“RHAMM knockout” mice express a truncated RHAMM protein that promotes pancreatic cancer progression with dysfunctional p53
Editorial Commentary

RHAMM knockout” mice express a truncated RHAMM protein that promotes pancreatic cancer progression with dysfunctional p53

Xiang Chen, Yi-Chieh Nancy Du

Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA

Correspondence to: Yi-Chieh Nancy Du. Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA. Email: nad2012@med.cornell.edu.

Comment on: Lin A, Feng J, Chen X, et al. High levels of truncated RHAMM cooperate with dysfunctional p53 to accelerate the progression of pancreatic cancer. Cancer Lett 2021;514:79-89.


Received: 09 June 2022; Accepted: 15 June 2022; Published: 10 July 2022.

doi: 10.21037/apc-2022-1


Pancreatic cancer, which lacks effective treatment, has the highest mortality rate of all major cancers (1). A recent study by Lin et al. published in Cancer Letters (2) sought out to investigate whether RHAMM is a therapeutic target in pancreatic cancer using a Rhamm-/- mouse strain. Surprisingly, a truncated HMMR∆exon8-16 protein expressed at higher levels than wild-type RHAMM protein was found in this “knockout” strain and HMMR∆exon8-16 accelerated pancreatic cancer progression in genetic engineered mouse models.

Pancreatic cancer is projected to become the second leading cause of cancer-related death by 2030 (3). The most common type of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC). Pancreatic neuroendocrine tumor (PNET) is the second common malignancy of pancreas and its incidence is increasing (4). Pancreatic cancer patients are often diagnosed at advanced stages. Despite intense efforts in improved diagnostic methods and development of targeted therapies, the overall survival for pancreatic cancer has changed little. It is critical to understand the biology of pancreatic cancer and identify novel therapeutic targets for this devastating disease.

Receptor for hyaluronan-mediated motility (RHAMM; gene name: HMMR) was identified as a protein that binds to hyaluronic acid (HA) (5). RHAMM promotes HA-induced motility, modulates cytoskeletal organization, and activates extracellular-regulated kinase (Erk) (6-8). RHAMM protein expression is limited in normal human tissues and not expressed in adult pancreas (9,10). In contrast, RHAMM protein is overexpressed in pancreatic cancer and many other cancer types (9-13). RHAMM encodes 18 exons. RHAMMB (known as RHAMMv3) is the most prominent alternative splicing RHAMM isoform found in pancreatic cancer patients (10). Human RHAMMB overexpression promotes metastasis and knockdown of RHAMM by shRNA suppresses metastasis in mouse models of PNET (10,14). High RHAMMB mRNA levels correlate with inferior survival of PDAC patients in TCGA cohort (10).

Lin et al. investigated whether genetic ablation of RHAMM is effective to impair pancreatic cancer progression using “RHAMM knockout” mice. The Rhamm-/- mouse was previously generated by deleting exons 8~16 of the HMMR gene through homologous recombination in embryonic stem cells (15). However, the detailed molecular and histological characterization identified a truncated RHAMM (HMMR∆exon8-16) of 239 amino acids with a molecular weight of ~27 kDa in the Rhamm-/- mice and the HMMR∆exon8-16 protein was more abundant than the full-length protein in RHAMM wild-type mice. This Rhamm-/- strain should be renamed as HMMRexon8-16/exon8-16.

This study further demonstrated that while HMMR∆exon8-16 by itself did not promote the progression of pancreatic intraepithelial neoplasia (PanIN) to PDAC in the p48-Cre; LSL-KRASG12D mice, the combination of HMMR∆exon8-16 and heterozygous p53 loss significantly promoted earlier onset of invasive PDAC formation and shortened survival of p48-Cre; LSL-KRASG12D; p53lox/+ mice. Moreover, HMMR∆exon8-16 decreased survival of RIP-Tag PNET mice, in which p53 was inhibited by SV40 T antigen. Importantly, pancreatic cancer patients with mutant TP53 or loss of one copy of TP53 had higher RHAMM expression, which, combined, predicted worse survival outcomes.

In summary, Lin et al. have shown that high levels of the N-terminal RHAMM protein, HMMR∆exon8-16 collaborated dysfunctional p53 to promote progression of PDAC and PNET in mouse models, and HMMR∆exon8-16, possessed the oncogenic function. The unexpected discovery of a truncated RHAMM protein in the “RHAMM knockout” mouse provides critical insights for the re-evaluation of previous work using this mouse strain and cells derived from it (15-21). A true RHAMM knockout will be needed to assess the therapeutic value of targeting RHAMM for pancreatic cancer treatment in pre-clinical mouse models.


Acknowledgments

Funding: This article was supported in part by NIH R01CA204916-01A1, DoD W81XWH-16-1-0619, and STARR I12-0043.


Footnote

Provenance and Peer Review: This article was commissioned and reviewed by the Associate Editor, Min Li, PhD (The University of Oklahoma Health Sciences Center, Stanton L. Young Biomedical Research Center, Oklahoma City, Oklahoma, USA) and the Editor-in-Chief, Lei Zheng, MD, PhD (Departments of Oncology and Surgery, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA).

