Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity

Tumor protein phosphorylation analysis may provide insight into intracellular signaling networks underlying tumor behavior, revealing diagnostic, prognostic or therapeutic information. Human tumors collected by The Cancer Genome Atlas program potentially offer the opportunity to characterize activat...

Full description

Bibliographic Details
Main Authors: Slebos, R. J. C., Shaddox, K., Wiles, K., Washington, M. K., Herline, A. J., Levine, D. A., Liebler, D. C., Gajadhar, Aaron, Johnson, Hannah, White, Forest M.
Other Authors: Massachusetts Institute of Technology. Department of Biological Engineering
Format: Article
Language:en_US
Published: American Association for Cancer Research 2017
Online Access:http://hdl.handle.net/1721.1/107688
https://orcid.org/0000-0002-5782-9544
https://orcid.org/0000-0002-1545-1651
_version_ 1811071230790336512
author Slebos, R. J. C.
Shaddox, K.
Wiles, K.
Washington, M. K.
Herline, A. J.
Levine, D. A.
Liebler, D. C.
Gajadhar, Aaron
Johnson, Hannah
White, Forest M.
author2 Massachusetts Institute of Technology. Department of Biological Engineering
author_facet Massachusetts Institute of Technology. Department of Biological Engineering
Slebos, R. J. C.
Shaddox, K.
Wiles, K.
Washington, M. K.
Herline, A. J.
Levine, D. A.
Liebler, D. C.
Gajadhar, Aaron
Johnson, Hannah
White, Forest M.
author_sort Slebos, R. J. C.
collection MIT
description Tumor protein phosphorylation analysis may provide insight into intracellular signaling networks underlying tumor behavior, revealing diagnostic, prognostic or therapeutic information. Human tumors collected by The Cancer Genome Atlas program potentially offer the opportunity to characterize activated networks driving tumor progression, in parallel with the genetic and transcriptional landscape already documented for these tumors. However, a critical question is whether cellular signaling networks can be reliably analyzed in surgical specimens, where freezing delays and spatial sampling disparities may potentially obscure physiologic signaling. To quantify the extent of these effects, we analyzed the stability of phosphotyrosine (pTyr) sites in ovarian and colon tumors collected under conditions of controlled ischemia and in the context of defined intratumoral sampling. Cold-ischemia produced a rapid, unpredictable, and widespread impact on tumor pTyr networks within 5 minutes of resection, altering up to 50% of pTyr sites by more than 2-fold. Effects on adhesion and migration, inflammatory response, proliferation, and stress response pathways were recapitulated in both ovarian and colon tumors. In addition, sampling of spatially distinct colon tumor biopsies revealed pTyr differences as dramatic as those associated with ischemic times, despite uniform protein expression profiles. Moreover, intratumoral spatial heterogeneity and pTyr dynamic response to ischemia varied dramatically between tumors collected from different patients. Overall, these findings reveal unforeseen phosphorylation complexity, thereby increasing the difficulty of extracting physiologically relevant pTyr signaling networks from archived tissue specimens. In light of this data, prospective tumor pTyr analysis will require appropriate sampling and collection protocols to preserve in vivo signaling features.
first_indexed 2024-09-23T08:47:58Z
format Article
id mit-1721.1/107688
institution Massachusetts Institute of Technology
language en_US
last_indexed 2024-09-23T08:47:58Z
publishDate 2017
publisher American Association for Cancer Research
record_format dspace
spelling mit-1721.1/1076882022-09-23T14:38:33Z Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity Slebos, R. J. C. Shaddox, K. Wiles, K. Washington, M. K. Herline, A. J. Levine, D. A. Liebler, D. C. Gajadhar, Aaron Johnson, Hannah White, Forest M. Massachusetts Institute of Technology. Department of Biological Engineering Koch Institute for Integrative Cancer Research at MIT Gajadhar, Aaron Johnson, Hannah White, Forest M Tumor protein phosphorylation analysis may provide insight into intracellular signaling networks underlying tumor behavior, revealing diagnostic, prognostic or therapeutic information. Human tumors collected by The Cancer Genome Atlas program potentially offer the opportunity to characterize activated networks driving tumor progression, in parallel with the genetic and transcriptional landscape already documented for these tumors. However, a critical question is whether cellular signaling networks can be reliably analyzed in surgical specimens, where freezing delays and spatial sampling disparities may potentially obscure physiologic signaling. To quantify the extent of these effects, we analyzed the stability of phosphotyrosine (pTyr) sites in ovarian and colon tumors collected under conditions of controlled ischemia and in the context of defined intratumoral sampling. Cold-ischemia produced a rapid, unpredictable, and widespread impact on tumor pTyr networks within 5 minutes of resection, altering up to 50% of pTyr sites by more than 2-fold. Effects on adhesion and migration, inflammatory response, proliferation, and stress response pathways were recapitulated in both ovarian and colon tumors. In addition, sampling of spatially distinct colon tumor biopsies revealed pTyr differences as dramatic as those associated with ischemic times, despite uniform protein expression profiles. Moreover, intratumoral spatial heterogeneity and pTyr dynamic response to ischemia varied dramatically between tumors collected from different patients. Overall, these findings reveal unforeseen phosphorylation complexity, thereby increasing the difficulty of extracting physiologically relevant pTyr signaling networks from archived tissue specimens. In light of this data, prospective tumor pTyr analysis will require appropriate sampling and collection protocols to preserve in vivo signaling features. National Institutes of Health (U.S.) (Grant U24 CA159988) 2017-03-24T14:46:25Z 2017-03-24T14:46:25Z 2016-09 2016-01 Article http://purl.org/eprint/type/JournalArticle 0008-5472 1538-7445 http://hdl.handle.net/1721.1/107688 Gajadhar, A. S. et al. “Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity.” Cancer Research 75.7 (2015): 1495–1503. https://orcid.org/0000-0002-5782-9544 https://orcid.org/0000-0002-1545-1651 en_US http://dx.doi.org/10.1158/0008-5472.can-14-2309 Cancer Research Creative Commons Attribution-Noncommercial-Share Alike http://creativecommons.org/licenses/by-nc-sa/4.0/ application/pdf American Association for Cancer Research PMC
spellingShingle Slebos, R. J. C.
Shaddox, K.
Wiles, K.
Washington, M. K.
Herline, A. J.
Levine, D. A.
Liebler, D. C.
Gajadhar, Aaron
Johnson, Hannah
White, Forest M.
Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity
title Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity
title_full Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity
title_fullStr Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity
title_full_unstemmed Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity
title_short Phosphotyrosine Signaling Analysis in Human Tumors Is Confounded by Systemic Ischemia-Driven Artifacts and Intra-Specimen Heterogeneity
title_sort phosphotyrosine signaling analysis in human tumors is confounded by systemic ischemia driven artifacts and intra specimen heterogeneity
url http://hdl.handle.net/1721.1/107688
https://orcid.org/0000-0002-5782-9544
https://orcid.org/0000-0002-1545-1651
work_keys_str_mv AT slebosrjc phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT shaddoxk phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT wilesk phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT washingtonmk phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT herlineaj phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT levineda phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT lieblerdc phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT gajadharaaron phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT johnsonhannah phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity
AT whiteforestm phosphotyrosinesignalinganalysisinhumantumorsisconfoundedbysystemicischemiadrivenartifactsandintraspecimenheterogeneity