24 results on '"Julie Prigent"'
Search Results
2. Conformational Plasticity in Broadly Neutralizing HIV-1 Antibodies Triggers Polyreactivity
- Author
-
Julie Prigent, Annaëlle Jarossay, Cyril Planchais, Caroline Eden, Jérémy Dufloo, Ayrin Kök, Valérie Lorin, Oxana Vratskikh, Thérèse Couderc, Timothée Bruel, Olivier Schwartz, Michael S. Seaman, Oliver Ohlenschläger, Jordan D. Dimitrov, and Hugo Mouquet
- Subjects
antibody ,B cells ,HIV-1 ,polyreactivity ,autoreactivity ,Biology (General) ,QH301-705.5 - Abstract
Human high-affinity antibodies to pathogens often recognize unrelated ligands. The molecular origin and the role of this polyreactivity are largely unknown. Here, we report that HIV-1 broadly neutralizing antibodies (bNAbs) are frequently polyreactive, cross-reacting with non-HIV-1 molecules, including self-antigens. Mutating bNAb genes to increase HIV-1 binding and neutralization also results in de novo polyreactivity. Unliganded paratopes of polyreactive bNAbs with improved HIV-1 neutralization exhibit a conformational flexibility, which contributes to enhanced affinity of bNAbs to various HIV-1 envelope glycoproteins and non-HIV antigens. Binding adaptation of polyreactive bNAbs to the divergent ligands mainly involves hydrophophic interactions. Plasticity of bNAbs’ paratopes may, therefore, facilitate accommodating divergent viral variants, but it simultaneously triggers promiscuous binding to non-HIV-1 antigens. Thus, a certain level of polyreactivity can be a mark of adaptable antibodies displaying optimal pathogens’ recognition.
- Published
- 2018
- Full Text
- View/download PDF
3. HIV-1 Envelope Recognition by Polyreactive and Cross-Reactive Intestinal B Cells
- Author
-
Cyril Planchais, Ayrin Kök, Alexia Kanyavuz, Valérie Lorin, Timothée Bruel, Florence Guivel-Benhassine, Tim Rollenske, Julie Prigent, Thierry Hieu, Thierry Prazuck, Laurent Lefrou, Hedda Wardemann, Olivier Schwartz, Jordan D. Dimitrov, Laurent Hocqueloux, and Hugo Mouquet
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Mucosal immune responses to HIV-1 involve the recognition of the viral envelope glycoprotein (gp)160 by tissue-resident B cells and subsequent secretion of antibodies. To characterize the B cells “sensing” HIV-1 in the gut of infected individuals, we probed monoclonal antibodies produced from single intestinal B cells binding to recombinant gp140 trimers. A large fraction of mucosal B cell antibodies were polyreactive and showed only low affinity to HIV-1 envelope glycoproteins, particularly the gp41 moiety. A few high-affinity gp140 antibodies were isolated but lacked neutralizing, potent ADCC, and transcytosis-blocking capacities. Instead, they displayed cross-reactivity with defined self-antigens. Specifically, intestinal HIV-1 gp41 antibodies targeting the heptad repeat 2 region (HR2) cluster II cross-reacted with the p38α mitogen-activated protein kinase 14 (MAPK14). Hence, physiologic polyreactivity of intestinal B cells and molecular mimicry-based self-reactivity of HIV-1 antibodies are two independent phenomena, possibly diverting and/or impairing mucosal humoral immunity to HIV-1. : Antibodies produced in mucosa after sexual transmission of HIV-1 could affect viral propagation. Planchais et al. show that intestinal B cells from HIV-1-infected individuals that recognize the HIV-1 envelope (Env) proteins are mainly low affinity and polyreactive and that rare, high-affinity antibodies to HIV-1 Env lack potent antiviral capacities and cross-react with self-antigens. Keywords: HIV-1, antibodies, B cells, mucosa, polyreactivity, cross-reactivity, MAPK14, intestine
- Published
- 2019
- Full Text
- View/download PDF
4. Ricin Antibodies’ Neutralizing Capacity against Different Ricin Isoforms and Cultivars
- Author
-
Maria Lucia Orsini Delgado, Arnaud Avril, Julie Prigent, Julie Dano, Audrey Rouaix, Sylvia Worbs, Brigitte G. Dorner, Clémence Rougeaux, François Becher, François Fenaille, Sandrine Livet, Hervé Volland, Jean-Nicolas Tournier, and Stéphanie Simon
- Subjects
ricin ,neutralizing antibodies ,recombinant antibodies ,monoclonal antibodies ,ricin isoforms ,Ricinus communis cultivars ,Medicine - Abstract
Ricin, a highly toxic protein from Ricinus communis, is considered a potential biowarfare agent. Despite the many data available, no specific treatment has yet been approved. Due to their ability to provide immediate protection, antibodies (Abs) are an approach of choice. However, their high specificity might compromise their capacity to protect against the different ricin isoforms (D and E) found in the different cultivars. In previous work, we have shown the neutralizing potential of different Abs (43RCA-G1 (anti ricin A-chain) and RB34 and RB37 (anti ricin B-chain)) against ricin D. In this study, we evaluated their protective capacity against both ricin isoforms. We show that: (i) RB34 and RB37 recognize exclusively ricin D, whereas 43RCA-G1 recognizes both isoforms, (ii) their neutralizing capacity in vitro varies depending on the cultivar, and (iii) there is a synergistic effect when combining RB34 and 43RCA-G1. This effect is also demonstrated in vivo in a mouse model of intranasal intoxication with ricin D/E (1:1), where approximately 60% and 40% of mice treated 0 and 6 h after intoxication, respectively, are protected. Our results highlight the importance of evaluating the effectiveness of the Abs against different ricin isoforms to identify the treatment with the broadest spectrum neutralizing effect.
- Published
- 2021
- Full Text
- View/download PDF
5. Adult-Derived Human Liver Stem/Progenitor Cells Infused 3 Days Postsurgery Improve Liver Regeneration in a Mouse Model of Extended Hepatectomy
- Author
-
Astrid Herrero, Julie Prigent, Catherine Lombard, Valérie Rosseels, Martine Daujat-Chavanieu, Karine Breckpot, Mustapha Najimi, Gisèle Deblandre, and Etienne M. Sokal
- Subjects
Medicine - Abstract
There is growing evidence that cell therapy constitutes a promising strategy for liver regenerative medicine. In the setting of hepatic cancer treatments, cell therapy could prove a useful therapeutic approach for managing the acute liver failure that occurs following extended hepatectomy. In this study, we examined the influence of delivering adult-derived human liver stem/progenitor cells (ADHLSCs) at two different early time points in an immunodeficient mouse model ( Rag2 −/- IL2R g -/- ) that had undergone a 70% hepatectomy procedure. The hepatic mesenchymal cells were intrasplenically infused either immediately after surgery ( n = 26) or following a critical 3-day period ( n = 26). We evaluated the cells' capacity to engraft at day 1 and day 7 following transplantation by means of human Alu qPCR quantification, along with histological assessment of human albumin and α-smooth muscle actin. In addition, cell proliferation (anti-mouse and human Ki-67 staining) and murine liver weight were measured in order to evaluate liver regeneration. At day 1 posttransplantation, the ratio of human to mouse cells was similar in both groups, whereas 1 week posttransplantation this ratio was significantly improved ( p < 0.016) in mice receiving ADHLSC injection at day 3 posthepatectomy (1.7%), compared to those injected at the time of surgery (1%). On the basis of liver weight, mouse liver regeneration was more extensive 1 week posttransplantation in mice transplanted with ADHLSCs (+65.3%) compared to that of mice from the sham vehicle group (+42.7%). In conclusion, infusing ADHLSCs 3 days after extensive hepatectomy improves the cell engraftment and murine hepatic tissue regeneration, thereby confirming that ADHLSCs could be a promising cell source for liver cell therapy and hepatic tissue repair.
