Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts

Research output: Contribution to journalLetterResearchpeer-review

Standard

Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts. / Eckert, Mark A; Coscia, Fabian; Chryplewicz, Agnieszka; Chang, Jae Won; Hernandez, Kyle M; Pan, Shawn; Tienda, Samantha M; Nahotko, Dominik A; Li, Gang; Blaženović, Ivana; Lastra, Ricardo R; Curtis, Marion; Yamada, S Diane; Perets, Ruth; McGregor, Stephanie M; Andrade, Jorge; Fiehn, Oliver; Moellering, Raymond E; Mann, Matthias; Lengyel, Ernst.

In: Nature, Vol. 569, No. 7758, 2019, p. 723-728.

Research output: Contribution to journalLetterResearchpeer-review

Harvard

Eckert, MA, Coscia, F, Chryplewicz, A, Chang, JW, Hernandez, KM, Pan, S, Tienda, SM, Nahotko, DA, Li, G, Blaženović, I, Lastra, RR, Curtis, M, Yamada, SD, Perets, R, McGregor, SM, Andrade, J, Fiehn, O, Moellering, RE, Mann, M & Lengyel, E 2019, 'Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts', Nature, vol. 569, no. 7758, pp. 723-728. https://doi.org/10.1038/s41586-019-1173-8

APA

Eckert, M. A., Coscia, F., Chryplewicz, A., Chang, J. W., Hernandez, K. M., Pan, S., Tienda, S. M., Nahotko, D. A., Li, G., Blaženović, I., Lastra, R. R., Curtis, M., Yamada, S. D., Perets, R., McGregor, S. M., Andrade, J., Fiehn, O., Moellering, R. E., Mann, M., & Lengyel, E. (2019). Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts. Nature, 569(7758), 723-728. https://doi.org/10.1038/s41586-019-1173-8

Vancouver

Eckert MA, Coscia F, Chryplewicz A, Chang JW, Hernandez KM, Pan S et al. Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts. Nature. 2019;569(7758):723-728. https://doi.org/10.1038/s41586-019-1173-8

Author

Eckert, Mark A ; Coscia, Fabian ; Chryplewicz, Agnieszka ; Chang, Jae Won ; Hernandez, Kyle M ; Pan, Shawn ; Tienda, Samantha M ; Nahotko, Dominik A ; Li, Gang ; Blaženović, Ivana ; Lastra, Ricardo R ; Curtis, Marion ; Yamada, S Diane ; Perets, Ruth ; McGregor, Stephanie M ; Andrade, Jorge ; Fiehn, Oliver ; Moellering, Raymond E ; Mann, Matthias ; Lengyel, Ernst. / Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts. In: Nature. 2019 ; Vol. 569, No. 7758. pp. 723-728.

Bibtex

@article{2e42e2b6754e474fa7d6f657ffe8834e,
title = "Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts",
abstract = "High-grade serous carcinoma has a poor prognosis, owing primarily to its early dissemination throughout the abdominal cavity. Genomic and proteomic approaches have provided snapshots of the proteogenomics of ovarian cancer1,2, but a systematic examination of both the tumour and stromal compartments is critical in understanding ovarian cancer metastasis. Here we develop a label-free proteomic workflow to analyse as few as 5,000 formalin-fixed, paraffin-embedded cells microdissected from each compartment. The tumour proteome was stable during progression from in situ lesions to metastatic disease; however, the metastasis-associated stroma was characterized by a highly conserved proteomic signature, prominently including the methyltransferase nicotinamide N-methyltransferase (NNMT) and several of the proteins that it regulates. Stromal NNMT expression was necessary and sufficient for functional aspects of the cancer-associated fibroblast (CAF) phenotype, including the expression of CAF markers and the secretion of cytokines and oncogenic extracellular matrix. Stromal NNMT expression supported ovarian cancer migration, proliferation and in vivo growth and metastasis. Expression of NNMT in CAFs led to depletion of S-adenosyl methionine and reduction in histone methylation associated with widespread gene expression changes in the tumour stroma. This work supports the use of ultra-low-input proteomics to identify candidate drivers of disease phenotypes. NNMT is a central, metabolic regulator of CAF differentiation and cancer progression in the stroma that may be therapeutically targeted.",
author = "Eckert, {Mark A} and Fabian Coscia and Agnieszka Chryplewicz and Chang, {Jae Won} and Hernandez, {Kyle M} and Shawn Pan and Tienda, {Samantha M} and Nahotko, {Dominik A} and Gang Li and Ivana Bla{\v z}enovi{\'c} and Lastra, {Ricardo R} and Marion Curtis and Yamada, {S Diane} and Ruth Perets and McGregor, {Stephanie M} and Jorge Andrade and Oliver Fiehn and Moellering, {Raymond E} and Matthias Mann and Ernst Lengyel",
year = "2019",
doi = "10.1038/s41586-019-1173-8",
language = "English",
volume = "569",
pages = "723--728",
journal = "Nature",
issn = "0028-0836",
publisher = "nature publishing group",
number = "7758",

