Cargando…

Gradient Boosting Machine Learning Model for Defective Endometrial Receptivity Prediction by Macrophage-Endometrium Interaction Modules

BACKGROUND: A receptive endometrium is a prerequisite for successful embryo implantation. Mounting evidence shows that nearly one-third of infertility and implantation failures are caused by defective endometrial receptivity. This study pooled 218 subjects from multiple datasets to investigate the a...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Bohan, Duan, Hua, Wang, Sha, Wu, Jiajing, Li, Yazhu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9120433/
https://www.ncbi.nlm.nih.gov/pubmed/35603216
http://dx.doi.org/10.3389/fimmu.2022.842607
_version_ 1784710923622023168
author Li, Bohan
Duan, Hua
Wang, Sha
Wu, Jiajing
Li, Yazhu
author_facet Li, Bohan
Duan, Hua
Wang, Sha
Wu, Jiajing
Li, Yazhu
author_sort Li, Bohan
collection PubMed
description BACKGROUND: A receptive endometrium is a prerequisite for successful embryo implantation. Mounting evidence shows that nearly one-third of infertility and implantation failures are caused by defective endometrial receptivity. This study pooled 218 subjects from multiple datasets to investigate the association of the immune infiltration level with reproductive outcome. Additionally, macrophage-endometrium interaction modules were constructed to explore an accurate and cost-effective approach to endometrial receptivity assessment. METHODS: Immune-infiltration levels in 4 GEO datasets (n=218) were analyzed and validated through meta-analysis. Macrophage-endometrium interaction modules were selected based on the weighted gene co-expression network in GSE58144 and differentially expressed genes dominated by GSE19834 dataset. Xgboost, random forests, and regression algorithms were applied to predictive models. Subsequently, the efficacy of the models was compared and validated in the GSE165004 dataset. Forty clinical samples (RT-PCR and western blot) were performed for expression and model validation, and the results were compared to those of endometrial thickness in clinical pregnancy assessment. RESULTS: Altered levels of Mϕs infiltration were shown to critically influence embryo implantation. The three selected modules, manifested as macrophage-endometrium interactions, were enrichment in the immunoreactivity, decidualization, and signaling functions and pathways. Moreover, hub genes within the modules exerted significant reproductive prognostic effects. The xgboost algorithm showed the best performance among the machine learning models, with AUCs of 0.998 (95% CI 0.994-1) and 0.993 (95% CI 0.979-1) in GSE58144 and GSE165004 datasets, respectively. These results were significantly superior to those of the other two models (random forest and regression). Similarly, the model was significantly superior to ultrasonography (endometrial thickness) with a better cost-benefit ratio in the population. CONCLUSION: Successful embryo implantation is associated with infiltration levels of Mϕs, manifested in genetic modules involved in macrophage-endometrium interactions. Therefore, utilizing the hub genes in these modules can provide a platform for establishing excellent machine learning models to predict reproductive outcomes in patients with defective endometrial receptivity.
format Online
Article
Text
id pubmed-9120433
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-91204332022-05-21 Gradient Boosting Machine Learning Model for Defective Endometrial Receptivity Prediction by Macrophage-Endometrium Interaction Modules Li, Bohan Duan, Hua Wang, Sha Wu, Jiajing Li, Yazhu Front Immunol Immunology BACKGROUND: A receptive endometrium is a prerequisite for successful embryo implantation. Mounting evidence shows that nearly one-third of infertility and implantation failures are caused by defective endometrial receptivity. This study pooled 218 subjects from multiple datasets to investigate the association of the immune infiltration level with reproductive outcome. Additionally, macrophage-endometrium interaction modules were constructed to explore an accurate and cost-effective approach to endometrial receptivity assessment. METHODS: Immune-infiltration levels in 4 GEO datasets (n=218) were analyzed and validated through meta-analysis. Macrophage-endometrium interaction modules were selected based on the weighted gene co-expression network in GSE58144 and differentially expressed genes dominated by GSE19834 dataset. Xgboost, random forests, and regression algorithms were applied to predictive models. Subsequently, the efficacy of the models was compared and validated in the GSE165004 dataset. Forty clinical samples (RT-PCR and western blot) were performed for expression and model validation, and the results were compared to those of endometrial thickness in clinical pregnancy assessment. RESULTS: Altered levels of Mϕs infiltration were shown to critically influence embryo implantation. The three selected modules, manifested as macrophage-endometrium interactions, were enrichment in the immunoreactivity, decidualization, and signaling functions and pathways. Moreover, hub genes within the modules exerted significant reproductive prognostic effects. The xgboost algorithm showed the best performance among the machine learning models, with AUCs of 0.998 (95% CI 0.994-1) and 0.993 (95% CI 0.979-1) in GSE58144 and GSE165004 datasets, respectively. These results were significantly superior to those of the other two models (random forest and regression). Similarly, the model was significantly superior to ultrasonography (endometrial thickness) with a better cost-benefit ratio in the population. CONCLUSION: Successful embryo implantation is associated with infiltration levels of Mϕs, manifested in genetic modules involved in macrophage-endometrium interactions. Therefore, utilizing the hub genes in these modules can provide a platform for establishing excellent machine learning models to predict reproductive outcomes in patients with defective endometrial receptivity. Frontiers Media S.A. 2022-05-06 /pmc/articles/PMC9120433/ /pubmed/35603216 http://dx.doi.org/10.3389/fimmu.2022.842607 Text en Copyright © 2022 Li, Duan, Wang, Wu and Li 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
Li, Bohan
Duan, Hua
Wang, Sha
Wu, Jiajing
Li, Yazhu
Gradient Boosting Machine Learning Model for Defective Endometrial Receptivity Prediction by Macrophage-Endometrium Interaction Modules
title Gradient Boosting Machine Learning Model for Defective Endometrial Receptivity Prediction by Macrophage-Endometrium Interaction Modules
title_full Gradient Boosting Machine Learning Model for Defective Endometrial Receptivity Prediction by Macrophage-Endometrium Interaction Modules
title_fullStr Gradient Boosting Machine Learning Model for Defective Endometrial Receptivity Prediction by Macrophage-Endometrium Interaction Modules
title_full_unstemmed Gradient Boosting Machine Learning Model for Defective Endometrial Receptivity Prediction by Macrophage-Endometrium Interaction Modules
title_short Gradient Boosting Machine Learning Model for Defective Endometrial Receptivity Prediction by Macrophage-Endometrium Interaction Modules
title_sort gradient boosting machine learning model for defective endometrial receptivity prediction by macrophage-endometrium interaction modules
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9120433/
https://www.ncbi.nlm.nih.gov/pubmed/35603216
http://dx.doi.org/10.3389/fimmu.2022.842607
work_keys_str_mv AT libohan gradientboostingmachinelearningmodelfordefectiveendometrialreceptivitypredictionbymacrophageendometriuminteractionmodules
AT duanhua gradientboostingmachinelearningmodelfordefectiveendometrialreceptivitypredictionbymacrophageendometriuminteractionmodules
AT wangsha gradientboostingmachinelearningmodelfordefectiveendometrialreceptivitypredictionbymacrophageendometriuminteractionmodules
AT wujiajing gradientboostingmachinelearningmodelfordefectiveendometrialreceptivitypredictionbymacrophageendometriuminteractionmodules
AT liyazhu gradientboostingmachinelearningmodelfordefectiveendometrialreceptivitypredictionbymacrophageendometriuminteractionmodules