Your browser doesn't support javascript.
loading
NRF2 Mediates Cellular Resistance to Transformation, Radiation, and Inflammation in Mice.
Schaue, Dörthe; Micewicz, Ewa D; Ratikan, Josephine A; Iwamoto, Keisuke S; Vlashi, Erina; McDonald, J Tyson; McBride, William H.
Afiliación
  • Schaue D; Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095-1714, USA.
  • Micewicz ED; Biotts S.A., Ul. Wroclawska 44C, 55-040 Bielany Wroclawskie, Poland.
  • Ratikan JA; Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095-1714, USA.
  • Iwamoto KS; Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095-1714, USA.
  • Vlashi E; Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095-1714, USA.
  • McDonald JT; Department of Radiation Medicine, School of Medicine, Georgetown University, Washington, DC 20057, USA.
  • McBride WH; Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095-1714, USA.
Antioxidants (Basel) ; 11(9)2022 Aug 25.
Article en En | MEDLINE | ID: mdl-36139722
ABSTRACT
Nuclear factor erythroid 2-related factor 2 (NRF2) is recognized as a master transcription factor that regulates expression of numerous detoxifying and antioxidant cytoprotective genes. In fact, models of NRF2 deficiency indicate roles not only in redox regulation, but also in metabolism, inflammatory/autoimmune disease, cancer, and radioresistancy. Since ionizing radiation (IR) generates reactive oxygen species (ROS), it is not surprising it activates NRF2 pathways. However, unexpectedly, activation is often delayed for many days after the initial ROS burst. Here, we demonstrate that, as assayed by γ-H2AX staining, rapid DNA double strand break (DSB) formation by IR in primary mouse Nrf2-/- MEFs was not affected by loss of NRF2, and neither was DSB repair to any great extent. In spite of this, basal and IR-induced transformation was greatly enhanced, suggesting that NRF2 protects against late IR-induced genomic instability, at least in murine MEFs. Another possible IR- and NRF2-related event that could be altered is inflammation and NRF2 deficiency increased IR-induced NF-κB pro-inflammatory responses mostly late after exposure. The proclivity of NRF2 to restrain inflammation is also reflected in the reprogramming of tumor antigen-specific lymphocyte responses in mice where Nrf2 k.o. switches Th2 responses to Th1 polarity. Delayed NRF2 responses to IR may be critical for the immune transition from prooxidant inflammation to antioxidant healing as well as in driving cellular radioresistance and survival. Targeting NRF2 to reprogram immunity could be of considerable therapeutic benefit in radiation and immunotherapy.
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: Antioxidants (Basel) Año: 2022 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Bases de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: Antioxidants (Basel) Año: 2022 Tipo del documento: Article País de afiliación: Estados Unidos