- Published
- 2017
- Full Text
- View/download PDF
6. Human Progenitor Cell Quantification after Xenotransplantation in Rat and Mouse Models by a Sensitive qPCR Assay
- Author
-
Julie Prigent, Astrid Herrero, Jérôme Ambroise, Françoise Smets, Gisèle A. Deblandre, and Etienne M. Sokal
- Subjects
Medicine - Abstract
Xenotransplantation of human cells in animal models is an essential tool for evaluation of safety and efficacy of cell-based products for therapeutic use. Sensitive and reproducible methods are needed to detect and quantify human cells engrafted into the host tissue either in the targeted organ or in undesired locations. We developed a robust quantitative polymerase chain reaction (qPCR) assay based on amplification of human AluYb8 repeats, to assess the number of human cells present in rat or mouse tissues after transplantation. Standard curves of mixed human/rodent DNA and mixed human/rodent cells have been performed to determine the limit of detection and linear range of the assay. Standard curves from DNA mixing differed significantly from standard curves from cell mixing. We show here that the AluYb8 qPCR assay is highly reproducible and is able to quantify human cells in a rodent cell matrix over a large linear range that extends from 50% to 0.01% human cells. Short-term in vivo studies showed that human cells could be quantified in mouse liver up to 7 days after intrasplenic transplantation and in rat liver 4 h after intrahepatic transplantation.
- Published
- 2015
- Full Text
- View/download PDF
7. Neutralising antibodies against ricin toxin.
- Author
-
Julie Prigent, Laetitia Panigai, Patricia Lamourette, Didier Sauvaire, Karine Devilliers, Marc Plaisance, Hervé Volland, Christophe Créminon, and Stéphanie Simon
- Subjects
Medicine ,Science - Abstract
The Centers for Disease Control and Prevention have listed the potential bioweapon ricin as a Category B Agent. Ricin is a so-called A/B toxin produced by plants and is one of the deadliest molecules known. It is easy to prepare and no curative treatment is available. An immunotherapeutic approach could be of interest to attenuate or neutralise the effects of the toxin. We sought to characterise neutralising monoclonal antibodies against ricin and to develop an effective therapy. For this purpose, mouse monoclonal antibodies (mAbs) were produced against the two chains of ricin toxin (RTA and RTB). Seven mAbs were selected for their capacity to neutralise the cytotoxic effects of ricin in vitro. Three of these, two anti-RTB (RB34 and RB37) and one anti-RTA (RA36), when used in combination improved neutralising capacity in vitro with an IC(50) of 31 ng/ml. Passive administration of association of these three mixed mAbs (4.7 µg) protected mice from intranasal challenges with ricin (5 LD(50)). Among those three antibodies, anti-RTB antibodies protected mice more efficiently than the anti-RTA antibody. The combination of the three antibodies protected mice up to 7.5 hours after ricin challenge. The strong in vivo neutralising capacity of this three mAbs combination makes it potentially useful for immunotherapeutic purposes in the case of ricin poisoning or possibly for prevention.
- Published
- 2011
- Full Text
- View/download PDF
8. Production and characterisation of a neutralising chimeric antibody against botulinum neurotoxin A.
- Author
-
Julie Prigent, Christelle Mazuet, Didier Boquet, Patricia Lamourette, Hervé Volland, Michel R Popoff, Christophe Créminon, and Stéphanie Simon
- Subjects
Medicine ,Science - Abstract
Botulinum neurotoxins, produced by Clostridium botulinum bacteria, are the causative agent of botulism. This disease only affects a few hundred people each year, thus ranking it among the orphan diseases. However, botulinum toxin type A (BoNT/A) is the most potent toxin known to man. Due to their potency and ease of production, these toxins were classified by the Centers for Disease Control and Prevention (CDC) as Category A biothreat agents. For several biothreat agents, like BoNT/A, passive immunotherapy remains the only possible effective treatment allowing in vivo neutralization, despite possible major side effects. Recently, several mouse monoclonal antibodies directed against a recombinant fragment of BoNT/A were produced in our laboratory and most efficiently neutralised the neurotoxin. In the present work, the most powerful one, TA12, was selected for chimerisation. The variable regions of this antibody were thus cloned and fused with the constant counterparts of human IgG1 (kappa light and gamma 1 heavy chains). Chimeric antibody production was evaluated in mammalian myeloma cells (SP2/0-Ag14) and insect cells (Sf9). After purifying the recombinant antibody by affinity chromatography, the biochemical properties of chimeric and mouse antibody were compared. Both have the same very low affinity constant (close to 10 pM) and the chimeric antibody exhibited a similar capacity to its parent counterpart in neutralising the toxin in vivo. Its strong affinity and high neutralising potency make this chimeric antibody interesting for immunotherapy treatment in humans in cases of poisoning, particularly as there is a probable limitation of the immunological side effects observed with classical polyclonal antisera from heterologous species.
- Published
- 2010
- Full Text
- View/download PDF
9. Abstract 2954: MAIT engagers: An efficacious novel modality in the field of T-cell engagers for the treatment of solid tumors
- Author
-
Simon Edward Plyte, Marie Fraudeau, Jonathan Grivel, Paloma Hougron, Katja Klausz, Dorothee Winterberg, Britta von Below, Alexandre Ivagnes, Claire Germain, Sebastian Amigorena, Eugene Zhukovsky, Matthias Peipp, Pierre-Emmanuel Gerard, Olivier Lantz, and Julie Prigent
- Subjects
Cancer Research ,Oncology - Abstract
Background: Mucosal-Associated Invariant T cells (MAITs) are an abundant subset of non-conventional T-cells with potent cytotoxic capacity (up to 20% of circulating T-cells) that are naturally resident in many tissues and solid tumors. T-cell redirection is a clinically validated approach to treating haematological cancers but has limited success in solid tumors. Classical T-cell engagers (TCE) bind the epsilon chain of the TCR leading to activation of all T-cells. MAIT cells utilize a semi-invariant TCR and recognize bacterial metabolites presented in the context of the MR1 protein. Biomunex has generated bispecific antibodies that bind the MAIT semi-invariant TCR (iTCR) and the HER2 receptor tyrosine kinase expressed on tumor cells. This enables the formation of an efficient immunological synapse and exclusive redirection of MAIT cells to directly kill cancer cells. Methods: Using the Biomunex proprietary BiXAb® platform, bispecific, tetravalent antibodies were generated that target the MAIT iTCR and HER2. Specific binding to the two targets was demonstrated by ELISA, DUAL ELISA and in FACS. In general, BiXAb®-mediated MAIT-cell activation, proliferation and degranulation were followed by gating on MAIT cells within a purified CD8 cell population. Tumor cell lines expressing varying amounts of HER2 were co-cultured with MAIT cells (peripheral and tumor-resident) and the BiXAb®s in several cytotoxic assays which were evaluated by measuring Chromium release, LDH release and FACS. Results: The iTCR x HER2 BiXAb® efficiently binds both target proteins with similar affinities to the parental Mabs and can bind both simultaneously, as judged by Dual ELISA. The iTCR x HER2 BiXAb® binds the MAIT-cell TCR and can bind cancer cells over a wide range of HER2 expression. BiXAb® engagement of MAIT cells and cancer cells leads to rapid activation, proliferation and degranulation of MAIT cells. In a population of PBMCs, only MAIT cells are activated by the BiXAb®. Even at low effector to target ratios (E:T = 2:1), MAIT cells efficiently kill engaged cancer cells in a HER2-dependent manner (over 50% cytotoxicity in 18 hrs). The MAIT-directed BiXAb® does not activate other T-cell subsets and hence has significantly reduced cytokine release when compared to a classical T-cell engager. Conclusions: BiXAb® mediated MAIT cell redirection leads to efficient killing of cancer cells and is a promising new approach for the treatment of solid tumors. The iTCR x HER2 BiXAb® has no impact on the general CD8 or CD4 T-cell population which may address some of the clinical limitations of classical TCEs. Citation Format: Simon Edward Plyte, Marie Fraudeau, Jonathan Grivel, Paloma Hougron, Katja Klausz, Dorothee Winterberg, Britta von Below, Alexandre Ivagnes, Claire Germain, Sebastian Amigorena, Eugene Zhukovsky, Matthias Peipp, Pierre-Emmanuel Gerard, Olivier Lantz, Julie Prigent. MAIT engagers: An efficacious novel modality in the field of T-cell engagers for the treatment of solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2954.
