primer sequences. The remaining seven reads were identical over a shortened length of 939 bp that excludes the vicinity of primers at both ends. Interestingly these reads were obtained from each of the 15 SMRT cells. Thus, the SMRT sequencing method and the IMGT/HighV-QUEST functionality for scFv provides a straightforward protocol for characterization of full-length scFv from combinatorial phage libraries.

11.
Oncotarget ; 7(24): 37013-37029, 2016 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-27203743

RÉSUMÉ

We characterized the mechanism of action of the neuregulin-non-competitive anti-HER3 therapeutic antibody 9F7-F11 that blocks the PI3K/AKT pathway, leading to cell cycle arrest and apoptosis in vitro and regression of pancreatic and breast cancer in vivo. We found that 9F7-F11 induces rapid HER3 down-regulation. Specifically, 9F7-F11-induced HER3 ubiquitination and degradation in pancreatic, breast and prostate cancer cell lines was driven mainly by the itchy E3 ubiquitin ligase (ITCH/AIP4). Overexpression of the ITCH/AIP4 inhibitor N4BP1 or small-interfering RNA-mediated knockdown of ITCH/AIP4 inhibited HER3 ubiquitination/degradation and PI3K/AKT signaling blockade induced by 9F7-F11. Moreover, 9F7-F11-mediated JNK1/2 phosphorylation led to ITCH/AIP4 activation and recruitment to HER3 for receptor ubiquitination and degradation. ITCH/AIP4 activity was activated by the deubiquitinases USP8 and USP9X, as demonstrated by RNA interference. Taken together, our results suggest that 9F7-F11-induced HER3 ubiquitination and degradation in cancer cells mainly occurs through JNK1/2-dependent ITCH/AIP4 activation.


Sujet(s)
Anticorps monoclonaux d'origine murine/pharmacologie , Anticorps monoclonaux/pharmacologie , Antinéoplasiques/pharmacologie , Système de signalisation des MAP kinases/physiologie , Récepteur ErbB-3/antagonistes et inhibiteurs , Protéines de répression/métabolisme , Ubiquitin-protein ligases/métabolisme , Lignée cellulaire tumorale , Régulation négative , Humains , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinase 8/métabolisme , Mitogen-Activated Protein Kinase 9/métabolisme , Protéines de répression/effets des médicaments et des substances chimiques , Ubiquitin-protein ligases/effets des médicaments et des substances chimiques , Ubiquitination
12.
Biochim Biophys Acta ; 1860(6): 1071-8, 2016 Jun.
Article de Anglais | MEDLINE | ID: mdl-26922828

RÉSUMÉ

BACKGROUND: Transforming growth factor beta inducible early gene-1 (TIEG-1), a member of the Krüppel-like factor, was identified as a primary response gene for TGF-ß. The role of TIEG-1 in skin repair has been mainly addressed in vivo on TIEG-1 null mice model and the mechanism remains unexplored. METHODS: We investigated the modulation of TIEG-1 expression in normal human skin fibroblasts by either down-expressing or overexpressing the gene. We evaluated reactive oxygen species production and the cell viability of treated cells. The effect of TIEG-1 overexpression was monitored by wound healing assay and immunofluorescence staining of actin fibers organization and alpha-smooth muscle actin (α-SMA). Western blots were carried out to identify the level of expression or phosphorylation of key proteins such as cofilin, Rho GTPases, and p38 mitogen-activated protein kinase (p38 MAPK). RESULTS: TIEG-1 down-regulation had a deleterious effect on the cell viability. It was significantly reduced (65±5%) and exposure to ultraviolet further increased this effect (47±3%). By contrast, cells overexpressing TIEG-1 had a reduced reactive oxygen species production (75%) compared to control and mock-transfected cells. This overexpression also resulted in formation of actin stress fibers and increased α-SMA expression and an enhanced wound healing feature. RhoB GTPase was upregulated and phosphorylation of cofilin and p38 MAPK was observed. CONCLUSION: TIEG-1 overexpression in normal human skin fibroblasts results in improved resistance to oxidative stress, myofibroblast-like conversion that involved RhoB signaling pathway with cofilin and p38 MAPK proteins activation. GENERAL SIGNIFICANCE: This study enlightens the role of TIEG-1 role in skin biology.


