59 research outputs found

    Segmental Duplications Arise from Pol32-Dependent Repair of Broken Forks through Two Alternative Replication-Based Mechanisms

    Get PDF
    The propensity of segmental duplications (SDs) to promote genomic instability is of increasing interest since their involvement in numerous human genomic diseases and cancers was revealed. However, the mechanism(s) responsible for their appearance remain mostly speculative. Here, we show that in budding yeast, replication accidents, which are most likely transformed into broken forks, play a causal role in the formation of SDs. The Pol32 subunit of the major replicative polymerase Polδ is required for all SD formation, demonstrating that SDs result from untimely DNA synthesis rather than from unequal crossing-over. Although Pol32 is known to be required for classical (Rad52-dependant) break-induced replication, only half of the SDs can be attributed to this mechanism. The remaining SDs are generated through a Rad52-independent mechanism of template switching between microsatellites or microhomologous sequences. This new mechanism, named microhomology/microsatellite-induced replication (MMIR), differs from all known DNA double-strand break repair pathways, as MMIR-mediated duplications still occur in the combined absence of homologous recombination, microhomology-mediated, and nonhomologous end joining machineries. The interplay between these two replication-based pathways explains important features of higher eukaryotic genomes, such as the strong, but not strict, association between SDs and transposable elements, as well as the frequent formation of oncogenic fusion genes generating protein innovations at SD junctions

    Analysis of the Globose Basal Cell Compartment in Rat Olfactory Epithelium Using GBC-1, a New Monoclonal Antibody against Globose Basal Cells

    No full text
    The olfactory epithelium (OE) supports ongoing neurogenesis throughout life and regenerates after experimental injury. Although evidence indicates that proliferative cells within the population of globose (light) basal cells (GBCs) give rise to new neurons, little is known about the biology of GBCs. Because GBCs have been identifiable only by an absence of staining with reagents that mark other cell types in the epithelium, we undertook to isolate antibodies that specifically react against GBCs and to characterize the GBC compartment in normal and regenerating OE. Monoclonal antibodies were produced using mice immunized with regenerating rat OE, and a monoclonal antibody designated GBC-1, which reacts against GBCs of the rat OE, was isolated. In immunohistochemical analyses, antibody GBC-1 was found to label GBCs in both normal and regenerating OE as we are currently able to define them: basal cells that incorporate the mitotic tracer bromodeoxyuridine and fail to express cytokeratins or neural cell adhesion molecule. During epithelial reconstitution after direct experimental injury with methyl bromide, expression of the GBC-1 antigen overlaps to a limited extent with expression of cell-specific markers for horizontal basal cells, Bowman’s gland and sustentacular cells, and neurons. These data suggest that GBC-1 may mark multipotent cells residing in the GBC compartment, which are prominent during regeneration. However, a limited number of cells in the regenerating OE with other phenotypic characteristics of GBCs lack expression of the GBC-1 antigen. GBC-1 has revealed novel aspects of GBC biology and will be useful for studying the process of olfactory neurogenesis

    Differential expression of components of the retinoic acid signaling pathway in the adult mouse olfactory epithelium

    No full text
    Position within a tissue often correlates with cellular phenotype, for example, differential expression of odorant receptors and cell adhesion molecules across the olfactory mucosa (OM). The association between position and phenotype is often paralleled by gradations in the concentration of retinoic acid (RA), caused by differential expression of the RA synthetic enzymes, the retinaldehyde dehydrogenases (RALDH). We show here that RALDH-1, -2, and -3 are enriched in the sustentacular cells, deep fibroblasts of the lamina propria, and the superficial fibroblasts, respectively, of the ventral and lateral OM as compared to the dorsomedial OM. The shift from high to low expression of the RALDHs matches the boundary defined by the differential expression of OCAM/mamFasII. Further, we found that RA-binding proteins are expressed in the epithelium overlying the RALDH-3 expressing fibroblasts of the lamina propria. Both findings suggest that local alterations in RA concentration may be more important than a gradient of RA across the epithelial plane, per se. In addition, RALDH-3 is found in a small population of basal cells in the ventral and lateral epithelium, which expand and contribute to the neuronal lineage following MeBr lesion. Indeed, transduction with a retrovirus expressing a dominant negative form of retinoic acid receptor type alpha blocks the reappearance of mature, olfactory marker protein (OMP) (+) olfactory neurons as compared to empty vector. These results support the notion of a potential role for RA, both in maintaining the spatial organization of the normal olfactory epithelium and in reestablishing the neuronal population during regeneration after injury

    Ascl1 (Mash1) knockout perturbs differentiation of nonneuronal cells in olfactory epithelium

    Get PDF
    The embryonic olfactory epithelium (OE) generates only a very few olfactory sensory neurons when the basic helix-loop-helix transcription factor, ASCL1 (previously known as MASH1) is eliminated by gene mutation. We have closely examined the structure and composition of the OE of knockout mice and found that the absence of neurons dramatically affects the differentiation of multiple other epithelial cell types as well. The most prominent effect is observed within the two known populations of stem and progenitor cells of the epithelium. The emergence of horizontal basal cells, a multipotent progenitor population in the adult epithelium, is anomalous in the Ascl1 knockout mice. The differentiation of globose basal cells, another multipotent progenitor population in the adult OE, is also aberrant. All of the persisting globose basal cells are marked by SOX2 expression, suggesting a prominent role for SOX2 in progenitors upstream of Ascl1. However, NOTCH1-expressing basal cells are absent from the knockout; since NOTCH1 signaling normally acts to suppress Ascl1 via HES1 and drives sustentacular (Sus) cell differentiation during adult epithelial regeneration, its absence suggests reciprocity between neurogenesis and the differentiation of Sus cells. Indeed, the Sus cells of the mutant mice express a markedly lower level of HES1, strengthening that notion of reciprocity. Duct/gland development appears normal. Finally, the expression of cKIT by basal cells is also undetectable, except in those small patches where neurogenesis escapes the effects of Ascl1 knockout and neurons are born. Thus, persistent neurogenic failure distorts the differentiation of multiple other cell types in the olfactory epithelium
    corecore