19 research outputs found

    Mature oligodendrocytes actively increase in vivo cytoskeletal plasticity following CNS damage

    Get PDF
    Background: Oligodendrocytes are myelinating cells of the central nervous system which support functionally, structurally, and metabolically neurons. Mature oligodendrocytes are generally believed to be mere targets of destruction in the context of neuroinflammation and tissue damage, but their real degree of in vivo plasticity has become a matter of debate. We thus investigated the in vivo dynamic, actin-related response of these cells under different kinds of demyelinating stress. Methods: We used a novel mouse model (oLucR) expressing luciferase in myelin oligodendrocyte glycoproteinpositive oligodendrocytes under the control of a beta-actin promoter. Activity of this promoter served as surrogate for dynamics of the cytoskeleton gene transcription through recording of in vivo bioluminescence following diphtheria toxin-induced oligodendrocyte death and autoimmune demyelination. Cytoskeletal gene expression was quantified from mature oligodendrocytes directly isolated from transgenic animals through cell sorting. Results: Experimental demyelinating setups augmented oligodendrocyte-specific in vivo bioluminescence. These changes in luciferase signal were confirmed by further ex vivo analysis of the central nervous system tissue from oLucR mice. Increase in bioluminescence upon autoimmune inflammation was parallel to an oligodendrocytespecific increased transcription of beta-tubulin. Conclusions: Mature oligodendrocytes acutely increase their cytoskeletal plasticity in vivo during demyelination. They are therefore not passive players under demyelinating conditions but can rather react dynamically to external insults

    DNA damage-induced interaction between a lineage addiction oncogenic transcription factor and the MRN complex shapes a tissue-specific DNA Damage Response and cancer predisposition

    Get PDF
    Since genome instability can drive cancer initiation and progression, cells have evolved highly effective and ubiquitous DNA Damage Response (DDR) programs. However, some cells, in skin for example, are normally exposed to high levels of DNA damaging agents. Whether such high-risk cells possess lineage-specific mechanisms that tailor DNA repair to the tissue remains largely unknown. Here we show, using melanoma as a model, that the microphthalmia-associated transcription factor MITF, a lineage addition oncogene that coordinates many aspects of melanocyte and melanoma biology, plays a non-transcriptional role in shaping the DDR. On exposure to DNA damaging agents, MITF is phosphorylated by ATM/DNA-PKcs, and unexpectedly its interactome is dramatically remodelled; most transcription (co)factors dissociate, and instead MITF interacts with the MRE11-RAD50-NBS1 (MRN) complex. Consequently, cells with high MITF levels accumulate stalled replication forks, and display defects in homologous recombination-mediated repair associated with impaired MRN recruitment to DNA damage. In agreement, high MITF levels are associated with increased SNV burden in melanoma. Significantly, the SUMOylation-defective MITF-E318K melanoma predisposition mutation recapitulates the effects of ATM/DNA-PKcs-phosphorylated MITF. Our data suggest that a non-transcriptional function of a lineage-restricted transcription factor contributes to a tissue-specialised modulation of the DDR that can impact cancer initiation

    DNA damage remodels the MITF interactome to increase melanoma genomic instability

    Get PDF
    Since genome instability can drive cancer initiation and progression, cells have evolved highly effective and ubiquitous DNA damage response (DDR) programs. However, some cells (for example, in skin) are normally exposed to high levels of DNA-damaging agents. Whether such high-risk cells possess lineage-specific mechanisms that tailor DNA repair to the tissue remains largely unknown. Using melanoma as a model, we show here that the microphthalmia-associated transcription factor MITF, a lineage addition oncogene that coordinates many aspects of melanocyte and melanoma biology, plays a nontranscriptional role in shaping the DDR. On exposure to DNA-damaging agents, MITF is phosphorylated at S325, and its interactome is dramatically remodeled; most transcription cofactors dissociate, and instead MITF interacts with the MRE11–RAD50–NBS1 (MRN) complex. Consequently, cells with high MITF levels accumulate stalled replication forks and display defects in homologous recombination-mediated repair associated with impaired MRN recruitment to DNA damage. In agreement with this, high MITF levels are associated with increased single-nucleotide and copy number variant burdens in melanoma. Significantly, the SUMOylation-defective MITF-E318K melanoma predisposition mutation recapitulates the effects of DNA-PKcs-phosphorylated MITF. Our data suggest that a nontranscriptional function of a lineage-restricted transcription factor contributes to a tissue-specialized modulation of the DDR that can impact cancer initiation

