14 results on '"Gabrielle S. Le Provost"'
Search Results
2. Supplementary Legends for Figures 1-4 from The HIF-1–Inducible Lysyl Oxidase Activates HIF-1 via the Akt Pathway in a Positive Regulation Loop and Synergizes with HIF-1 in Promoting Tumor Cell Growth
- Author
-
Caroline Reynaud, Jacques Pouysségur, Pascal Sommer, Philippe Clézardin, Blandine Deux, Gabrielle S. Le Provost, Géraldine Aimond, Bastien Kaniewski, Jérome Durivault, Frédéric Dayan, and Floriane Pez
- Abstract
Supplementary Legends for Figures 1-4 from The HIF-1–Inducible Lysyl Oxidase Activates HIF-1 via the Akt Pathway in a Positive Regulation Loop and Synergizes with HIF-1 in Promoting Tumor Cell Growth
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 1 from The HIF-1–Inducible Lysyl Oxidase Activates HIF-1 via the Akt Pathway in a Positive Regulation Loop and Synergizes with HIF-1 in Promoting Tumor Cell Growth
- Author
-
Caroline Reynaud, Jacques Pouysségur, Pascal Sommer, Philippe Clézardin, Blandine Deux, Gabrielle S. Le Provost, Géraldine Aimond, Bastien Kaniewski, Jérome Durivault, Frédéric Dayan, and Floriane Pez
- Abstract
Supplementary Figure 1 from The HIF-1–Inducible Lysyl Oxidase Activates HIF-1 via the Akt Pathway in a Positive Regulation Loop and Synergizes with HIF-1 in Promoting Tumor Cell Growth
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure 4 from The HIF-1–Inducible Lysyl Oxidase Activates HIF-1 via the Akt Pathway in a Positive Regulation Loop and Synergizes with HIF-1 in Promoting Tumor Cell Growth
- Author
-
Caroline Reynaud, Jacques Pouysségur, Pascal Sommer, Philippe Clézardin, Blandine Deux, Gabrielle S. Le Provost, Géraldine Aimond, Bastien Kaniewski, Jérome Durivault, Frédéric Dayan, and Floriane Pez
- Abstract
Supplementary Figure 4 from The HIF-1–Inducible Lysyl Oxidase Activates HIF-1 via the Akt Pathway in a Positive Regulation Loop and Synergizes with HIF-1 in Promoting Tumor Cell Growth
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Methods from The HIF-1–Inducible Lysyl Oxidase Activates HIF-1 via the Akt Pathway in a Positive Regulation Loop and Synergizes with HIF-1 in Promoting Tumor Cell Growth
- Author
-
Caroline Reynaud, Jacques Pouysségur, Pascal Sommer, Philippe Clézardin, Blandine Deux, Gabrielle S. Le Provost, Géraldine Aimond, Bastien Kaniewski, Jérome Durivault, Frédéric Dayan, and Floriane Pez
- Abstract
Supplementary Methods from The HIF-1–Inducible Lysyl Oxidase Activates HIF-1 via the Akt Pathway in a Positive Regulation Loop and Synergizes with HIF-1 in Promoting Tumor Cell Growth
- Published
- 2023
- Full Text
- View/download PDF
6. An approved in vitro approach to preclinical safety and efficacy evaluation of engineered T cell receptor anti-CD3 bispecific (ImmTAC) molecules.