- Published
- 2023
10. Abstract 2899: Targeting regulatory T cells with HFB101110, a novel anti-human CCR8 antibody for the treatment of solid tumors
- Author
-
Roshan M. Kumar, Julie Prigent, Hombline Poullain, Ayrin Kok, Carine George, Sami Ellouze, Yun-Yueh Lu, Rebecca Silver, Clarisse Monchecourt, Ross B. Fulton, Qian Zhang, Nicola Beltraminelli, Bernhard Moser, Francisco Adrian, and Liang Schweizer
- Subjects
Cancer Research ,Oncology - Abstract
Regulatory T cells (Tregs) contribute to immunosuppression within the tumor microenvironment and have been associated with poor outcomes in a range of cancers. Targeted depletion of tumor-infiltrating Tregs (TITRs) is an attractive therapeutic strategy with the potential to enhance antitumor immunity in patients who do not respond to current treatments. However, such approaches have been limited to date by a lack of markers that are highly specific for TITRs. CCR8 is a chemokine G protein-coupled receptor (GPCR) that has recently been shown to be specifically expressed on TITRs as compared to Tregs in the periphery or other T cells within tumors. Here, we report the development and characterization of HFB101110, a humanized monoclonal antibody against CCR8 with potent and specific antibody-dependent cellular cytotoxicity (ADCC) activity. HFB101110 specifically recognizes an epitope on the N-terminus extracellular domain of CCR8 and does not recognize the closely related chemokine receptor CCR4. HFB101110 acts through a dual mechanism of action, by both depleting CCR8+ cells via ADCC and blocking binding of the CCL1 chemokine to its receptor CCR8. Blockade by HFB101110 inhibited calcium flux and chemotaxis induced by the interaction between CCL1 and CCR8. Furthermore, HFB101110 showed potent single agent anti-tumor activity associated with depletion of intratumoral Tregs in a human CCR8 knock-in mouse model. HFB101110 mediated specific ex vivo killing of Tregs from primary patient tumor samples both in the presence, and in some cases the absence, of allogeneic NK cells. HFB101110 showed favorable pharmacokinetic properties and a favorable developability profile, and was well-tolerated in both wild-type mice and cynomolgus monkeys. HFB101110 is currently being developed as a novel immunotherapy for the treatment of solid tumors coupled with a patient biomarker strategy derived from HiFiBiO’s Drug Intelligent Science (DIS™) single-cell immune profiling platform. Citation Format: Roshan M. Kumar, Julie Prigent, Hombline Poullain, Ayrin Kok, Carine George, Sami Ellouze, Yun-Yueh Lu, Rebecca Silver, Clarisse Monchecourt, Ross B. Fulton, Qian Zhang, Nicola Beltraminelli, Bernhard Moser, Francisco Adrian, Liang Schweizer. Targeting regulatory T cells with HFB101110, a novel anti-human CCR8 antibody for the treatment of solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2899.
- Published
- 2022
11. Ricin Antibodies' Neutralizing Capacity against Different Ricin Isoforms and Cultivars
- Author
-
Stéphanie Simon, Hervé Volland, Arnaud Avril, François Becher, Clémence Rougeaux, Sandrine Livet, François Fenaille, Jean-Nicolas Tournier, Julie Dano, Sylvia Worbs, Julie Prigent, Brigitte G. Dorner, Maria Lucia Orsini Delgado, Audrey Rouaix, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), and Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)
- Subjects
Health, Toxicology and Mutagenesis ,Antidotes ,lcsh:Medicine ,Pharmacology ,Toxicology ,chemistry.chemical_compound ,Jurkat Cells ,ricin isoforms ,Antibody Specificity ,[SDV.IDA]Life Sciences [q-bio]/Food engineering ,Protein Isoforms ,Ricinus communis cultivars ,ComputingMilieux_MISCELLANEOUS ,0303 health sciences ,Mice, Inbred BALB C ,biology ,treatment ,Poisoning ,030302 biochemistry & molecular biology ,Ricinus ,3. Good health ,Ricin ,[SDV.TOX]Life Sciences [q-bio]/Toxicology ,Drug Therapy, Combination ,Female ,monoclonal antibodies ,Antibody ,Gene isoform ,endocrine system ,medicine.drug_class ,Cell Survival ,mouse model ,recombinant antibodies ,Monoclonal antibody ,Article ,Lethal Dose 50 ,03 medical and health sciences ,In vivo ,medicine ,Animals ,Humans ,neutralizing antibodies ,030304 developmental biology ,pulmonary intoxication ,lcsh:R ,biology.organism_classification ,Antibodies, Neutralizing ,In vitro ,carbohydrates (lipids) ,enzymes and coenzymes (carbohydrates) ,chemistry ,biology.protein ,Nasal administration - Abstract
Ricin, a highly toxic protein from Ricinus communis, is considered a potential biowarfare agent. Despite the many data available, no specific treatment has yet been approved. Due to their ability to provide immediate protection, antibodies (Abs) are an approach of choice. However, their high specificity might compromise their capacity to protect against the different ricin isoforms (D and E) found in the different cultivars. In previous work, we have shown the neutralizing potential of different Abs (43RCA-G1 (anti ricin A-chain) and RB34 and RB37 (anti ricin B-chain)) against ricin D. In this study, we evaluated their protective capacity against both ricin isoforms. We show that: (i) RB34 and RB37 recognize exclusively ricin D, whereas 43RCA-G1 recognizes both isoforms, (ii) their neutralizing capacity in vitro varies depending on the cultivar, and (iii) there is a synergistic effect when combining RB34 and 43RCA-G1. This effect is also demonstrated in vivo in a mouse model of intranasal intoxication with ricin D/E (1:1), where approximately 60% and 40% of mice treated 0 and 6 h after intoxication, respectively, are protected. Our results highlight the importance of evaluating the effectiveness of the Abs against different ricin isoforms to identify the treatment with the broadest spectrum neutralizing effect.
