1
|
Qin CC, Tong XZ, Su C, Zhou ZH, Zheng D. ITGAM Upregulation in Acute Myeloid Leukemia Leads to Poor Prognosis Associated With Infiltration of Inhibitory Innate Immune Cells. Int J Lab Hematol 2025; 47:481-490. [PMID: 40054851 DOI: 10.1111/ijlh.14444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/25/2024] [Accepted: 01/19/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a prevalent and potentially fatal hematologic malignancy with limited improvements in survival rates over the past few decades. ITGAM, encoding CD11b, is a significant integrin component in leukocytes, involved in various biological processes. However, its role in AML prognosis and immune cell infiltration remains poorly understood. METHODS This study investigated the prognostic significance and potential function of ITGAM in AML using comprehensive bioinformatic analyses. RESULTS ITGAM expression was markedly upregulated in AML patients, correlating with advanced age (> 60 years), French-American-British (FAB) classification subtypes M4 and M5, and intermediate or poor cytogenetic risk. Gene enrichment analysis revealed that ITGAM was involved in immune system regulation and was positively associated with the infiltration of neutrophils, immature dendritic cells, and macrophages in AML. Notably, the expression of ITGAM was linked to increased infiltration of immunosuppressive subsets of these innate immune cells, which may partially explain its association with a poorer prognosis. CONCLUSION These findings suggest that ITGAM could serve as a valuable prognostic biomarker in AML, reflecting an adverse prognosis associated with enhanced infiltration of immunosuppressive innate immune cells.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/pathology
- Prognosis
- Immunity, Innate
- CD11b Antigen/genetics
- CD11b Antigen/immunology
- Up-Regulation
- Middle Aged
- Male
- Female
- Gene Expression Regulation, Leukemic
- Biomarkers, Tumor
- Aged
Collapse
Affiliation(s)
- Chen-Chen Qin
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiu-Zhen Tong
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chang Su
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen-Hai Zhou
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dong Zheng
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Cao L, Shao M, Gu Y, Jia D, Lu W, Liang C, Liu X, Pan Z, Zhang Y, Hu J, Peng P. Calceolarioside B targets MMP12 in the tumor microenvironment to inhibit M2 macrophage polarization and suppress hepatocellular carcinoma progression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156805. [PMID: 40347889 DOI: 10.1016/j.phymed.2025.156805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/01/2025] [Accepted: 04/25/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are crucial in hepatocellular carcinoma (HCC) progression and prognosis, making them promising immunotherapy targets. In traditional Chinese medicine (TCM), qi stagnation and blood stasis are linked to the HCC tumor microenvironment (TME), but few studies explore the effects of related TCM herbs on the TME. Calceolarioside B, a key phenylethanoid glycoside in Akebiae Fructus, has not been well studied for its pharmacological activities or molecular targets, and its role in HCC remains unclear. PURPOSE This study aimed to investigate the effects of Calceolarioside B on TAMs in HCC and clarify its potential targets and regulatory mechanisms. METHODS Murine intrahepatic transplantation HCC models and macrophage-HCC cell co-culture systems were used to investigate the effects of Calceolarioside B on M2-like TAMs polarization and infiltration, and tumor growth. Cellular thermal shift assay, small molecular pull-down assay and surface plasmon resonance were utilized to identify the potential targets regulating M2-like TAMs. Single-cell RNA sequencing and TCGA dataset analyses clarified the differential expression, prognosis, and TAMs association of the potential targets in HCC. RESULTS Calceolarioside B reduces M2-like TAMs polarization and infiltration in the TME by binding to and inhibiting matrix metallopeptidase-12 (MMP12) form both macrophages and HCC cells, thereby preventing immunosuppressive effects. Public database analysis revealed that MMP12 overexpression promoted macrophage infiltration, with MMP12+ macrophages preferentially aggregating in primary and metastatic HCC tumors. CONCLUSION Calceolarioside B is identified as a novel MMP12 inhibitor modulating TAMs in the TME, offering a potential TAM-targeting strategy for HCC therapy.
Collapse
Affiliation(s)
- Linna Cao
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Miaomiao Shao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yifei Gu
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Dongwei Jia
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenli Lu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chao Liang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaomei Liu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhiqiang Pan
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yiwei Zhang
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Jinquan Hu
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| | - Peike Peng
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
3
|
Liu T, Cui Y, Ouyang Y, Wang M, Yue S. Exosomal CCT3 as a biomarker for diagnosis and immune therapy response in patients diagnosed with hepatocellular carcinoma. Dig Liver Dis 2025:S1590-8658(25)00301-9. [PMID: 40221386 DOI: 10.1016/j.dld.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/19/2024] [Accepted: 03/21/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the dominant type of liver cancer and is associated with a high mortality rate. However, HCC lacks biomarkers for diagnosis and immune therapy response. Tumor-derived exosomes (TDEs) carcinogen-specific molecules have been used for screening multiple biomarkers. This study aimed to identify new biomarkers for the diagnosis of HCC and response to immune checkpoint blockade (ICB) therapy. METHODS Analysis of differentially expressed genes (DEGs) in HCC and normal tissues was integrated using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and ExoCarta datasets. The expression of CCT3 was validated in samples from patients with HCC using quantitative polymerase chain reaction (qPCR), Western blotting, and immunohistochemistry (IHC) techniques. RESULTS Exosomal CCT3 was identified as a potential biomarker with significant impact. The expression of CCT3 in different tumor stages and normal tissues adjacent to the tumors (NATs) was validated using qPCR, western blotting, and IHC. CCT3 expression significantly increased the number of activated natural killer cells in HCC, as confirmed by qPCR and IHC. CCT3 expression significantly increases the expression of immune checkpoints in HCC. HCC-derived exosomes significantly increase the enrichment of CCT3. CONCLUSION Exosomal CCT3 is a biomarker for diagnosis and ICB therapy of HCC via MYC pathway activation and immune infiltration.
Collapse
Affiliation(s)
- Tiange Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Yanyan Cui
- The Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Yiben Ouyang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Meilin Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shijing Yue
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
4
|
Zhao R, Pan Z, Qiu J, Li B, Qi Y, Gao Z, Qiu W, Tang W, Guo X, Deng L, Li G, Xue H. Blocking ITGA5 potentiates the efficacy of anti-PD-1 therapy on glioblastoma by remodeling tumor-associated macrophages. Cancer Commun (Lond) 2025. [PMID: 40084746 DOI: 10.1002/cac2.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is largely refractory to antibodies against programmed cell death 1 (anti-PD-1) therapy. Fully understanding the cellular heterogeneity and immune adaptations in response to anti-PD-1 therapy is necessary to design more effective immunotherapies for GBM. This study aimed to dissect the molecular mechanisms of specific immunosuppressive subpopulations to drive anti-PD-1 resistance in GBM. METHODS We systematically analysed single-cell RNA sequencing and spatial transcriptomics data from GBM tissues receiving anti-PD-1 therapy to characterize the microenvironment alterations. The biological functions of a novel circular RNA (circRNA) were validated both in vitro and in vivo. Mechanically, co-immunoprecipitation, RNA immunoprecipitation and pull-down assays were conducted. RESULTS Mesenchymal GBM (MES-GBM) cells, which were associated with a poor prognosis, and secreted phosphoprotein 1 (SPP1)+ myeloid-derived macrophages (SPP1+ MDMs), a unique subpopulation of MDMs with complex functions, preferentially accumulated in non-responders to anti-PD-1 therapy, indicating that MES-GBM cells and SPP1+ MDMs were the main anti-PD-1-resistant cell subpopulations. Functionally, we determined that circular RNA succinate dehydrogenase complex assembly factor 2 (circSDHAF2), which was positively associated with the abundance of these two anti-PD-1-resistant cell subpopulations, facilitated the formation of a regional MES-GBM and SPP1+ MDM cell interaction loop, resulting in a spatially specific adaptive immunosuppressive microenvironment. Mechanically, we found that circSDHAF2 promoted MES-GBM cell formation by stabilizing the integrin alpha 5 (ITGA5) protein through N-glycosylation. Meanwhile, the N-glycosylation of the ITGA5 protein facilitated its translocation into exosomes and subsequent delivery to MDMs to induce the formation of SPP1+ MDMs, which in turn maintained the MES-GBM cell status and induced T-cell dysfunction via the SPP1-ITGA5 pathway, ultimately promoting GBM immune escape. Importantly, our findings demonstrated that antibody-mediated ITGA5 blockade enhanced anti-PD-1-mediated antitumor immunity. CONCLUSIONS This work elucidated the potential tissue adaptation mechanism of intratumoral dynamic interactions between MES-GBM cells, MDMs and T cells in anti-PD-1 non-responders and identified the therapeutic potential of targeting ITGA5 to reduce anti-PD-1 resistance in GBM.