- Author
-
Jane Harper, Katherine J Adams, Giovanna Bossi, Debbie E Wright, Andrea R Stacey, Nicole Bedke, Ruth Martinez-Hague, Dan Blat, Laure Humbert, Hazel Buchanan, Gabrielle S Le Provost, Zoe Donnellan, Ricardo J Carreira, Samantha J Paston, Luise U Weigand, Martina Canestraro, Joseph P Sanderson, Sophie Botta Gordon-Smith, Kate L Lowe, Karolina A Rygiel, Alex S Powlesland, Annelise Vuidepot, Namir J Hassan, Brian J Cameron, Bent K Jakobsen, and Joseph Dukes
- Subjects
Medicine ,Science - Abstract
Robust preclinical testing is essential to predict clinical safety and efficacy and provide data to determine safe dose for first-in-man studies. There are a growing number of examples where the preclinical development of drugs failed to adequately predict clinical adverse events in part due to their assessment with inappropriate preclinical models. Preclinical investigations of T cell receptor (TCR)-based immunotherapies prove particularly challenging as these biologics are human-specific and thus the conventional testing in animal models is inadequate. As these molecules harness the full force of the immune system, and demonstrate tremendous potency, we set out to design a preclinical package that would ensure adequate evaluation of these therapeutics. Immune Mobilising Monoclonal TCR Against Cancer (ImmTAC) molecules are bi-specific biologics formed of an affinity-enhanced TCR fused to an anti-CD3 effector function. ImmTAC molecules are designed to activate human T lymphocytes and target peptides within the context of a human leukocyte antigen (HLA), thus require an intact human immune system and peptidome for suitable preclinical screening. Here we draw upon the preclinical testing of four ImmTAC molecules, including IMCgp100, the first ImmTAC molecule to reach the clinic, to present our comprehensive, informative and robust approach to in vitro preclinical efficacy and safety screening. This package comprises a broad range of cellular and molecular assays using human tissues and cultured cells to test efficacy, safety and specificity, and hence predict human responses in clinical trials. We propose that this entirely in vitro package offers a potential model to be applied to screening other TCR-based biologics.
- Published
- 2018
- Full Text
- View/download PDF
7. 624 IFNγ secreted by tebentafusp (IMCgp100)-redirected T cells inhibits expression of melanin synthesis pathway genes in healthy melanocytes
- Author
-
Jane Houghton, Laura Collins, Koustubh Ranade, Adel Benlahrech, Gabrielle S. Le Provost, Camille Britton-Rivet, Jane Harper, David Depoil, and Mariantonella Vardeu
- Subjects
Granzyme B production ,business.industry ,Melanoma ,medicine.medical_treatment ,T cell ,Vitiligo ,Melanocyte ,medicine.disease ,Melanin ,medicine.anatomical_structure ,Cytokine ,Antigen ,Cancer research ,medicine ,business - Abstract
Background Tebentafusp (IMCgp100) is a bispecific T cell redirector comprised of an affinity-enhanced TCR recognising melanocyte lineage antigen gp100 and a T cell engaging anti-CD3 scFv domain. Tebentafusp has shown activity as monotherapy in advanced cutaneous and uveal melanoma (Middleton et al., ASCO 2019), and we have previously reported that over half of uveal melanoma patients treated with tebentafusp display melanocyte-related adverse events (MRAE). These include vitiligo/skin hypopigmentation, leukotrichia, and hyperpigmentation and, collectively, are associated with better overall survival in uveal patients receiving tebentafusp (Orloff et al, AACR 2020). In this study, we dissected the mechanisms by which tebentafusp may induce MRAE and highlight the potential clinical significance. Methods In vitro studies were conducted to assess the direct and indirect effects of tebentafusp on epidermal melanocytes from healthy donors. Expression of gp100 and the gp100:HLA*02:01 target complex by melanocytes were quantified at the mRNA level and on the cell surface by confocal microscopy, respectively. Melanocytes co-cultured with PBMC and increasing concentrations of tebentafusp were assessed for their susceptibility to lysis and/or ability to stimulate cytokine production. These readouts were compared to gp100-positive and negative melanoma cancer cell lines. Melanin production by melanocytes was quantified and the melanin synthesis pathway interrogated at the mRNA and protein level following exposure to secretomes from tebentafusp-redirected PBMC against melanoma cancer cells. Results Healthy melanocytes expressed 2 to 3-fold lower levels of gp100 peptide-HLA complexes on their surface compared to gp100-positive melanoma cell lines. In the presence of tebentafusp, this lower target expression translated into 3–6 fold lower levels of IFNγ and more than 100 fold lower granzyme B production by redirected T cells and these melanocytes were resistant to direct tebentafusp-induced killing (EC50 for melanocytes greater than 1nM vs EC50 melanoma cell lines of 23–50 pM). Supernatants from T cells activated