- Published
- 2020
12. Abstract 1884: HFB9-2, a novel Galectin 9 neutralizing antibody for the treatment of AML and other cancers
- Author
-
Xing Cai, Liang Schweizer, Philippe Busson, Wenhua Xu, Mingfang Feng, Francisco Adrian, Mingjie Chen, Dean Lee, He Zhou, Stephanie Beq, Véronique Saada, Julia Delahousse, Nicola Beltraminelli, Jia Wu, Angelo Paci, Stéphane de Botton, Julie Prigent, Andreas Raue, Joyce Pi, Germain Margall, Muriel D. David, and Yun-Yueh Lu
- Subjects
Cancer Research ,Tumor microenvironment ,biology ,medicine.drug_class ,Regulatory T cell ,business.industry ,medicine.medical_treatment ,CD44 ,Cancer ,Immunosuppression ,Monoclonal antibody ,medicine.disease ,medicine.anatomical_structure ,Immune system ,Oncology ,biology.protein ,medicine ,Cancer research ,Antibody ,business - Abstract
Monoclonal antibodies targeting immune checkpoints have demonstrated clinical success in a range of tumor types; however sustained responses are achieved in a fraction of patients due to primary or secondary resistance to treatment. Recent evidence has implicated the pleiotropic immunosuppressive modulator Galactoside-binding lectin Galectin 9 (Gal-9) as a key factor in the tumor microenvironment that renders tumors resistant to current chemo and immunotherapies. High Gal-9 expression has been reported in different types of cancer, including hematological malignancies such as AML and ALL, and multiple solid tumors. Previously, we have described the characterization of an anti-human Gal-9 antibody, HFB9-2, with sub-nanomolar affinity for human, murine and cynomolgous Gal-9. HFB9-2 blocks the interaction of Gal-9 with its receptors TIM3 and CD44, leading to inhibition of Gal-9 mediated Th1 cell apoptosis and regulatory T cell expansion. HFB9-2 demonstrated significant anti-tumor efficacy and increased survival in a WEHI-164 syngeneic model as a single agent and in combination with anti-PD1 antibody. HFB9-2 is well tolerated in cynomolgous monkeys after administered by intravenous infusion as a single dose of 10, 50, 200 mg/kg, with no adverse effects and a NOAEL of 200 mg/kg. HFB9-2 developability profile assessment does not anticipate any stability and manufacturing issues. We hypothesize that targeting Gal-9 represents a valuable strategy to reduce immunosuppression and improve clinical response in selected cancer patients, in particular AML. Gal-9 has been reported to play a dual role in AML as both a self-renewal factor for leukemic stem cells and a suppressor of anti-cancer immunity. Analysis of AML patient serum samples demonstrated that Gal-9 expression was significantly higher than in healthy controls and that dropped at complete remission. Higher levels of Gal-9 were found in French-America-British (FAB) type M4 and M5 AML samples, and the lowest levels were observed in M3. To better understand the role for Gal-9 in the development of AML, we implanted primary human AML cells from different donors in M-NSG mice (NOD-Prkdcscid Il2rgem1). We demonstrate that the engraftment of primary human AML cells in these mice resulted in high levels of human Gal-9 in their serum that were detectable 2 weeks after intravenous implantation. The serum levels of Gal-9 increased over time following the increase of hCD45+ AML cells. Further analysis to evaluate the effect of Gal-9 neutralization in AML progression after primary and secondary engraftment with HFB9-2 are currently ongoing. The data presented here provide evidence that neutralization of Gal-9 with HFB9-2 blocks key immunosuppressive mechanisms known to favor cancer progression and to limit the efficacy of current immunotherapies, and together with the data obtained in AML patient samples position Gal-9 neutralization as a promising anticancer approach. Citation Format: Germain Margall, Muriel David, Julie Prigent, Dean Lee, Wenhua Xu, Joyce Pi, Xing Cai, He Zhou, Andreas Raue, Nicola Beltraminelli, Mingjie Chen, Jia Wu, Mingfang Feng, Angelo Paci, Julia Delahousse, Véronique Saada, Stéphane de Botton, Pierre Busson, Stephanie Beq, Francisco Adrian, Liang Schweizer, Yun-Yueh Lu. HFB9-2, a novel Galectin 9 neutralizing antibody for the treatment of AML and other cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1884.
- Published
- 2021
13. Scarcity of autoreactive human blood IgA+ memory B cells
- Author
-
Hugo Mouquet, Ayrin Kök, Salomé Bourgeau, Valérie Lorin, Julie Prigent, and Thierry Hieu
- Subjects
0301 basic medicine ,Immunoglobulin A ,Immunoglobulin gene ,Immunology ,Adaptive immunity ,Antibody Affinity ,Autoimmunity ,Biology ,IgA Antibodies ,Polyreactivity ,Autoantigens ,Immunoglobulin G ,Affinity maturation ,03 medical and health sciences ,Immune system ,Antigen ,Immunology and Allergy ,Humans ,Basic ,Clonal Selection, Antigen-Mediated ,Research Articles ,Autoantibodies ,B-Lymphocytes ,Immunoglobulin genes ,Memory B cells ,Immunoglobulin Class Switching ,3. Good health ,Clone Cells ,030104 developmental biology ,Immunoglobulin class switching ,Autoreactivity ,Antibody Formation ,biology.protein ,Research Article|Basic ,Antibody ,Single-Cell Analysis ,Immunologic Memory ,Antibody Diversity - Abstract
Class-switched memory B cells are key components of the "reactive" humoral immunity, which ensures a fast and massive secretion of high-affinity antigen-specific antibodies upon antigenic challenge. In humans, IgA class-switched (IgA+ ) memory B cells and IgA antibodies are abundant in the blood. Although circulating IgA+ memory B cells and their corresponding secreted immunoglobulins likely possess major protective and/or regulatory immune roles, little is known about their specificity and function. Here, we show that IgA+ and IgG+ memory B-cell antibodies cloned from the same healthy humans share common immunoglobulin gene features. IgA and IgG memory antibodies have comparable lack of reactivity to vaccines, common mucosa-tropic viruses and commensal bacteria. However, the IgA+ memory B-cell compartment contains fewer polyreactive clones and importantly, only rare self-reactive clones compared to IgG+ memory B cells. Self-reactivity of IgAs is acquired following B-cell affinity maturation but not antibody class switching. Together, our data suggest the existence of different regulatory mechanisms for removing autoreactive clones from the IgG+ and IgA+ memory B-cell repertoires, and/or different maturation pathways potentially reflecting the distinct nature and localization of the cognate antigens recognized by individual B-cell populations.
- Published
- 2016
14. Abstract 6532: HFB9-2: A novel Gal-9 neutralizing antibody to reverse immune suppression in the tumor microenvironment
- Author
-
He Zhou, Francisco Adrian, Stephanie Beq, Nicola Beltraminelli, Roshan Kumar, Rachel Pacherie, Julie Prigent, Andreas Raue, Germain Margall, and Liang Schweizer
- Subjects
Cancer Research ,Tumor microenvironment ,biology ,business.industry ,medicine.drug_class ,CD44 ,Cancer ,Monoclonal antibody ,medicine.disease ,Immune system ,Oncology ,biology.protein ,Cancer research ,Medicine ,Stem cell ,business ,Neutralizing antibody ,Galectin - Abstract
Although monoclonal antibodies targeting immune checkpoints have demonstrated clinical success in a range of tumor types, sustained responses are only observed in a fraction of patients due to primary or secondary resistance to treatment. Recent evidence has implicated the pleiotropic immunosuppressive modulator Galactoside-binding lectin Galectin 9 (Gal-9) as a key factor present in the tumor microenvironment that renders tumors resistant to current immunotherapies. High Gal-9 expression has been reported in different types of cancer including hematological malignancies such as AML and ALL, and multiple solid tumors. We hypothesize that targeting Gal-9 may represent a valuable strategy to overcome resistance and improve clinical response in selected cancer patients. We present a monoclonal antibody, HFB9-2, that specifically binds to human Gal-9 with sub-nanomolar affinity, recognizes recombinant Gal-9 and Gal-9 produced by human tumor cells, and is cross-reactive with mouse and monkey Gal-9 orthologs. HFB9-2 blocks the interaction of Gal-9 with its receptors TIM3 and CD44 in a dose dependent manner. These two receptors have been described to mediate Gal-9-immunosuppressive signals in effector and regulatory T cells. Treatment of human PBMCs from healthy donors with HFB9-2 prevents Gal-9-induced Th1 cell apoptosis and suppresses the expansion of regulatory T cells. A humanized variant of HFB9-2, HFB9-2hz11, was generated and further characterized for its stability and pharmacokinetic profile. HFB9-2hz11 has a favorable developability profile. It demonstrated stability for at least 14 days at 40°C, as well as for several hours at low pH, and following several freeze-thaw cycles. High plasma exposures following a single dose administration to mice were observed. Furthermore, several preclinical in vivo studies are ongoing to demonstrate the efficacy of HFB-2hz11. Gal-9 has been reported to play a dual role in AML as both a self-renewal factor for leukemic stem cells and a suppressor of anti-cancer immunity, suggesting that Gal-9 neutralization represents an attractive therapeutic approach for treatment of AML patients. To explore this hypothesis, we have initiated a predictive biomarker discovery effort, using HiFiBiO's proprietary Drug Intelligent Science (DIS™) platform, using AML primary patient cells and integrating single-cell technology to identify patient subpopulations likely to respond to HFB9-2hz11. Potential novel biomarkers for patient stratification, that could be applied to other tumor indications, will be identified for AML patients. Altogether, the data presented here provide evidence that neutralization of Gal-9 with HFB9-2hz11 blocks key immunosuppressive mechanisms known to limit the efficacy of current immunotherapies and position HFB9-2hz11 as a drug candidate for clinical exploration in AML and other indications. Citation Format: Germain Margall, Rachel Pacherie, Julie Prigent, He Zhou, Francisco Adrian, Liang Schweizer, Andreas Raue, Roshan Kumar, Nicola Beltraminelli, Stéphanie Beq. HFB9-2: A novel Gal-9 neutralizing antibody to reverse immune suppression in the tumor microenvironment [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6532.
