27 research outputs found

    Identification of the kinetochore generated "Wait- Anaphase" signal of the mitotic checkpoint

    No full text
    To ensure accurate segregation, the major cell cycle control mechanism in mitosis, the mitotic checkpoint, delays anaphase onset until all chromosomes have properly attached to spindle microtubules. This is achieved through the production of a "wait anaphase" inhibitor(s) that blocks recognition of cyclin B and securin by Cdc20- activated APC/C, an E3 ubiquitin ligase which targets them for destruction. Using physiologically relevant levels of Mad2, Bub3, BubR1, and Cdc20, unattached kinetochores on purified chromosomes are demonstrated to catalyze generation of a soluble Cdc20 inhibitor or inhibition of Cdc20 already bound to APC/C. Antibody inhibition of Mad1 and dimerization deficient Mad2 are used to demonstrate that the chromosome-produced inhibitor requires both recruitment of Mad2 by Mad1 stably bound at unattached kinetochores and dimerization competent Mad2. By acting directly on Mad2, but not BubR1, purified chromosomes promote BubR1 binding to Cdc20 and APC/C, supporting a model in which immobilized Mad1/Mad2 at kinetochores provides a template for initial assembly of Mad2 bound to Cdc20 that is then converted to BubR1-Cdc20 as sequentially produced mitotic checkpoint inhibitor

    Epidermal development, growth control, and homeostasis in the face of centrosome amplification

    No full text
    As nucleators of the mitotic spindle and primary cilium, centrosomes play crucial roles in equal segregation of DNA content to daughter cells, coordination of growth and differentiation, and transduction of homeostatic cues. Whereas the majority of mammalian cells carry no more than two centrosomes per cell, exceptions to this rule apply in certain specialized tissues and in select disease states, including cancer. Centrosome amplification, or the condition of having more than two centrosomes per cell, has been suggested to contribute to instability of chromosomes, imbalance in asymmetric divisions, and reorganization of tissue architecture; however, the degree to which these conditions are a direct cause of or simply a consequence of human disease is poorly understood. Here we addressed this issue by generating a mouse model inducing centrosome amplification in a naturally proliferative epithelial tissue by elevating Polo-like kinase 4 (Plk4) expression in the skin epidermis. By altering centrosome numbers, we observed multiciliated cells, spindle orientation errors, and chromosome segregation defects within developing epidermis. None of these defects was sufficient to impart a proliferative advantage within the tissue, however. Rather, impaired mitoses led to p53-mediated cell death and contributed to defective growth and stratification. Despite these abnormalities, mice remained viable and healthy, although epidermal cells with centrosome amplification were still appreciable. Moreover, these abnormalities were insufficient to disrupt homeostasis and initiate or enhance tumorigenesis, underscoring the powerful surveillance mechanisms in the skin
    corecore