Mutations that disrupt PHOXB interaction with the neuronal calcium sensor HPCAL1 impede cellular differentiation in neuroblastoma



Wang, W, Zhong, Q, Teng, L, Bhatnagar, N, Sharma, B, Zhang, X, Luther, W, Haynes, LP ORCID: 0000-0002-1296-0338, Burgoyne, RD ORCID: 0000-0002-9219-0387, Vidal, M
et al (show 3 more authors) (2014) Mutations that disrupt PHOXB interaction with the neuronal calcium sensor HPCAL1 impede cellular differentiation in neuroblastoma. Oncogene, 33 (25). pp. 3316-3324.

Access the full-text of this item by clicking on the Open Access link.

Abstract

Heterozygous germline mutations in PHOX2B, a transcriptional regulator of sympathetic neuronal differentiation, predispose to diseases of the sympathetic nervous system, including neuroblastoma and congenital central hypoventilation syndrome (CCHS). Although the PHOX2B variants in CCHS largely involve expansions of the second polyalanine repeat within the C-terminus of the protein, those associated with neuroblastic tumors are nearly always frameshift and truncation mutations. To test the hypothesis that the neuroblastoma-associated variants exert their effects through loss or gain of protein–protein interactions, we performed a large-scale yeast two-hybrid screen using both wild-type (WT) and six different mutant PHOX2B proteins against over 10 000 human genes. The neuronal calcium sensor protein HPCAL1 (VILIP-3) exhibited strong binding to WT PHOX2B and a CCHS-associated polyalanine expansion mutant but only weakly or not at all to neuroblastoma-associated frameshift and truncation variants. We demonstrate that both WT PHOX2B and the neuroblastoma-associated R100L missense and the CCHS-associated alanine expansion variants induce nuclear translocation of HPCAL1 in a Ca2+-independent manner, while the neuroblastoma-associated 676delG frameshift and K155X truncation mutants impair subcellular localization of HPCAL1, causing it to remain in the cytoplasm. HPCAL1 did not appreciably influence the ability of WT PHOX2B to transactivate the DBH promoter, nor did it alter the decreased transactivation potential of PHOX2B variants in 293T cells. Abrogation of the PHOX2B–HPCAL1 interaction by shRNA knockdown of HPCAL1 in neuroblastoma cells expressing PHOX2B led to impaired neurite outgrowth with transcriptional profiles indicative of inhibited sympathetic neuronal differentiation. Our results suggest that certain PHOX2B variants associated with neuroblastoma pathogenesis, because of their inability to bind to key interacting proteins such as HPCAL1, may predispose to this malignancy by impeding the differentiation of immature sympathetic neurons.

Item Type: Article
Additional Information: Wang, W Zhong, Q Teng, L Bhatnagar, N Sharma, B Zhang, X Luther, W 2nd Haynes, L P Burgoyne, R D Vidal, M Volchenboum, S Hill, D E George, R E ENG P50 HG004233/HG/NHGRI NIH HHS/ R01 HG001715/HG/NHGRI NIH HHS/ R33 CA132073/CA/NCI NIH HHS/ 2013/07/23 06:00 Oncogene. 2013 Jul 22. doi: 10.1038/onc.2013.290. ## TULIP Type: Articles/Papers (Journal) ##
Uncontrolled Keywords: CNS cancer, differentiation, mutation
Depositing User: Symplectic Admin
Date Deposited: 28 Apr 2016 10:13
Last Modified: 19 Jan 2023 07:37
DOI: 10.1038/onc.2013.290
Open Access URL: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC404033...
Related URLs:
URI: https://livrepository.liverpool.ac.uk/id/eprint/3000890