Phosphodiesterase 1A physically interacts with YTHDF2 and reinforces the progression of non-small cell lung cancer

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife Assessment

    This manuscript provides valuable mechanistic insight into NSCLC progression, both in terms of tumour metastasis and the development of chemoresistance. The authors draw upon a range of techniques and assays and although the evidence shown is solid, suggestions by the two reviewers will strengthen the message. The work presented will be of interest to cancer biologists and more broadly to those interested in NSCLC translational studies.

This article has been Reviewed by the following groups

Read the full article

Abstract

Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer, and the prognosis is poor due to distant metastasis and drug resistance. Thus, there is an urgent need to discover novel therapeutic targets and strategies to overcome cisplatin resistance and metastasis. A series of in vitro and in vivo phenotype experiments were performed to investigate the role of PDE1A in NSCLC. The RIP assay, mRNA stability assay and LC- MS/MS were performed to investigate the molecular mechanisms of PDE1A in NSCLC progression. We demonstrated that phosphodiesterase 1A (PDE1A) promoted metastasis and EMT progression of NSCLC. In addition, NSCLC cells overexpressing PDE1A promoted angiogenesis by regulating exosome release. IL-6/JAK/STAT3 signaling pathway was highly enriched in PDE1A- coexpresssed genes, and PDE1A promoted NSCLC metastasis by activating the STAT3 pathway. GO enrichment analysis of PDE1A-interacting genes showed that PDE1A might interact with YTHDF2 and participate in m6A- containing RNA binding. The binding between PDE1A and YTHDF2 was verified, and PDE1A regulated the STAT3 pathway by interacting with YTHDF2. The mechanism of YTHDF2/PDE1A complex in regulating STAT3 pathway was predicted by overlapping YTHDF2-interacting-RNAs, and genes coexpressed with YTHDF2 and STAT3. The interactions between YTHDF2 and target mRNAs were predicted, and there were three predicted targets of YTHDF2 with high scores: NRF2, SOCS2, and MET. Indeed, PDE1A interacted with YTHDF2, destabilized SOCS2, and activated STAT3 pathway. Moreover, PDE1A suppression sensitized anti-NSCLC activity of cisplatin via regulating NRF2 and MET. This work not only uncovers a novel PDE1A/YTHDF2/STAT3 pathway in NSCLC progression but also provides therapeutic strategies for treating NSCLC patients with metastasis or cisplatin- resistance.

Article activity feed

  1. eLife Assessment

    This manuscript provides valuable mechanistic insight into NSCLC progression, both in terms of tumour metastasis and the development of chemoresistance. The authors draw upon a range of techniques and assays and although the evidence shown is solid, suggestions by the two reviewers will strengthen the message. The work presented will be of interest to cancer biologists and more broadly to those interested in NSCLC translational studies.

  2. Reviewer #1 (Public review):

    Summary:

    The manuscript entitled "Phosphodiesterase 1A Physically Interacts with YTHDF2 and Reinforces the Progression of Non-Small Cell Lung Cancer" explores the role of PDE1A in promoting NSCLC progression by binding to the m6A reader YTHDF2 and regulating the mRNA stability of several novel target genes, consequently activating the STAT3 pathway and leading to metastasis and drug resistance.

    Strengths:

    The study addresses a novel mechanism involving PDE1A and YTHDF2 interaction in NSCLC, contributing to our understanding of cancer progression.

    Weaknesses:

    The following issues should be addressed:

    (1) The body weight changes and/or survival times of each group in the in vivo metastasis studies should be provided.

    (2) In Figure 7, the direct binding between YTHDF2 and the potential target genes should be further validated by silencing YTHDF2 to observe the half-life of the mRNA levels of target genes, in addition to silencing PDE1A.

    (3) In Figure 7, the potential methylation sites of "A" on the target genes such as SOCS2 should be verified by mutation analysis, followed by m6A IP or reporter assays.

    (4) In Figure 6G, the correlation between the mRNA levels of STAT3 and YTHDF2 needs clarification. According to the authors' mechanism, the STAT3 pathway is activated, rather than upregulation of mRNA levels (or protein levels, as shown in Figure 6F). Figure 7 does not provide evidence that STAT3 is a bona fide target gene regulated by YTHDF2.

    (5) The final figure, which discusses sensitization to cisplatin by PDE1A suppression, does not appear to be closely related to the interaction or regulation of PDE1A/YTHDF2. If the authors claim this is an m6A-associated event, additional evidence is needed. Otherwise, this part could be removed from the manuscript.

  3. Reviewer #2 (Public review):

    This manuscript aims to investigate the biological impact and mechanisms of phosphodiesterase 1A (PDE1A) in promoting non-small cell lung cancer (NSCLC) progression. They first analyzed several databases and used three established NSCLC cell lines and a normal cell line to demonstrate that PDE1A is overexpressed in lung cancer and its expression negatively correlated with the outcomes of patients. Based on this data, they suggested PDE1A could be considered as a novel prognostic predictor in lung cancer treatment and progression. To study the biological function of PDE1A in NSCLC, they focused on testing the effect of inhibition of PDE1A genetically and pharmacologically on cell proliferation, migration, and invasion in vitro. They also used an experimental metastasis model via tail vein injection of H1299 cells to test if PDE1A promoted metastasis. By database analysis, they also decided to investigate if PDE1A promoted angiogenesis by co-culturing NSCLC cells with HUVECs as well as assessing the tumors from the subcutaneous xenograft model. However, in this model, whether PDE1A modulation impacted tumor metastasis was not examined. To address the mechanism of how PDE1A promotes metastasis, the authors again performed a bioinformatic and GSEA enrichment analysis and confirmed PDE1A indeed activated STAT3 signaling to promote migration. In combination with IP followed by Mass spectrometry, they found PDE1A is a partner of YTHDF2, the cooperation of PDE1A and YTHDF2 negatively regulated SOCS2 mRNA as demonstrated by RIP assay, and ultimately activated STAT3 signaling. Finally, the authors shifted the direction from metastasis to chemoresistance, specifically, they found that PDEA1 inhibitions sensitized NSCLC cells to cisplatin through MET and NRF2 signaling.

    Strength:

    Overall, the manuscript was well-written and the majority of the data supported the conclusions. The authors used a series of methods including cell lines, animal models, and database analysis to demonstrate the novel roles and mechanism of how PDE1 promotes NSCLC invasion and metastasis as well as cisplatin sensitivity. Given that PDE1A inhibitors have been perused to use in clinic, this study provided valuable findings that have the translational potential for NSCLC treatment.

    Weaknesses:

    The role of YTHDF2 in PDE1A-promoted tumor metastasis was not investigated. To make the findings more clinical and physiologically relevant, it would be interesting to test if inhibition of PDE1A impacts metastasis using lung cancer orthotopic and patient-derived xenograft models. It is also important to use a cisplatin-resistant NSCLC cell line to test if a PDE1A inhibitor has the potential to sensitize cisplatin in vitro and in vivo. Furthermore, this study relied heavily on different database analyses, although providing novel and compelling data that was followed up and confirmed in the paper, it is critical to have detailed statistical description section on data acquisition throughout the manuscript.