}

RIS

TY - JOUR

T1 - Proteomics reveals NNMT as a master metabolic regulator of cancer-associated fibroblasts

AU - Eckert, Mark A

AU - Coscia, Fabian

AU - Chryplewicz, Agnieszka

AU - Chang, Jae Won

AU - Hernandez, Kyle M

AU - Pan, Shawn

AU - Tienda, Samantha M

AU - Nahotko, Dominik A

AU - Li, Gang

AU - Blaženović, Ivana

AU - Lastra, Ricardo R

AU - Curtis, Marion

AU - Yamada, S Diane

AU - Perets, Ruth

AU - McGregor, Stephanie M

AU - Andrade, Jorge

AU - Fiehn, Oliver

AU - Moellering, Raymond E

AU - Mann, Matthias

AU - Lengyel, Ernst

PY - 2019

Y1 - 2019

N2 - High-grade serous carcinoma has a poor prognosis, owing primarily to its early dissemination throughout the abdominal cavity. Genomic and proteomic approaches have provided snapshots of the proteogenomics of ovarian cancer1,2, but a systematic examination of both the tumour and stromal compartments is critical in understanding ovarian cancer metastasis. Here we develop a label-free proteomic workflow to analyse as few as 5,000 formalin-fixed, paraffin-embedded cells microdissected from each compartment. The tumour proteome was stable during progression from in situ lesions to metastatic disease; however, the metastasis-associated stroma was characterized by a highly conserved proteomic signature, prominently including the methyltransferase nicotinamide N-methyltransferase (NNMT) and several of the proteins that it regulates. Stromal NNMT expression was necessary and sufficient for functional aspects of the cancer-associated fibroblast (CAF) phenotype, including the expression of CAF markers and the secretion of cytokines and oncogenic extracellular matrix. Stromal NNMT expression supported ovarian cancer migration, proliferation and in vivo growth and metastasis. Expression of NNMT in CAFs led to depletion of S-adenosyl methionine and reduction in histone methylation associated with widespread gene expression changes in the tumour stroma. This work supports the use of ultra-low-input proteomics to identify candidate drivers of disease phenotypes. NNMT is a central, metabolic regulator of CAF differentiation and cancer progression in the stroma that may be therapeutically targeted.

AB - High-grade serous carcinoma has a poor prognosis, owing primarily to its early dissemination throughout the abdominal cavity. Genomic and proteomic approaches have provided snapshots of the proteogenomics of ovarian cancer1,2, but a systematic examination of both the tumour and stromal compartments is critical in understanding ovarian cancer metastasis. Here we develop a label-free proteomic workflow to analyse as few as 5,000 formalin-fixed, paraffin-embedded cells microdissected from each compartment. The tumour proteome was stable during progression from in situ lesions to metastatic disease; however, the metastasis-associated stroma was characterized by a highly conserved proteomic signature, prominently including the methyltransferase nicotinamide N-methyltransferase (NNMT) and several of the proteins that it regulates. Stromal NNMT expression was necessary and sufficient for functional aspects of the cancer-associated fibroblast (CAF) phenotype, including the expression of CAF markers and the secretion of cytokines and oncogenic extracellular matrix. Stromal NNMT expression supported ovarian cancer migration, proliferation and in vivo growth and metastasis. Expression of NNMT in CAFs led to depletion of S-adenosyl methionine and reduction in histone methylation associated with widespread gene expression changes in the tumour stroma. This work supports the use of ultra-low-input proteomics to identify candidate drivers of disease phenotypes. NNMT is a central, metabolic regulator of CAF differentiation and cancer progression in the stroma that may be therapeutically targeted.

U2 - 10.1038/s41586-019-1173-8

DO - 10.1038/s41586-019-1173-8

M3 - Letter

C2 - 31043742

VL - 569

SP - 723

EP - 728

JO - Nature

JF - Nature

SN - 0028-0836

IS - 7758

ER -

ID: 226913415