Collapse
Affiliation(s)
- Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Ziwen Pan
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Jiawei Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Boyan Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Yanhua Qi
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Zijie Gao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Wei Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Weijie Tang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Xiaofan Guo
- Department of Neurology, Loma Linda University Health, Loma Linda, California, USA
| | - Lin Deng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, P. R. China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, P. R. China
| |
Collapse
|
5
|
Zhang M, Huang K, Yin Q, Wu X, Zhu M, Li M. Spatial heterogeneity of the hepatocellular carcinoma microenvironment determines the efficacy of immunotherapy. Discov Oncol 2025; 16:15. [PMID: 39775241 PMCID: PMC11706828 DOI: 10.1007/s12672-025-01747-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a global health challenge owing to its widespread incidence and high mortality. HCC has a specific immune tolerance function because of its unique physiological structure, which limits the efficacy of chemotherapy, radiotherapy, and molecular targeting. In recent years, new immune approaches, including adoptive cell therapy, tumor vaccines, and oncolytic virus therapy, have shown great potential. As the efficacy of immunotherapy mainly depends on the spatial heterogeneity of the tumor immune microenvironment, it is necessary to elucidate the crosstalk between the composition of the liver cancer immune environment, from which potential therapeutic targets can be selected to provide more appropriate individualized treatment programs. The role of spatial heterogeneity of immune cells in the microenvironment of HCC in the progression and influence of immunotherapy on improving the treatment and prognosis of HCC were comprehensively analyzed, providing new inspiration for the subsequent clinical treatment of liver cancer.
Collapse
Affiliation(s)
- Minni Zhang
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China
- The First Affiliated Hospital, Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, The Hainan Branch of National Clinical Research Center for Cancer, Hainan Medical University, Haikou, 570102, Hainan, People's Republic of China
| | - Kailin Huang
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China
| | - Qiushi Yin
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China
| | - Xueqin Wu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China
| | - Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China.
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China.
- Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical University, Haikou, 570023, Hainan, People's Republic of China.
- Key Laboratory of Tropical Translational Medicine, Ministry of Education, Hainan Medical University, Haikou, 571199, Hainan, People's Republic of China.
| |
Collapse
|
6
|
Wada Y, Matsugaki S, Nagao Y, Taniwaki S, Okuda K, Morimitsu Y. Hematogenous metastasis to the colon from hepatocellular carcinoma: A case report. Int J Surg Case Rep 2025; 126:110491. [PMID: 39662369 PMCID: PMC11697117 DOI: 10.1016/j.ijscr.2024.110491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction and importance: Most extrahepatic metastases of hepatocellular carcinoma (HCC) are to the lungs and bones, metastases to the colon are rare. In the present study, we experienced a case of metastasis to the ascending colon during repeated treatment for HCC. Case Presentation: A 63-year-old man was diagnosed with multiple HCCs (T4N0M0 stage IIIB) associated with portal vein invasion. Transcatheter arterial chemoembolization (TACE), transarterial infusion (TAI) and radiofrequency ablation (RFA) were performed, and partial response was achieved, but the main nodule at S6 lesion subsequently recurred to protrude outside of the liver. A partial hepatic S6 resection was performed for local control 1.5 years after the initial treatment. IVR was then performed again, but approximately 8 months after hepatic resection, an abdominal computed toography (CT) showed a mass lesion in the ascending colon. After a total colonoscopy and biopsy, a diagnosis of colorectal metastasis of HCC was made. A right hemicolectomy was performed for local control. The patient had a good post-operative course, but developed liver failure due to rapid growth of the tumor thrombus of the main portal vein and died of primary disease approximately 3.5 months after the colon resection. Clinical Discussion: The metastasis of HCC to the colon is an extremely rare occurrence. Conclusion: One possible reason for this rarity is that portal vein tumor thrombosis (PVTT) results in colorectal metastasis via trans-portal retrograde metastasis.
Collapse
Affiliation(s)
- Yoshito Wada
- Department of Surgery, Tobata Kyoritsu Hospital, 2-5-1 Sawami, Tobata-ku, Kitakyushu city, Fukuoka Prefecture 804-0093, Japan.
| | - Satoru Matsugaki
- Department of Internal Medicine, Tobata Kyoritsu Hospital, 2-5-1 Sawami, Tobata-ku, Kitakyushu city, Fukuoka Prefecture 804-0093,Japan
| | - Yuichi Nagao
- Department of Surgery, Tobata Kyoritsu Hospital, 2-5-1 Sawami, Tobata-ku, Kitakyushu city, Fukuoka Prefecture 804-0093, Japan.
| | - Satoshi Taniwaki
- Department of Surgery, Tobata Kyoritsu Hospital, 2-5-1 Sawami, Tobata-ku, Kitakyushu city, Fukuoka Prefecture 804-0093, Japan.
| | - Koji Okuda
- Department of Surgery, Tobata Kyoritsu Hospital, 2-5-1 Sawami, Tobata-ku, Kitakyushu city, Fukuoka Prefecture 804-0093, Japan
| | - Yosuke Morimitsu
- Department of Pathology, Tobata Kyoritsu Hospital, 2-5-1 Sawami, Tobata-ku, Kitakyushu city, Fukuoka Prefecture 804-0093, Japan
| |
Collapse
|
7
|
Wang B, Cheng H, Ji Z, Jiang Z, Wang R, Ding Y, Ni J. Synergistic Target-Attacking Tumor Cells and M2 Macrophages via a Triple-Responsive Nanoassembly for Complete Metastasis Blocking. Adv Healthc Mater 2025; 14:e2304096. [PMID: 39663738 DOI: 10.1002/adhm.202304096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/24/2024] [Indexed: 12/13/2024]
Abstract
Collaboration of cancerous cells and microenvironment is the root for tumor spreading, leading to difficulty in complete metastasis blockage via mono-intervention. Herein, a triple-responsive nanoassembly is designed for orienting tumor cells and migration-driving M2 tumor associated macrophages (TAMs) in microenvironment for efficient anti-metastatic therapy. Structurally, a reactive oxygen species (ROS)-responsive crosslinked short-chain polyquaternium is synthesized to bridge graphene oxide (GO) scaffold with apolipoprotein A-I crown via borate-crosslinking, electrostatic adherence, and coordinative coupling. The protein-crowning polymeric GO nanoparticles could give multimodal shielding and triple-responsive release of doxorubicin and Snail-targeted siRNA. Tailor-made apolipoprotein A-I crown fulfills nanoparticles synergistically attacking tumor cells and M2 TAMs via binding with overexpressed scavenger receptors. The findings witness the targeted accumulation and potent cytotoxicity of the hybrid nanoparticles for M2 TAMs and tumor cells; especially, elimination of M2 TAMs in tumor microenvironment holds back Snail-enhancing transforming growth factor (TGF)-β signal pathway, which collaborates with Snail silencing in tumor cells to reverse epithelial mesenchymal transition (EMT) and metastasis-promoting niche. Collectively, the synergistic targeting therapeutic platform could provide a promising solution for metastatic tumor treatment.
Collapse
Affiliation(s)
- Bei Wang
- Institute of Integration of Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| | - Hao Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhongsheng Ji
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zijun Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Rong Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiang Ni
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| |
Collapse
|
8
|
Tripathi S, Sharma Y, Kumar D. Biological Cargo: Exosomes and their Role in Cancer Progression and Metastasis. Curr Top Med Chem 2025; 25:263-285. [PMID: 38984577 DOI: 10.2174/0115680266304636240626055711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/11/2024]
Abstract
Cancer cells are among the many types of cells that release exosomes, which are nanovesicles. Because of their many potential applications, exosomes have recently garnered much attention from cancer researchers. The bioactive substances that exosomes release as cargo have been the subject of several investigations. The substances in question may operate as biomarkers for diagnosis or affect apoptosis, the immune system, the development and spread of cancer, and other processes. Others have begun to look at exosomes in experimental therapeutic trials because they believe they may be useful in the treatment of cancer. This review started with a short description of exosome biogenesis and key features. Next, the potential of tumor-derived exosomes and oncosomes to influence the immune system throughout the development of cancer, as well as alter tumor microenvironments (TMEs) and pre-metastatic niche creation, was investigated. Finally, there was talk of exosomes' possible use in cancer treatment. Furthermore, there is emerging consensus about the potential application of exosomes to be biological reprogrammers of cancer cells, either as carriers of naturally occurring chemicals, including anticancer medications, or as carriers of anticancer vaccines for immunotherapy as well as boron neutron capture therapy (BNCT). We briefly review the key ideas and logic behind this intriguing therapy recommendation.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| |
Collapse
|
9
|
Yang Z, Li H, Wang J, Gao W, Zhao Q, Meng Q, Huang J, Xi Q, Wei J, Yang X. CCL2/CCR2 axis promotes perineural invasion of salivary adenoid cystic carcinoma via ITGβ5-mediated nerve-tumor interaction. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167484. [PMID: 39222826 DOI: 10.1016/j.bbadis.2024.167484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Perineural invasion (PNI) is a notorious feature of salivary adenoid cystic carcinoma (SACC) and other neurotropic tumors. The pathogenesis of PNI that involves the molecular communication between the tumor and the suffered nerve is elusive. The in vitro co-culture assays of SACC cells with dorsal root ganglia (DRG) or neural cells showed that nerve-derived CCL2 activated CCR2 expression in SACC cells, promoting the proliferation, adhesion, migration, and invasion of SACC cells via the ERK1/2/ITGβ5 pathway. Meanwhile, SACC-derived exosomes delivered ITGβ5 to promote the neurite outgrowth of neural cells or DRG. Blocking of CCL2/CCR2 axis or ITGβ5 inhibited the PNI of SACC cells in models in vitro by 3D co-culture of DRG with SACC cells and in vivo by xenografting SACC cells onto the murine sciatic nerve. High levels of ITGβ5 in tissues or plasma exosomes were significantly correlated with CCL2 and CCR2 expression in the tissues and associated with PNI and poor prognosis of SACC cases. Our findings revealed a novel reciprocal loop between neural and tumor cells driven by the CCL2/CCR2 axis and exosomal ITGβ5 during PNI of SACC. The present study may provide a prospective diagnostic and anti-PNI treatment strategy for SACC patients via targeting the nerve-tumor interactions.