- Published
- 2020
15. HIV-1 Envelope Recognition by Polyreactive and Cross-Reactive Intestinal B Cells
- Author
-
Thierry Prazuck, Hedda Wardemann, Alexia Kanyavuz, Olivier Schwartz, Tim Rollenske, Valérie Lorin, Laurent Lefrou, Hugo Mouquet, Jordan D. Dimitrov, Ayrin Kök, Timothée Bruel, Florence Guivel-Benhassine, Laurent Hocqueloux, Cyril Planchais, Thierry Hieu, Julie Prigent, Immunologie humorale - Humoral Immunology, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Sorbonne Université (SU), Université Sorbonne Paris Cité (USPC), Virus et Immunité, Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris], German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), Centre Hospitalier Régional d'Orléans (CHRO), Work in the O.S. lab is funded by the Institut Pasteur, ANRS, Sidaction, the Vaccine Research Institute (ANR-10-LABX-77), the Labex IBEID (ANR-10-IHUB-0002), the 'TIMTAMDEN' ANR-14-CE14-0029, the 'CHIKV-Viro-Immuno' ANR-14-CE14-0015-01, and the Gilead HIV Cure program. H.M. received core grants from the G5 Institut Pasteur program, the Milieu Intérieur program (ANR-10-LABX-69-01), and INSERM. This work was funded by the European Research Council (ERC)-Seventh Framework Program (ERC-2013-StG 337146). C.P., A.K., and T.H. were supported by the ERC-2013-StG 337146 program and J.P. by an ANRS postdoctoral fellowship. J.D.D. was supported by an ERC starting grant (ERC-2015-StG 678905)., ANR-10-LABX-0077,VRI,Initiative for the creation of a Vaccine Research Institute(2010), ANR-14-CE14-0029,TIMTAMDEN,Rôle des récepteurs TIM et TAM dans l'infection des cellules cibles par le virus de la dengue(2014), ANR-14-CE14-0015,CHIKV-Viro-Immuno,Multiplication et Relation avec l'hôte du virus Chikungunya(2014), ANR-10-LABX-0069,MILIEU INTERIEUR,GENETIC & ENVIRONMENTAL CONTROL OF IMMUNE PHENOTYPE VARIANCE: ESTABLISHING A PATH TOWARDS PERSONALIZED MEDICINE(2010), European Project: 337146,EC:FP7:ERC,ERC-2013-StG,HUMANTIVIRUSES(2014), European Project: 678905,H2020-EU.1.1., ERC-StG-2015,CoBABATI(2016), École pratique des hautes études (EPHE)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Régional d'Orléans (CHR), ANR-10-LABX-0077/10-LABX-0077,VRI,Initiative for the creation of a Vaccine Research Institute(2010), ANR: 10-LABX-0069,MILIEU INTERIEUR,GENETIC & ENVIRONMENTAL CONTROL OF IMMUNE PHENOTYPE VARIANCE: ESTABLISHING A PATH TOWARDS PERSONALIZED MEDICINE(2010), Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), École Pratique des Hautes Études (EPHE), and Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,medicine.drug_class ,Cross Reactions ,Gp41 ,medicine.disease_cause ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Viral envelope ,medicine ,Humans ,lcsh:QH301-705.5 ,B cell ,Antibody-dependent cell-mediated cytotoxicity ,B-Lymphocytes ,biology ,Chemistry ,virus diseases ,Molecular biology ,3. Good health ,Intestines ,Molecular mimicry ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,[SDV.IMM.IA]Life Sciences [q-bio]/Immunology/Adaptive immunology ,Humoral immunity ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,biology.protein ,HIV-1 ,Antibody ,030217 neurology & neurosurgery - Abstract
Summary: Mucosal immune responses to HIV-1 involve the recognition of the viral envelope glycoprotein (gp)160 by tissue-resident B cells and subsequent secretion of antibodies. To characterize the B cells “sensing” HIV-1 in the gut of infected individuals, we probed monoclonal antibodies produced from single intestinal B cells binding to recombinant gp140 trimers. A large fraction of mucosal B cell antibodies were polyreactive and showed only low affinity to HIV-1 envelope glycoproteins, particularly the gp41 moiety. A few high-affinity gp140 antibodies were isolated but lacked neutralizing, potent ADCC, and transcytosis-blocking capacities. Instead, they displayed cross-reactivity with defined self-antigens. Specifically, intestinal HIV-1 gp41 antibodies targeting the heptad repeat 2 region (HR2) cluster II cross-reacted with the p38α mitogen-activated protein kinase 14 (MAPK14). Hence, physiologic polyreactivity of intestinal B cells and molecular mimicry-based self-reactivity of HIV-1 antibodies are two independent phenomena, possibly diverting and/or impairing mucosal humoral immunity to HIV-1. : Antibodies produced in mucosa after sexual transmission of HIV-1 could affect viral propagation. Planchais et al. show that intestinal B cells from HIV-1-infected individuals that recognize the HIV-1 envelope (Env) proteins are mainly low affinity and polyreactive and that rare, high-affinity antibodies to HIV-1 Env lack potent antiviral capacities and cross-react with self-antigens. Keywords: HIV-1, antibodies, B cells, mucosa, polyreactivity, cross-reactivity, MAPK14, intestine
- Published
- 2018
16. Conformational Plasticity in Broadly Neutralizing HIV-1 Antibodies Triggers Polyreactivity
- Author
-
Valérie Lorin, Jordan D. Dimitrov, Oxana Vratskikh, Julie Prigent, Thérèse Couderc, Caroline Eden, Hugo Mouquet, Cyril Planchais, Michael S. Seaman, Olivier Schwartz, Annaëlle Jarossay, Ayrin Kök, Timothée Bruel, Jérémy Dufloo, Oliver Ohlenschläger, Réponse humorale aux pathogènes, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Icahn School of Medicine at Mount Sinai [New York] (MSSM), Immunopathologie Virale, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Biologie des Infections - Biology of Infection, Virus et Immunité - Virus and immunity (CNRS-UMR3569), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Pasteur [Paris], Université Paris Diderot - Paris 7 (UPD7)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Virus et Immunité - Virus and immunity, Université Paris Diderot - Paris 7 (UPD7)-École pratique des hautes études (EPHE)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Virus et Immunité, and Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris]
- Subjects
0301 basic medicine ,HIV Antigens ,Protein Conformation ,autoreactivity ,polyreactivity ,antibody ,B cells ,HIV-1 ,Human immunodeficiency virus (HIV) ,Biology ,Cross Reactions ,HIV Antibodies ,medicine.disease_cause ,Autoantigens ,General Biochemistry, Genetics and Molecular Biology ,Neutralization ,Article ,03 medical and health sciences ,Immunoglobulin Fab Fragments ,0302 clinical medicine ,Antigen ,Neutralization Tests ,medicine ,Humans ,lcsh:QH301-705.5 ,Gene ,chemistry.chemical_classification ,env Gene Products, Human Immunodeficiency Virus ,Antibodies, Neutralizing ,3. Good health ,Cell biology ,030104 developmental biology ,lcsh:Biology (General) ,chemistry ,[SDV.IMM.IA]Life Sciences [q-bio]/Immunology/Adaptive immunology ,biology.protein ,Thermodynamics ,Binding Sites, Antibody ,Antibody ,Glycoprotein ,Hydrophobic and Hydrophilic Interactions ,030215 immunology - Abstract
