15 research outputs found

    CD95 recruits PLCγ1 to trigger a calcium response promoting Th17 accumulation in inflamed organs of lupus mice

    Get PDF
    CD95 ligand (CD95L) is expressed by immune cells and triggers apoptotic death. Metalloprotease-cleaved CD95L (cl-CD95L) is released into the bloodstream but does not trigger apoptotic signaling. Hence, the pathophysiological role of cl-CD95L remains unclear. We observed that skin-derived endothelial cells from systemic lupus erythematosus (SLE) patients expressed CD95L, and that after cleavage, cl-CD95L promoted T helper 17 (Th17) lymphocyte transmigration across the endothelial barrier at the expense of T regulatory cells. T cell migration relied on a direct interaction between the CD95 domain called calcium-inducing domain (CID) and the Src homology 3 domain of phospholipase Cγ1. Th17 cells stimulated with cl-CD95L produced sphingosine-1-phosphate (S1P), which promoted endothelial transmigration by activating the S1P receptor 3. We generated a cell-penetrating CID peptide that prevented Th17 cell transmigration and alleviated clinical symptoms in lupus mice. Therefore, neutralizing the CD95 non-apoptotic signaling pathway may be attractive therapeutic approach for SLE treatment

    CD95-mediated calcium signaling promotes T helper 17 trafficking to inflamed organs in lupus-prone mice

    Get PDF
    CD95 ligand (CD95L) is expressed by immune cells and triggers apoptotic death. Metalloprotease-cleaved CD95L (cl-CD95L) is released into the bloodstream but does not trigger apoptotic signaling. Hence, the pathophysiological role of cl-CD95L remains unclear. We observed that skin-derived endothelial cells from systemic lupus erythematosus (SLE) patients expressed CD95L and that after cleavage, cl-CD95L promoted T helper 17 (Th17) lymphocyte transmigration across the endothelial barrier at the expense of T regulatory cells. T cell migration relied on a direct interaction between the CD95 domain called calcium-inducing domain (CID) and the Src homology 3 domain of phospholipase Cγ1. Th17 cells stimulated with cl-CD95L produced sphingosine-1-phosphate (S1P), which promoted endothelial transmigration by activating the S1P receptor 3. We generated a cell-penetrating CID peptide that prevented Th17 cell transmigration and alleviated clinical symptoms in lupus mice. Therefore, neutralizing the CD95 non-apoptotic signaling pathway could be an attractive therapeutic approach for SLE treatment

    Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models

    Get PDF
    Cytotoxic chemotherapy is an effective treatment for invasive breast cancer. However, experimental studies in mice also suggest that chemotherapy has pro-metastatic effects. Primary tumours release extracellular vesicles (EVs), including exosomes, that can facilitate the seeding and growth of metastatic cancer cells in distant organs, but the effects of chemotherapy on tumour-derived EVs remain unclear. Here we show that two classes of cytotoxic drugs broadly employed in pre-operative (neoadjuvant) breast cancer therapy, taxanes and anthracyclines, elicit tumour-derived EVs with enhanced pro-metastatic capacity. Chemotherapy-elicited EVs are enriched in annexin A6 (ANXA6), a Ca2+-dependent protein that promotes NF-κB-dependent endothelial cell activation, Ccl2 induction and Ly6C+CCR2+ monocyte expansion in the pulmonary pre-metastatic niche to facilitate the establishment of lung metastasis. Genetic inactivation of Anxa6 in cancer cells or Ccr2 in host cells blunts the prometastatic effects of chemotherapy-elicited EVs. ANXA6 is detected, and potentially enriched, in the circulating EVs of breast cancer patients undergoing neoadjuvant chemotherapy

    CD95/CD95L signaling pathway : implications in Systemic Lupus Erythematosus and development of targeted therapeutic tools