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://apc.amegroups.com/article/view/10.21037/apc-2022-1/coif). XC and YCND are supported by NIH R01CA204916-01A1, DoD W81XWH-16-1-0619, and STARR I12-0043.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7-30. [Crossref] [PubMed]
  2. Lin A, Feng J, Chen X, et al. High levels of truncated RHAMM cooperate with dysfunctional p53 to accelerate the progression of pancreatic cancer. Cancer Lett 2021;514:79-89. [Crossref] [PubMed]
  3. Rahib L, Smith BD, Aizenberg R, et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 2014;74:2913-21. [Crossref] [PubMed]
  4. Buicko JL, Finnerty BM, Zhang T, et al. Insights into the biology and treatment strategies of pancreatic neuroendocrine tumors. Ann Pancreat Cancer 2019;2:12. [Crossref] [PubMed]
  5. Turley EA. Purification of a hyaluronate-binding protein fraction that modifies cell social behavior. Biochem Biophys Res Commun 1982;108:1016-24. [Crossref] [PubMed]
  6. Hall CL, Lange LA, Prober DA, et al. pp60(c-src) is required for cell locomotion regulated by the hyaluronanreceptor RHAMM. Oncogene 1996;13:2213-24. [PubMed]
  7. Zhang S, Chang MC, Zylka D, et al. The hyaluronan receptor RHAMM regulates extracellular-regulated kinase. J Biol Chem 1998;273:11342-8. [Crossref] [PubMed]
  8. Assmann V, Jenkinson D, Marshall JF, et al. The intracellular hyaluronan receptor RHAMM/IHABP interacts with microtubules and actin filaments. J Cell Sci 1999;112:3943-54. [Crossref] [PubMed]
  9. Chen YT, Chen Z, Du YN. Immunohistochemical analysis of RHAMM expression in normal and neoplastic human tissues: a cell cycle protein with distinctive expression in mitotic cells and testicular germ cells. Oncotarget 2018;9:20941-52. [Crossref] [PubMed]
  10. Choi S, Wang D, Chen X, et al. Function and clinical relevance of RHAMM isoforms in pancreatic tumor progression. Mol Cancer 2019;18:92. [Crossref] [PubMed]
  11. Cheng XB, Sato N, Kohi S, et al. Receptor for Hyaluronic Acid-Mediated Motility is Associated with Poor Survival in Pancreatic Ductal Adenocarcinoma. J Cancer 2015;6:1093-8. [Crossref] [PubMed]
  12. Wang D, Narula N, Azzopardi S, et al. Expression of the receptor for hyaluronic acid mediated motility (RHAMM) is associated with poor prognosis and metastasis in non-small cell lung carcinoma. Oncotarget 2016;7:39957-69. [Crossref] [PubMed]
  13. Schatz-Siemers N, Chen YT, Chen Z, et al. Expression of the Receptor for Hyaluronic Acid-Mediated Motility (RHAMM) in Endometrial Cancer is Associated With Adverse Histologic Parameters and Tumor Progression. Appl Immunohistochem Mol Morphol 2020;28:453-9. [Crossref] [PubMed]
  14. Du YC, Chou CK, Klimstra DS, et al. Receptor for hyaluronan-mediated motility isoform B promotes liver metastasis in a mouse model of multistep tumorigenesis and a tail vein assay for metastasis. Proc Natl Acad Sci U S A 2011;108:16753-8. [Crossref] [PubMed]
  15. Tolg C, Poon R, Fodde R, et al. Genetic deletion of receptor for hyaluronan-mediated motility (Rhamm) attenuates the formation of aggressive fibromatosis (desmoid tumor). Oncogene 2003;22:6873-82. [Crossref] [PubMed]
  16. Tolg C, Hamilton SR, Nakrieko KA, et al. Rhamm-/- fibroblasts are defective in CD44-mediated ERK1,2 motogenic signaling, leading to defective skin wound repair. J Cell Biol 2006;175:1017-28. [Crossref] [PubMed]
  17. Bahrami SB, Tolg C, Peart T, et al. Receptor for hyaluronan mediated motility (RHAMM/HMMR) is a novel target for promoting subcutaneous adipogenesis. Integr Biol (Camb) 2017;9:223-37. [Crossref] [PubMed]
  18. Tolg C, Hamilton SR, Morningstar L, et al. RHAMM promotes interphase microtubule instability and mitotic spindle integrity through MEK1/ERK1/2 activity. J Biol Chem 2010;285:26461-74. [Crossref] [PubMed]
  19. Wu KY, Kim S, Liu VM, et al. Function-Blocking RHAMM Peptides Attenuate Fibrosis and Promote Antifibrotic Adipokines in a Bleomycin-Induced Murine Model of Systemic Sclerosis. J Invest Dermatol 2021;141:1482-1492.e4. [Crossref] [PubMed]
  20. Tolg C, Liu M, Cousteils K, et al. Cell-specific expression of the transcriptional regulator RHAMM provides a timing mechanism that controls appropriate wound re-epithelialization. J Biol Chem 2020;295:5427-48. [Crossref] [PubMed]
  21. Tolg C, Hamilton SR, Zalinska E, et al. A RHAMM mimetic peptide blocks hyaluronan signaling and reduces inflammation and fibrogenesis in excisional skin wounds. Am J Pathol 2012;181:1250-70. [Crossref] [PubMed]
doi: 10.21037/apc-2022-1
Cite this article as: Chen X, Du YCN. “RHAMM knockout” mice express a truncated RHAMM protein that promotes pancreatic cancer progression with dysfunctional p53. Ann Pancreat Cancer 2022;5:7.

Download Citation