Summary Human high-affinity antibodies to pathogens often recognize unrelated ligands. The molecular origin and the role of this polyreactivity are largely unknown. Here, we report that HIV-1 broadly neutralizing antibodies (bNAbs) are frequently polyreactive, cross-reacting with non-HIV-1 molecules, including self-antigens. Mutating bNAb genes to increase HIV-1 binding and neutralization also results in de novo polyreactivity. Unliganded paratopes of polyreactive bNAbs with improved HIV-1 neutralization exhibit a conformational flexibility, which contributes to enhanced affinity of bNAbs to various HIV-1 envelope glycoproteins and non-HIV antigens. Binding adaptation of polyreactive bNAbs to the divergent ligands mainly involves hydrophophic interactions. Plasticity of bNAbs’ paratopes may, therefore, facilitate accommodating divergent viral variants, but it simultaneously triggers promiscuous binding to non-HIV-1 antigens. Thus, a certain level of polyreactivity can be a mark of adaptable antibodies displaying optimal pathogens’ recognition., Graphical Abstract, Highlights • Most HIV-1 bNAbs are polyreactive and often cross-react with self-antigens • Polyreactivity of bNAbs involves hydrophobic interactions • Mutations enhancing HIV-1 bNAbs’ capacities also induce de novo polyreactivity • Enhanced neutralization and polyreactivity co-emerge by antibody conformational flexibility, HIV-1 bNAbs are frequently polyreactive and bind to self-antigens. Prigent et al. show that specific mutations in bNAbs that enhance their neutralizing capacity create an intrinsic structural flexibility of the antibody paratope. This promotes the conformational adaptation that facilitates binding to HIV-1 variants and polyreactivity to topologically distinct non-HIV-1 molecules.
- Published
- 2018
17. Long-Term In Vivo Monitoring of Adult-Derived Human Liver Stem/Progenitor Cells by Bioluminescence Imaging, Positron Emission Tomography, and Contrast-Enhanced Computed Tomography
- Author
-
Lionel Larbanoix, Vincent Grégoire, Mei-Ju Hsu, Mustapha Najimi, Julie Prigent, Catherine Lombard, Gaetan Van Simaeys, Gisèle Deblandre, Etienne Sokal, Serge Goldman, Anne Bol, Joachim Ravau, and Pierre-Edouard Dollet
- Subjects
0301 basic medicine ,Adult ,Biodistribution ,Contrast Media ,Biology ,Green fluorescent protein ,03 medical and health sciences ,Mice ,In vivo ,medicine ,Bioluminescence imaging ,Animals ,Humans ,Tissue Distribution ,Progenitor cell ,Liver injury ,medicine.diagnostic_test ,Stem Cells ,Cell Biology ,Hematology ,medicine.disease ,Molecular biology ,Luminescent Proteins ,030104 developmental biology ,Liver ,Positron emission tomography ,Cell Tracking ,Positron-Emission Tomography ,Luminescent Measurements ,Stem cell ,Developmental Biology - Abstract
Adult-derived human liver stem/progenitor cells (ADHLSCs) have the potential to alleviate liver injury. However, the optimal delivery route and long-term biodistribution of ADHLSCs remain unclear. In this article, we used a triple fusion reporter system to determine the kinetic differences in the biodistribution of ADHLSCs following intrasplenic (IS) and intrahepatic (IH) administration in severe combined immunodeficiency/beige mice. ADHLSCs were transduced with a lentiviral vector expressing a triple fusion reporter comprising renilla luciferase, monomeric red fluorescent protein, and truncated HSV-1 thymidine kinase. The stability and duration of the transgenes, and the effects of transduction on the cell properties were evaluated in vitro. The acute retention and long-term engraftment in vivo were revealed by positron emission tomography and bioluminescence imaging (BLI), respectively, followed by histochemical analysis. We showed that ADHLSCs can be safely transduced with the triple fusion reporter. Radiolabeled ADHLSCs showed acute cell retention at the sites of injection. The IH group showed a confined BLI signal at the injection site, while the IS group displayed a dispersed distribution at the upper abdominal liver area, and a more intense signal. In conclusion, ADHLSCs could be monitored by BLI for up to 4 weeks with a spread out biodistribution following IS injection.
- Published
- 2017
18. Adult-Derived Human Liver Stem/Progenitor Cells Infused 3 Days Postsurgery Improve Liver Regeneration in a Mouse Model of Extended Hepatectomy
- Author
-
Mustapha Najimi, Julie Prigent, Astrid Herrero, Catherine Lombard, Valérie Rosseels, Martine Daujat-Chavanieu, Karine Breckpot, Etienne Sokal, Gisèle Deblandre, Laboratory of Molecullar and Cellular Therapy, Basic (bio-) Medical Sciences, Centre Hospitalier Régional Universitaire [Montpellier] ( CHRU Montpellier ), Cellules Souches, Plasticité Cellulaire, Médecine Régénératrice et Immunothérapies (IRMB), CHU Saint-Eloi-Centre Hospitalier Régional Universitaire [Montpellier] ( CHRU Montpellier ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Université de Montpellier ( UM ), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain = Catholic University of Louvain (UCL), Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Vrije Universiteit Brussel [Bruxelles] (VUB), This study was supported by both the Région Wallonne, as part of the Regenestem project (Convention No. 1017264), and the Université Catholique de Louvain. The authors would like to thank Jonathan Evraerts and Charles de Pierpont for their technical help, and Dr. Julie Sainz for review-ing the manuscript., Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), and Herrada, Anthony
- Subjects
0301 basic medicine ,Male ,[SDV.MHEP.CHI] Life Sciences [q-bio]/Human health and pathology/Surgery ,Liver cytology ,medicine.medical_treatment ,Mesenchymal cells ,lcsh:Medicine ,Cell therapy ,Mesoderm ,Mice ,Hepatic stem/progenitor cells ,biomedical engineering ,[ SDV.MHEP.CHI ] Life Sciences [q-bio]/Human health and pathology/Surgery ,Liver cell ,Liver Diseases ,Stem Cells ,Alanine Transaminase ,Liver regeneration ,3. Good health ,Liver ,Xenotransplantation ,Stem cell ,engraftment ,Mice, Transgenic ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,[SDV.MHEP.CHI]Life Sciences [q-bio]/Human health and pathology/Surgery ,Biology ,Mesenchymal Stem Cell Transplantation ,Article ,Andrology ,Alu qPCR ,03 medical and health sciences ,medicine ,Animals ,Hepatectomy ,Humans ,Aspartate Aminotransferases ,Progenitor cell ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Cell Proliferation ,[ SDV.BC ] Life Sciences [q-bio]/Cellular Biology ,lcsh:R ,Cell Biology ,Liver Regeneration ,Transplantation ,Disease Models, Animal ,030104 developmental biology ,Immunology ,Hepatocytes ,Liver Failure ,transplantation - Abstract
There is growing evidence that cell therapy constitutes a promising strategy for liver regenerative medicine. In the setting of hepatic cancer treatments, cell therapy could prove a useful therapeutic approach for managing the acute liver failure that occurs following extended hepatectomy. In this study, we examined the influence of delivering adult-derived human liver stem/progenitor cells (ADHLSCs) at two different early time points in an immunodeficient mouse model ( Rag2−/-IL2Rg -/-) that had undergone a 70% hepatectomy procedure. The hepatic mesenchymal cells were intrasplenically infused either immediately after surgery ( n = 26) or following a critical 3-day period ( n = 26). We evaluated the cells' capacity to engraft at day 1 and day 7 following transplantation by means of human Alu qPCR quantification, along with histological assessment of human albumin and α-smooth muscle actin. In addition, cell proliferation (anti-mouse and human Ki-67 staining) and murine liver weight were measured in order to evaluate liver regeneration. At day 1 posttransplantation, the ratio of human to mouse cells was similar in both groups, whereas 1 week posttransplantation this ratio was significantly improved ( p < 0.016) in mice receiving ADHLSC injection at day 3 posthepatectomy (1.7%), compared to those injected at the time of surgery (1%). On the basis of liver weight, mouse liver regeneration was more extensive 1 week posttransplantation in mice transplanted with ADHLSCs (+65.3%) compared to that of mice from the sham vehicle group (+42.7%). In conclusion, infusing ADHLSCs 3 days after extensive hepatectomy improves the cell engraftment and murine hepatic tissue regeneration, thereby confirming that ADHLSCs could be a promising cell source for liver cell therapy and hepatic tissue repair.