Collapse
Affiliation(s)
- Zihui Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Huan Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Jun Wang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Wanpeng Gao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Qi Zhao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Qingzhe Meng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Junhong Huang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Qi Xi
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Jianhua Wei
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China.
| | - Xinjie Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
10
|
Saleh RO, Hamad HA, Najim MA, Menon SV, Kaur M, Sivaprasad GV, Abohassan M, Juan WT, Husseen B, Mustafa YF. Exosome-mediated Transfer of lncRNA in Liver Associated Diseases; Uncovered Truths. Cell Biochem Biophys 2024:10.1007/s12013-024-01617-x. [PMID: 39567423 DOI: 10.1007/s12013-024-01617-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
Exosomes are extracellular vesicles with a diameter ranging from 40 to 160 nm. They are produced by hepatocytes, cholangiocytes, hepatic stellate cells (HSCs), liver sinusoidal endothelial cells (LSECs) and Kupffer cells in liver tissue. The secretion of exosomes might vary in quantity and composition in reaction to multiple triggers and various stages of disease. They transport various payloads, such as proteins, DNAs, and RNAs, and enable cell interaction to regulate myriad physiological and pathological processes in liver tissue. Long non-coding RNAs (lncRNAs) are a crucial component of exosomes with an excellent capability to regulate multiple cellular activities such as differentiation, development, metabolism, proliferation, apoptosis, and activation. With the advancements in transcriptomic and genomic study methods and database management technology, the functions and mechanisms of exosomal lncRNAs in liver diseases have been well-studied. This article delves into the detailed role of exosomal lncRNAs in liver disease onset and progression, ranging from hepatocellular carcinoma (HCC) to liver fibrosis drug-induced liver damage (DILI) and steatotic liver diseases.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al Maarif, Anbar, Iraq.
| | - Hamad Ali Hamad
- Department of Pathological Analysis, Collage of Applied Sciences, University of Fallujah, Fallujah, Iraq
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Malaysia
| | | | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Wen-Tau Juan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Beneen Husseen
- Medical Laboratory Technique college, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique college, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique college, The Islamic University of Babylon, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
11
|
Saadh MJ, Khalifehsoltani A, Hussein AHA, Allela OQB, Sameer HN, Rizaev J, Hameed HG, Idan AH, Alsaikhan F. Exosomal microRNAs in cancer metastasis: A bridge between tumor micro and macroenvironment. Pathol Res Pract 2024; 263:155666. [PMID: 39476605 DOI: 10.1016/j.prp.2024.155666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/10/2024]
Abstract
Malignant tumors are complicated structures of cancer cells that are constantly in communication with their local and distant environment. Exosomes are released by tumor cells and can facilitate the cell-cell interaction within the local microenvironment and the primary tumor. In fact, exosomes are secreted by both tumor and non-tumor cells, to provide a mutual communication network between cells and their micro- and/or macro-environments. Exososmes can contain a variety of biological cargos mostly based on their originated cells. Uptake of these exosomes by their recipient cells results in the alterations that their cargo can exert. MicroRNAs are identified as one of the most critical exosomal components, considering their pivotal regulatory roles in distinct biological process, including metastasis. Release and absorbance of exosomal microRNAs is possible by various cells within the host, and can have distinct biological consequences. Therefore, in this review we will discuss the role of exosomal microRNAs derived from tumor cells and untransformed cells within their micro- and macroenvironment in cancer progression and metastasis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | | | | | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | - Jasur Rizaev
- Department of Public health and Healthcare management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia; School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| |
Collapse
|
12
|
Guan Y, Li J, Sun B, Xu K, Zhang Y, Ben H, Feng Y, Liu M, Wang S, Gao Y, Duan Z, Zhang Y, Chen D, Wang Y. HBx-induced upregulation of MAP1S drives hepatocellular carcinoma proliferation and migration via MAP1S/Smad/TGF-β1 loop. Int J Biol Macromol 2024; 281:136327. [PMID: 39374711 DOI: 10.1016/j.ijbiomac.2024.136327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 09/12/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024]
Abstract
Hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC), has a significantly higher risk of recurrence. However, the exact mechanism by which HBV prompts HCC recurrence remains largely unknown. In this study liver microarray test revealed significant upregulation of microtubule associated protein 1S (MAP1S) in metastatic HCC compared to control. MAP1S knockdown suppressed growth of HCCLM3 cells in vitro and in vivo. Mechanistically, HBV-encoded X protein (HBx) upregulates MAP1S, which enhances microtubule (MT) acetylation by promoting the degradation of histone deacetylase 6 (HDAC6), and facilitates the nuclear translocation of Smad complex, and thereby enhancing downstream TGF-β signaling. Smad complex, in turn, increases MAP1S, establishing a feedback loop of MAP1S/Smad/TGF-β1. Finally, survival analysis of 150 HBV-associated HCC patients demonstrated both increased MAP1S and decreased HDAC6 were significantly associated with shorter relapse-free survival. Collectively, this study reveals a unique mechanism whereby HBx-induced upregulation of MAP1S drives HBV-related HCC proliferation and migration through the MAP1S/Smad/TGF-β1 feedback loop. TEASER: MAP1S is a key link between HBV infection and a higher risk of metastatic recurrence of HCC.
Collapse
Affiliation(s)
- Yuanyue Guan
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Tsinghua Changgung Hospital, School Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Jiaxi Li
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Bin Sun
- Clinical Center for Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Intervention Therapy Center of Tumor and Liver Diseases, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Kaikun Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yonghong Zhang
- Clinical Center for Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Intervention Therapy Center of Tumor and Liver Diseases, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Haijing Ben
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Yingmei Feng
- Department of Science and Development, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Mengcheng Liu
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Shanshan Wang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Yuxue Gao
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Zhongping Duan
- Clinical Center for Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Artificial Liver Center, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Yang Zhang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China.
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China.
| | - Yanjun Wang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
13
|
Hsu CY, Ahmed AT, Bansal P, Hjazi A, Al-Hetty HRAK, Qasim MT, Sapaev I, Deorari M, Mustafa YF, Elawady A. MicroRNA-enriched exosome as dazzling dancer between cancer and immune cells. J Physiol Biochem 2024; 80:811-829. [PMID: 39316240 DOI: 10.1007/s13105-024-01050-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/11/2024] [Indexed: 09/25/2024]
Abstract
Exosomes are widely recognized for their roles in numerous biological processes and as intercellular communication mediators. Human cancerous and normal cells can both produce massive amounts of exosomes. They are extensively dispersed in tumor-modeling animals' pleural effusions, ascites, and plasma from people with cancer. Tumor cells interact with host cells by releasing exosomes, which allow them to interchange various biological components. Tumor growth, invasion, metastasis, and even tumorigenesis can all be facilitated by this delicate and complex system by modifying the nearby and remote surroundings. Due to the existence of significant levels of biomolecules like microRNA, exosomes can modulate the immune system's stimulation or repression, which in turn controls tumor growth. However, the role of microRNA in exosome-mediated communication between immunological and cancer cells is still poorly understood. This study aims to get the most recent information on the "yin and yang" of exosomal microRNA in the regulation of tumor immunity and immunotherapy, which will aid current cancer treatment and diagnostic techniques.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona 85004, USA
| | - Abdulrahman T Ahmed
- Department of Nursing, Al-Maarif University College, Ramadi, AL-Anbar Governorate, Iraq
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, 560069, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | | | - Maytham T Qasim
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar, 64001, Iraq
| | - Ibrokhim Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan
- School of Engineering, Central Asian University, Tashkent, 111221, Uzbekistan
- Western Caspian University, Scientific researcher, Baku, Azerbaijan
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Ahmed Elawady
- College of Technical Engineering, the Islamic University, Najaf, Iraq
- College of Technical Engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
14
|
Yu M, Yu H, Wang H, Xu X, Sun Z, Chen W, Yu M, Liu C, Jiang M, Zhang X. Tumor‑associated macrophages activated in the tumor environment of hepatocellular carcinoma: Characterization and treatment (Review). Int J Oncol 2024; 65:100. [PMID: 39239752 PMCID: PMC11387121 DOI: 10.3892/ijo.2024.5688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/08/2024] [Indexed: 09/07/2024] Open
Abstract
Hepatocellular carcinoma (HCC) tissue is rich in dendritic cells, T cells, B cells, macrophages, natural killer cells and cellular stroma. Together they form the tumor microenvironment (TME), which is also rich in numerous cytokines. Tumor‑associated macrophages (TAMs) are involved in the regulation of tumor development. TAMs in HCC receive stimuli in different directions, polarize in different directions and release different cytokines to regulate the development of HCC. TAMs are mostly divided into two cell phenotypes: M1 and M2. M1 TAMs secrete pro‑inflammatory mediators, and M2 TAMs secrete a variety of anti‑inflammatory and pro‑tumorigenic substances. The TAM polarization in HCC tumors is M2. Both direct and indirect methods for TAMs to regulate the development of HCC are discussed. TAMs indirectly support HCC development by promoting peripheral angiogenesis and regulating the immune microenvironment of the TME. In terms of the direct regulation between TAMs and HCC cells, the present review mainly focuses on the molecular mechanism. TAMs are involved in both the proliferation and apoptosis of HCC cells to regulate the quantitative changes of HCC, and stimulate the related invasive migratory ability and cell stemness of HCC cells. The present review aims to identify immunotherapeutic options based on the mechanisms of TAMs in the TME of HCC.
