Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses. / Troike, Katie M.; Wang, Sabrina Z.; Silver, Daniel J.; Lee, Juyeun; Mulkearns-Hubert, Erin E.; Hajdari, Nicole; Ghosh, Prabar K.; Kay, Kristen E.; Beilis, Julia L.; Mitchell, Sofia E.; Bishop, Christopher W.; Hong, Ellen S.; Artomov, Mykyta; Hubert, Christopher G.; Rajappa, Prajwal; Connor, James R.; Fox, Paul L.; Kristensen, Bjarne W.; Lathia, Justin D.

In: Neuro-Oncology Advances, Vol. 6, No. 1, vdad154, 2024.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Troike, KM, Wang, SZ, Silver, DJ, Lee, J, Mulkearns-Hubert, EE, Hajdari, N, Ghosh, PK, Kay, KE, Beilis, JL, Mitchell, SE, Bishop, CW, Hong, ES, Artomov, M, Hubert, CG, Rajappa, P, Connor, JR, Fox, PL, Kristensen, BW & Lathia, JD 2024, 'Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses', Neuro-Oncology Advances, vol. 6, no. 1, vdad154. https://doi.org/10.1093/noajnl/vdad154

APA

Troike, K. M., Wang, S. Z., Silver, D. J., Lee, J., Mulkearns-Hubert, E. E., Hajdari, N., Ghosh, P. K., Kay, K. E., Beilis, J. L., Mitchell, S. E., Bishop, C. W., Hong, E. S., Artomov, M., Hubert, C. G., Rajappa, P., Connor, J. R., Fox, P. L., Kristensen, B. W., & Lathia, J. D. (2024). Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses. Neuro-Oncology Advances, 6(1), [vdad154]. https://doi.org/10.1093/noajnl/vdad154

Vancouver

Troike KM, Wang SZ, Silver DJ, Lee J, Mulkearns-Hubert EE, Hajdari N et al. Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses. Neuro-Oncology Advances. 2024;6(1). vdad154. https://doi.org/10.1093/noajnl/vdad154

Author

Troike, Katie M. ; Wang, Sabrina Z. ; Silver, Daniel J. ; Lee, Juyeun ; Mulkearns-Hubert, Erin E. ; Hajdari, Nicole ; Ghosh, Prabar K. ; Kay, Kristen E. ; Beilis, Julia L. ; Mitchell, Sofia E. ; Bishop, Christopher W. ; Hong, Ellen S. ; Artomov, Mykyta ; Hubert, Christopher G. ; Rajappa, Prajwal ; Connor, James R. ; Fox, Paul L. ; Kristensen, Bjarne W. ; Lathia, Justin D. / Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses. In: Neuro-Oncology Advances. 2024 ; Vol. 6, No. 1.

Bibtex

@article{0e427d75a04b4d0799674ce7a6559be4,
title = "Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses",
abstract = "Background: Glioblastoma (GBM) displays alterations in iron that drive proliferation and tumor growth. Iron regulation is complex and involves many regulatory mechanisms, including the homeostatic iron regulator (HFE) gene, which encodes the homeostatic iron regulatory protein. While HFE is upregulated in GBM and correlates with poor survival outcomes, the function of HFE in GBM remains unclear. Methods: We interrogated the impact of cell-intrinsic Hfe expression on proliferation and survival of intracranially implanted animals through genetic gain- and loss-of-function approaches in syngeneic mouse glioma models, along with in vivo immune assessments. We also determined the expression of iron-associated genes and their relationship to survival in GBM using public data sets and used transcriptional profiling to identify differentially expressed pathways in control compared to Hfe-knockdown cells. Results: Overexpression of Hfe accelerated GBM proliferation and reduced animal survival, whereas suppression of Hfe induced apoptotic cell death and extended survival, which was more pronounced in females and associated with attenuation of natural killer cells and CD8+ T cell activity. Analysis of iron gene signatures in Hfe-knockdown cells revealed alterations in the expression of several iron-associated genes, suggesting global disruption of intracellular iron homeostasis. Further analysis of differentially expressed pathways revealed oxidative stress as the top pathway upregulated following Hfe loss. Hfe knockdown indeed resulted in enhanced 55Fe uptake and generation of reactive oxygen species. Conclusions: These findings reveal an essential function for HFE in GBM cell growth and survival, as well as a sex-specific interaction with the immune response. ",
keywords = "glioblastoma, HFE, iron, reactive oxygen species, sex difference",
author = "Troike, {Katie M.} and Wang, {Sabrina Z.} and Silver, {Daniel J.} and Juyeun Lee and Mulkearns-Hubert, {Erin E.} and Nicole Hajdari and Ghosh, {Prabar K.} and Kay, {Kristen E.} and Beilis, {Julia L.} and Mitchell, {Sofia E.} and Bishop, {Christopher W.} and Hong, {Ellen S.} and Mykyta Artomov and Hubert, {Christopher G.} and Prajwal Rajappa and Connor, {James R.} and Fox, {Paul L.} and Kristensen, {Bjarne W.} and Lathia, {Justin D.}",
note = "Publisher Copyright: {\textcopyright} 2023 The Author(s). Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.",
year = "2024",
doi = "10.1093/noajnl/vdad154",
language = "English",
volume = "6",
journal = "Neuro-Oncology Advances",
issn = "2632-2498",
publisher = "Oxford University Press",
number = "1",

}

RIS

TY - JOUR

T1 - Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses

AU - Troike, Katie M.