Sujet(s)
Cytosquelette d'actine/composition chimique , Facteurs de transcription EGR/physiologie , Fibroblastes/métabolisme , Facteurs de transcription Krüppel-like/physiologie , Stress oxydatif , Facteurs de dépolymérisation de l'actine/métabolisme , Mouvement cellulaire , Cellules cultivées , Humains , Phosphorylation , Peau/cytologie , Cicatrisation de plaie , p38 Mitogen-Activated Protein Kinases/métabolisme
14.
J Cosmet Dermatol ; 14(2): 152-60, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25817264

RÉSUMÉ

This study was conducted to establish a new methodology for evaluating elements of dermal extracellular matrix (ECM), of epidermal-dermal junction (EDJ), and effects of molecules which can modulate their synthesis. This methodology is based on matrix-assisted laser desorption/ionization mass spectrometric imaging (MALDI-MSI). In vivo reflectance confocal microscopy (in vivo RCM) and echography were also used. Using immunohistochemistry methods on explants, age-related modification data were obtained for selected dermal ECM and EDJ proteins (collagen I, collagen IV, collagen VII, collagen XVII, nidogen I, decorin/decorunt) and used as reference for MALDI-MSI studies. A methodology was developed with MALDI-MSI to map epidermis and dermis proteins. Then MALDI-MSI was used to study age modifications. In vivo RCM and high-frequency ultrasounds were used to evaluate ECM and EDJ undulation modifications caused by aging. Anti-aging molecule evaluations were performed with a blend of palmitoyl oligopeptide and palmitoyl tetrapeptide-7. Immunohistochemistry studies demonstrated that the selected proteins were found to be less abundant in aged group explants vs. young group except for decorin. MALDI-MSI studies correlated the results obtained for decorin. In vivo RCM measurements indicated a decrease of EDJ undulation depth with age and ECM modifications in the upper part of dermis. Echography demonstrated that the peptide blend reduced subepidermal low-echogenic band thickness and improved its density. In vivo RCM studies indicated that the peptides improved the ECM structure vs. placebo. This preliminary MALDI-MSI study raised some technical difficulties that were overcome. Further studies will be conducted to identify more proteins and to demonstrate the interest of this method for cosmetic evaluations.


Sujet(s)
Vieillissement/métabolisme , Derme/métabolisme , Épiderme/métabolisme , Protéines de la matrice extracellulaire/métabolisme , Matrice extracellulaire/métabolisme , Adulte , Sujet âgé , Collagène/métabolisme , Décorine/métabolisme , Test ELISA , Femelle , Humains , Immunohistochimie , Glycoprotéines membranaires/métabolisme , Microscopie confocale , Adulte d'âge moyen , Spectrométrie de masse MALDI , Échographie
15.
Front Immunol ; 6: 39, 2015.
Article de Anglais | MEDLINE | ID: mdl-25699055

RÉSUMÉ

Despite the reasonably long half-life of immunoglogulin G (IgGs), market pressure for higher patient convenience while conserving efficacy continues to drive IgG half-life improvement. IgG half-life is dependent on the neonatal Fc receptor (FcRn), which among other functions, protects IgG from catabolism. FcRn binds the Fc domain of IgG at an acidic pH ensuring that endocytosed IgG will not be degraded in lysosomal compartments and will then be released into the bloodstream. Consistent with this mechanism of action, several Fc-engineered IgG with increased FcRn affinity and conserved pH dependency were designed and resulted in longer half-life in vivo in human FcRn-transgenic mice (hFcRn), cynomolgus monkeys, and recently in healthy humans. These IgG variants were usually obtained by in silico approaches or directed mutagenesis in the FcRn-binding site. Using random mutagenesis, combined with a pH-dependent phage display selection process, we isolated IgG variants with improved FcRn-binding, which exhibited longer in vivo half-life in hFcRn mice. Interestingly, many mutations enhancing Fc/FcRn interaction were located at a distance from the FcRn-binding site validating our random molecular approach. Directed mutagenesis was then applied to generate new variants to further characterize our IgG variants and the effect of the mutations selected. Since these mutations are distributed over the whole Fc sequence, binding to other Fc effectors, such as complement C1q and FcγRs, was dramatically modified, even by mutations distant from these effectors' binding sites. Hence, we obtained numerous IgG variants with increased FcRn-binding and different binding patterns to other Fc effectors, including variants without any effector function, providing distinct "fit-for-purpose" Fc molecules. We therefore provide evidence that half-life and effector functions should be optimized simultaneously as mutations can have unexpected effects on all Fc receptors that are critical for IgG therapeutic efficacy.

16.
J Appl Toxicol ; 35(1): 48-58, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-24496914

RÉSUMÉ

Most animal experiments on cosmetics safety are prohibited and since March 2013, this obligation includes sensitization tests. However, until now there has been no validated alternative in vitro method. In this work, 400 compounds used in the cosmetic industry were selected to cover the greatest diversity of structures, biological activities and sensitizing potential. These molecules were submitted to a series of tests aimed at reproducing essential steps in sensitization and to distinguish between sensitization and irritations, i.e., transcutaneous permeation (factor A), haptenation (factor B), sensitization cytokines production (factor C) and acute toxicity (factor D). The transcutaneous diffusion was measured on human skin explants using Franz cells. Haptenation was tested in solution on human serum albumin. Sensitization cytokine production was investigated by measurement of interleukin-18 release by keratinocytes. Acute toxicity was determined using an 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(75) cell viability test. As only sufficiently stable, soluble and detectable compounds are usable, 33, 72, 68 and 68 molecules were finally tested on factors A, B, C and D, respectively, and 32 were completely screened by the four factors. The individual correlation of the four factors with the reference in vivo tests was limited but the combination of these factors led to a correlation between in vivo and in vitro assays of 81.2% and the safety of the test (risk of false negative) reached 96.8%. The techniques employed are simple and inexpensive and this model of four tests appears as a promising technique to evaluate in vitro the skin sensitization potential of unknown molecules.