    Premigratory and Migratory Neural Crest Cells are Multipotent In Vivo and The Roles of Sall4 in Melanoma

    Full text link
    The neural crest (NC) is a transient embryonic stem/progenitor cell population and a hallmark of vertebrate development. The NC is induced during neurulation at the border between the neuronal and non-neuronal ectoderm. Upon induction NC cells undergo an epithelial to mesenchymal transition (EMT), delaminate from the neural folds and migrate extensively through the developing embryo. NC cells give rise to a broad variety of cell types including the sensory and autonomic neurons of the peripheral nervous system, the myelinating Schwann cells and the melanocytes, among others. Despite the broad differentiation potential of NC cells it has been highly debated whether the NC consisted of multipotent cells or whether it was rather a heterogeneous population of restricted progenitors. In fact, although several earlier studies have described multipotency of NC cells, the existence of multipotent NC cells has been questioned both by cell culture experiments and in recent in vivo studies performed in chick embryos. In this study, we solved a longstanding controversy regarding a pivotal question in the field of stem cells. Using genetic lineage tracing in the mouse, we revealed for the first time the broad developmental potential of NC cells in a mammalian system and we demonstrated that the majority of premigratory and migratory NC cells are multipotent in vivo. Embryogenesis and tumorigenesis share several mechanisms in common and it has become more and more evident that knowledge in developmental biology can provide further insights into tumor biology. For instance, melanoma, a malignancy of NC-derived melanocytes, can exploit various NC developmental programs for disease progression. Malignant melanoma cells can indeed aberrantly activate EMT master regulators, such as members of the Snail, Twist and Zeb families, which are normally activated by NC cells during migration in the embryo. In the course of melanoma progression malignant cells brake through the basament membrane, invade the underlying mesenchyme, reach blood and lymphatic vessels and metastasize to distant organs. Moreover, similar to NC cells, melanoma cells can possess multipotency features and be able to express different lineage markers. We discovered that the zinc-finger transcription factor Sall4 is expressed in NC cells and it is downregulated upon differentiation. Sall4 is a crucial factor for the maintenance of self-renewal and pluripotency of embryonic stem cells. Moreover, SALL4 has been associated with tumorigenesis and to worse patient outcome in various cancer types. However, whether SALL4 may also play a role in melanoma 2 formation and progression has not been addressed so far. We observed that SALL4 was mostly expressed in human proliferative melanoma cell lines, while it was absent in more invasive melanoma cell lines or upon EMT induction. Interestingly, SALL4 downregulation induced, in turn, an EMT signature in a proliferative melanoma cell line, suggesting that there may be a regulatory feedback loop. In vivo we induced Sall4 loss in the Tyr::NRasQ61K Ink4a-/- Tyr::CreERT2 Sall4lox/lox R26R::GFP melanoma mouse model and could observed that primary tumor formation was impaired. However, Sall4 loss was linked to a reduced survival of the knock out animals and recombined cells were detected in lymph nodes and in some lungs. Further investigations now urge to be performed to prove whether Sall4 loss is necessary and sufficient for EMT induction

    Harmony in chaos: understanding cancer through the lenses of developmental biology

    No full text
    When we think about cancer, the link to development might not immediately spring to mind. Yet, many foundational concepts in cancer biology trace their roots back to developmental processes. Several defining traits of cancer were indeed initially observed and studied within developing embryos. As our comprehension of embryonic mechanisms deepens, it not only illuminates how and why cancer cells hijack these processes but also spearheads the emergence of innovative technologies for modeling and comprehending tumor biology. Among these technologies are stem cell‐based models, made feasible through our grasp of fundamental mechanisms related to embryonic development. The intersection between cancer and stem cell research is evolving into a tangible synergy that extends beyond the concepts of cancer stem cells and cell‐of‐origin, offering novel tools to unravel the mechanisms of cancer initiation and progression