    No full text
    Le Lupus Erythémateux Systémique est une pathologie inflammatoire chronique. L’étiologie de cette maladie auto-immune est encore méconnue bien que certains facteurs génétiques et environnementaux aggravants aient été mis en évidence. Les traitements proposés aux patients ont pour but de réduire les symptômes et aucun remède curatif n’a encore été mis au point. Nous avons observé de forts taux de sCD95L dans le sérum de patients atteints de LES comparé à celui de sujets sains. Nos données indiquent que ce facteur soluble agit comme une cytokine pro-inflammatoire et promeut la transmigration des lymphocytes T CD4+ Th17 dans les organes, au détriment des lymphocytes T régulateurs (Treg). L’accumulation de ces cellules Th17 est responsable du maintien d’une réponse inflammatoire chronique chez les patients lupiques. Nous mettons en évidence qu’il existe une interaction directe entre le récepteur CD95 et la Phospholipase Cγ1, par l’identification du Calcium Inducing Domain impliqué dans ce recrutement. L’identification du couple CD95/CD95L comme acteur aggravant le LES et la mise en évidences des mécanismes cellulaires et moléculaires sous-jacents nous ont conduit à l’élaboration de stratégies thérapeutiques innovantes. En collaboration avec les chimistes et modélisateurs de notre Unité, nous avons généré une petite librairie d’inhibiteurs, composée de peptides et peptidomimétiques sélectifs. Parmi ces composés, le TAT-CID est une protéine piège comprenant la zone d’interaction de CD95 (domaine CID des aa 175 à 210) à la PLCγ1. L’injection de ce peptide dans un modèle de souris lupiques restaure la fonction biologique rénale de ces souris et diminue la production d’auto-anticorps (anti-DNA) et de complexes immuns, marqueurs biologiques associés à la progression de la pathologie. En parallèle, le criblage d’une librairie chimique commerciale constituée de médicaments approuvés par la FDA et l’EMA a permis d’identifier un inhibiteur efficace de notre interaction. In vivo, ce composé est capable de réduire drastiquement les signes cliniques de la pathologie lupique. Ces nouvelles données enrichissent notre compréhension du processus mis en place par le système CD95/CD95L dans l’aggravation du LES, et nous permettent de proposer des outils thérapeutiques interessants. Ces molécules pourraient représenter de nouvelles options thérapeutiques originales et attrayantes pour prévenir l’inflammation dans les pathologies inflammatoires chroniques.Systemic Lupus Erythematosus (SLE) is a chronic inflammatory disease. The etiology of this autoimmune disease is still unknown although some aggravating genetic and environmental factors have been identified. The treatments used for patients are intended to reduce the symptoms and no curative one has been developed yet. We observed high levels of sCD95L in the serum of patients with SLE compared to healthy subjects. Our data indicate that this soluble factor acts as a pro-inflammatory cytokine and promotes the transmigration of CD4 + Th17 T lymphocytes to the detriment of regulatory T lymphocytes (Treg) in the enflammed organs of patients. The accumulation of these Th17 cells is responsible for maintaining a chronic inflammatory response in lupus patients. We show that there is a direct interaction between the CD95 receptor and the phospholipase Cγ1, by the identification of the Calcium Inducing Domain involved in this recruitment. The identification of the CD95/CD95L couple as an aggravating factor in SLE context and the underlying cellular and molecular mechanisms led us to the development of innovative therapeutic strategies. In collaboration with the chemists and modellers of our research Unit, we have generated a small library of inhibitors, composed of selective peptides and peptidomimetics. Among these compounds, TAT-CID is a decoy peptide comprising the interaction zone of CD95 (CID domain 175 to 210 aa) with PLCγ1. Repeated treatments of lupus-prone mice with this peptide restore the biological function of these mice and decrease the production of autoantibodies (anti-DNA) and immune complexes, biological markers associated with the progression of the pathology. In parallel, the screening of a commercial chemical library consisting of FDA and EMA-approved drugs allowed us to identify an effective inhibitor of our targeted interaction. In vivo, this compound is able to drastically reduce the clinical signs of lupus pathology. These new data enrich our understanding of the process implemented by the CD95/CD95L system in the aggravation of SLE, and allow us to propose interesting therapeutic tools. These molecules could represent novel and attractive therapeutic options for preventing inflammation in chronic inflammatory pathologies

    Boyden Chamber Assay to Study of Cell Migration Induced by Metalloprotease Cleaved-CD95L

