Your browser doesn't support javascript.
loading
Boolean model of growth signaling, cell cycle and apoptosis predicts the molecular mechanism of aberrant cell cycle progression driven by hyperactive PI3K.
Sizek, Herbert; Hamel, Andrew; Deritei, Dávid; Campbell, Sarah; Ravasz Regan, Erzsébet.
Afiliación
  • Sizek H; Biochemistry and Molecular Biology, The College of Wooster, Wooster, OH, United States of America.
  • Hamel A; Biochemistry and Molecular Biology, The College of Wooster, Wooster, OH, United States of America.
  • Deritei D; Department of Physics, Pennsylvania State University, State College, PA, United States of America.
  • Campbell S; Department of Network and Data Science, Central European University, Budapest, Hungary.
  • Ravasz Regan E; Biochemistry and Molecular Biology, The College of Wooster, Wooster, OH, United States of America.
PLoS Comput Biol ; 15(3): e1006402, 2019 03.
Article en En | MEDLINE | ID: mdl-30875364
ABSTRACT
The PI3K/AKT signaling pathway plays a role in most cellular functions linked to cancer progression, including cell growth, proliferation, cell survival, tissue invasion and angiogenesis. It is generally recognized that hyperactive PI3K/AKT1 are oncogenic due to their boost to cell survival, cell cycle entry and growth-promoting metabolism. That said, the dynamics of PI3K and AKT1 during cell cycle progression are highly nonlinear. In addition to negative feedback that curtails their activity, protein expression of PI3K subunits has been shown to oscillate in dividing cells. The low-PI3K/low-AKT1 phase of these oscillations is required for cytokinesis, indicating that oncogenic PI3K may directly contribute to genome duplication. To explore this, we construct a Boolean model of growth factor signaling that can reproduce PI3K oscillations and link them to cell cycle progression and apoptosis. The resulting modular model reproduces hyperactive PI3K-driven cytokinesis failure and genome duplication and predicts the molecular drivers responsible for these failures by linking hyperactive PI3K to mis-regulation of Polo-like kinase 1 (Plk1) expression late in G2. To do this, our model captures the role of Plk1 in cell cycle progression and accurately reproduces multiple effects of its loss G2 arrest, mitotic catastrophe, chromosome mis-segregation / aneuploidy due to premature anaphase, and cytokinesis failure leading to genome duplication, depending on the timing of Plk1 inhibition along the cell cycle. Finally, we offer testable predictions on the molecular drivers of PI3K oscillations, the timing of these oscillations with respect to division, and the role of altered Plk1 and FoxO activity in genome-level defects caused by hyperactive PI3K. Our model is an important starting point for the predictive modeling of cell fate decisions that include AKT1-driven senescence, as well as the non-intuitive effects of drugs that interfere with mitosis.
Asunto(s)

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Simulación por Computador / Transducción de Señal / Ciclo Celular / Apoptosis / Fosfatidilinositol 3-Quinasas Tipo de estudio: Prognostic_studies / Risk_factors_studies Límite: Humans Idioma: En Revista: PLoS Comput Biol Asunto de la revista: BIOLOGIA / INFORMATICA MEDICA Año: 2019 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Simulación por Computador / Transducción de Señal / Ciclo Celular / Apoptosis / Fosfatidilinositol 3-Quinasas Tipo de estudio: Prognostic_studies / Risk_factors_studies Límite: Humans Idioma: En Revista: PLoS Comput Biol Asunto de la revista: BIOLOGIA / INFORMATICA MEDICA Año: 2019 Tipo del documento: Article País de afiliación: Estados Unidos