    Ezh2 is required for neural crest-derived cartilage and bone formation

    Full text link
    The emergence of craniofacial skeletal elements, and of the jaw in particular, was a crucial step in the evolution of higher vertebrates. Most facial bones and cartilage are generated during embryonic development by cranial neural crest cells, while an osteochondrogenic fate is suppressed in more posterior neural crest cells. Key players in this process are Hox genes, which suppress osteochondrogenesis in posterior neural crest derivatives. How this specific pattern of osteochondrogenic competence is achieved remains to be elucidated. Here we demonstrate that Hox gene expression and osteochondrogenesis are controlled by epigenetic mechanisms. Ezh2, which is a component of polycomb repressive complex 2 (PRC2), catalyzes trimethylation of lysine 27 in histone 3 (H3K27me3), thereby functioning as transcriptional repressor of target genes. Conditional inactivation of Ezh2 does not interfere with localization of neural crest cells to their target structures, neural development, cell cycle progression or cell survival. However, loss of Ezh2 results in massive derepression of Hox genes in neural crest cells that are usually devoid of Hox gene expression. Accordingly, craniofacial bone and cartilage formation is fully prevented in Ezh2 conditional knockout mice. Our data indicate that craniofacial skeleton formation in higher vertebrates is crucially dependent on epigenetic regulation that keeps in check inhibitors of an osteochondrogenic differentiation program

    HDAC1 and HDAC2 Control the specification of neural crest cells into peripheral glia

    Get PDF
    Schwann cells, the myelinating glia of the peripheral nervous system (PNS), originate from multipotent neural crest cells that also give rise to other cells, including neurons, melanocytes, chondrocytes, and smooth muscle cells. The transcription factor Sox10 is required for peripheral glia specification. However, all neural crest cells express Sox10 and the mechanisms directing neural crest cells into a specific lineage are poorly understood. We show here that histone deacetylases 1 and 2 (HDAC1/2) are essential for the specification of neural crest cells into Schwann cell precursors and satellite glia, which express the early determinants of their lineage myelin protein zero (P0) and/or fatty acid binding protein 7 (Fabp7). In neural crest cells, HDAC1/2 induced expression of the transcription factor Pax3 by binding and activating the Pax3 promoter. In turn, Pax3 was required to maintain high Sox10 levels and to trigger expression of Fabp7. In addition, HDAC1/2 were bound to the P0 promoter and activated P0 transcription. Consistently, in vivo genetic deletion of HDAC1/2 in mouse neural crest cells led to strongly decreased Sox10 expression, no detectable Pax3, virtually no satellite glia, and no Schwann cell precursors in dorsal root ganglia and peripheral nerves. Similarly, in vivo ablation of Pax3 in the mouse neural crest resulted in strongly reduced expression of Sox10 and Fabp7. Therefore, by controlling the expression of Pax3 and the concerted action of Pax3 and Sox10 on their target genes, HDAC1/2 direct the specification of neural crest cells into peripheral glia

    Epigenetic control of melanoma cell invasiveness by the stem cell factor SALL4

    Full text link
    Melanoma cells rely on developmental programs during tumor initiation and progression. Here we show that the embryonic stem cell (ESC) factor Sall4 is re-expressed in the Tyr::NrasQ61K^{Q61K}; Cdkn2a−/−^{−/−} melanoma model and that its expression is necessary for primary melanoma formation. Surprisingly, while Sall4 loss prevents tumor formation, it promotes micrometastases to distant organs in this melanoma-prone mouse model. Transcriptional profiling and in vitro assays using human melanoma cells demonstrate that SALL4 loss induces a phenotype switch and the acquisition of an invasive phenotype. We show that SALL4 negatively regulates invasiveness through interaction with the histone deacetylase (HDAC) 2 and direct co-binding to a set of invasiveness genes. Consequently, SALL4 knock down, as well as HDAC inhibition, promote the expression of an invasive signature, while inhibition of histone acetylation partially reverts the invasiveness program induced by SALL4 loss. Thus, SALL4 appears to regulate phenotype switching in melanoma through an HDAC2-mediated mechanism

    Lipid droplets are a metabolic vulnerability in melanoma

    Get PDF
    Abstract Melanoma exhibits numerous transcriptional cell states including neural crest-like cells as well as pigmented melanocytic cells. How these different cell states relate to distinct tumorigenic phenotypes remains unclear. Here, we use a zebrafish melanoma model to identify a transcriptional program linking the melanocytic cell state to a dependence on lipid droplets, the specialized organelle responsible for lipid storage. Single-cell RNA-sequencing of these tumors show a concordance between genes regulating pigmentation and those involved in lipid and oxidative metabolism. This state is conserved across human melanoma cell lines and patient tumors. This melanocytic state demonstrates increased fatty acid uptake, an increased number of lipid droplets, and dependence upon fatty acid oxidative metabolism. Genetic and pharmacologic suppression of lipid droplet production is sufficient to disrupt cell cycle progression and slow melanoma growth in vivo. Because the melanocytic cell state is linked to poor outcomes in patients, these data indicate a metabolic vulnerability in melanoma that depends on the lipid droplet organelle
    corecore