Abstract

Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer-related death, with a 5-year survival of <10% and severely limited treatment options. PDAC hallmarks include profound metabolic acid production and aggressive local proliferation and invasiveness. This phenotype is supported by upregulated net acid extrusion and epithelial-to-mesenchymal transition (EMT), the latter typically induced by aberrant transforming growth factor-β (TGFβ) signaling. It is, however, unknown whether TGFβ-induced EMT and upregulation of acid extrusion are causally related. Here, we show that mRNA and protein expression of the net acid extruding transporters Na+/H+ exchanger 1 (NHE1, SLC9A1) and Na+, HCO cotransporter 1 (NBCn1, SLC4A7) are increased in a panel of human PDAC cell lines compared to immortalized human pancreatic ductal epithelial (HPDE) cells. Treatment of Panc-1 cells (which express SMAD4, required for canonical TGFβ signaling) with TGFβ-1 for 48 h elicited classical EMT with down- and upregulation of epithelial and mesenchymal markers, respectively, in a manner inhibited by SMAD4 knockdown. Accordingly, less pronounced EMT was induced in BxPC-3 cells, which do not express SMAD4. TGFβ-1 treatment elicited a SMAD4-dependent increase in NHE1 expression, and a smaller, SMAD4-independent increase in NBCn1 in Panc-1 cells. Consistent with this, TGFβ-1 treatment led to elevated intracellular pH and increased net acid extrusion capacity in Panc-1 cells, but not in BxPC-3 cells, in an NHE1-dependent manner. Proliferation was increased in Panc-1 cells and decreased in BxPC-3 cells, upon TGFβ-1 treatment, and this, as well as EMT per se, was unaffected by NHE1- or NBCn1 inhibition. TGFβ-1-induced EMT was associated with a 4-fold increase in Panc-1 cell invasiveness, which further increased ~10-fold upon knockdown of the tumor suppressor Merlin (Neurofibromatosis type 2). Knockdown of NHE1 or NBCn1 abolished Merlin-induced invasiveness, but not that induced by TGFβ-1 alone. In conclusion, NHE1 and NBCn1 expression and NHE-dependent acid extrusion are upregulated during TGFβ-1-induced EMT of Panc-1 cells. NHE1 upregulation is SMAD4-dependent, and SMAD4-deficient BxPC-3 cells show no change in pHi regulation. NHE1 and NBCn1 are not required for EMT per se or EMT-associated proliferation changes, but are essential for the potentiation of invasiveness induced by Merlin knockdown.

Highlights

  • Pancreatic ductal adenocarcinoma (PDAC) is one of the most devastating cancers globally [1]

  • Our previous in silico analysis of RNA sequencing data from pancreatic cancer patient tissue indicated that a number of SLC9 and SLC4 family transporters are upregulated in PDAC patient tumor tissue compared to normal pancreatic epithelium [29]

  • We selected for widely expressed net acid extruders for initial analyses: two Na+/H+ exchangers, NHE1 and−2 (SLC9A1-2), and two Na+,HCO−3 cotransporters (NBCs), NBCe2 (SLC4A5) and NBCn1 (SLC4A7)

Read more

Summary

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is one of the most devastating cancers globally [1]. While PDAC genotypes are highly complex, the most widely characterized driver mutations are activating KRAS mutations, inactivating p53 tumor suppressor mutations, and inactivation or loss of the cyclin-dependent kinase inhibitor 2A (CDKN2A, P16INK4) and the transforming growth factor β (TGF-β) effector, SMAD4 [4, 5]. TGFβ signaling involves the binding of a TGFβ dimer (TGFβ1,−2, or−3, of which TGFβ-1 is most ubiquitous) to the TGFβ receptor types I and II (TGFβRI and –II; the former known as ALK5) This results in formation of a heterotetrameric receptor complex, where TGFβRII phosphorylates and activates TGFβRI. TGFβRI in turn phosphorylates the transcription factors SMAD2/3, which bind to the co-SMAD, SMAD4, to form a hetero-trimeric protein complex that enters the nucleus to control gene expression. TGFβ ligands signal through SMAD-independent pathways, including mitogen-activated protein kinases, small GTPases, and the phosphatidyl-inositol-3-kinase (PI3K)-AKT-mTOR pathway [6, 7]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call