Cargando…

Distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy

BACKGROUND: Endometriosis (EMs) is a chronic inflammatory condition that is highly heterogeneous. Current clinical staging fails to accurately predict drug responses and prognosis. In this study, we aimed to reveal the heterogeneity of ectopic lesions and investigate the possible underlying mechanis...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yuning, Chen, Yun, Xiao, Yinping, Ruan, Jingyao, Tian, Qi, Cheng, Qi, Chang, Kaikai, Yi, Xiaofang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10324653/
https://www.ncbi.nlm.nih.gov/pubmed/37426659
http://dx.doi.org/10.3389/fimmu.2023.1133672
_version_ 1785069188949213184
author Wang, Yuning
Chen, Yun
Xiao, Yinping
Ruan, Jingyao
Tian, Qi
Cheng, Qi
Chang, Kaikai
Yi, Xiaofang
author_facet Wang, Yuning
Chen, Yun
Xiao, Yinping
Ruan, Jingyao
Tian, Qi
Cheng, Qi
Chang, Kaikai
Yi, Xiaofang
author_sort Wang, Yuning
collection PubMed
description BACKGROUND: Endometriosis (EMs) is a chronic inflammatory condition that is highly heterogeneous. Current clinical staging fails to accurately predict drug responses and prognosis. In this study, we aimed to reveal the heterogeneity of ectopic lesions and investigate the possible underlying mechanisms using transcriptomic data and clinical information. METHODS: The EMs microarray dataset GSE141549 was obtained from the Gene Expression Omnibus database. Unsupervised hierarchical clustering was performed to identify EMs subtypes, which was followed by the functional enrichment analysis and estimation of immune infiltrates. Subtype-associated gene signatures were identified and further validated in other independent datasets, including GSE25628, E-MTAB-694, and GSE23339. Additionally, tissue microarrays (TMAs) were generated from premenopausal patients with EMs to investigate the potential clinical implications of the two identified subtypes. RESULTS: The unsupervised clustering analysis revealed that ectopic EMs lesions can be classified into two distinct subtypes: stroma-enriched (S1) and immune-enriched (S2). The functional analysis revealed that S1 correlated with fibroblast activation and extracellular matrix remodeling in the ectopic milieu, whereas S2 was characterized by the upregulation of immune pathways and a higher positive correlation with the immunotherapy response. Moreover, we identified a subtype signature composed of FHL1 and SORBS1, and constructed a subtype diagnostic model. Based on the cohort data from the TMAs, we found that S2 was strongly associated with the failure of/intolerance to hormone therapy. CONCLUSIONS: This study identified two distinct subtypes that are varyingly associated with hormone resistance, stroma-immunity, and molecular features, thereby highlighting the importance of this stromal-immune heterogeneity in identifying EMs subtypes and providing novel insights into future personalized hormone-free therapy in EMs.
format Online
Article
Text
id pubmed-10324653
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-103246532023-07-07 Distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy Wang, Yuning Chen, Yun Xiao, Yinping Ruan, Jingyao Tian, Qi Cheng, Qi Chang, Kaikai Yi, Xiaofang Front Immunol Immunology BACKGROUND: Endometriosis (EMs) is a chronic inflammatory condition that is highly heterogeneous. Current clinical staging fails to accurately predict drug responses and prognosis. In this study, we aimed to reveal the heterogeneity of ectopic lesions and investigate the possible underlying mechanisms using transcriptomic data and clinical information. METHODS: The EMs microarray dataset GSE141549 was obtained from the Gene Expression Omnibus database. Unsupervised hierarchical clustering was performed to identify EMs subtypes, which was followed by the functional enrichment analysis and estimation of immune infiltrates. Subtype-associated gene signatures were identified and further validated in other independent datasets, including GSE25628, E-MTAB-694, and GSE23339. Additionally, tissue microarrays (TMAs) were generated from premenopausal patients with EMs to investigate the potential clinical implications of the two identified subtypes. RESULTS: The unsupervised clustering analysis revealed that ectopic EMs lesions can be classified into two distinct subtypes: stroma-enriched (S1) and immune-enriched (S2). The functional analysis revealed that S1 correlated with fibroblast activation and extracellular matrix remodeling in the ectopic milieu, whereas S2 was characterized by the upregulation of immune pathways and a higher positive correlation with the immunotherapy response. Moreover, we identified a subtype signature composed of FHL1 and SORBS1, and constructed a subtype diagnostic model. Based on the cohort data from the TMAs, we found that S2 was strongly associated with the failure of/intolerance to hormone therapy. CONCLUSIONS: This study identified two distinct subtypes that are varyingly associated with hormone resistance, stroma-immunity, and molecular features, thereby highlighting the importance of this stromal-immune heterogeneity in identifying EMs subtypes and providing novel insights into future personalized hormone-free therapy in EMs. Frontiers Media S.A. 2023-06-22 /pmc/articles/PMC10324653/ /pubmed/37426659 http://dx.doi.org/10.3389/fimmu.2023.1133672 Text en Copyright © 2023 Wang, Chen, Xiao, Ruan, Tian, Cheng, Chang and Yi https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Wang, Yuning
Chen, Yun
Xiao, Yinping
Ruan, Jingyao
Tian, Qi
Cheng, Qi
Chang, Kaikai
Yi, Xiaofang
Distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy
title Distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy
title_full Distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy
title_fullStr Distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy
title_full_unstemmed Distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy
title_short Distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy
title_sort distinct subtypes of endometriosis identified based on stromal-immune microenvironment and gene expression: implications for hormone therapy
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10324653/
https://www.ncbi.nlm.nih.gov/pubmed/37426659
http://dx.doi.org/10.3389/fimmu.2023.1133672
work_keys_str_mv AT wangyuning distinctsubtypesofendometriosisidentifiedbasedonstromalimmunemicroenvironmentandgeneexpressionimplicationsforhormonetherapy
AT chenyun distinctsubtypesofendometriosisidentifiedbasedonstromalimmunemicroenvironmentandgeneexpressionimplicationsforhormonetherapy
AT xiaoyinping distinctsubtypesofendometriosisidentifiedbasedonstromalimmunemicroenvironmentandgeneexpressionimplicationsforhormonetherapy
AT ruanjingyao distinctsubtypesofendometriosisidentifiedbasedonstromalimmunemicroenvironmentandgeneexpressionimplicationsforhormonetherapy
AT tianqi distinctsubtypesofendometriosisidentifiedbasedonstromalimmunemicroenvironmentandgeneexpressionimplicationsforhormonetherapy
AT chengqi distinctsubtypesofendometriosisidentifiedbasedonstromalimmunemicroenvironmentandgeneexpressionimplicationsforhormonetherapy
AT changkaikai distinctsubtypesofendometriosisidentifiedbasedonstromalimmunemicroenvironmentandgeneexpressionimplicationsforhormonetherapy
AT yixiaofang distinctsubtypesofendometriosisidentifiedbasedonstromalimmunemicroenvironmentandgeneexpressionimplicationsforhormonetherapy