Collapse
Affiliation(s)
- Mingkai Yu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Haixia Yu
- Pharmacy College, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xiaoya Xu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Zhaoqing Sun
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Wenshuai Chen
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Miaomiao Yu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Chunhua Liu
- Department of Physiology and Neurobiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Mingchun Jiang
- Department of Physiology and Neurobiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Xiaowei Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, P.R. China
| |
Collapse
|
15
|
Wang X, Sun R, Che N, Zhang D, Li Y, Zhao N. Overexpression of NDRG1 leads to poor prognosis in hepatocellular carcinoma through mediating immune infiltration and EMT. Dig Liver Dis 2024; 56:1382-1399. [PMID: 38290958 DOI: 10.1016/j.dld.2024.01.182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 12/31/2023] [Accepted: 01/07/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND NDRG1, the first member of the NDRG family, is a multifunctional protein associated with carcinogenesis. Its function in human cancer is currently poorly understood. The aim of this study was to explore the importance of NDRG1 in tumor immune cell infiltration and epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma. METHODS NDRG1 expression in various cancers was analyzed using TIMER 2.0, the Human Protein Atlas (HPA), UALCAN and PrognoScan. Wound healing, Transwell, MTT and colony formation assays were performed to confirm the effects of NDRG1 on the metastasis and proliferation of HCC cells. Western blotting was used to study the effect of NDRG1 on the expression of EMT-related proteins. Signaling networks were constructed using LinkedOmics and Metascape. TIMER2.0 and TISIDB were used for comprehensive analysis of tumor-infiltrating immune cells and tumor-infiltrating lymphocytes (TILs). RESULT NDRG1 expression was higher in HCC tissue than in normal liver tissue at both the mRNA and protein levels. Overexpression of NDRG1 is associated with poor prognosis in HCC patients. Genomic analysis suggests that NDRG1 promoter hypermethylation leads to enhanced transcription, which may be one mechanism for NDRG1 upregulation in HCC. The overexpression of NDRG1 promotes the invasion, migration, and proliferation of HCC cells and induces the expression of EMT-related proteins. Immunoinfiltration analysis suggests that NDRG1 is involved in the recruitment of immune cells. CONCLUSIONS The present study showed that NDRG1 may induce metastasis and invasion through EMT and immune cell infiltration. NDRG1 could be used as a biomarker for the diagnosis and prognosis of HCC and could be a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Ran Sun
- Hospital of Integrated Chinese and Western Medicine , Tianjin 300100, China
| | - Na Che
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Dpartment of Pathology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Dpartment of Pathology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Yanlei Li
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Dpartment of Pathology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China; Dpartment of Pathology, General Hospital of Tianjin Medical University, Tianjin 300052, China.
| |
Collapse
|
16
|
Du G, Dou C, Sun P, Wang S, Liu J, Ma L. Regulatory T cells and immune escape in HCC: understanding the tumor microenvironment and advancing CAR-T cell therapy. Front Immunol 2024; 15:1431211. [PMID: 39136031 PMCID: PMC11317284 DOI: 10.3389/fimmu.2024.1431211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Liver cancer, which most commonly manifests as hepatocellular carcinoma (HCC), is the sixth most common cancer in the world. In HCC, the immune system plays a crucial role in the growth and proliferation of tumor cells. HCC achieve immune escape through the tumor microenvironment, which significantly promotes the development of this cancer. Here, this article introduces and summarizes the functions and effects of regulatory T cells (Tregs) in the tumor microenvironment, highlighting how Tregs inhibit and regulate the functions of immune and tumor cells, cytokines, ligands and receptors, etc, thereby promoting tumor immune escape. In addition, it discusses the mechanism of CAR-T therapy for HCC and elaborate on the relationship between CAR-T and Tregs.
Collapse
Affiliation(s)
- Guangtan Du
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Cunmiao Dou
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Peng Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Leina Ma
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| |
Collapse
|
17
|
Ren B, Li X, Zhang Z, Tai S, Yu S. Exosomes: a significant medium for regulating drug resistance through cargo delivery. Front Mol Biosci 2024; 11:1379822. [PMID: 39135913 PMCID: PMC11317298 DOI: 10.3389/fmolb.2024.1379822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/17/2024] [Indexed: 08/15/2024] Open
Abstract
Exosomes are small lipid nanovesicles with a diameter of 30-150 nm. They are present in all body fluids and are actively secreted by the majority of cells through the process of exocytosis. Exosomes play an essential role in intercellular communication and act as significant molecular carriers in regulating various physiological and pathological processes, such as the emergence of drug resistance in tumors. Tumor-associated exosomes transfer drug resistance to other tumor cells by releasing substances such as multidrug resistance proteins and miRNAs through exosomes. These substances change the cell phenotype, making it resistant to drugs. Tumor-associated exosomes also play a role in impacting drug resistance in other cells, like immune cells and stromal cells. Exosomes alter the behavior and function of these cells to help tumor cells evade immune surveillance and form a tumor niche. In addition, exosomes also export substances such as tumoricidal drugs and neutralizing antibody drugs to help tumor cells resist drug therapy. In this review, we summarize the mechanisms of exosomes in promoting drug resistance by delivering cargo in the context of the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Bixuan Ren
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoqing Li
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhihua Zhang
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Sheng Tai
- Department of Hepatic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shan Yu
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
18
|
Li D, Zhang T, Guo Y, Bi C, Liu M, Wang G. Biological impact and therapeutic implication of tumor-associated macrophages in hepatocellular carcinoma. Cell Death Dis 2024; 15:498. [PMID: 38997297 PMCID: PMC11245522 DOI: 10.1038/s41419-024-06888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024]
Abstract
The tumor microenvironment is a complex space comprised of normal, cancer and immune cells. The macrophages are considered as the most abundant immune cells in tumor microenvironment and their function in tumorigenesis is interesting. Macrophages can be present as M1 and M2 polarization that show anti-cancer and oncogenic activities, respectively. Tumor-associated macrophages (TAMs) mainly have M2 polarization and they increase tumorigenesis due to secretion of factors, cytokines and affecting molecular pathways. Hepatocellular carcinoma (HCC) is among predominant tumors of liver that in spite of understanding its pathogenesis, the role of tumor microenvironment in its progression still requires more attention. The presence of TAMs in HCC causes an increase in growth and invasion of HCC cells and one of the reasons is induction of glycolysis that such metabolic reprogramming makes HCC distinct from normal cells and promotes its malignancy. Since M2 polarization of TAMs stimulates tumorigenesis in HCC, molecular networks regulating M2 to M1 conversion have been highlighted and moreover, drugs and compounds with the ability of targeting TAMs and suppressing their M2 phenotypes or at least their tumorigenesis activity have been utilized. TAMs increase aggressive behavior and biological functions of HCC cells that can result in development of therapy resistance. Macrophages can provide cell-cell communication in HCC by secreting exosomes having various types of biomolecules that transfer among cells and change their activity. Finally, non-coding RNA transcripts can mainly affect polarization of TAMs in HCC.