in response to melanoma cancer cells by tebentafusp downregulated the melanin content of healthy melanocytes (20–30% reduction). Western blotting revealed 30–40% inhibition of two key components of the melanin synthesis pathway; the tyrosinase-related protein (TRP)-1 and TRP-2. This inhibition was reversed by blocking IFNγ in supernatants from activated T cells. Conclusions MRAEs, especially vitiligo, associated with response to tebentafusp, may be explained, at least in part, by the downregulation of melanin biosynthesis pathway genes by IFNγ secreted by tebentafusp-activated T cells. Ethics Approval The study was approved by the South Central - Oxford A Research Ethics Committee (UK), REC reference 13/SC/0226 References Middleton, et al., Relationship between clinical efficacy and AEs of IMCgp100, a novel bispecific TCR–anti-CD3, in patients with advanced melanoma. Journal of Clinical Oncology. 2019. Orloff, et al., Vitiligo and other clinical melanocyte-related adverse events following tebentafusp (IMCgp100) exposure in patients with uveal melanoma. AACR (American Association for Cancer Research), 2020.
- Published
- 2020
- Full Text
- View/download PDF
8. Abstract 5572: IMC-F106C, a novel and potent immunotherapy approach to treat PRAME expressing solid and hematologic tumors
- Author
-
Florence Schlosser, Laure Humbert, Gabrielle S. Le Provost, Amanda M. Williams, Jane Harper, Jacob Robinson, Greg Dobrynin, Alessio Vantellini, Sylvie Moureau, Athiva Shankar, and David Berman
- Subjects
Cancer Research ,PRAME ,business.industry ,T cell ,Melanoma ,medicine.medical_treatment ,Cancer ,Immunotherapy ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Antigen ,medicine ,Cancer research ,Adenocarcinoma ,business ,Triple-negative breast cancer - Abstract
Background: Bispecific immunotherapies have been validated for the treatment of hematologic tumors but none have yet been approved for solid tumor indications, including high prevalent tumors such as non-small-cell lung carcinoma (NSCLC). Immunocore is developing ImmTAC® molecules, a new class of TCR/anti-CD3 bispecific fusion protein, that target intracellularly derived peptides presented at the tumor cell surface in complex with human leukocyte antigen (HLA). The ImmTAC IMC-F106C is in development for the treatment of advanced cancers that are positive for Preferentially Expressed Antigen in Melanoma (PRAME). PRAME is a cancer-testis antigen (CTA) that is highly expressed in normal testis and a range of solid and hematologic malignancies. The aim of this study was to characterise the expression of PRAME in a variety of human malignancies (mRNA and protein) and demonstrate that IMC-F106C can potently redirect T cells to eliminate indication-relevant tumor cells in vitro. Method: FFPE tumor samples were analysed by RT-qPCR and IHC for a number of tumor indications, to determine levels of PRAME mRNA and protein expression in patient samples for each indication. The activity of IMC-F106C was investigated in cellular assays using healthy donor PBMCs as effectors, targeting a variety of indication-relevant tumor cell lines expressing a PRAME-specific peptide complexed with HLA-A*02:01, including NSCLC non-small-cell lung carcinoma, ovarian carcinoma, and acute myeloid leukemia cell lines. T cell activation was assessed by cytokine release and T cell-mediated target-cell killing was evaluated by measurement of cell death (xCELLigence). Results: PRAME mRNA and protein expression was highly prevalent in samples of NSCLC, including both the adenocarcinoma and squamous cell carcinoma subtypes, SCLC, melanoma, ovarian, endometrial carcinoma samples, and in triple negative breast cancer (TNBC). Over 60% of samples demonstrated some level of PRAME expression by IHC and RT-qPCR in these 6 cancer indications. In the PRAME positive HLA-A*02:01 positive cell lines, IMC-F106C redirected donor effector cells to release IFNγ and GrB and kill tumor cells in a dose-dependent manner, with activity demonstrated as low as < 1 pM. By contrast, cell lines negative for PRAME or HLA-A*02:01 expression failed to induce responses < 1 nM of IMC-F106C. Conclusion: These data indicate that PRAME is expressed in a number of solid tumors, and is highly prevalent in lung tumours, irrespective of EGFR status, as well as female-oriented cancers. In conjunction, IMC-F106C efficiently redirects T cell activity against tumor cell lines that express PRAME across a range of tumor indications. Taken together, IMC-F106C could prove to be a highly effective immunotherapy option for HLA*02:01 positive patients with PRAME positive tumors. Citation Format: Sylvie Moureau, Alessio Vantellini, Florence Schlosser, Jacob Robinson, Jane Harper, Athiva Shankar, Greg Dobrynin, Gabrielle Le Provost, Amanda Williams, David Berman, Laure Humbert. IMC-F106C, a novel and potent immunotherapy approach to treat PRAME expressing solid and hematologic tumors [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 5572.
