4.7 Article

RASSF1A disrupts the NOTCH signaling axis via SNURF/RNF4-mediated ubiquitination of HES1

Journal

EMBO REPORTS
Volume 23, Issue 2, Pages -

Publisher

WILEY
DOI: 10.15252/embr.202051287

Keywords

cancer stemness; Hippo-Notch signaling crosstalk; RASSF1A-HES1-SNURF/RNF4 complex; gamma-secretase inhibitors (GSIs)

Funding

  1. CRUK [A19277]
  2. MRC
  3. Kidani Memorial Trust
  4. Federal Agency for Scientific Organizations (FASO Russia) [0324-2016-0008]
  5. Wellcome Trust
  6. Science Foundation Ireland [15/CDA/3495]
  7. European Union (European Social Fund- ESF) through the Operational Program <> [MIS-5033021]
  8. Hellenic Foundation for Research and Innovation (HFRI)
  9. General Secretariat for Research and Innovation (GSRI) [775]
  10. HFRI
  11. GSRI [3782]
  12. European Union [722729]
  13. National Public Investment Program of the Ministry of Development and Investment / General Secretariat for Research and Technology [2020SigmaE01300001]
  14. Welfare Foundation for Social & Cultural Sciences (KIKPE), Athens, Greece
  15. NKUA-SARG grants [70/3/8916, 70/3/12128]
  16. Science Foundation Ireland (SFI) [15/CDA/3495] Funding Source: Science Foundation Ireland (SFI)

Ask authors/readers for more resources

The interaction between RASSF1A and HES1 affects tumor differentiation and response to drug treatment, which is crucial for clinical therapy.
RASSF1A promoter methylation has been correlated with tumor dedifferentiation and aggressive oncogenic behavior. Nevertheless, the underlying mechanism of RASSF1A-dependent tumor dedifferentiation remains elusive. Here, we show that RASSF1A directly uncouples the NOTCH-HES1 axis, a key suppressor of differentiation. Interestingly, the crosstalk of RASSF1A with HES1 occurs independently from the signaling route connecting RASSF1A with the Hippo pathway. At the molecular level, we demonstrate that RASSF1A acts as a scaffold essential for the SUMO-targeted E3 ligase SNURF/RNF4 to target HES1 for degradation. The reciprocal relationship between RASSF1A and HES1 is evident across a wide range of human tumors, highlighting the clinical significance of the identified pathway. We show that HES1 upregulation in a RASSF1A-depleted environment renders cells non-responsive to the downstream effects of gamma-secretase inhibitors (GSIs) which restrict signaling at the level of the NOTCH receptor. Taken together, we report a mechanism through which RASSF1A exerts autonomous regulation of the critical Notch effector HES1, thus classifying RASSF1A expression as an integral determinant of the clinical effectiveness of Notch inhibitors.

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