Collapse
Affiliation(s)
- Deming Li
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, PR China
| | - Ting Zhang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, 110001, PR China
| | - Ye Guo
- Department of Intervention, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, PR China
| | - Cong Bi
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, PR China.
| | - Ming Liu
- Department of Oral Radiology, School of Stomatology, China Medical University, Shenyang, Liaoning, 110002, PR China.
| | - Gang Wang
- Department of Intervention, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, PR China.
| |
Collapse
|
19
|
Zhou J, Li L, Han Y, Ge G, Ji Q, Li H. RNA binding protein RALY facilitates colorectal cancer metastasis via enhancing exosome biogenesis in m6A dependent manner. Int J Biol Macromol 2024; 273:133112. [PMID: 38880454 DOI: 10.1016/j.ijbiomac.2024.133112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Tumor metastasis is the leading cause of cancer-related death in patients with colorectal cancer (CRC). Heterogeneous nuclear ribonucleoproteins (hnRNPs) are RNA-binding proteins, involved in the tumorigenesis and metastasis of various cancers. However, the molecular mechanisms of hnRNPs in CRC metastasis remain unclear. This study aims to uncover the pivotal roles and molecular mechanisms of hnRNPs in CRC metastasis. Clinical database analysis suggested that the expression of hnRNP-Associated with Lethal Yellow (RALY, an important member of hnRNPs) was strongly correlated with the aggressiveness and survival of CRC patients. Gain- and loss-of-function studies demonstrated that RALY promotes the production of exosomes by increasing the formation of multivesicular bodies (MVBs) and enhancing the fusion of MVBs with the plasma membrane. Notably, RALY directly interacts with phospholipase D2 (PLD2) to enable exosome biogenesis, and cooperates with RBM15b to control PLD2 mRNA stability in an m6A-dependent manner. RALY-mediated exosome secretion activates pro-tumor macrophages and further facilitates CRC metastasis, while rescue experiments in vivo further confirmed that RALY-mediated exosome biogenesis facilitates CRC metastasis. Collectively, our findings demonstrate that RALY promotes exosome biogenesis and facilitates colorectal cancer metastasis by upregulating PLD2 and enhancing exosome production in an m6A-dependent manner, suggesting potential therapeutic strategies for combating CRC metastasis.
Collapse
Affiliation(s)
- Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ling Li
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yicun Han
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Qing Ji
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China.
| |
Collapse
|
20
|
Li Z, Duan D, Li L, Peng D, Ming Y, Ni R, Liu Y. Tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for hepatocellular carcinoma: recent research progress. Front Pharmacol 2024; 15:1382256. [PMID: 38957393 PMCID: PMC11217528 DOI: 10.3389/fphar.2024.1382256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the cancers that seriously threaten human health. Immunotherapy serves as the mainstay of treatment for HCC patients by targeting the programmed cell death protein 1/programmed cell death 1 ligand 1 (PD-1/PD-L1) axis. However, the effectiveness of anti-PD-1/PD-L1 treatment is limited when HCC becomes drug-resistant. Tumor-associated macrophages (TAMs) are an important factor in the negative regulation of PD-1 antibody targeted therapy in the tumor microenvironment (TME). Therefore, as an emerging direction in cancer immunotherapy research for the treatment of HCC, it is crucial to elucidate the correlations and mechanisms between TAMs and PD-1/PD-L1-mediated immune tolerance. This paper summarizes the effects of TAMs on the pathogenesis and progression of HCC and their impact on HCC anti-PD-1/PD-L1 immunotherapy, and further explores current potential therapeutic strategies that target TAMs in HCC, including eliminating TAMs in the TME, inhibiting TAMs recruitment to tumors and functionally repolarizing M2-TAMs (tumor-supportive) to M1-TAMs (antitumor type).
Collapse
Affiliation(s)
| | | | | | | | | | - Rui Ni
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Yao Liu
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
21
|
Guo N, Wang Y, Wen Z, Fan X. Promising nanotherapeutics of stem cell extracellular vesicles in liver regeneration. Regen Ther 2024; 26:1037-1047. [PMID: 39569342 PMCID: PMC11576938 DOI: 10.1016/j.reth.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/22/2024] [Accepted: 09/26/2024] [Indexed: 11/22/2024] Open
Abstract
Extracellular vesicles (EVs) have gainedsignificant attention due totheir crucialroles invarious biological systems. This review aims to explore the functions of EVs in both in physiological and pathological states of the liver, with a specific focus on the potential mechanisms and concrete evidence of EVs in liver regeneration processes. The review begins by emphasizing the importance of EVs in maintaining liver health and their involvement in different pathological conditions, starting from the liver's own EVs. Reviewing the role of EVs in liver diseases to reveal the impact of EVs in pathological processes (e.g., hepatitis, liver fibrosis, and cirrhosis) and elucidate their signaling functions at the molecular level. Subsequently, the work concentrates on the functions of EVs in liver regeneration, revealing their key role in repair and regeneration following liver injury by carrying growth factors, nucleic acids, and other bioactive molecules. This part not only theoretically clarifies the mechanisms of EVs in liver regeneration but also experimentally demonstrates their role in promoting liver cell proliferation, inhibiting apoptosis, regulating immune responses, and fostering angiogenesis, laying the groundwork for future clinical applications. Moreover, this work provides a comprehensive analysis of the challenges faced by existing EV-based therapies in liver regeneration and offers prospects for future research directions. It highlights that despite the tremendous potential of EVs in treating liver diseases, there are still technical challenges (e.g., EV isolation and purification, dosage control, and targeted delivery). To overcome these challenges, the review suggests improvements to current technologies and the development of new methods to realize the clinical application of EVs in treating liver diseases.
Collapse
Affiliation(s)
- Na Guo
- Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Hexi Yuelu District, Changsha, Hunan, 410000, China
| | - Yan Wang
- Department of Basic Medicine, Cangzhou Medical College, No.39, West Jiuhe Road, Cangzhou, 061001, China
| | - Zhaofeng Wen
- Heze Medical College, No.1950, Daxue Road, Heze Shandong, 274000, China
| | - Xiaofei Fan
- Shandong Medical College, No.5460, Second Ring South Road, Jinan, Shandong, 250002, China
| |
Collapse
|
22
|
Liang C, Zhang Y, Wang S, Jiao W, Guo J, Zhang N, Liu X. Nanomaterials in modulating tumor-associated macrophages and enhancing immunotherapy. J Mater Chem B 2024; 12:4809-4823. [PMID: 38695349 DOI: 10.1039/d4tb00230j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Tumor-associated macrophages (TAMs) are predominantly present in the tumor microenvironment (TME) and play a crucial role in shaping the efficacy of tumor immunotherapy. These TAMs primarily exhibit a tumor-promoting M2-like phenotype, which is associated with the suppression of immune responses and facilitation of tumor progression. Interestingly, recent research has highlighted the potential of repolarizing TAMs from an M2 to a pro-inflammatory M1 status-a shift that has shown promise in impeding tumor growth and enhancing immune responsiveness. This concept is particularly intriguing as it offers a new dimension to cancer therapy by targeting the tumor microenvironment, which is a significant departure from traditional approaches that focus solely on tumor cells. However, the clinical application of TAM-modulating agents is often challenged by issues such as insufficient tumor accumulation and off-target effects, limiting their effectiveness and safety. In this regard, nanomaterials have emerged as a novel solution. They serve a dual role: as delivery vehicles that can enhance the accumulation of therapeutic agents in the tumor site and as TAM-modulators. This dual functionality of nanomaterials is a significant advancement as it addresses the key limitations of current TAM-modulating strategies and opens up new avenues for more efficient and targeted therapies. This review provides a comprehensive overview of the latest mechanisms and strategies involving nanomaterials in modulating macrophage polarization within the TME. It delves into the intricate interactions between nanomaterials and macrophages, elucidating how these interactions can be exploited to drive macrophage polarization towards a phenotype that is more conducive to anti-tumor immunity. Additionally, the review explores the burgeoning field of TAM-associated nanomedicines in combination with tumor immunotherapy. This combination approach is particularly promising as it leverages the strengths of both nanomedicine and immunotherapy, potentially leading to synergistic effects in combating cancer.
Collapse
Affiliation(s)
- Chen Liang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Yihan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Siyao Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Wangbo Jiao
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Jingyi Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Nan Zhang
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiaoli Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
23
|
Ran XM, Yang J, Wang ZY, Xiao LZ, Deng YP, Zhang KQ. M2 macrophage-derived exosomal circTMCO3 acts through miR-515-5p and ITGA8 to enhance malignancy in ovarian cancer. Commun Biol 2024; 7:583. [PMID: 38755265 PMCID: PMC11098810 DOI: 10.1038/s42003-024-06095-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 03/22/2024] [Indexed: 05/18/2024] Open
Abstract
Tumor-associated macrophages of the M2 phenotype promote cancer initiation and progression. Importantly, M2 macrophage-derived exosomes play key roles in the malignancy of cancer cells. Here, we report that circTMCO3 is upregulated in ovarian cancer patients, and its high expression indicates poor survival. M2-derived exosomes promote proliferation, migration, and invasion in ovarian cancer, but these effects are abolished by knockdown of circTMCO3. Furthermore, circTMCO3 functions as a competing endogenous RNA for miR-515-5p to reduce its abundance, thus upregulating ITGA8 in ovarian cancer. miR-515-5p inhibits ovarian cancer malignancy via directly downregulating ITGA8. The decreased oncogenic activity of circTMCO3-silencing exosomes is reversed by miR-515-5p knockdown or ITGA8 overexpression. Exosomal circTMCO3 promotes ovarian cancer progression in nude mice. Thus, M2 macrophage-derived exosomes promote malignancy by delivering circTMCO3 and targeting the miR-515-5p/ITGA8 axis in ovarian cancer. Our findings not only provide mechanistic insights into ovarian cancer progression, but also suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Xiao-Min Ran
- Department of Gynecologic Oncology Ward 5, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Juan Yang
- Department of Gynecologic Oncology Ward 5, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Zi-Yi Wang
- Department of Gynecologic Oncology Ward 5, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Ling-Zhi Xiao
- Department of Gynecologic Oncology Ward 5, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Yu-Ping Deng
- Department of Gynecologic Oncology Ward 5, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Ke-Qiang Zhang
- Department of Gynecologic Oncology Ward 5, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, PR China.
