4.6 Article

In Vivo Application of CRISPR/Cas9 Revealed Implication of Foxa1 and Foxp1 in Prostate Cancer Proliferation and Epithelial Plasticity

Journal

CANCERS
Volume 14, Issue 18, Pages -

Publisher

MDPI
DOI: 10.3390/cancers14184381

Keywords

CRISPR; prostate cancer; mouse models; Forkhead box proteins; cell plasticity

Categories

Funding

  1. Danish Cancer Society [R204-A12490]
  2. Dagmar Marshall Fond
  3. Aarhus University Research Foundation, Denmark
  4. P.A. Messerschmidt og Hustrus Fond

Ask authors/readers for more resources

This study reveals two distinct functions of Forkhead box proteins in prostate cancer, with Foxp1 acting as a tumor suppressor by controlling proliferation and genes regulated by the androgen receptor, while Foxa1 controls cell plasticity.
Simple Summary Prostate cancer is diagnosed in one out of eight men, with large implications on life quality. Forkhead box proteins are often found mutated in prostate cancer but their functions are still not fully understood. In this study, we applied CRISPR to investigate the function of two Forkhead box proteins, Foxa1 and Foxp1, in the mouse prostate in combination with loss of Pten. Our results reveal that Foxp1 is a tumor suppressor in prostate cancer progression by controlling proliferation and genes regulated by the androgen receptor. Foxa1 controls cell plasticity, as loss of Foxa1 converted the prostatic luminal cells to basal cells. Hereby, this study sheds light on two distinct functions of Forkhead box proteins in prostate cancer. Prostate cancer is the most common cancer in men in the Western world and the number is rising. Prostate cancer is notoriously heterogeneous, which makes it hard to generate and study in pre-clinical models. The family of Forkhead box (FOX) transcription factors are often altered in prostate cancer with especially high mutation burden in FOXA1 and FOXP1. FOXA1 harbors loss or gain of function mutations in 8% of prostate cancer, which increases to 14% in metastatic samples. FOXP1 predominately occurs with loss of function mutations in 7% of primary tumors, and similar incidents are found in metastatic samples. Here, we applied in vivo CRISPR editing, to study the loss of functions of these two FOX transcription factors, in murine prostate in combination with loss of Pten. Deficiency of Foxp1 increased proliferation in combination with loss of Pten. In contrast, proliferation was unchanged when androgen was deprived. The expression of Tmprss2 was increased when Foxp1 was mutated in vivo, showing that Foxp1 is a repressor for this androgen-regulated target. Furthermore, analysis of FOXP1 and TMPRSS2 expression in a human prostate cancer data set revealed a negative correlation. Mutation of Foxa1 in the murine prostate induces cell plasticity to luminal cells. Here, epithelial cells with loss of Foxa1 were transdifferentiated to cells with expression of the basal markers Ck5 and p63. Interestingly, these cells were located in the lumen and did not co-express Ck8. Overall, this study reveals that loss of Foxp1 increases cell proliferation, whereas loss of Foxa1 induces epithelial plasticity in prostate 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

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available