- Published
- 2020
- Full Text
- View/download PDF
9. Lysyl Oxidase silencing impairs keratinocyte differentiation in a reconstructed-epidermis model
- Author
-
Pascal Sommer, Valerie Andre, Valerie Cenizo, Gabrielle S. Le Provost, Géraldine Aimond, Bastien Kaniewski, Floriane Pez, and Romain Debret
- Subjects
0303 health sciences ,Cell type ,integumentary system ,Epidermis (botany) ,Cell growth ,Lysyl oxidase ,Dermatology ,Biology ,Biochemistry ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,RNA interference ,030220 oncology & carcinogenesis ,Extracellular ,medicine ,Gene silencing ,Keratinocyte ,Molecular Biology ,030304 developmental biology - Abstract
Lysyl Oxidase (LOX) is an extracellular enzyme involved in the maturation of connective tissues. It also acts in many cell types as a regulator of cell behaviour and phenotype through intracellular signalling pathways. Recently, LOX was shown to be present in human epidermis where its precise functions remain unclear. We showed here that in confluent monolayer cultures of normal human keratinocytes (KCs) and N/TERT-1-immortalized KCs, LOX expression was induced during the first differentiation steps. Moreover, the silencing of LOX by stable RNA interference disrupted the expression of early differentiation markers. In a reconstructed-epidermis model, LOX silencing did not impair the stratification process nor the formation of the first differentiated layers. However, terminal differentiation was strongly impaired, as shown by a decreased expression of late differentiation proteins and by the absence of stratum corneum. Nonetheless, inhibition of LOX enzymatic activity by β-aminopropionitrile did not affect the differentiation process. Therefore, LOX protein acts during the first steps of KC differentiation and is important for subsequent commitment into terminal differentiation. Taken together, these results suggest that a finely regulated expression of LOX is necessary for normal KC differentiation and thus for maintenance of epidermal homeostasis.