| |
Collapse
|
24
|
Meng Y, Zhang M, Li X, Wang X, Dong Q, Zhang H, Zhai Y, Song Q, He F, Tian C, Sun A. Myeloid cell-expressed MNDA enhances M2 polarization to facilitate the metastasis of hepatocellular carcinoma. Int J Biol Sci 2024; 20:2814-2832. [PMID: 38904028 PMCID: PMC11186364 DOI: 10.7150/ijbs.91877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 04/27/2024] [Indexed: 06/22/2024] Open
Abstract
Stable infiltration of myeloid cells, especially tumor-associated M2 macrophages, acts as one of the essential features of the tumor immune microenvironment by promoting the malignant progression of hepatocellular carcinoma (HCC). However, the factors affecting the infiltration of M2 macrophages are not fully understood. In this study, we found the molecular subtypes of HCC with the worst prognosis are characterized by immune disorders dominated by myeloid cell infiltration. Myeloid cell nuclear differentiation antigen (MNDA) was significantly elevated in the most aggressive subtype and exhibited a positively correlation with M2 infiltration and HCC metastasis. Moreover, MNDA functioned as an independent prognostic predictor and has a good synergistic effect with some existing prognostic clinical indicators. We further confirmed that MNDA was primarily expressed in tumor M2 macrophages and contributed to the enhancement of its polarization by upregulating the expression of the M2 polarization enhancers. Furthermore, MNDA could drive the secretion of M2 macrophage-derived pro-metastasis proteins to accelerate HCC cells metastasis both in vivo and in vitro. In summary, MNDA exerts a protumor role by promoting M2 macrophages polarization and HCC metastasis, and can serve as a potential biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Yanru Meng
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, China
| | - Mengxin Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xinli Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xinxin Wang
- Department of Pathology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, China
| | - Qian Dong
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Hu Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yuanjun Zhai
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Qin Song
- College of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Fuchu He
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
- Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chunyan Tian
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
- Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, China
- College of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Aihua Sun
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
- Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, China
- College of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| |
Collapse
|
25
|
Wang X, Niu R, Yang H, Lin Y, Hou H, Yang H. Fibroblast activation protein promotes progression of hepatocellular carcinoma via regulating the immunity. Cell Biol Int 2024; 48:577-593. [PMID: 38501437 DOI: 10.1002/cbin.12154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/26/2023] [Accepted: 11/13/2023] [Indexed: 03/20/2024]
Abstract
Fibroblast activation protein (FAP) has been indicated to express in cancer-associated fibroblasts (CAFs) in most cancers. This work was dedicated to exploring FAP's effects on hepatocellular carcinoma (HCC). The data were extracted from The Cancer Genome Atlas, Gene Expression Omnibus, ImmPort, and Reactome databases. The correlation between FAP and HCC patients' prognosis was explored via survival analysis. The qRT-PCR and western blot analysis were used to analyze the FAP mRNA and protein expression levels, respectively. The cell proliferation and apoptosis were determined using the cell counting kit-8 assay kit and Annexin V-FITC/PI apoptosis kit, respectively. The HCC patients with FAP overexpression displayed a worse prognosis. The FAP expression was positively associated with the infiltration levels of tumor purity, B cell, CD8 + T cell, CD4 + T cell, macrophage, neutrophil, and dendritic cell. The optimal nine immune related genes were screened between two groups (FAP high vs. low). Moreover, we identified 24 energy metabolism related genes (FAP high vs. low) and these 24 genes were highly expressed in the high FAP expression group. The FAP expression had a significant positive correlation with the expression of PD-1, CTLA4, PDL-1, and PDL-2. The FAP overexpression promoted proliferation and migration while inhibiting the apoptosis of HCC cells. The FAP overexpression promoted the progression of HCC by regulating the immunity to affect the prognosis of HCC patients, thereby serving as a poor prognostic marker for HCC patients.
Collapse
Affiliation(s)
- Xiangcheng Wang
- Department of Nuclear Medicine, Shenzhen People's Hospital, Shenzhen, P.R. China
| | - Ruilong Niu
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, P.R. China
| | - Hao Yang
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital & Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, P.R. China
| | - Yu Lin
- Department of Radiation Oncology, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, P.R. China
| | - Hui Hou
- Department of Paediatrics, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, P.R. China
| | - Hong Yang
- Department of Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, P.R. China
| |
Collapse
|
26
|
Zhou X, Kong X, Lu J, Wang H, Liu M, Zhao S, Xia Z, Liu Q, Sun H, Gao X, Ma C, Niu Z, Yang F, Song X, Gao H, Zhang S, Zhu H. Circulating tumor cell-derived exosome-transmitted long non-coding RNA TTN-AS1 can promote the proliferation and migration of cholangiocarcinoma cells. J Nanobiotechnology 2024; 22:191. [PMID: 38637832 PMCID: PMC11025154 DOI: 10.1186/s12951-024-02459-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 04/04/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Exosomes assume a pivotal role as essential mediators of intercellular communication within tumor microenvironments. Within this context, long noncoding RNAs (LncRNAs) have been observed to be preferentially sorted into exosomes, thus exerting regulatory control over the initiation and progression of cancer through diverse mechanisms. RESULTS Exosomes were successfully isolated from cholangiocarcinoma (CCA) CTCs organoid and healthy human serum. Notably, the LncRNA titin-antisense RNA1 (TTN-AS1) exhibited a conspicuous up-regulation within CCA CTCs organoid derived exosomes. Furthermore, a significant elevation of TTN-AS1 expression was observed in tumor tissues, as well as in blood and serum exosomes from patients afflicted with CCA. Importantly, this hightened TTN-AS1 expression in serum exosomes of CCA patients manifested a strong correlation with both lymph node metastasis and TNM staging. Remarkably, both CCA CTCs organoid-derived exosomes and CCA cells-derived exosomes featuring pronounced TTN-AS1 expression demonstrated the capability to the proliferation and migratory potential of CCA cells. Validation of these outcomes was conducted in vivo experiments. CONCLUSIONS In conclusion, our study elucidating that CCA CTCs-derived exosomes possess the capacity to bolster the metastasis tendencies of CCA cells by transporting TTN-AS1. These observations underscore the potential of TTN-AS1 within CTCs-derived exosomes to serve as a promising biomarker for the diagnosis and therapeutic management of CCA.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Xiaohan Kong
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Jun Lu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Heng Wang
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Meng Liu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Shuchao Zhao
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Zhaozhi Xia
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Qinggong Liu
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Hongrui Sun
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Xin Gao
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Chaoqun Ma
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Zheyu Niu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Faji Yang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Xie Song
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Hengjun Gao
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Shizhe Zhang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Huaqiang Zhu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hepatobiliary Surgery, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
27
|
Khoushab S, Aghmiuni MH, Esfandiari N, Sarvandani MRR, Rashidi M, Taheriazam A, Entezari M, Hashemi M. Unlocking the potential of exosomes in cancer research: A paradigm shift in diagnosis, treatment, and prevention. Pathol Res Pract 2024; 255:155214. [PMID: 38430814 DOI: 10.1016/j.prp.2024.155214] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024]
Abstract
Exosomes, which are tiny particles released by cells, have the ability to transport various molecules, including proteins, lipids, and genetic material containing non-coding RNAs (ncRNAs). They are associated with processes like cancer metastasis, immunity, and tissue repair. Clinical trials have shown exosomes to be effective in treating cancer, inflammation, and chronic diseases. Mesenchymal stem cells (MSCs) and dendritic cells (DCs) are common sources of exosome production. Exosomes have therapeutic potential due to their ability to deliver cargo, modulate the immune system, and promote tissue regeneration. Bioengineered exosomes could revolutionize disease treatment. However, more research is needed to understand exosomes in tumor growth and develop new therapies. This paper provides an overview of exosome research, focusing on cancer and exosome-based therapies including chemotherapy, radiotherapy, and vaccines. It explores exosomes as a drug delivery system for cancer therapy, highlighting their advantages. The article discusses using exosomes for various therapeutic agents, including drugs, antigens, and RNAs. It also examines challenges with engineered exosomes. Analyzing exosomes for clinical purposes faces limitations in sensitivity, specificity, and purification. On the other hand, Nanotechnology offers solutions to overcome these challenges and unlock exosome potential in healthcare. Overall, the article emphasizes the potential of exosomes for personalized and targeted cancer therapy, while acknowledging the need for further research.
