Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity

  1. Scott W. Lowe1,7,8,10
  1. 1Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
  2. 2Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
  3. 3Ministry of Education (MOE) Key Laboratory of Bioinformatics, Bioinformatics Division, Tsinghua National Laboratory for Information Science and Technology (TNLIST), Department of Automation, Tsinghua University, Beijing 10084, China;
  4. 4Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, USA;
  5. 5Calibrant, Gaithersburg, Maryland 20878, USA;
  6. 6Department of Laboratory Medicine, Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA;
  7. 7Howard Hughes Medical Institute, Cold Spring Harbor, New York 11724, USA;
  8. 8Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
    1. 9 These authors contributed equally to this work.

    Abstract

    Cellular senescence acts as a potent barrier to tumorigenesis and contributes to the anti-tumor activity of certain chemotherapeutic agents. Senescent cells undergo a stable cell cycle arrest controlled by RB and p53 and, in addition, display a senescence-associated secretory phenotype (SASP) involving the production of factors that reinforce the senescence arrest, alter the microenvironment, and trigger immune surveillance of the senescent cells. Through a proteomics analysis of senescent chromatin, we identified the nuclear factor-κB (NF-κB) subunit p65 as a major transcription factor that accumulates on chromatin of senescent cells. We found that NF-κB acts as a master regulator of the SASP, influencing the expression of more genes than RB and p53 combined. In cultured fibroblasts, NF-κB suppression causes escape from immune recognition by natural killer (NK) cells and cooperates with p53 inactivation to bypass senescence. In a mouse lymphoma model, NF-κB inhibition bypasses treatment-induced senescence, producing drug resistance, early relapse, and reduced survival. Our results demonstrate that NF-κB controls both cell-autonomous and non-cell-autonomous aspects of the senescence program and identify a tumor-suppressive function of NF-κB that contributes to the outcome of cancer therapy.

    Keywords

    Footnotes

    • Received June 22, 2011.
    • Accepted September 1, 2011.
    | Table of Contents

    Life Science Alliance