- Published
- 2017
19. Human Liver Progenitor Cells for Liver Repair
- Author
-
Catherine Lombard, Etienne Sokal, and Julie Prigent
- Subjects
business.industry ,Liver cell ,Cell ,Review ,Bioinformatics ,medicine.disease ,Regenerative medicine ,Cryopreservation ,Cell therapy ,Transplantation ,Liver disease ,medicine.anatomical_structure ,General Earth and Planetary Sciences ,Medicine ,Progenitor cell ,business ,General Environmental Science - Abstract
Because of their high proliferative capacity, resistance to cryopreservation, and ability to differentiate into hepatocyte-like cells, stem and progenitor cells have recently emerged as attractive cell sources for liver cell therapy, a technique used as an alternative to orthotopic liver transplantation in the treatment of various hepatic ailments ranging from metabolic disorders to end-stage liver disease. Although stem and progenitor cells have been isolated from various tissues, obtaining them from the liver could be an advantage for the treatment of hepatic disorders. However, the techniques available to isolate these stem/progenitor cells are numerous and give rise to cell populations with different morphological and functional characteristics. In addition, there is currently no established consensus on the tests that need to be performed to ensure the quality and safety of these cells when used clinically. The purpose of this review is to describe the different types of liver stem/progenitor cells currently reported in the literature, discuss their suitability and limitations in terms of clinical applications, and examine how the culture and transplantation techniques can potentially be improved to achieve a better clinical outcome.
- Published
- 2016
20. The stringent response of Bacillus anthracis contributes to sporulation but not to virulence
- Author
-
Willem van Schaik, Agnès Fouet, Julie Prigent, Toxines et Pathogénie Bactérienne, Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris], and Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Stringent response ,Bacillus subtilis ,MESH: Virulence ,medicine.disease_cause ,MESH: Down-Regulation ,Mice ,MESH: Spores, Bacterial ,MESH: Transcription Factor RelA ,MESH: Animals ,MESH: Bacterial Proteins ,Pathogen ,Spores, Bacterial ,2. Zero hunger ,MESH: Gene Expression Regulation, Bacterial ,0303 health sciences ,Virulence ,biology ,Effector ,MESH: Bacillus anthracis ,Bacillus anthracis ,RNA, Bacterial ,Female ,MESH: RNA, Bacterial ,Anthrax toxin ,Down-Regulation ,Guanosine Tetraphosphate ,Microbiology ,Anthrax ,03 medical and health sciences ,Bacterial Proteins ,MESH: Anthrax ,medicine ,Animals ,MESH: Mice ,Escherichia coli ,030304 developmental biology ,030306 microbiology ,Transcription Factor RelA ,MESH: Guanosine Tetraphosphate ,Gene Expression Regulation, Bacterial ,biology.organism_classification ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,Culture Media ,MESH: Gene Deletion ,MESH: Culture Media ,MESH: Female ,Amino Acids, Branched-Chain ,Gene Deletion ,MESH: Amino Acids, Branched-Chain - Abstract
International audience; The Gram-positive, spore-forming pathogen Bacillus anthracis is the aetiological agent of anthrax. Its main virulence factors are two toxins and an anti-phagocytic capsule. When B. anthracis is grown in laboratory culture, the highest expression of the anthrax toxin genes occurs during entry into stationary phase, suggesting that nutrient limitation is an environmental cue which induces toxin production. A common bacterial response to starvation is the so-called stringent response, in which the hyperphosphorylated guanosine nucleotide (p)ppGpp is the effector molecule. In Escherichia coli, Bacillus subtilis and other bacteria, accumulation of this molecule leads to down-regulation of stable RNA synthesis and upregulation of the expression of genes involved in survival under nutrient-poor conditions. This study focuses on the stringent response of B. anthracis. We show that in B. anthracis the relA gene is responsible for the synthesis of (p)ppGpp and the stringent down-regulation of stable RNA synthesis upon starvation for the essential amino acids isoleucine, leucine and valine. The deletion of relA did not affect the expression of the virulence gene pagA or virulence in a mouse model of infection. In contrast, spore counts upon growth and sporulation in a defined medium were approximately 10,000-fold lower for the relA deletion mutant than for the parental strain. The contribution of the stringent response to efficient sporulation of B. anthracis is notable, as this suggests that the stringent response may contribute to the persistence of B. anthracis in the natural environment.
- Published
- 2007
21. Human Progenitor Cell Quantification after Xenotransplantation in Rat and Mouse Models by a Sensitive qPCR Assay
- Author
-
Jérôme Ambroise, Gisèle Deblandre, Françoise Smets, Julie Prigent, Etienne Sokal, Astrid Herrero, and Université Catholique de Louvain = Catholic University of Louvain (UCL)
- Subjects
Liver cytology ,Xenotransplantation ,medicine.medical_treatment ,Transplantation, Heterologous ,Cell ,Biomedical Engineering ,Heterologous ,lcsh:Medicine ,Biology ,Real-Time Polymerase Chain Reaction ,Hepatic progenitor cells ,Mice ,Alu Elements ,In vivo ,Alu repeat sequences ,medicine ,Animals ,Humans ,Cell Lineage ,Rats, Wistar ,Progenitor cell ,Quantitative polymerase chain reaction (qPCR) ,Mice, Knockout ,Transplantation ,Stem Cells ,lcsh:R ,DNA ,Cell Biology ,Molecular biology ,Engraftment analysis ,Rats ,3. Good health ,DNA-Binding Proteins ,Real-time polymerase chain reaction ,medicine.anatomical_structure ,Liver ,Models, Animal ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Interleukin Receptor Common gamma Subunit - Abstract
International audience; Xenotransplantation of human cells in animal models is an essential tool for evaluation of safety and efficacy of cell-based products for therapeutic use. Sensitive and reproducible methods are needed to detect and quantify human cells engrafted into the host tissue either in the targeted organ or in undesired locations. We developed a robust quantitative polymerase chain reaction (qPCR) assay based on amplification of human AluYb8 repeats, to assess the number of human cells present in rat or mouse tissues after transplantation. Standard curves of mixed human/rodent DNA and mixed human/rodent cells have been performed to determine the limit of detection and linear range of the assay. Standard curves from DNA mixing differed significantly from standard curves from cell mixing. We show here that the AluYb8 qPCR assay is highly reproducible and is able to quantify human cells in a rodent cell matrix over a large linear range that extends from 50% to 0.01% human cells. Short-term in vivo studies showed that human cells could be quantified in mouse liver up to 7 days after intrasplenic transplantation and in rat liver 4 h after intrahepatic transplantation.
