Your browser doesn't support javascript.
loading
Combinatorial regulation by ERK1/2 and CK1δ protein kinases leads to HIF-1α association with microtubules and facilitates its symmetrical distribution during mitosis.
Arseni, Christina; Samiotaki, Martina; Panayotou, George; Simos, George; Mylonis, Ilias.
  • Arseni C; Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500, Larissa, Greece.
  • Samiotaki M; Institute for Bio-Innovation, BSRC "Alexander Fleming", 16672, Vari, Greece.
  • Panayotou G; Institute for Bio-Innovation, BSRC "Alexander Fleming", 16672, Vari, Greece.
  • Simos G; Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500, Larissa, Greece. simos@med.uth.gr.
  • Mylonis I; Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, Canada. simos@med.uth.gr.
Cell Mol Life Sci ; 81(1): 72, 2024 Feb 01.
Article en En | MEDLINE | ID: mdl-38300329
ABSTRACT
Hypoxia-inducible factor-1 (HIF-1) is the key transcriptional mediator of the cellular response to hypoxia and is also involved in cancer progression. Regulation of its oxygen-sensitive HIF-1α subunit involves post-translational modifications that control its stability, subcellular localization, and activity. We have previously reported that phosphorylation of the HIF-1α C-terminal domain by ERK1/2 promotes HIF-1α nuclear accumulation and stimulates HIF-1 activity while lack of this modification triggers HIF-1α nuclear export and its association with mitochondria. On the other hand, modification of the N-terminal domain of HIF-1α by CK1δ impairs HIF-1 activity by obstructing the formation of a HIF-1α/ARNT heterodimer. Investigation of these two antagonistic events by expressing double phospho-site mutants in HIF1A-/- cells under hypoxia revealed independent and additive phosphorylation effects that can create a gradient of HIF-1α subcellular localization and transcriptional activity. Furthermore, modification by CK1δ caused mitochondrial release of the non-nuclear HIF-1α form and binding to microtubules via its N-terminal domain. In agreement, endogenous HIF-1α could be shown to co-localize with mitotic spindle microtubules and interact with tubulin, both of which were inhibited by CK1δ silencing or inhibition. Moreover, CK1δ expression was necessary for equal partitioning of mother cell-produced HIF-1α to the daughter cell nuclei at the end of mitosis. Overall, our results suggest that phosphorylation by CK1δ stimulates the association of non-nuclear HIF-1α with microtubules, which may serve as a means to establish a symmetric distribution of HIF-1α during cell division under low oxygen conditions.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Proteínas Quinasas / Sistema de Señalización de MAP Quinasas Tipo de estudio: Risk_factors_studies Límite: Humans Idioma: En Año: 2024 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Proteínas Quinasas / Sistema de Señalización de MAP Quinasas Tipo de estudio: Risk_factors_studies Límite: Humans Idioma: En Año: 2024 Tipo del documento: Article