Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencing

Single-cell RNA-sequencing (scRNA-seq) presents better insights into cell behavior in the context of a complex tumor microenvironment by profiling single-cell populations. However, the mechanisms underlying treatment failure in hepatocellular carcinoma (HCC) are poorly understood. In this study, we...

Full description

Bibliographic Details
Main Authors: Xiaopei Hao, Zhiying Zheng, Hanyuan Liu, Yao Zhang, Junwei Kang, Xiangyi Kong, Dawei Rong, Guangshun Sun, Guoqiang Sun, Li Liu, Haibo Yu, Weiwei Tang, Xuehao Wang
Format: Article
Language:English
Published: Elsevier 2022-10-01
Series:Redox Biology
Subjects:
Online Access:http://www.sciencedirect.com/science/article/pii/S221323172200235X
_version_ 1798035921948377088
author Xiaopei Hao
Zhiying Zheng
Hanyuan Liu
Yao Zhang
Junwei Kang
Xiangyi Kong
Dawei Rong
Guangshun Sun
Guoqiang Sun
Li Liu
Haibo Yu
Weiwei Tang
Xuehao Wang
author_facet Xiaopei Hao
Zhiying Zheng
Hanyuan Liu
Yao Zhang
Junwei Kang
Xiangyi Kong
Dawei Rong
Guangshun Sun
Guoqiang Sun
Li Liu
Haibo Yu
Weiwei Tang
Xuehao Wang
author_sort Xiaopei Hao
collection DOAJ
description Single-cell RNA-sequencing (scRNA-seq) presents better insights into cell behavior in the context of a complex tumor microenvironment by profiling single-cell populations. However, the mechanisms underlying treatment failure in hepatocellular carcinoma (HCC) are poorly understood. In this study, we performed deep scRNA-seq on immune cells under the isolation in peripheral blood, cancer tissues, and nearby common tissues of four HCC cases and two non-cancer controls, and 212,494 cells were included in the analysis. We identified distinct immune cell subtypes, enriched pathways for differential genes, and delineated associated developmentally relevant trajectories. APOC1 was found over-expressed in tumor-associated macrophages (TAMs) of HCC tissues than in normal tissues. Inhibition of APOC1 reversed the M2 phenotype to the M1 phenotype via the ferroptosis pathway in TAMs from HCC. Tumors in APOC1 −/− C57BL/6 mice demonstrated consistent attenuation compared to wild-type (WT) mice. Mass spectrometry results revealed that the relative proportion of M2 macrophages, B cells, and CD4+ T cells in the APOC1 −/− group exhibited a downward expression compared with the WT group, whereas CD8+ T cells, M1 macrophages, and NK cells exhibited an upward trend. Finally, APOC1 was found to be negatively correlated with the expression of PD1/PD-L1 in human HCC samples. In conclusion, the present study demonstrated that inhibiting APOC1 can promote the transformation of M2 macrophages into M1 macrophages via the ferroptosis pathway, thereby reshaping the tumor immune microenvironment and improving the anti-PD1 immunotherapy for HCC, providing a new strategy for improving the therapeutic effect of anti-PD1, and bringing new hope to HCC patients.
first_indexed 2024-04-11T21:05:09Z
format Article
id doaj.art-9e92ec0b3aaa45f1813c3d7eae007166
institution Directory Open Access Journal
issn 2213-2317
language English
last_indexed 2024-04-11T21:05:09Z
publishDate 2022-10-01
publisher Elsevier
record_format Article
series Redox Biology
spelling doaj.art-9e92ec0b3aaa45f1813c3d7eae0071662022-12-22T04:03:23ZengElsevierRedox Biology2213-23172022-10-0156102463Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencingXiaopei Hao0Zhiying Zheng1Hanyuan Liu2Yao Zhang3Junwei Kang4Xiangyi Kong5Dawei Rong6Guangshun Sun7Guoqiang Sun8Li Liu9Haibo Yu10Weiwei Tang11Xuehao Wang12Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, ChinaDepartment of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, ChinaDepartment of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, ChinaHepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, ChinaDepartment of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, ChinaHepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, ChinaHepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, ChinaDepartment of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, ChinaDepartment of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, ChinaFirst Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Corresponding author.Department of Hepatobiliary Surgery, People's Hospital of Zhengzhou University, Zhengzhou, China; Corresponding author.Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China; Corresponding author.Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China; Corresponding author.Single-cell RNA-sequencing (scRNA-seq) presents better insights into cell behavior in the context of a complex tumor microenvironment by profiling single-cell populations. However, the mechanisms underlying treatment failure in hepatocellular carcinoma (HCC) are poorly understood. In this study, we performed deep scRNA-seq on immune cells under the isolation in peripheral blood, cancer tissues, and nearby common tissues of four HCC cases and two non-cancer controls, and 212,494 cells were included in the analysis. We identified distinct immune cell subtypes, enriched pathways for differential genes, and delineated associated developmentally relevant trajectories. APOC1 was found over-expressed in tumor-associated macrophages (TAMs) of HCC tissues than in normal tissues. Inhibition of APOC1 reversed the M2 phenotype to the M1 phenotype via the ferroptosis pathway in TAMs from HCC. Tumors in APOC1 −/− C57BL/6 mice demonstrated consistent attenuation compared to wild-type (WT) mice. Mass spectrometry results revealed that the relative proportion of M2 macrophages, B cells, and CD4+ T cells in the APOC1 −/− group exhibited a downward expression compared with the WT group, whereas CD8+ T cells, M1 macrophages, and NK cells exhibited an upward trend. Finally, APOC1 was found to be negatively correlated with the expression of PD1/PD-L1 in human HCC samples. In conclusion, the present study demonstrated that inhibiting APOC1 can promote the transformation of M2 macrophages into M1 macrophages via the ferroptosis pathway, thereby reshaping the tumor immune microenvironment and improving the anti-PD1 immunotherapy for HCC, providing a new strategy for improving the therapeutic effect of anti-PD1, and bringing new hope to HCC patients.http://www.sciencedirect.com/science/article/pii/S221323172200235XAPOC1Hepatocellular carcinomaSingle-cell RNA sequencingFerroptosisMacrophages
spellingShingle Xiaopei Hao
Zhiying Zheng
Hanyuan Liu
Yao Zhang
Junwei Kang
Xiangyi Kong
Dawei Rong
Guangshun Sun
Guoqiang Sun
Li Liu
Haibo Yu
Weiwei Tang
Xuehao Wang
Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencing
Redox Biology
APOC1
Hepatocellular carcinoma
Single-cell RNA sequencing
Ferroptosis
Macrophages
title Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencing
title_full Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencing
title_fullStr Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencing
title_full_unstemmed Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencing
title_short Inhibition of APOC1 promotes the transformation of M2 into M1 macrophages via the ferroptosis pathway and enhances anti-PD1 immunotherapy in hepatocellular carcinoma based on single-cell RNA sequencing
title_sort inhibition of apoc1 promotes the transformation of m2 into m1 macrophages via the ferroptosis pathway and enhances anti pd1 immunotherapy in hepatocellular carcinoma based on single cell rna sequencing
topic APOC1
Hepatocellular carcinoma
Single-cell RNA sequencing
Ferroptosis
Macrophages
url http://www.sciencedirect.com/science/article/pii/S221323172200235X
work_keys_str_mv AT xiaopeihao inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT zhiyingzheng inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT hanyuanliu inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT yaozhang inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT junweikang inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT xiangyikong inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT daweirong inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT guangshunsun inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT guoqiangsun inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT liliu inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT haiboyu inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT weiweitang inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing
AT xuehaowang inhibitionofapoc1promotesthetransformationofm2intom1macrophagesviatheferroptosispathwayandenhancesantipd1immunotherapyinhepatocellularcarcinomabasedonsinglecellrnasequencing