    No full text
    International audienceCD95 receptor, also called Fas or Apo-1, is a member of the tumor necrosis factor receptors (TNF-R) superfamily (Itoh and Nagata, J Biol Chem 268:10932-10937, 1993). Its cognate ligand, CD95L, is a transmembrane cytokine, which can be cleaved by metalloproteases (Matsuno et al., J Rheumatol 28:22-28, 2001; Vargo-Gogola et al., Arch Biochem Biophys 408:155-161, 2002; Kiaei et al., Exp Neurol 205:74-81, 2007; Schulte et al., Cell Death Differ 14:1040-1049, 2007) releasing a soluble ligand into the bloodstream. Recent work has shown that this metalloprotease-cleaved CD95L (cl-CD95L) is involved in carcinogenesis (Malleter et al., Cancer Res 73:6711-6721, 2013). Cl-CD95L also fuels the inflammatory process in patients affected by systemic lupus erythematosus by promoting the accumulation of activated T lymphocytes in enflamed organs (Tauzin et al., PLoS Biol 9:e1001090, 2011). This chapter aims at describing the methodology used to measure the chemoattractive effect of cl-CD95L on human cancer cells and lymphocytes

    CD95/Fas, Non-Apoptotic Signaling Pathways, and Kinases

    No full text
    Endothelial cells lining new blood vessels that develop during inflammatory disorders or cancers act as doors that either allow or block access to the tumor or inflamed organ. Recent data show that these endothelial cells in cancer tissues and inflamed tissues of lupus patients overexpress CD95L, the biological role of which is a subject of debate. The receptor CD95 (also named Fas or apoptosis antigen 1) belongs to the tumor necrosis factor (TNF) receptor superfamily. Its cognate ligand, CD95L, is implicated in immune homeostasis and immune surveillance. Because mutations of this receptor or its ligand lead to autoimmune disorders such as systemic lupus erythematosus (SLE) and cancers, CD95 and CD95L were initially thought to play a role in immune homeostasis and tumor elimination via apoptotic signaling pathways. However, recent data reveal that CD95 also evokes non-apoptotic signals, promotes inflammation, and contributes to carcinogenesis; therefore, it is difficult to dissect its apoptotic effects from its non-apoptotic effects during pathogenesis of disease. CD95L is cleaved by metalloproteases and so exists in two different forms: a transmembrane form and a soluble ligand (s-CD95L). We recently observed that the soluble ligand is overexpressed in serum from patients with triple-negative breast cancer or SLE, in whom it contributes to disease severity by activating non-apoptotic signaling pathways and promoting either metastatic dissemination or accumulation of certain T cell subsets in damaged organs. Here, we discuss the roles of CD95 in modulating immune functions via induction of mainly non-apoptotic signaling pathways

    Tumor analysis: freeze–thawing cycle of triple-negative breast cancer cells alters tumor CD24/CD44 profiles and the percentage of tumor-infiltrating immune cells

    No full text
    Abstract Objective The use of novel methods to characterize living tumor cells relies on well-conceived biobanks. Herein, we raised the question of whether the composition of fresh and freeze/thawed dissociated tumor samples is comparable in terms of quantitative and qualitative profiling. Results Breast cancer is a heterogeneous disease, encompassing luminal A and B, basal/triple-negative breast cancer (TNBC), and ERBB2-like tumors. We examined living cells dissociated from TNBC and found that a classical freeze/thaw protocol leads to a marked reduction in the number of CD45−CD44LowCD24Low tumor cells. This, in turn, changed the percentage of tumor cells with certain CD44/CD24 expression patterns and changed the percentage of tumor-infiltrating immune cells. These cryopreservation-driven alterations in cellular phenotype make it impossible to compare fresh and frozen samples from the same patient directly. Moreover, the freeze/thaw process changed the transcriptomic signatures of triple-negative cancer stem cells in such a manner that hierarchical clustering no longer ranked them according to expected inter-individual differences. Overall, this study suggests that all analyses of living tumor cells should be conducted only using freshly dissociated tumors if we are to generate a robust scoring system for prognostic/predictive markers
    corecore