Lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapy

Abstract Background Platinum-drugs based chemotherapy in clinic increases the potency of tumor cells to produce M2 macrophages, thus leading to poor anti-metastatic activity and immunosuppression. Lysosome metabolism is critical for cancer cell migration and invasion, but how it promotes antitumor i...

Full description

Bibliographic Details
Main Authors: Jing Ma, Ruijuan Ma, Xueke Zeng, Liming Zhang, Jianing Liu, Wei Zhang, Tao Li, Hanjing Niu, Guochen Bao, Chaojie Wang, Peng George Wang, Jiajia Wang, Xia Li, Taotao Zou, Songqiang Xie
Format: Article
Language:English
Published: BMC 2023-08-01
Series:Journal of Experimental & Clinical Cancer Research
Online Access:https://doi.org/10.1186/s13046-023-02768-0
_version_ 1797451021468827648
author Jing Ma
Ruijuan Ma
Xueke Zeng
Liming Zhang
Jianing Liu
Wei Zhang
Tao Li
Hanjing Niu
Guochen Bao
Chaojie Wang
Peng George Wang
Jiajia Wang
Xia Li
Taotao Zou
Songqiang Xie
author_facet Jing Ma
Ruijuan Ma
Xueke Zeng
Liming Zhang
Jianing Liu
Wei Zhang
Tao Li
Hanjing Niu
Guochen Bao
Chaojie Wang
Peng George Wang
Jiajia Wang
Xia Li
Taotao Zou
Songqiang Xie
author_sort Jing Ma
collection DOAJ
description Abstract Background Platinum-drugs based chemotherapy in clinic increases the potency of tumor cells to produce M2 macrophages, thus leading to poor anti-metastatic activity and immunosuppression. Lysosome metabolism is critical for cancer cell migration and invasion, but how it promotes antitumor immunity in tumours and macrophages is poorly understood and the underlying mechanisms are elusive. The present study aimed to explore a synergistic strategy to dismantle the immunosuppressive microenvironment of tumours and metallodrugs discovery by using the herent metabolic plasticity. Methods Naphplatin was prepared by coordinating an active alkaline moiety to cisplatin, which can regulate the lysosomal functions. Colorectal carcinoma cells were selected to perform the in vivo biological assays. Blood, tumour and spleen tissues were collected and analyzed by flow cytometry to further explore the relationship between anti-tumour activity and immune cells. Transformations of bone marrow derived macrophage (BMDM) and M2-BMDM to the M1 phenotype was confirmed after treatment with naphplatin. The key mechanisms of lysosome-mediated mucolipin-1(Mcoln1) and mitogen-activated protein kinase (MAPK) activation in M2 macrophage polarization have been unveiled. RNA sequencing (RNA-seq) was used to further explore the key mechanism underlying high-mobility group box 1(HMGB1)-mediated Cathepsin L(CTSL)-lysosome function blockade. Results We demonstrated that naphplatin induces divergent lysosomal metabolic programs and reprograms macrophages in tumor cells to terminate the vicious tumour-associated macrophages (TAMs)-MDSCs-Treg triangle. Mechanistically, macrophages treated with naphplatin cause lysosome metabolic activation by triggering Ca2+ release via Mcoln1, which induces the activation of p38 and nuclear factor-κB (NF-κB) and finally results in polarizing M2 macrophages. In contrast, HMGB1-mediated lysosome metabolic blockade in cancer cells is strongly linked to antitumor effects by promoting cytoplasmic translocation of HMGB1. Conclusions This study reveals the crucial strategies of macrophage-based metallodrugs discovery that are able to treat both immunologically “hot” and “cold” cancers. Different from traditional platinum-based antitumour drugs by inhibition of DNAs, we also deliver a strong antitumour strategy by targeting lysosome to induce divergent metabolic programs in macrophages and tumours for cancer immunotherapy.
first_indexed 2024-03-09T14:48:56Z
format Article
id doaj.art-002eb2bdab89473f94a351ef49dadf5c
institution Directory Open Access Journal
issn 1756-9966
language English
last_indexed 2024-03-09T14:48:56Z
publishDate 2023-08-01
publisher BMC
record_format Article
series Journal of Experimental & Clinical Cancer Research
spelling doaj.art-002eb2bdab89473f94a351ef49dadf5c2023-11-26T14:34:31ZengBMCJournal of Experimental & Clinical Cancer Research1756-99662023-08-0142111510.1186/s13046-023-02768-0Lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapyJing Ma0Ruijuan Ma1Xueke Zeng2Liming Zhang3Jianing Liu4Wei Zhang5Tao Li6Hanjing Niu7Guochen Bao8Chaojie Wang9Peng George Wang10Jiajia Wang11Xia Li12Taotao Zou13Songqiang Xie14School of Pharmacy, Institute of Chemical Biology, Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, State key Laboratory of Antiviral Drugs, School of Pharmacy, Henan UniversitySchool of Pharmacy, Institute of Chemical Biology, Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, State key Laboratory of Antiviral Drugs, School of Pharmacy, Henan UniversityJoint National Laboratory for Antibody Drug Engineering, Henan UniversityJoint National Laboratory for Antibody Drug Engineering, Henan UniversitySchool of Pharmacy, Institute of Chemical Biology, Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, State key Laboratory of Antiviral Drugs, School of Pharmacy, Henan UniversitySchool of Pharmacy, Institute of Chemical Biology, Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, State key Laboratory of Antiviral Drugs, School of Pharmacy, Henan UniversitySchool of Pharmacy, Institute of