- Published
- 2010
- Full Text
- View/download PDF
10. Direct molecular mimicry enables off-target cardiovascular toxicity by an enhanced affinity TCR designed for cancer immunotherapy
- Author
-
Marine C C Raman, Pierre J Rizkallah, Ruth Simmons, Zoe Donnellan, Joseph Dukes, Giovanna Bossi, Gabrielle S Le Provost, Penio Todorov, Emma Baston, Emma Hickman, Tara Mahon, Namir Hassan, Annelise Vuidepot, Malkit Sami, David K Cole, and Bent K. Jakobsen
- Subjects
Models, Molecular ,Antigen Presentation ,Protein Conformation ,Receptors, Antigen, T-Cell, alpha-beta ,Molecular Mimicry ,Receptors, Antigen, T-Cell ,Antigen-Presenting Cells ,Epitopes, T-Lymphocyte ,T-Cell Antigen Receptor Specificity ,Cross Reactions ,R1 ,Cardiotoxicity ,Peptide Fragments ,Article ,Cell Line ,Neoplasm Proteins ,Antigens, Neoplasm ,T-Lymphocyte Subsets ,Mutation ,Humans ,Connectin ,Immunotherapy ,Genetic Engineering ,Protein Binding - Abstract
Natural T-cell responses generally lack the potency to eradicate cancer. Enhanced affinity T-cell receptors (TCRs) provide an ideal approach to target cancer cells, with emerging clinical data showing significant promise. Nevertheless, the risk of off target reactivity remains a key concern, as exemplified in a recent clinical report describing fatal cardiac toxicity, following administration of MAGE-A3 specific TCR-engineered T-cells, mediated through cross-reactivity with an unrelated epitope from the Titin protein presented on cardiac tissue. Here, we investigated the structural mechanism enabling TCR cross-recognition of MAGE-A3 and Titin, and applied the resulting data to rationally design mutants with improved antigen discrimination, providing a proof-of-concept strategy for altering the fine specificity of a TCR towards an intended target antigen. This study represents the first example of direct molecular mimicry leading to clinically relevant fatal toxicity, mediated by a modified enhanced affinity TCR designed for cancer immunotherapy. Furthermore, these data demonstrate that self-antigens that are expressed at high levels on healthy tissue should be treated with extreme caution when designing immuno-therapeutics.
- Published
- 2015
11. β2AR antagonists and β2AR gene deletion both promote skin wound repair processes
- Author
-
Roslyn Rivkah Isseroff, Andrew P. O'Leary, Brian S. Baier, Christine E. Pullar, Sian Evans, and Gabrielle S. Le Provost
- Subjects
Keratinocytes ,Vascular Endothelial Growth Factor A ,Time Factors ,Angiogenesis ,Chick Embryo ,Biochemistry ,Mice ,0302 clinical medicine ,Adrenergic beta-2 Receptor Antagonists ,Receptors ,Aorta ,Zebrafish ,Skin ,0303 health sciences ,integumentary system ,Neovascularization, Pathologic ,3. Good health ,Cell biology ,Adrenergic ,030220 oncology & carcinogenesis ,Knockout mouse ,Original Article ,Development of treatments and therapeutic interventions ,1.1 Normal biological development and functioning ,Clinical Sciences ,Oncology and Carcinogenesis ,beta-2 ,Enzyme-Linked Immunosorbent Assay ,Dermatology ,Biology ,Dermal fibroblast ,03 medical and health sciences ,Paracrine signalling ,In vivo ,Underpinning research ,Animals ,Autocrine signalling ,Molecular Biology ,Neovascularization ,030304 developmental biology ,Inflammation ,Pathologic ,Wound Healing ,5.2 Cellular and gene therapies ,Dermatology & Venereal Diseases ,Cell Biology ,Fibroblasts ,Rats ,Immunology ,Receptors, Adrenergic, beta-2 ,Wound healing ,Ex vivo ,Gene Deletion - Abstract
Skin wound healing is a complex process requiring the coordinated, temporal orchestration of numerous cell types and biological processes to regenerate damaged tissue. Previous work has demonstrated that a functional β-adrenergic receptor autocrine/paracrine network exists in skin, but the role of β2-adrenergic receptor (β2AR) in wound healing is unknown. A range of in vitro (single-cell migration, immunoblotting, ELISA, enzyme immunoassay), ex vivo (rat aortic ring assay), and in vivo (chick chorioallantoic membrane assay, zebrafish, murine wild-type, and β2AR knockout excisional skin wound models) models were used to demonstrate that blockade or loss of β2AR gene deletion promoted wound repair, a finding that is, to our knowledge, previously unreported. Compared with vehicle-only controls, β2AR antagonism increased angiogenesis, dermal fibroblast function, and re-epithelialization, but had no effect on wound inflammation in vivo. Skin wounds in β2AR knockout mice contracted and re-epithelialized faster in the first few days of wound repair in vivo. β2AR antagonism enhanced cell motility through distinct intracellular signalling mechanisms and increased vascular endothelial growth factor secretion from keratinocytes. β2AR antagonism promoted wound repair processes in the early stages of wound repair, revealing a possible new avenue for therapeutic intervention.
