4.7 Article

Senescence-Inflammatory Regulation of Reparative Cellular Reprogramming in Aging and Cancer

Journal

Publisher

FRONTIERS MEDIA SA
DOI: 10.3389/fcell.2017.00049

Keywords

reprogramming; aging; cancer; senescence; inflammation

Funding

  1. Ministerio de Ciencia e Innovacion [SAF2016-80639-P]
  2. Plan Nacional de I+ D+ I, Spain
  3. Agencia de Gestio d'Ajuts Universitaris i de Recerca (AGAUR) [2014 SGR229]
  4. Departament d'Economia i Coneixement, Catalonia, Spain
  5. MINECO [MTM2015-71509-C2-1-R, 2014SGR1307, MDM-2014-0445]
  6. AGAUR [MTM2015-71509-C2-1-R, 2014SGR1307]
  7. Obra Social La Caixa Foundation on Collaborative Mathematics
  8. CERCA Programme of the Generalitat de Catalunya
  9. Armangue family (Girona, Catalonia)

Ask authors/readers for more resources

The inability of adult tissues to transitorily generate cells with functional stem cell-like properties is a major obstacle to tissue self-repair. Nuclear reprogramming-like phenomena that induce a transient acquisition of epigenetic plasticity and phenotype malleability may constitute a reparative route through which human tissues respond to injury, stress, and disease. However, tissue rejuvenation should involve not only the transient epigenetic reprogramming of differentiated cells, but also the committed re-acquisition of the original or alternative committed cell fate. Chronic or unrestrained epigenetic plasticity would drive aging phenotypes by impairing the repair or the replacement of damaged cells: such uncontrolled phenomena of in vivo reprogramming might also generate cancer-like cellular states. We herein propose that the ability of senescence-associated inflammatory signaling to regulate in vivo reprogramming cycles of tissue repair outlines a threshold model of aging and cancer. The degree of senescence/inflammation-associated deviation from the homeostatic state may delineate a type of thresholding algorithm distinguishing beneficial from deleterious effects of in vivo reprogramming. First, transient activation of NF-kappa B-related innate immunity and senescence-associated inflammatory components (e.g., IL-6) might facilitate reparative cellular reprogramming in response to acute inflammatory events. Second, para-inflammation switches might promote long-lasting but reversible refractoriness to reparative cellular reprogramming. Third, chronic senescence-associated inflammatory signaling might lock cells in highly plastic epigenetic states disabled for reparative differentiation. The consideration of a cellular reprogramming-centered view of epigenetic plasticity as a fundamental element of a tissue's capacity to undergo successful repair, aging degeneration or malignant transformation should provide challenging stochastic insights into the current deterministic genetic paradigm for most chronic diseases, thereby increasing the spectrum of therapeutic approaches for physiological aging and cancer.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available