Chemical Biology, Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, State key Laboratory of Antiviral Drugs, School of Pharmacy, Henan UniversitySchool of Pharmacy, Institute of Chemical Biology, Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, State key Laboratory of Antiviral Drugs, School of Pharmacy, Henan UniversityInstitute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology SydneyThe Key Laboratory of Natural Medicine and Immuno-Engineering, Henan UniversitySchool of Medicine, The Southern University of Science and TechnologyJoint National Laboratory for Antibody Drug Engineering, Henan UniversityJoint National Laboratory for Antibody Drug Engineering, Henan UniversitySchool of Pharmaceutical Sciences Sun Yat, Sen UniversitySchool of Pharmacy, Institute of Chemical Biology, Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, State key Laboratory of Antiviral Drugs, School of Pharmacy, Henan UniversityAbstract Background Platinum-drugs based chemotherapy in clinic increases the potency of tumor cells to produce M2 macrophages, thus leading to poor anti-metastatic activity and immunosuppression. Lysosome metabolism is critical for cancer cell migration and invasion, but how it promotes antitumor immunity in tumours and macrophages is poorly understood and the underlying mechanisms are elusive. The present study aimed to explore a synergistic strategy to dismantle the immunosuppressive microenvironment of tumours and metallodrugs discovery by using the herent metabolic plasticity. Methods Naphplatin was prepared by coordinating an active alkaline moiety to cisplatin, which can regulate the lysosomal functions. Colorectal carcinoma cells were selected to perform the in vivo biological assays. Blood, tumour and spleen tissues were collected and analyzed by flow cytometry to further explore the relationship between anti-tumour activity and immune cells. Transformations of bone marrow derived macrophage (BMDM) and M2-BMDM to the M1 phenotype was confirmed after treatment with naphplatin. The key mechanisms of lysosome-mediated mucolipin-1(Mcoln1) and mitogen-activated protein kinase (MAPK) activation in M2 macrophage polarization have been unveiled. RNA sequencing (RNA-seq) was used to further explore the key mechanism underlying high-mobility group box 1(HMGB1)-mediated Cathepsin L(CTSL)-lysosome function blockade. Results We demonstrated that naphplatin induces divergent lysosomal metabolic programs and reprograms macrophages in tumor cells to terminate the vicious tumour-associated macrophages (TAMs)-MDSCs-Treg triangle. Mechanistically, macrophages treated with naphplatin cause lysosome metabolic activation by triggering Ca2+ release via Mcoln1, which induces the activation of p38 and nuclear factor-κB (NF-κB) and finally results in polarizing M2 macrophages. In contrast, HMGB1-mediated lysosome metabolic blockade in cancer cells is strongly linked to antitumor effects by promoting cytoplasmic translocation of HMGB1. Conclusions This study reveals the crucial strategies of macrophage-based metallodrugs discovery that are able to treat both immunologically “hot” and “cold” cancers. Different from traditional platinum-based antitumour drugs by inhibition of DNAs, we also deliver a strong antitumour strategy by targeting lysosome to induce divergent metabolic programs in macrophages and tumours for cancer immunotherapy.https://doi.org/10.1186/s13046-023-02768-0
spellingShingle Jing Ma
Ruijuan Ma
Xueke Zeng
Liming Zhang
Jianing Liu
Wei Zhang
Tao Li
Hanjing Niu
Guochen Bao
Chaojie Wang
Peng George Wang
Jiajia Wang
Xia Li
Taotao Zou
Songqiang Xie
Lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapy
Journal of Experimental & Clinical Cancer Research
title Lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapy
title_full Lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapy
title_fullStr Lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapy
title_full_unstemmed Lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapy
title_short Lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapy
title_sort lysosome blockade induces divergent metabolic programs in macrophages and tumours for cancer immunotherapy
url https://doi.org/10.1186/s13046-023-02768-0
work_keys_str_mv AT jingma lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT ruijuanma lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT xuekezeng lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT limingzhang lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT jianingliu lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT weizhang lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT taoli lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT hanjingniu lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT guochenbao lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT chaojiewang lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT penggeorgewang lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT jiajiawang lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT xiali lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT taotaozou lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy
AT songqiangxie lysosomeblockadeinducesdivergentmetabolicprogramsinmacrophagesandtumoursforcancerimmunotherapy