AU - Wang, Sabrina Z.

AU - Silver, Daniel J.

AU - Lee, Juyeun

AU - Mulkearns-Hubert, Erin E.

AU - Hajdari, Nicole

AU - Ghosh, Prabar K.

AU - Kay, Kristen E.

AU - Beilis, Julia L.

AU - Mitchell, Sofia E.

AU - Bishop, Christopher W.

AU - Hong, Ellen S.

AU - Artomov, Mykyta

AU - Hubert, Christopher G.

AU - Rajappa, Prajwal

AU - Connor, James R.

AU - Fox, Paul L.

AU - Kristensen, Bjarne W.

AU - Lathia, Justin D.

N1 - Publisher Copyright: © 2023 The Author(s). Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.

PY - 2024

Y1 - 2024

N2 - Background: Glioblastoma (GBM) displays alterations in iron that drive proliferation and tumor growth. Iron regulation is complex and involves many regulatory mechanisms, including the homeostatic iron regulator (HFE) gene, which encodes the homeostatic iron regulatory protein. While HFE is upregulated in GBM and correlates with poor survival outcomes, the function of HFE in GBM remains unclear. Methods: We interrogated the impact of cell-intrinsic Hfe expression on proliferation and survival of intracranially implanted animals through genetic gain- and loss-of-function approaches in syngeneic mouse glioma models, along with in vivo immune assessments. We also determined the expression of iron-associated genes and their relationship to survival in GBM using public data sets and used transcriptional profiling to identify differentially expressed pathways in control compared to Hfe-knockdown cells. Results: Overexpression of Hfe accelerated GBM proliferation and reduced animal survival, whereas suppression of Hfe induced apoptotic cell death and extended survival, which was more pronounced in females and associated with attenuation of natural killer cells and CD8+ T cell activity. Analysis of iron gene signatures in Hfe-knockdown cells revealed alterations in the expression of several iron-associated genes, suggesting global disruption of intracellular iron homeostasis. Further analysis of differentially expressed pathways revealed oxidative stress as the top pathway upregulated following Hfe loss. Hfe knockdown indeed resulted in enhanced 55Fe uptake and generation of reactive oxygen species. Conclusions: These findings reveal an essential function for HFE in GBM cell growth and survival, as well as a sex-specific interaction with the immune response.

AB - Background: Glioblastoma (GBM) displays alterations in iron that drive proliferation and tumor growth. Iron regulation is complex and involves many regulatory mechanisms, including the homeostatic iron regulator (HFE) gene, which encodes the homeostatic iron regulatory protein. While HFE is upregulated in GBM and correlates with poor survival outcomes, the function of HFE in GBM remains unclear. Methods: We interrogated the impact of cell-intrinsic Hfe expression on proliferation and survival of intracranially implanted animals through genetic gain- and loss-of-function approaches in syngeneic mouse glioma models, along with in vivo immune assessments. We also determined the expression of iron-associated genes and their relationship to survival in GBM using public data sets and used transcriptional profiling to identify differentially expressed pathways in control compared to Hfe-knockdown cells. Results: Overexpression of Hfe accelerated GBM proliferation and reduced animal survival, whereas suppression of Hfe induced apoptotic cell death and extended survival, which was more pronounced in females and associated with attenuation of natural killer cells and CD8+ T cell activity. Analysis of iron gene signatures in Hfe-knockdown cells revealed alterations in the expression of several iron-associated genes, suggesting global disruption of intracellular iron homeostasis. Further analysis of differentially expressed pathways revealed oxidative stress as the top pathway upregulated following Hfe loss. Hfe knockdown indeed resulted in enhanced 55Fe uptake and generation of reactive oxygen species. Conclusions: These findings reveal an essential function for HFE in GBM cell growth and survival, as well as a sex-specific interaction with the immune response.

KW - glioblastoma

KW - HFE

KW - iron

KW - reactive oxygen species

KW - sex difference

U2 - 10.1093/noajnl/vdad154

DO - 10.1093/noajnl/vdad154

M3 - Journal article

C2 - 38239626

AN - SCOPUS:85182752963

VL - 6

JO - Neuro-Oncology Advances

JF - Neuro-Oncology Advances

SN - 2632-2498

IS - 1

M1 - vdad154

ER -

ID: 381059167