Collapse
Affiliation(s)
- Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Hobabi Aghmiuni
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esfandiari
- Department of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
28
|
Mu Z, Shen T, Deng H, Zeng B, Huang C, Mao Z, Xie Y, Pei Y, Guo L, Hu R, Chen L, Zhou Y. Enantiomer-Dependent Supramolecular Immunosuppressive Modulation for Tissue Reconstruction. ACS NANO 2024; 18:5051-5067. [PMID: 38306400 DOI: 10.1021/acsnano.3c11601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Abstract
Modulating the properties of biomaterials in terms of the host immune response is critical for tissue repair and regeneration. However, it is unclear how the preference for the cellular microenvironment manipulates the chiral immune responses under physiological or pathological conditions. Here, we reported that in vivo and in vitro oligopeptide immunosuppressive modulation was achieved by manipulation of macrophage polarization using chiral tetrapeptide (Ac-FFFK-OH, marked as FFFK) supramolecular polymers. The results suggested that chiral FFFK nanofibers can serve as a defense mechanism in the restoration of tissue homeostasis by upregulating macrophage M2 polarization via the Src-STAT6 axis. More importantly, transiently acting STAT6, insufficient to induce a sustained polarization program, then passes the baton to EGR2, thereby continuously maintaining the M2 polarization program. It is worth noting that the L-chirality exhibits a more potent effect in inducing macrophage M2 polarization than does the D-chirality, leading to enhanced tissue reconstruction. These findings elucidate the crucial molecular signals that mediate chirality-dependent supramolecular immunosuppression in damaged tissues while also providing an effective chiral supramolecular strategy for regulating macrophage M2 polarization and promoting tissue injury repair based on the self-assembling chiral peptide design.
Collapse
Affiliation(s)
- Zhixiang Mu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Tianxi Shen
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Hui Deng
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Bairui Zeng
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Chen Huang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Zhengjin Mao
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Yuyu Xie
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, P. R. China
| | - Yu Pei
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, P. R. China
| | - Liting Guo
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325000, P. R. China
| | - Rongdang Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Limin Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, P. R. China
| | - Yunlong Zhou
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, P. R. China
| |
Collapse
|
29
|
Zhang R, Chen X, Miao C, Chen Y, Li Y, Shen J, Yuan M, Chen M, Cheng J, Liu S, Sun Q, Wu J. Tumor-associated macrophage-derived exosomal miR-513b-5p is a target of jianpi yangzheng decoction for inhibiting gastric cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117013. [PMID: 37572927 DOI: 10.1016/j.jep.2023.117013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/16/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jianpi Yangzheng decoction (JPYZ) possesses a potential anti-tumor activity in gastric cancer. However, potential effect of JPYZ on regulating tumor-associated macrophage (TAM)-derived exosomes to affect gastric cancer is still unclear. AIM OF STUDY We aimed to clarify the role of tumor-associated macrophage derived exosomes (TAM-exos) in invasive and metastasis of gastric cancer and the mechanism of JPYZ regulate TAM-exos against gastric cancer. MATERIALS AND METHODS Flow cytometry was performed to demonstrate whether JPYZ involved in TAM polarization. After JPYZ treatment, TAM conditioned medium (TAM-CM)/TAM-exos were co-cultured with gastric cancer cells and were detected by wound healing and transwell assay. Transcriptome sequencing and bioinformatics analysis predicted the exosomal miRNA after JPYZ intervention in TAM. miRNA mimic and inhibitor were used to verify the effect of miRNA in exosomes on gastric cancer cells. Q-PCR and luciferase reporter assay were employed to clarify the targeting relationship between miRNA and target gene. Western blot assay detected the expression levels of epithelial-mesenchymal transition (EMT) markers and related signaling pathways proteins. RESULTS We firstly demonstrated that TAM-CM intervened by JPYZ significantly inhibited the invasion and migration of gastric cancer. Furthermore, exosomes in TAM supernatants play a key role in migration of gastric cancer. Meanwhile, transcriptome sequencing and q-PCR revealed that miR-513b-5p expression was significantly reduced in TAM-exos intervened by JPYZ. And miR-513b-5p in TAM aggravated TAM-exos mediated invasion and migration of gastric cancer cells, the inhibitor of miR-513b-5p reversed TAM-exos mediated promotion. Bioinformatics analysis and luciferase reporter assay confirmed that PTEN was a direct target of miR-513b-5p in gastric cancer. MiR-513b-5p inhibited PTEN to activate AKT/mTOR signaling pathway thus promoting gastric cancer invasion and metastasis in vivo and in vitro. Importantly, JPYZ inhibited TAM derived exosomal miR-513b-5p, and alleviated AKT/mTOR activation by PTEN depended manner in gastric cancer. CONCLUSION TAM-exos containing miR-513b-5p lead to gastric cancer invasion and migration. Our findings clarify a novel TAM-exos mechanism of JPYZ for inhibiting gastric cancer progression.
Collapse
Affiliation(s)
- Ruijuan Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Xu Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Chunrun Miao
- Department of Gastroenterology, Dongtai Hospital of Traditional Chinese Medicine, Dongtai, Jiangsu, 224299, China
| | - Yuxuan Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Yaqi Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Junyu Shen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Mengyun Yuan
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Menglin Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China; No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Jian Cheng
- BD Bioscience, Becton, Dickinson and Company, Shanghai, 201200, China
| | - Shenlin Liu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Qingmin Sun
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Jian Wu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
30
|
Yang C, Xue Y, Duan Y, Mao C, Wan M. Extracellular vesicles and their engineering strategies, delivery systems, and biomedical applications. J Control Release 2024; 365:1089-1123. [PMID: 38065416 DOI: 10.1016/j.jconrel.2023.11.057] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 01/07/2024]
Abstract
Extracellular vesicles are nanoscale vesicles that can be secreted by all cell types, are intracellular in origin and have the same composition as their parent cells, play a key role in intercellular communication in organismal health and disease, and are now often used as biomarkers of disease and therapeutic agents in biomedical research. When injected locally or systemically, they have the ability to provide a variety of therapeutic effects, for example, regeneration of skin damage or restoration of cardiac function. However, direct injection of extracellular vesicles may result in their rapid clearance from the injection site.In order to maintain the biological activity of extracellular vesicles and to control the release of effective concentrations for better therapeutic efficacy during long-term disease treatment, the design of an optimized drug delivery system is necessary and different systems for the continuous delivery of extracellular vesicles have been developed. This paper first provides an overview of the biogenesis, composition and physiological function of extracellular vesicles, followed by a review of different strategies for extracellular vesicle isolation and methods for engineering extracellular vesicles. In addition, this paper reviews the latest extracellular vesicle delivery platforms such as micro-nanoparticles, injectable hydrogels, microneedles and scaffold patches. At the same time, the research progress and key cases of extracellular vesicle delivery systems in the field of biomedical therapeutics are described. Finally, the challenges and future trends of extracellular vesicle delivery are discussed.
Collapse
Affiliation(s)
- Chunhao Yang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yunxin Xue
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yu Duan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
31
|
Sungu M, Isik M, Güler Ü, Eylem CC, Eskizengin H, Nemutlu E, Salih B, Derkus B. Manipulating macrophage polarization with nanoparticles to control metastatic behavior in heterotypic breast cancer micro-tissues via exosome signaling. NANOSCALE 2023; 16:394-410. [PMID: 38073471 DOI: 10.1039/d3nr04980a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
This study aimed to investigate the effects of nanoparticles on macrophage polarization and their subsequent influence on post-tumorigenic behavior. Initially, seven different nanoparticles were applied to macrophages, and Zn-Ni-FeO (100 nm) and palladium nanoparticles (PdNPs, ∼25 nm) were found to induce M1-polarization in macrophages. A co-culture experiment was then conducted to examine the effects of macrophages on MCF-7 breast cancer micro-tissues. The M2-macrophages promoted tumor proliferation, while M1- and PdNPs-induced macrophages showed anti-tumor effects by suppressing cell proliferation. To reveal the mechanisms of effect, exosomes isolated from M1 (M1-Exo), M0 (M0-Exo), M2 (M2-Exo), and PdNPs-induced (PdNPs-Exo) macrophages were applied to the heterotypic tumor micro-tissues including MCF-7, human umbilical vein endothelial cells (HUVECs), and primary human dermal fibroblasts (phDFs). M2-Exo was seen to promote the migration of cancer cells and induce epithelial-mesenchymal transition (EMT), while M1-Exo suppressed these behaviors. PdNPs-Exo was effective in suppressing the aggressive nature of breast cancer cells similar to M1-Exo, moreover, the efficacy of 5-fluorouracil (5-FU) was increased in combination with PdNPs-Exo in both MCF-7 and heterotypic micro-tissues. In conclusion, PdNPs-Exo has potential anti-tumor effects, can be used as a combination therapy to enhance the efficacy of anti-cancer drugs, as well as innovative implants for breast cancer treatment.
Collapse
Affiliation(s)
- Mustafa Sungu
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey.
| | - Melis Isik
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey.
| | - Ülkü Güler
- Department of Chemistry, Faculty of Science, Hacettepe University, 06800 Ankara, Turkey
| | - Cemil Can Eylem
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey
| | - Hakan Eskizengin
- Department of Biology, Faculty of Science, Ankara University, 06560 Ankara, Turkey
| | - Emirhan Nemutlu
- Analytical Chemistry Division, Faculty of Pharmacy, Hacettepe University, Ankara 06230, Turkey
| | - Bekir Salih
- Department of Chemistry, Faculty of Science, Hacettepe University, 06800 Ankara, Turkey
| | - Burak Derkus
- Stem Cell Research Lab, Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey.
