LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1–TINCR-USP20-PD-L1 axis

Abstract Although programmed death-ligand 1 (PD-L1) inhibitors have achieved some therapeutic success in breast cancer, their efficacy is limited by low therapeutic response rates, which is closely related to the immune escape of breast cancer cells. Tissue differentiation inducing non-protein codin...

Full description

Bibliographic Details
Main Authors: Qin Wang, Guozheng Li, Xin Ma, Lei Liu, Jiena Liu, Yanling Yin, Hui Li, Yihai Chen, Xin Zhang, Lei Zhang, Liyang Sun, Jing Ai, Shouping Xu
Format: Article
Language:English
Published: Nature Publishing Group 2023-02-01
Series:Cell Death and Disease
Online Access:https://doi.org/10.1038/s41419-023-05609-2
_version_ 1811171482943881216
author Qin Wang
Guozheng Li
Xin Ma
Lei Liu
Jiena Liu
Yanling Yin
Hui Li
Yihai Chen
Xin Zhang
Lei Zhang
Liyang Sun
Jing Ai
Shouping Xu
author_facet Qin Wang
Guozheng Li
Xin Ma
Lei Liu
Jiena Liu
Yanling Yin
Hui Li
Yihai Chen
Xin Zhang
Lei Zhang
Liyang Sun
Jing Ai
Shouping Xu
author_sort Qin Wang
collection DOAJ
description Abstract Although programmed death-ligand 1 (PD-L1) inhibitors have achieved some therapeutic success in breast cancer, their efficacy is limited by low therapeutic response rates, which is closely related to the immune escape of breast cancer cells. Tissue differentiation inducing non-protein coding RNA (TINCR), a long non-coding RNA, as an oncogenic gene associated with the progression of various malignant tumors, including breast cancer; however, the role of TINCR in tumor immunity, especially in breast cancer, remains unclear. We confirmed that TINCR upregulated PD-L1 expression in vivo and in vitro, and promoted the progression of breast cancer. Next, we revealed that TINCR knockdown can significantly improve the therapeutic effect of PD-L1 inhibitors in breast cancer in vivo. Mechanistically, TINCR recruits DNMT1 to promote the methylation of miR-199a-5p loci and inhibit its transcription. Furthermore, in the cytoplasm, TINCR potentially acts as a molecular sponge of miR-199a-5p and upregulates the stability of USP20 mRNA through a competing endogenous RNA (ceRNA) regulatory mechanism, thus promoting PD-L1 expression by decreasing its ubiquitination level. IFN-γ stimulation activates STAT1 by phosphorylation, which migrates into the nucleus to promote TINCR transcription. This is the first study to describe the regulatory role of TINCR in breast cancer tumor immunity, broadening the current paradigm of the functional diversity of TINCR in tumor biology. In addition, our study provides new research directions and potential therapeutic targets for PD-L1 inhibitors in breast cancer.
first_indexed 2024-04-10T17:15:45Z
format Article
id doaj.art-0599f9cbed6740339bf6ce19e9f387c1
institution Directory Open Access Journal
issn 2041-4889
language English
last_indexed 2024-04-10T17:15:45Z
publishDate 2023-02-01
publisher Nature Publishing Group
record_format Article
series Cell Death and Disease
spelling doaj.art-0599f9cbed6740339bf6ce19e9f387c12023-02-05T12:25:29ZengNature Publishing GroupCell Death and Disease2041-48892023-02-0114211210.1038/s41419-023-05609-2LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1–TINCR-USP20-PD-L1 axisQin Wang0Guozheng Li1Xin Ma2Lei Liu3Jiena Liu4Yanling Yin5Hui Li6Yihai Chen7Xin Zhang8Lei Zhang9Liyang Sun10Jing Ai11Shouping Xu12Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical UniversityDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Breast Surgery, Harbin Medical University Cancer HospitalDepartment of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical UniversityDepartment of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical UniversityHeilongjiang Academy of Medical SciencesAbstract Although programmed death-ligand 1 (PD-L1) inhibitors have achieved some therapeutic success in breast cancer, their efficacy is limited by low therapeutic response rates, which is closely related to the immune escape of breast cancer cells. Tissue differentiation inducing non-protein coding RNA (TINCR), a long non-coding RNA, as an oncogenic gene associated with the progression of various malignant tumors, including breast cancer; however, the role of TINCR in tumor immunity, especially in breast cancer, remains unclear. We confirmed that TINCR upregulated PD-L1 expression in vivo and in vitro, and promoted the progression of breast cancer. Next, we revealed that TINCR knockdown can significantly improve the therapeutic effect of PD-L1 inhibitors in breast cancer in vivo. Mechanistically, TINCR recruits DNMT1 to promote the methylation of miR-199a-5p loci and inhibit its transcription. Furthermore, in the cytoplasm, TINCR potentially acts as a molecular sponge of miR-199a-5p and upregulates the stability of USP20 mRNA through a competing endogenous RNA (ceRNA) regulatory mechanism, thus promoting PD-L1 expression by decreasing its ubiquitination level. IFN-γ stimulation activates STAT1 by phosphorylation, which migrates into the nucleus to promote TINCR transcription. This is the first study to describe the regulatory role of TINCR in breast cancer tumor immunity, broadening the current paradigm of the functional diversity of TINCR in tumor biology. In addition, our study provides new research directions and potential therapeutic targets for PD-L1 inhibitors in breast cancer.https://doi.org/10.1038/s41419-023-05609-2
spellingShingle Qin Wang
Guozheng Li
Xin Ma
Lei Liu
Jiena Liu
Yanling Yin
Hui Li
Yihai Chen
Xin Zhang
Lei Zhang
Liyang Sun
Jing Ai
Shouping Xu
LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1–TINCR-USP20-PD-L1 axis
Cell Death and Disease
title LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1–TINCR-USP20-PD-L1 axis
title_full LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1–TINCR-USP20-PD-L1 axis
title_fullStr LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1–TINCR-USP20-PD-L1 axis
title_full_unstemmed LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1–TINCR-USP20-PD-L1 axis
title_short LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1–TINCR-USP20-PD-L1 axis
title_sort lncrna tincr impairs the efficacy of immunotherapy against breast cancer by recruiting dnmt1 and downregulating mir 199a 5p via the stat1 tincr usp20 pd l1 axis
url https://doi.org/10.1038/s41419-023-05609-2
work_keys_str_mv AT qinwang lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT guozhengli lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT xinma lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT leiliu lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT jienaliu lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT yanlingyin lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT huili lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT yihaichen lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT xinzhang lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT leizhang lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT liyangsun lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT jingai lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis
AT shoupingxu lncrnatincrimpairstheefficacyofimmunotherapyagainstbreastcancerbyrecruitingdnmt1anddownregulatingmir199a5pviathestat1tincrusp20pdl1axis