Cargando…

Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets

BACKGROUND: As a rare pathologic subtype, small cell carcinoma of the cervix (SCCC) is characterized by extensive aggressiveness and resistance to current therapies. To date, our knowledge of SCCC origin and progression is limited and sometimes even controversial. Herein, we explored the whole-prote...

Descripción completa

Detalles Bibliográficos
Autores principales: Qiu, Haifeng, Su, Ning, Wang, Jing, Yan, Shuping, Li, Jing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10082492/
https://www.ncbi.nlm.nih.gov/pubmed/37031178
http://dx.doi.org/10.1186/s12014-023-09408-x
_version_ 1785021324299599872
author Qiu, Haifeng
Su, Ning
Wang, Jing
Yan, Shuping
Li, Jing
author_facet Qiu, Haifeng
Su, Ning
Wang, Jing
Yan, Shuping
Li, Jing
author_sort Qiu, Haifeng
collection PubMed
description BACKGROUND: As a rare pathologic subtype, small cell carcinoma of the cervix (SCCC) is characterized by extensive aggressiveness and resistance to current therapies. To date, our knowledge of SCCC origin and progression is limited and sometimes even controversial. Herein, we explored the whole-protein expression profiles in a panel of SCCC cases, aiming to provide more evidence for the precise diagnosis and targeting therapy. METHODS: Eighteen SCCC samples and six matched normal cervix tissues were collected from January 2013 to December 2017. Data independent acquisition mass spectrometry (DIA) was performed to discriminate the different proteins (DEPs) associated with SCCC. The expression of CDN2A and SYP in corresponding SCCC tissues was verified using immunohistochemistry. GO and KEGG enrichment analyses were used to identify the key DEPs related to SCCC development and tumor recurrence. RESULTS: As a result, 1311 DEPs were identified in SCCC tissues (780 up-regulated and 531 down-regulated). In up-regulated DEPs, both GO analysis and KEGG analysis showed the most enriched were related to DNA replication (including nuclear DNA replication, DNA-dependent DNA replication, and cell cycle DNA replication), indicating the prosperous proliferation in SCCC. As for the down-regulated DEPs, GO analysis showed that the most enriched functions were associated with extracellular matrix collagen-containing extracellular matrix. KEGG analysis revealed that the DEPs were enriched in Complement and coagulation cascades, proteoglycans in cancer, and focal adhesion-related pathways. Down-regulation of these proteins could enhance the mobility of cancer cells and establish a favorable microenvironment for tumor metastasis, which might be accounted for the frequent local and distant metastasis in SCCC. Surprisingly, the blood vessels and circulatory system exhibit a down-regulation in SCCC, which might be partly responsible for its resistance to anti-angiogenic regimens. In the stratification analysis of early-stage tumors, a group of enzymes involved in the cancer metabolism was discriminated in these recurrence cases. CONCLUSIONS: Using quantitative proteomics analysis, we first reported the whole-protein expression profiles in SCCC. Significant alterations were found in proteins associated with the enhancement of DNA replication and cellular motility. Besides the association with mitosis, a unique metabolic feature was detected in cases with tumor recurrence. These findings provided novel targets for disease surveillance and treatments, which warranted further validation in the future. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12014-023-09408-x.
format Online
Article
Text
id pubmed-10082492
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-100824922023-04-09 Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets Qiu, Haifeng Su, Ning Wang, Jing Yan, Shuping Li, Jing Clin Proteomics Research BACKGROUND: As a rare pathologic subtype, small cell carcinoma of the cervix (SCCC) is characterized by extensive aggressiveness and resistance to current therapies. To date, our knowledge of SCCC origin and progression is limited and sometimes even controversial. Herein, we explored the whole-protein expression profiles in a panel of SCCC cases, aiming to provide more evidence for the precise diagnosis and targeting therapy. METHODS: Eighteen SCCC samples and six matched normal cervix tissues were collected from January 2013 to December 2017. Data independent acquisition mass spectrometry (DIA) was performed to discriminate the different proteins (DEPs) associated with SCCC. The expression of CDN2A and SYP in corresponding SCCC tissues was verified using immunohistochemistry. GO and KEGG enrichment analyses were used to identify the key DEPs related to SCCC development and tumor recurrence. RESULTS: As a result, 1311 DEPs were identified in SCCC tissues (780 up-regulated and 531 down-regulated). In up-regulated DEPs, both GO analysis and KEGG analysis showed the most enriched were related to DNA replication (including nuclear DNA replication, DNA-dependent DNA replication, and cell cycle DNA replication), indicating the prosperous proliferation in SCCC. As for the down-regulated DEPs, GO analysis showed that the most enriched functions were associated with extracellular matrix collagen-containing extracellular matrix. KEGG analysis revealed that the DEPs were enriched in Complement and coagulation cascades, proteoglycans in cancer, and focal adhesion-related pathways. Down-regulation of these proteins could enhance the mobility of cancer cells and establish a favorable microenvironment for tumor metastasis, which might be accounted for the frequent local and distant metastasis in SCCC. Surprisingly, the blood vessels and circulatory system exhibit a down-regulation in SCCC, which might be partly responsible for its resistance to anti-angiogenic regimens. In the stratification analysis of early-stage tumors, a group of enzymes involved in the cancer metabolism was discriminated in these recurrence cases. CONCLUSIONS: Using quantitative proteomics analysis, we first reported the whole-protein expression profiles in SCCC. Significant alterations were found in proteins associated with the enhancement of DNA replication and cellular motility. Besides the association with mitosis, a unique metabolic feature was detected in cases with tumor recurrence. These findings provided novel targets for disease surveillance and treatments, which warranted further validation in the future. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12014-023-09408-x. BioMed Central 2023-04-08 /pmc/articles/PMC10082492/ /pubmed/37031178 http://dx.doi.org/10.1186/s12014-023-09408-x Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Qiu, Haifeng
Su, Ning
Wang, Jing
Yan, Shuping
Li, Jing
Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets
title Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets
title_full Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets
title_fullStr Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets
title_full_unstemmed Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets
title_short Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets
title_sort quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10082492/
https://www.ncbi.nlm.nih.gov/pubmed/37031178
http://dx.doi.org/10.1186/s12014-023-09408-x
work_keys_str_mv AT qiuhaifeng quantitativeproteomicsanalysisinsmallcellcarcinomaofcervixrevealsnoveltherapeutictargets
AT suning quantitativeproteomicsanalysisinsmallcellcarcinomaofcervixrevealsnoveltherapeutictargets
AT wangjing quantitativeproteomicsanalysisinsmallcellcarcinomaofcervixrevealsnoveltherapeutictargets
AT yanshuping quantitativeproteomicsanalysisinsmallcellcarcinomaofcervixrevealsnoveltherapeutictargets
AT lijing quantitativeproteomicsanalysisinsmallcellcarcinomaofcervixrevealsnoveltherapeutictargets