Please use this identifier to cite or link to this item: https://doi.org/10.7150/thno.72297
DC FieldValue
dc.titleFAM3C in circulating tumor-derived extracellular vesicles promotes non-small cell lung cancer growth in secondary sites
dc.contributor.authorThuya, WL
dc.contributor.authorKong, LR
dc.contributor.authorSyn, NL
dc.contributor.authorDing, LW
dc.contributor.authorCheow, ESH
dc.contributor.authorWong, RTX
dc.contributor.authorWang, T
dc.contributor.authorGoh, RMWJ
dc.contributor.authorSong, H
dc.contributor.authorJayasinghe, MK
dc.contributor.authorLe, MTN
dc.contributor.authorHu, JC
dc.contributor.authorYong, WP
dc.contributor.authorLee, SC
dc.contributor.authorWong, ALA
dc.contributor.authorSethi, G
dc.contributor.authorHung, HT
dc.contributor.authorHo, PCL
dc.contributor.authorThiery, JP
dc.contributor.authorSze, SK
dc.contributor.authorGuo, T
dc.contributor.authorSoo, RA
dc.contributor.authorYang, H
dc.contributor.authorLim, YC
dc.contributor.authorWang, L
dc.contributor.authorGoh, BC
dc.date.accessioned2023-03-01T09:18:03Z
dc.date.available2023-03-01T09:18:03Z
dc.date.issued2023-01-01
dc.identifier.citationThuya, WL, Kong, LR, Syn, NL, Ding, LW, Cheow, ESH, Wong, RTX, Wang, T, Goh, RMWJ, Song, H, Jayasinghe, MK, Le, MTN, Hu, JC, Yong, WP, Lee, SC, Wong, ALA, Sethi, G, Hung, HT, Ho, PCL, Thiery, JP, Sze, SK, Guo, T, Soo, RA, Yang, H, Lim, YC, Wang, L, Goh, BC (2023-01-01). FAM3C in circulating tumor-derived extracellular vesicles promotes non-small cell lung cancer growth in secondary sites. Theranostics 13 (2) : 621-638. ScholarBank@NUS Repository. https://doi.org/10.7150/thno.72297
dc.identifier.issn1838-7640
dc.identifier.urihttps://scholarbank.nus.edu.sg/handle/10635/237759
dc.description.abstractRationale: Metastasis is a complex process with a molecular underpinning that remains unclear. We hypothesize that cargo proteins conducted by extracellular vesicles (EVs) released from tumors may confer growth and metastasis potential on recipient cells. Here, we report that a cytokine-like secreted protein, FAM3C, contributes to late-stage lung tumor progression. Methods: EV protein profiling was conducted with an unbiased proteomic mass spectrometry analysis on non-small cell lung cancer (NSCLC) and normal lung fibroblast cell lines. Expression of FAM3C was confirmed in a panel of NSCLC cell lines, and correlated to the invasive and metastatic potentials. Functional phenotype of endogenous FAM3C and tumor-derived EVs (TDEs) were further investigated using various biological approaches in RNA and protein levels. Metastasis potential of TDEs secreted by FAM3C-overexpressing carcinoma cells was validated in mouse models. Results: Transcriptomic meta-analysis of pan-cancer datasets confirmed the overexpression of FAM3C - a gene encoding for interleukin-like EMT inducer (ILEI) - in NSCLC tumors, with strong association with poor patient prognosis and cancer metastasis. Aberrant expression of FAM3C in lung carcinoma cells enhances cellular transformation and promotes distant lung tumor colonization. In addition, higher FAM3C concentrations were detected in EVs extracted from plasma samples of NSCLC patients compared to those of healthy subjects. More importantly, we defined a hitherto-unknown mode of microenvironmental crosstalk involving FAM3C in EVs, whereby the delivery and uptake of FAM3C via TDEs enhances oncogenic signaling - in recipient cells that phenocopies the cell-endogenous overexpression of FAM3C. The oncogenicity transduced by FAM3C is executed via a novel interaction with the Ras-related protein RalA, triggering the downstream activation of the Src/Stat3 signaling cascade. Conclusions: Our study describes a novel mechanism for FAM3C-driven carcinogenesis and shed light on EV FAM3C as a driver for metastatic lung tumors that could be exploited for cancer therapeutics.
dc.publisherIvyspring International Publisher
dc.sourceElements
dc.subjectFAM3C
dc.subjectnon-small cell lung carcinoma
dc.subjectpredictive biomarker
dc.subjecttumor metastasis.
dc.subjecttumor-derived extracellular vesicles
dc.subjectAnimals
dc.subjectHumans
dc.subjectMice
dc.subjectCarcinoma, Non-Small-Cell Lung
dc.subjectCell Line, Tumor
dc.subjectExtracellular Vesicles
dc.subjectLung Neoplasms
dc.subjectProteomics
dc.subjectCarcinogenesis
dc.typeArticle
dc.date.updated2023-03-01T06:35:56Z
dc.contributor.departmentCANCER SCIENCE INSTITUTE OF SINGAPORE
dc.contributor.departmentMEDICINE
dc.contributor.departmentPHARMACOLOGY
dc.contributor.departmentPHARMACY
dc.contributor.departmentBIOCHEMISTRY
dc.contributor.departmentBIOLOGY (NU)
dc.description.doi10.7150/thno.72297
dc.description.sourcetitleTheranostics
dc.description.volume13
dc.description.issue2
dc.description.page621-638
dc.published.statePublished
dc.description.redepositcompleted
Appears in Collections:Staff Publications
Elements

Show simple item record
Files in This Item:
File Description SizeFormatAccess SettingsVersion 
FAM3C in circulating tumor-derived extracellular vesicles promotes non-small cell lung cancer growth in secondary sites.pdf10.05 MBAdobe PDF

OPEN

NoneView/Download
FAM3C in circulating tumor-derived extracellular vesicles promotes non-small cell lung cancer growth in secondary sites.pdf10.05 MBAdobe PDF

OPEN

NoneView/Download

Google ScholarTM

Check

Altmetric


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.