- Published
- 2012
12. The HIF-1-inducible Lysyl Oxidase activates HIF-1 via the Akt pathway in a positive regulation loop and synergizes with HIF-1 in promoting tumor cell growth
- Author
-
Philippe Clézardin, Jacques Pouysségur, Blandine Deux, Caroline Reynaud, Frédéric Dayan, Jérôme Durivault, Floriane Pez, Pascal Sommer, Géraldine Aimond, Gabrielle S. Le Provost, Bastien Kaniewski, Institut de signalisation, biologie du développement et cancer (ISBDC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), Laboratoire Jean Alexandre Dieudonné (JAD), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS), Institute of Developmental Biology and Cancer (IBDC), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA)
- Subjects
Cancer Research ,Mice, Nude ,Lysyl oxidase ,Biology ,Protein-Lysine 6-Oxidase ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Gene silencing ,Animals ,Humans ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Cell Proliferation ,Regulation of gene expression ,Feedback, Physiological ,0303 health sciences ,integumentary system ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,food and beverages ,Hypoxia-Inducible Factor 1, alpha Subunit ,Immunohistochemistry ,Cell Hypoxia ,Cell biology ,Gene Expression Regulation, Neoplastic ,enzymes and coenzymes (carbohydrates) ,Oncology ,Biochemistry ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,Signal transduction ,Colorectal Neoplasms ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Adaptation to hypoxia is a driving force for tumor progression that leads to therapy resistance and poor clinical outcome. Hypoxic responses are mainly mediated by hypoxia-inducible transcription factor-1 (HIF-1). One critical HIF-1 target mediating tumor progression is lysyl oxidase (LOX), which catalyzes cross-linking of collagens and elastin in the extracellular matrix, thereby regulating tissue tensile strength. Paradoxically, LOX has been reported to be both upregulated and downregulated in cancer cells, especially in colorectal cancer. Thus, we hypothesized that LOX might regulate expression of HIF-1 to create a self-timing regulatory circuit. Using human colorectal carcinoma cell lines in which HIF-1 and LOX expression could be modulated, we showed that LOX induction enhanced HIF-1 expression, whereas LOX silencing reduced it. Mechanistic investigations revealed that LOX activated the PI3K (phosphoinositide 3-kinase)–Akt signaling pathway, thereby upregulating HIF-1α protein synthesis in a manner requiring LOX-mediated hydrogen peroxide production. Consistent with these results, cancer cell proliferation was stimulated by secreted and active LOX in an HIF-1α–dependent fashion. Furthermore, nude mice xenograft assays established that HIF-1 potentiated LOX action on tumor growth in vivo. Taken together, these findings provide compelling evidence that LOX and HIF-1 act in synergy to foster tumor formation, and they suggest that HIF-1/LOX mutual regulation is a pivotal mechanism in the adaptation of tumor cells to hypoxia. Cancer Res; 71(5); 1647–57. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
13. Lysyl Oxidase silencing impairs keratinocyte differentiation in a reconstructed-epidermis model
- Author
-
Gabrielle S, Le Provost, Romain, Debret, Valérie, Cenizo, Géraldine, Aimond, Floriane, Pez, Bastien, Kaniewski, Valérie, André, Pascal, Sommer, Laboratoire de Biologie Tissulaire et d'ingénierie Thérapeutique UMR 5305 (LBTI), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM), and BASF Beauty Care Solutions France
- Subjects
Keratinocytes ,Transglutaminases ,integumentary system ,Cell Survival ,[SDV]Life Sciences [q-bio] ,Gene Expression ,Membrane Proteins ,Cell Differentiation ,Fibroblasts ,Filaggrin Proteins ,Keratin-10 ,Coculture Techniques ,Up-Regulation ,Protein-Lysine 6-Oxidase ,Epidermal Cells ,Intermediate Filament Proteins ,Aminopropionitrile ,Humans ,RNA Interference ,Enzyme Inhibitors ,Epidermis ,Protein Precursors ,Cells, Cultured ,Cell Line, Transformed ,Cell Proliferation - Abstract