Sujet(s)
Allergènes/toxicité , Cosmétiques/toxicité , Eczéma de contact allergique/étiologie , Kératinocytes/effets des médicaments et des substances chimiques , Peau/effets des médicaments et des substances chimiques , Peau/immunologie , Allergènes/composition chimique , Alternatives à l'expérimentation animale , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cosmétiques/composition chimique , Eczéma de contact allergique/immunologie , Chambres de culture à diffusion , Haptènes/métabolisme , Humains , Techniques in vitro , Interleukine-18/biosynthèse , Interleukine-18/immunologie , Irritants/composition chimique , Irritants/toxicité , Kératinocytes/immunologie , Kératinocytes/anatomopathologie , Modèles statistiques , Analyse multifactorielle , Peau/anatomopathologie , Absorption cutanée/effets des médicaments et des substances chimiques , Tests de toxicité aigüe/méthodes
17.
MAbs ; 6(2): 422-36, 2014.
Article de Anglais | MEDLINE | ID: mdl-24492301

RÉSUMÉ

While glyco-engineered monoclonal antibodies (mAbs) with improved antibody-dependent cell-mediated cytotoxicity (ADCC) are reaching the market, extensive efforts have also been made to improve their pharmacokinetic properties to generate biologically superior molecules. Most therapeutic mAbs are human or humanized IgG molecules whose half-life is dependent on the neonatal Fc receptor FcRn. FcRn reduces IgG catabolism by binding to the Fc domain of endocytosed IgG in acidic lysosomal compartments, allowing them to be recycled into the blood. Fc-engineered mAbs with increased FcRn affinity resulted in longer in vivo half-life in animal models, but also in healthy humans. These Fc-engineered mAbs were obtained by alanine scanning, directed mutagenesis or in silico approach of the FcRn binding site. In our approach, we applied a random mutagenesis technology (MutaGen™) to generate mutations evenly distributed over the whole Fc sequence of human IgG1. IgG variants with improved FcRn-binding were then isolated from these Fc-libraries using a pH-dependent phage display selection process. Two successive rounds of mutagenesis and selection were performed to identify several mutations that dramatically improve FcRn binding. Notably, many of these mutations were unpredictable by rational design as they were located distantly from the FcRn binding site, validating our random molecular approach. When produced on the EMABling(®) platform allowing effector function increase, our IgG variants retained both higher ADCC and higher FcRn binding. Moreover, these IgG variants exhibited longer half-life in human FcRn transgenic mice. These results clearly demonstrate that glyco-engineering to improve cytotoxicity and protein-engineering to increase half-life can be combined to further optimize therapeutic mAbs.


Sujet(s)
Anticorps monoclonaux/pharmacocinétique , Antigènes d'histocompatibilité de classe I/métabolisme , Immunoglobuline G/métabolisme , Immunothérapie/méthodes , Ingénierie des protéines/méthodes , Récepteur Fc/métabolisme , Animaux , Anticorps monoclonaux/génétique , Cytotoxicité à médiation cellulaire dépendante des anticorps/génétique , Techniques d'exposition à la surface cellulaire , Cytotoxicité immunologique/génétique , Glycosylation , Période , Antigènes d'histocompatibilité de classe I/génétique , Humains , Immunoglobuline G/génétique , Immunothérapie/tendances , Souris , Souris transgéniques , Mutagenèse dirigée , Mutation/génétique , Récepteur Fc/génétique , Récepteurs du fragment Fc des IgG/antagonistes et inhibiteurs , Récepteurs du fragment Fc des IgG/immunologie , Récepteurs du fragment Fc des IgG/métabolisme
18.
Neoplasia ; 15(3): 335-47, 2013 Mar.
Article de Anglais | MEDLINE | ID: mdl-23479511

