Activating KRAS, NRAS, and BRAF mutants enhance proteasome capacity and reduce endoplasmic reticulum stress in multiple myeloma

Fazal Shirazi, Richard J. Jones, Ram K. Singh, Jianxuan Zou, Isere Kuiatse, Zuzana Berkova, Hua Wang, Hans C. Lee, Samuel Hong, Larry Dick, Nibedita Chattopadhyay, Robert Z. Orlowski

Research output: Contribution to journalArticlepeer-review

42 Scopus citations

Abstract

KRAS, NRAS, and BRAF mutations which activate p44/42 mitogen-activated protein kinase (MAPK) signaling are found in half of myeloma patients and contribute to proteasome inhibitor (PI) resistance, but the underlying mechanisms are not fully understood. We established myeloma cell lines expressing wild-type (WT), constitutively active (CA) (G12V/G13D/Q61H), or dominant-negative (DN) (S17N)-KRAS and -NRAS, or BRAF-V600E. Cells expressing CA mutants showed increased proteasome maturation protein (POMP) and nuclear factor (erythroid-derived 2)-like 2 (NRF2) expression. This correlated with an increase in catalytically active proteasome subunit β (PSMB)-8, PSMB9, and PSMB10, which occurred in an ETS transcription factor-dependent manner. Proteasome chymotrypsin-like, trypsin-like, and caspase-like activities were increased, and this enhanced capacity reduced PI sensitivity, while DN-KRAS and DN-NRAS did the opposite. Pharmacologic RAF or MAPK kinase (MEK) inhibitors decreased proteasome activity, and sensitized myeloma cells to PIs. CA-KRAS, CA-NRAS, and CA-BRAF down-regulated expression of endoplasmic reticulum (ER) stress proteins, and reduced unfolded protein response activation, while DN mutations increased both. Finally, a bortezomib (BTZ)/ MEK inhibitor combination showed enhanced activity in vivo specifically in CA-NRAS models. Taken together, the data support the hypothesis that activating MAPK pathway mutations enhance PI resistance by increasing proteasome capacity, and provide a rationale for targeting such patients with PI/RAF or PI/MEK inhibitor combinations. Moreover, they argue these mutations promote myeloma survival by reducing cellular stress, thereby distancing plasma cells from the apoptotic threshold, potentially explaining their high frequency in myeloma.

Original languageEnglish (US)
Pages (from-to)20004-20014
Number of pages11
JournalProceedings of the National Academy of Sciences of the United States of America
Volume117
Issue number33
DOIs
StatePublished - Aug 2020

Keywords

  • BRAF
  • KRAS
  • NRAF
  • Proteasome capacity
  • Proteasome inhibitor sensitivity

ASJC Scopus subject areas

  • General

MD Anderson CCSG core facilities

  • Advanced Technology Genomics Core
  • Flow Cytometry and Cellular Imaging Facility
  • Research Animal Support Facility
  • Small Animal Imaging Facility
  • Cytogenetics and Cell Authentication Core

Fingerprint

Dive into the research topics of 'Activating KRAS, NRAS, and BRAF mutants enhance proteasome capacity and reduce endoplasmic reticulum stress in multiple myeloma'. Together they form a unique fingerprint.

Cite this