All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit the MPN Advocates Network.

The MPN Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your MPN Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The MPN Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the MPN Hub cannot guarantee the accuracy of translated content. The MPN Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2021-05-26T13:31:34.000Z

Mutant calreticulin is dependent on zinc to drive constitutive MPL activity

May 26, 2021
Share:

Bookmark this article

Mutations of the calreticulin gene (CALR) have been recognized as driver mutations involved in the development of myeloproliferative neoplasms (MPN) along with Janus kinase (JAK) or thrombopoietin receptor (MPL) mutations. CALR mutations in MPN are insertion-deletion mutations, and the most common is a 52-bp deletion (CALRdel52).

CALRdel52 homomultimers are responsible for binding to MPL and causing constitutive activation of JAK-STAT signaling. However, there is limited understanding of the mechanism of CALRdel52 interaction with MPL and its activation. 

Rivera et al. recently conducted a comprehensive mutagenesis analysis and explored the mechanism of CALRdel52 binding and activation of the MPL signal, including the role of the glycan-binding lectin motif, which associates with the extracellular domain of MPL and the zinc-binding domain.1 The results are published in Blood Advances, and the MPN Hub is pleased to summarize the key findings here.

Materials

293T cells were derived from the American Type Culture Collection, and MPL-expressing Ba/F3 cells were generated by retroviral transduction. Peripheral blood mononuclear cells were isolated from patients with MPN.

Results

Residues related with lectin and zinc-binding activity are essential for oncogenic activity based on a mutagenesis screen described below:

  • As expected, nine residues involved in the function of the lectin motif abrogated cytokine independence. Specifically C105A, K111A, G133A, or D135A mutations displayed complete nullification of cytokine independence, decreased STAT3/5 phosphorylation, and prevented co-immunoprecipitation of CALRdel52 with MPL.
  • Interestingly, tyrosine mutations Y109A and Y128A which inhibit normal CALR function, showed only partial loss of cytokine independence in CALRdel52 transfected cells but had no effect on STAT3/5 phosphorylation or MPL binding.
  • When investigating the role of zinc-binding histidines, it could be shown that single mutations in H99A, H145A, and H170A (1His) showed partial abrogation of cytokine independence, while binding to MPL was maintained.
  • However, loss of either two or all three histidine residues in combination (2His and 3His) led to significant impairment of cytokine-independent growth along with decreased pSTAT3/5 phosphorylation and loss of MPL binding.
  • By contrast, mutating a histidine not involved in zinc-binding, H42A, did not abrogate cytokine-independent growth and the capacity to promote STAT3/5 phosphorylation.
  • Using mCherry-tagged CALRdel52 and green fluorescent protein (GFP)-MPL, significant co-localization could be shown for MPL with CALRdel52 and with 1His-CALRdel52 variants, but not with wild-type CALR or 2His and 3His CALRdel52 variants.

Histidine-mutated CALRdel52 is incapable of homomultimerization:

  • FLAG-tagged CALRdel52 co-immunoprecipitated with a V5-tagged CALRdel52 but did not bind to wild-type CALR.
  • Multimerization was retained in lectin-deficient CALRdel52 and all three 1His- CALRdel52 variants. However, it was impaired in 2His- and 3His- CALRdel52 variants.
  • Lectin-deficient CALRdel52-D135A retained the ability to homomultimerize with another CALRdel52-D135A, but not with histidine-mutated 3His-CALRdel52.

 Zinc is essential for CALRdel52-MPL complex formation and downstream signaling:

  • Using affinity chromatography with a zinc resin, the authors found retention of CALRdel52 and 1His- CALRdel52 in the zinc-bound fraction, which was not the case for wild-type CALR, 2His-, and 3His- CALRdel52, suggesting that zinc may be essential for CALRdel52 multimerization.
  • This was further tested in co-precipitation experiments using 293T cells expressing FLAG- and V5-tagged CALRdel52 and MPL. Treatment with Tetrakis (2-pyridylmethyl)-ethylenediamine (TPEN), a zinc chelator, decreased CALRdel52 multimerization and impaired MPL binding, confirming that zinc was needed for CALRdel52 to multimerize and interact with MPL.
  • To understand if zinc chelation with TPEN could eradicate cytokine independence and JAK-STAT signalling, CALRdel52-expressing Ba/F3-MPL cells were investigated. Treatment with TPEN led to decreased viability and diminished STAT3/5 phosphorylation. Similar effects were seen using another zinc chelator, clioquinol.
  • Furthermore, there was reduced interaction between CALRdel52 and MPL in these cells as seen in FLAG pulldown experiments.
  • By contrast, Ba/F3 cells transformed with the oncogenic MPLW515L were less sensitive to TPEN-induced cytotoxicity and had no impairment in STAT3/5 phosphorylation.

TPEN preferentially eradicates CALR-mutant primary MPN cells:

  • No evidence of impaired growth of wild-type colonies were observed at TPEN dosages <3 µM.
  • When testing peripheral blood mononuclear cells (PBMCs) from five patients with CALRdel52-mutated myelofibrosis, there was clear indication that zinc chelation selectively affected the growth of CALRdel52-positive, patient-derived, burst-forming unit erythroid (BFU-E) colonies. Using supplementation with 2.5 µM TPEN, the authors demonstrated a decreased growth of CALRdel52-positive BFU-Es, but a normal growth of wild-type BFU-Es in four of five patients.

Conclusion

This study provides a better understanding of the molecular mechanism through which CALRdel52 exerts its oncogenic activity. The study identifies the zinc-binding histidines within CALRdel52 essential for the formation of homomultimers that interact with MPL to induce its constitutive activation. It also shows that homomultimerization and the MPL-binding of CALRdel52 are zinc-dependent and could therefore, be a potential target for treatment of CALR-mutant MPNs.   

  1. Rivera JF, Baral AJ, Nadat F, et al. Zinc-dependent multimerization of mutant calreticulin is required for MPL binding and MPN pathogenesis. Blood Adv. 2021;5(7):1922-1932. DOI: 1182/bloodadvances.2020002402

Newsletter

Subscribe to get the best content related to MPN delivered to your inbox