International audience; Lysyl Oxidase (LOX) is an extracellular enzyme involved in the maturation of connective tissues. It also acts in many cell types as a regulator of cell behaviour and phenotype through intracellular signalling pathways. Recently, LOX was shown to be present in human epidermis where its precise functions remain unclear. We showed here that in confluent monolayer cultures of normal human keratinocytes (KCs) and N/TERT-1-immortalized KCs, LOX expression was induced during the first differentiation steps. Moreover, the silencing of LOX by stable RNA interference disrupted the expression of early differentiation markers. In a reconstructed-epidermis model, LOX silencing did not impair the stratification process nor the formation of the first differentiated layers. However, terminal differentiation was strongly impaired, as shown by a decreased expression of late differentiation proteins and by the absence of stratum corneum. Nonetheless, inhibition of LOX enzymatic activity by β-aminopropionitrile did not affect the differentiation process. Therefore, LOX protein acts during the first steps of KC differentiation and is important for subsequent commitment into terminal differentiation. Taken together, these results suggest that a finely regulated expression of LOX is necessary for normal KC differentiation and thus for maintenance of epidermal homeostasis.
- Published
- 2010
- Full Text
- View/download PDF
14. β2-Adrenoceptor Activation Modulates Skin Wound Healing Processes to Reduce Scarring
- Author
-
Christine E. Pullar and Gabrielle S. Le Provost
- Subjects
Pathology ,medicine.medical_specialty ,Angiogenesis ,Sus scrofa ,Neovascularization, Physiologic ,Scars ,Inflammation ,Chick Embryo ,Dermatology ,Biology ,Fibroblast growth factor ,Biochemistry ,Chorioallantoic Membrane ,Dermal fibroblast ,Neovascularization ,Cicatrix ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Molecular Biology ,Cells, Cultured ,Zebrafish ,030304 developmental biology ,Focal Adhesions ,Wound Healing ,0303 health sciences ,integumentary system ,Dermis ,Cell Biology ,Fibroblasts ,Zebrafish Proteins ,Disease Models, Animal ,Chorioallantoic membrane ,Original Article ,Female ,Fibroblast Growth Factor 2 ,Receptors, Adrenergic, beta-2 ,medicine.symptom ,Wound healing - Abstract
During wound healing, excessive inflammation, angiogenesis, and differentiated human dermal fibroblast (HDF ) function contribute to scarring, whereas hyperpigmentation negatively affects scar quality. Over 100 million patients heal with a scar every year. To investigate the role of the beta 2 adrenergic receptor (β2AR) in wound scarring, the ability of beta 2 adrenergic receptor agonist (β2ARag) to alter HDF differentiation and function, wound inflammation, angiogenesis, and wound scarring was explored in HDFs, zebrafish, chick chorioallantoic membrane assay (CAM), and a porcine skin wound model, respectively. Here we identify a β2AR-mediated mechanism for scar reduction. β2ARag significantly reduced HDF differentiation, via multiple cAMP and/or fibroblast growth factor 2 or basic FGF (FGF2)-dependent mechanisms, in the presence of transforming growth factor betaβ1, reduced contractile function, and inhibited mRNA expression of a number of profibrotic markers. β2ARag also reduced inflammation and angiogenesis in zebrafish and CAMs in vivo, respectively. In Red Duroc pig full-thickness wounds, β2ARag reduced both scar area and hyperpigmentation by almost 50% and significantly improved scar quality. Indeed, mechanisms delineated in vitro and in other in vivo models were evident in the β2ARag-treated porcine scars in vivo. Both macrophage infiltration and angiogenesis were initially decreased, whereas DF function was impaired in the β2ARag-treated porcine wound bed. These data collectively reveal the potential of β2ARag to improve skin scarring.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.