- Published
- 2015
22. Human liver progenitor cells for liver repair
- Author
-
Catherine A. Lombard, Julie Prigent, and Etienne M. Sokal
- Subjects
General Earth and Planetary Sciences ,General Environmental Science - Published
- 2013
23. Neutralising antibodies against ricin toxin
- Author
-
Laetitia Panigai, Karine Devilliers, Hervé Volland, Marc Plaisance, Christophe Créminon, Didier Sauvaire, Julie Prigent, Patricia Lamourette, and Stéphanie Simon
- Subjects
Male ,Antibody Affinity ,Poison control ,lcsh:Medicine ,Lactose ,Pharmacology ,Toxicology ,medicine.disease_cause ,Jurkat Cells ,Mice ,chemistry.chemical_compound ,Antibody Specificity ,Toxin Binding ,lcsh:Science ,Multidisciplinary ,biology ,Poisoning ,Antibodies, Monoclonal ,Drug Synergism ,Ricin ,Immunotherapy ,Antibody ,Protein Binding ,Research Article ,endocrine system ,Cell Survival ,medicine.drug_class ,Blotting, Western ,Immunology ,Toxic Agents ,Immunoglobulins ,Monoclonal antibody ,Binding, Competitive ,In vivo ,medicine ,Animals ,Humans ,Immunoassays ,Biology ,Dose-Response Relationship, Drug ,Toxin ,business.industry ,lcsh:R ,Immunity ,Surface Plasmon Resonance ,Antibodies, Neutralizing ,Virology ,In vitro ,carbohydrates (lipids) ,Protein Subunits ,enzymes and coenzymes (carbohydrates) ,chemistry ,Immunologic Techniques ,biology.protein ,Nasal administration ,lcsh:Q ,business - Abstract
The Centers for Disease Control and Prevention have listed the potential bioweapon ricin as a Category B Agent. Ricin is a so-called A/B toxin produced by plants and is one of the deadliest molecules known. It is easy to prepare and no curative treatment is available. An immunotherapeutic approach could be of interest to attenuate or neutralise the effects of the toxin. We sought to characterise neutralising monoclonal antibodies against ricin and to develop an effective therapy. For this purpose, mouse monoclonal antibodies (mAbs) were produced against the two chains of ricin toxin (RTA and RTB). Seven mAbs were selected for their capacity to neutralise the cytotoxic effects of ricin in vitro. Three of these, two anti-RTB (RB34 and RB37) and one anti-RTA (RA36), when used in combination improved neutralising capacity in vitro with an IC(50) of 31 ng/ml. Passive administration of association of these three mixed mAbs (4.7 µg) protected mice from intranasal challenges with ricin (5 LD(50)). Among those three antibodies, anti-RTB antibodies protected mice more efficiently than the anti-RTA antibody. The combination of the three antibodies protected mice up to 7.5 hours after ricin challenge. The strong in vivo neutralising capacity of this three mAbs combination makes it potentially useful for immunotherapeutic purposes in the case of ricin poisoning or possibly for prevention.
- Published
- 2011
24. Production and characterisation of a neutralising chimeric antibody against botulinum neurotoxin A
- Author
-
Prigent, Julie, Mazuet, Christelle, Boquet, Didier, Lamourette, Patricia, Volland, Hervé, Popoff, Michel, Créminon, Christophe, Simon, Stéphanie, Moreno, Edgardo, Institut de Biologie et de Technologies de Saclay (IBITECS), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay, Bactéries anaérobies et Toxines, Institut Pasteur [Paris] (IP), Laboratoire d'Ingénierie des Anticorps pour la Santé (LIAS), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Universidad Nacional, Heredia (UNA), Escuela de Medicina Veterinaria, This study was supported by a grant from the PhD programme of the CEA (Commissariat à l'Energie Atomique et aux Energies Renouvelables) (Julie Prigent) and funded by the IMASSA (Institut de Médecine Aérospatiale du Service de Santé des Armées) and NRBC (Nuclear Radiological Biological and Chemical Risks) programmes., Institut Pasteur [Paris], Institut de Biologie et de Technologies de Saclay ( IBITECS ), Université Paris-Saclay-Commissariat à l'énergie atomique et aux énergies alternatives ( CEA ), Laboratoire d'Ingénierie des Anticorps pour la Santé ( LIAS ), Commissariat à l'énergie atomique et aux énergies alternatives ( CEA ), and Universidad Nacional, Heredia ( UNA )
- Subjects
MESH : Cell Line ,[ SDV.TOX ] Life Sciences [q-bio]/Toxicology ,lcsh:Medicine ,MESH : Blotting, Western ,medicine.disease_cause ,Antibody production ,Biochemistry ,Neutralization ,MESH: Antibodies, Neutralizing ,Infectious Diseases/Bacterial Infections ,Botulinum toxin ,[ SDV.IMM ] Life Sciences [q-bio]/Immunology ,Toxins ,Botulism ,MESH: Animals ,Botulinum Toxins, Type A ,Enzyme-linked immunoassays ,lcsh:Science ,0303 health sciences ,Multidisciplinary ,MESH: Spodoptera ,MESH : Antibodies, Neutralizing ,3. Good health ,[SDV.TOX]Life Sciences [q-bio]/Toxicology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Antibody ,Research Article ,MESH : Botulinum Toxins, Type A ,medicine.drug_class ,Immunology ,Blotting, Western ,Biology ,Spodoptera ,Monoclonal antibody ,MESH : Spodoptera ,Antibodies ,Cell Line ,03 medical and health sciences ,Affinity chromatography ,medicine ,Animals ,MESH: Blotting, Western ,030304 developmental biology ,Antiserum ,030306 microbiology ,Toxin ,MESH: Botulinum Toxins, Type A ,lcsh:R ,medicine.disease ,Virology ,Antibodies, Neutralizing ,MESH: Cell Line ,biology.protein ,Clostridium botulinum ,lcsh:Q ,MESH : Animals ,Antitoxins ,Cloning - Abstract
International audience; Botulinum neurotoxins, produced by Clostridium botulinum bacteria, are the causative agent of botulism. This disease only affects a few hundred people each year, thus ranking it among the orphan diseases. However, botulinum toxin type A (BoNT/A) is the most potent toxin known to man. Due to their potency and ease of production, these toxins were classified by the Centers for Disease Control and Prevention (CDC) as Category A biothreat agents. For several biothreat agents, like BoNT/A, passive immunotherapy remains the only possible effective treatment allowing in vivo neutralization, despite possible major side effects. Recently, several mouse monoclonal antibodies directed against a recombinant fragment of BoNT/A were produced in our laboratory and most efficiently neutralised the neurotoxin. In the present work, the most powerful one, TA12, was selected for chimerisation. The variable regions of this antibody were thus cloned and fused with the constant counterparts of human IgG1 (kappa light and gamma 1 heavy chains). Chimeric antibody production was evaluated in mammalian myeloma cells (SP2/0-Ag14) and insect cells (Sf9). After purifying the recombinant antibody by affinity chromatography, the biochemical properties of chimeric and mouse antibody were compared. Both have the same very low affinity constant (close to 10 pM) and the chimeric antibody exhibited a similar capacity to its parent counterpart in neutralising the toxin in vivo. Its strong affinity and high neutralising potency make this chimeric antibody interesting for immunotherapy treatment in humans in cases of poisoning, particularly as there is a probable limitation of the immunological side effects observed with classical polyclonal antisera from heterologous species.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.