Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancer

Breast cancer (BC) is one of the most common malignancies among women worldwide. It is necessary to search for improvement in diagnosis and treatment methods to improve the prognosis. Protein kinase, membrane associated tyrosine/threonine 1 (PKMYT1), a member of the Wee family of protein kinases, ha...

Full description

Bibliographic Details
Main Authors: Huihui Li, Li Wang, Wei Zhang, Youting Dong, Yefeng Cai, Xiaoli Huang, Xubin Dong
Format: Article
Language:English
Published: Frontiers Media S.A. 2023-01-01
Series:Frontiers in Oncology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fonc.2022.1002186/full
_version_ 1811175331422273536
author Huihui Li
Li Wang
Wei Zhang
Youting Dong
Yefeng Cai
Yefeng Cai
Xiaoli Huang
Xiaoli Huang
Xubin Dong
author_facet Huihui Li
Li Wang
Wei Zhang
Youting Dong
Yefeng Cai
Yefeng Cai
Xiaoli Huang
Xiaoli Huang
Xubin Dong
author_sort Huihui Li
collection DOAJ
description Breast cancer (BC) is one of the most common malignancies among women worldwide. It is necessary to search for improvement in diagnosis and treatment methods to improve the prognosis. Protein kinase, membrane associated tyrosine/threonine 1 (PKMYT1), a member of the Wee family of protein kinases, has been studied in some tumors except BC. This study has explored that PKMYT1 functional role by bioinformatics methods combined with local clinical samples and experiments. Comprehensive analysis showed that PKMYT1 expression was higher in BC tissues, especially in advanced patients than that in normal breast tissues. The expression of PKMYT1 was an independent determinant for BC patients’ prognosis when combined with the clinical features. In addition, based on multi-omics analysis, we found that the PKMYT1 expression was closely relevant to several oncogenic or tumor suppressor gene variants. The analysis of single-cell sequencing indicated that PKMYT1 expression was upregulated in triple-negative breast cancer (TNBC), consistent with the results of bulk RNA-sequencing. High PKMYT1 expression was correlated with a poor prognosis. Functional enrichment analysis revealed that PKMYT1 expression was associated with cell cycle-related, DNA replication-related, and cancer-related pathways. Further research revealed that PKMYT1 expression was linked to immune cell infiltration in the tumor microenvironment. Additionally, loss-of-function experiments in vitro were performed to investigate the role of PKMYT1. TNBC cell lines’ proliferation, migration, and invasion were inhibited when PKMYT1 expression was knock-down. Besides, the down-regulation of PKMYT1 induced apoptosis in vitro. As a result, PKMYT1 might be a biomarker for prognosis and a therapeutic target for TNBC.
first_indexed 2024-04-10T19:35:23Z
format Article
id doaj.art-3f93f922093045288f0342e2ad4660c4
institution Directory Open Access Journal
issn 2234-943X
language English
last_indexed 2024-04-10T19:35:23Z
publishDate 2023-01-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Oncology
spelling doaj.art-3f93f922093045288f0342e2ad4660c42023-01-30T07:28:31ZengFrontiers Media S.A.Frontiers in Oncology2234-943X2023-01-011210.3389/fonc.2022.10021861002186Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancerHuihui Li0Li Wang1Wei Zhang2Youting Dong3Yefeng Cai4Yefeng Cai5Xiaoli Huang6Xiaoli Huang7Xubin Dong8Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, ChinaDepartment of Gastroenterology, Wenzhou Central Hospital, Wenzhou, ChinaDepartment of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, ChinaShanghai Medical College, Fudan University, Shanghai, ChinaDepartment of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, ChinaDepartment of Thyroid Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, ChinaDepartment of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, ChinaDepartment of Thyroid Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, ChinaDepartment of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, ChinaBreast cancer (BC) is one of the most common malignancies among women worldwide. It is necessary to search for improvement in diagnosis and treatment methods to improve the prognosis. Protein kinase, membrane associated tyrosine/threonine 1 (PKMYT1), a member of the Wee family of protein kinases, has been studied in some tumors except BC. This study has explored that PKMYT1 functional role by bioinformatics methods combined with local clinical samples and experiments. Comprehensive analysis showed that PKMYT1 expression was higher in BC tissues, especially in advanced patients than that in normal breast tissues. The expression of PKMYT1 was an independent determinant for BC patients’ prognosis when combined with the clinical features. In addition, based on multi-omics analysis, we found that the PKMYT1 expression was closely relevant to several oncogenic or tumor suppressor gene variants. The analysis of single-cell sequencing indicated that PKMYT1 expression was upregulated in triple-negative breast cancer (TNBC), consistent with the results of bulk RNA-sequencing. High PKMYT1 expression was correlated with a poor prognosis. Functional enrichment analysis revealed that PKMYT1 expression was associated with cell cycle-related, DNA replication-related, and cancer-related pathways. Further research revealed that PKMYT1 expression was linked to immune cell infiltration in the tumor microenvironment. Additionally, loss-of-function experiments in vitro were performed to investigate the role of PKMYT1. TNBC cell lines’ proliferation, migration, and invasion were inhibited when PKMYT1 expression was knock-down. Besides, the down-regulation of PKMYT1 induced apoptosis in vitro. As a result, PKMYT1 might be a biomarker for prognosis and a therapeutic target for TNBC.https://www.frontiersin.org/articles/10.3389/fonc.2022.1002186/fullPKMYT1prognosismulti-omicsTNBC subtypesimmune infiltration
spellingShingle Huihui Li
Li Wang
Wei Zhang
Youting Dong
Yefeng Cai
Yefeng Cai
Xiaoli Huang
Xiaoli Huang
Xubin Dong
Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancer
Frontiers in Oncology
PKMYT1
prognosis
multi-omics
TNBC subtypes
immune infiltration
title Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancer
title_full Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancer
title_fullStr Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancer
title_full_unstemmed Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancer
title_short Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancer
title_sort overexpression of pkmyt1 associated with poor prognosis and immune infiltration may serve as a target in triple negative breast cancer
topic PKMYT1
prognosis
multi-omics
TNBC subtypes
immune infiltration
url https://www.frontiersin.org/articles/10.3389/fonc.2022.1002186/full
work_keys_str_mv AT huihuili overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer
AT liwang overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer
AT weizhang overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer
AT youtingdong overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer
AT yefengcai overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer
AT yefengcai overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer
AT xiaolihuang overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer
AT xiaolihuang overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer
AT xubindong overexpressionofpkmyt1associatedwithpoorprognosisandimmuneinfiltrationmayserveasatargetintriplenegativebreastcancer