RÉSUMÉ

Blockade of the human epidermal growth factor receptor 3 (HER3) and of the downstream phosphatidylinositide 3-kinase (PI3K)/AKT pathway is a prerequisite for overcoming drug resistance and to develop novel treatments for cancers that are not eligible for the currently approved targeted therapies. To this end, we generated specific antibodies (Abs) against domain 1 (D1) and domain 3 (D3) of HER3 that recognize epitopes that do not overlap with the neuregulin-binding site. The fully human H4B-121 Ab and the mouse monoclonal Abs 16D3-C1 and 9F7-F11 inhibited tumor growth in nude mice xenografted with epidermoid, pancreatic, or triple-negative breast cancer cells. The combination of one anti-HER3 Ab and trastuzumab improved tumor growth inhibition in mice xenografted with HER2(low) cancer cell lines, for which trastuzumab alone shows no or moderate efficiency. Ab-induced disruption of tumor growth was associated with G1 cell cycle arrest, proliferation inhibition, and apoptosis of cancer cells. Anti-HER3 Abs blocked HER2/HER3 heterodimerization and HER3 phosphorylation at the cell membrane, leading to inhibition of phosphorylation of the downstream AKT targets murine double minute 2, X-linked inhibitor of apoptosis, and forkhead box O1. This study demonstrates that anti-HER3 D1 and D3 Abs could represent a new option for immunotherapy of pancreatic and triple-negative breast cancers.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Facteurs de transcription Forkhead/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-mdm2/métabolisme , Récepteur ErbB-2/métabolisme , Récepteur ErbB-3/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Séquence d'acides aminés , Animaux , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/métabolisme , Anticorps monoclonaux humanisés/pharmacologie , Spécificité des anticorps , Apoptose/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Dimérisation , Épitopes/composition chimique , Épitopes/immunologie , Femelle , Protéine O1 à motif en tête de fourche , Humains , Souris , Données de séquences moléculaires , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Phosphorylation/effets des médicaments et des substances chimiques , Liaison aux protéines , Récepteur ErbB-2/composition chimique , Récepteur ErbB-3/composition chimique , Récepteur ErbB-3/immunologie , Trastuzumab , Charge tumorale/effets des médicaments et des substances chimiques
19.
Radiat Res ; 179(2): 232-42, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23289389

RÉSUMÉ

Skin exposure to ionizing radiation affects the normal wound healing process. We investigated the beneficial effects of a pharmacological treatment with geranylgeranylacetone (GGA) on keratinocytes using in vitro scratch wound injury assay in nonirradiated and irradiated conditions. Irradiation affected the wound closure of keratinocytes 24 h after scratch injury, whereas re-epithelialization was markedly accelerated after GGA treatment when compared to nontreated keratinocytes. We demonstrated that GGA treatment increased migration of human epidermal keratinocytes and this migratory property was not related to RhoA signaling. Interestingly, Western blot analysis revealed that GGA treatment down-regulated caspase 3 active form expression and up-regulated the activated phenotype by inducing both keratin 6 (K6) expression and interleukin-1ß (IL-1ß) release without modification of the differentiate phenotype. Finally, the proteomic profiling was performed on keratinocytes, showing that global protein changes occurred after irradiation of keratinocytes treated or untreated with GGA.


Sujet(s)
Diterpènes/pharmacologie , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/effets des radiations , Radioprotecteurs/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/effets des radiations , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des radiations , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des radiations , Cellules épithéliales/cytologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/effets des radiations , Humains , Kératinocytes/cytologie , Kératinocytes/métabolisme , Protéomique , Facteurs temps , Protéine G RhoA/métabolisme
20.
Methods Mol Biol ; 907: 451-61, 2012.
Article de Anglais | MEDLINE | ID: mdl-22907368

RÉSUMÉ

As a growing number of therapeutic antibodies are developed, robust methods to efficiently improve the affinity and/or specificity of antibody candidates are needed. Here we describe our powerful platform that combines scFv affinity maturation and IgG high-throughput screening. After creating diversity with our random mutagenesis technology (MutaGen™), the scFv libraries are fully cleaned using a fusion system introducing the beta-lactamase gene to select in-frame and stop codon free variants on the basis of ampicillin resistance. The high-quality scFv libraries thereby constructed are then selected on the target in vitro using phage display technology. Contrary to standard procedures, instead of producing a limited number of affinity matured scFv as IgG molecules, we developed a cloning system to directly transfer the entire pool of selected scFv into an IgG expression vector permitting rapid IgG small-scale production (96 wells) in mammalian cells. Our integrated process allows us to generate high-quality scFv libraries and test numerous IgG variants, increasing the chances to select the best therapeutic antibody candidate.


Sujet(s)
Affinité des anticorps/immunologie , Tests de criblage à haut débit/méthodes , Immunoglobuline G/isolement et purification , Banque de peptides , Anticorps à chaîne unique/biosynthèse , Ampicilline/pharmacologie , Affinité des anticorps/effets des médicaments et des substances chimiques , Lignée cellulaire , Clonage moléculaire , Vecteurs génétiques/génétique , Humains , Immunoglobuline G/biosynthèse , Immunoglobuline G/génétique , Cadres ouverts de lecture/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...