- Neuroscience and Neurotechnology Excellence Joint Application and Research Center (NEUROM), 06560 Ankara, Turkey
| |
Collapse
|
32
|
Wu D, Li Y. Application of adoptive cell therapy in hepatocellular carcinoma. Immunology 2023; 170:453-469. [PMID: 37435926 DOI: 10.1111/imm.13677] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/20/2023] [Indexed: 07/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) remains a global health challenge. Novel treatment modalities are urgently needed to extend the overall survival of patients. The liver plays an immunomodulatory function due to its unique physiological structural characteristics. Therefore, following surgical resection and radiotherapy, immunotherapy regimens have shown great potential in the treatment of hepatocellular carcinoma. Adoptive cell immunotherapy is rapidly developing in the treatment of hepatocellular carcinoma. In this review, we summarize the latest research on adoptive immunotherapy for hepatocellular carcinoma. The focus is on chimeric antigen receptor (CAR)-T cells and T cell receptor (TCR) engineered T cells. Then tumour-infiltrating lymphocytes (TILs), natural killer (NK) cells, cytokine-induced killer (CIK) cells, and macrophages are briefly discussed. The main overview of the application and challenges of adoptive immunotherapy in hepatocellular carcinoma. It aims to provide the reader with a comprehensive understanding of the current status of HCC adoptive immunotherapy and offers some strategies. We hope to provide new ideas for the clinical treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Dengqiang Wu
- Department of Clinical Laboratory, Ningbo No. 6 Hospital, Ningbo, China
| | - Yujie Li
- Clinical Laboratory of Ningbo Medical Centre Lihuili Hospital, Ningbo University, Zhejiang, Ningbo, China
| |
Collapse
|
33
|
Xiang SY, Deng KL, Yang DX, Yang P, Zhou YP. Function of macrophage-derived exosomes in chronic liver disease: From pathogenesis to treatment. World J Hepatol 2023; 15:1196-1209. [DOI: 10.4254/wjh.v15.i11.1196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023] Open
Abstract
Chronic liver disease (CLD) imposes a heavy burden on millions of people worldwide. Despite substantial research on the pathogenesis of CLD disorders, no optimal treatment is currently available for some diseases, such as liver cancer. Exosomes, which are extracellular vesicles, are composed of various cellular components. Exosomes have unique functions in maintaining cellular homeostasis and regulating cell communication, which are associated with the occurrence of disease. Furthermore, they have application potential in diagnosis and treatment by carrying diverse curative payloads. Hepatic macrophages, which are key innate immune cells, show extraordinary heterogeneity and polarization. Hence, macrophage-derived exosomes may play a pivotal role in the initiation and progression of various liver diseases. This review focuses on the effects of macrophage-derived exosomes on liver disease etiology and their therapeutic potential, which will provide new insights into alleviating the global pressure of CLD.
Collapse
Affiliation(s)
- Shi-Yi Xiang
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315020, Zhejiang Province, China
| | - Kai-Li Deng
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Dong-Xue Yang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315020, Zhejiang Province, China
- Institute of Digestive Disease of Ningbo University, Ningbo University, Ningbo 315020, Zhejiang Province, China
| | - Ping Yang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315020, Zhejiang Province, China
| | - Yu-Ping Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315020, Zhejiang Province, China
- Institute of Digestive Disease of Ningbo University, Ningbo University, Ningbo 315020, Zhejiang Province, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Ningbo Key Laboratory, Ningbo 315020, Zhejiang Province, China
| |
Collapse
|
34
|
Wang J, Zhang X, Ma X, Chen D, Cai M, Xiao L, Li J, Huang Z, Huang Y, Lian Y. Blockage of CacyBP inhibits macrophage recruitment and improves anti-PD-1 therapy in hepatocellular carcinoma. J Exp Clin Cancer Res 2023; 42:303. [PMID: 37968706 PMCID: PMC10652496 DOI: 10.1186/s13046-023-02885-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/03/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Despite remarkable advancements in cancer immunotherapy, the overall response rate to anti-programmed cell death-1 (anti-PD-1) therapy in hepatocellular carcinoma (HCC) patients remains low. Our previous study has demonstrated the critical role of CacyBP/SIP (Calcyclin-Binding Protein and Siah-1 Interacting Protein) as a regulator of HCC development and progression. However, the possible impact of CacyBP on the tumor immune microenvironment has not yet been clarified. METHODS The expressions of CacyBP and Myd88 in HCC cell lines and tissues was detected by bioinformatics analysis, real-time quantitative PCR, western blotting and immunohistochemistry. The interaction between CacyBP and Myd88 was measured using co-immunoprecipitation and immunofluorescence. In vitro and in vivo assays were used to investigate the regulation of CacyBP on tumor-associated macrophages (TAMs). RESULTS We identified that CacyBP was positively correlated with Myd88, a master regulator of innate immunity, and Myd88 was a novel binding substrate downstream of CacyBP in HCC. Additionally, CacyBP protected Myd88 from Siah-1-mediated proteasome-dependent degradation by competitively binding to its Toll/interleukin-1 receptor (TIR) domain. Inhibition of CacyBP-Myd88 signaling subsequently diminished HDAC1-mediated H3K9ac and H3K27ac modifications on the CX3CL1 promoter and reduced its transcription and secretion in HCC cells. Moreover, by using in vitro and in vivo strategies, we demonstrated that depletion of CacyBP impaired the infiltration of TAMs and the immunosuppressive state of the tumor microenvironment, further sensitizing HCC-bearing anti-PD-1 therapy. CONCLUSIONS Our findings suggest that targeting CacyBP may be a novel treatment strategy for improving the efficacy of anti-PD-1 immunotherapy in HCC.
Collapse
Affiliation(s)
- Jialiang Wang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China
| | - Xiaoyu Zhang
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China
| | - Xinyi Ma
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China
| | - Dongmei Chen
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China
| | - Meina Cai
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China
| | - Lexin Xiao
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China
| | - Jing Li
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China
| | - Zexuan Huang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China
| | - Yuehua Huang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China.
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China.
| | - Yifan Lian
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Rd., Guangzhou, 510630, China.
| |
Collapse
|
35
|
Li M, Che N, Liu X, Xuan Y, Jin Y. Dauricine regulates prostate cancer progression by inhibiting PI3K/AKT-dependent M2 polarization of macrophages. Biochem Pharmacol 2023; 217:115838. [PMID: 37778445 DOI: 10.1016/j.bcp.2023.115838] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023]
Abstract
M2 type tumor-associated macrophages, an essential component of the tumor microenvironment (TME), have been proved to contribute to tumor metastasis. Dauricine (Dau) has recently received widespread attention due to its multiple targets and low price. However, the effect of Dau on macrophage polarization of TME remains unclear. In this study, we investigated the effect of Dau on prostate cancer (PCa) metastasis and specifically its correlation to macrophage polarization. Our results showed that Dau efficiently suppressed M2 polarization of macrophages induced by interleukin (IL) -4 and IL-13. Mechanistically, Dau inhibited the activity of PI3K/AKT signaling pathway, which subsequently suppressed macrophage differentiation to M2 type. Importantly, our study indicated that Dau decreased the release of chitinase 3-like protein 1 (CHI3L1) from M2 macrophages, which ultimately inhibited the M2 macrophage-mediated progression of PCa cells in vitro and in vivo. Taken together, our data demonstrated that Dau suppressed M2 polarization of macrophages via downregulation of the PI3K/AKT signaling pathway, in turn, preventing proliferation, epithelial-mesenchymal transition, migration, and invasion of PCa cells. Thus, this study reveals a previously unrecognized function of Dau in inhibition of PCa progression via intervention in M2 polarization of macrophages.
Collapse
Affiliation(s)
- Mengxuan Li
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China
| | - Nan Che
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China
| | - Xingzhe Liu
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China; Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | - Yanhua Xuan
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China; Department of Pathology, Yanbian University College of Medicine, Yanji, China.
| | - Yu Jin
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China; Department of Human Anatomy and Histoembryology, Yanbian University College of Medicine, Yanji, China.
| |
Collapse
|
36
|
Wang X, Ye X, Chen Y, Lin J. Mechanism of M2 type macrophage-derived extracellular vesicles regulating PD-L1 expression via the MISP/IQGAP1 axis in hepatocellular carcinoma immunotherapy resistance. Int Immunopharmacol 2023; 124:110848. [PMID: 37633233 DOI: 10.1016/j.intimp.2023.110848] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a prevailing cancer affecting human health. M2 macrophages are essential in mediating immune responses in tumors. This study investigated the action of M2 macrophages in immune escape of HCC. METHODS Mitotic spindle positioning (MISP), IQ motif containing GTPase activating protein 1 (IQGAP1) and programmed cell death-1 (PD-L1) levels in primary HCC/tumor-adjacent tissues were determined by Western blot, followed by correlation analysis. M2 macrophage and CD3+CD8+T cell percentages were estimated by flow cytometry. Hep3B and HepG2 cells were treated with M2 macrophage conditioned medium (M2-CM) and M2 macrophage-derived extracellular vesicles (M2-EVs) and/or co-cultured with CD8+T cells, followed by assessment of cell viability and apoptosis. TNF-α and INF-γ levels were measured by ELISA. MISP and IQGAP1 overexpression plasmids were transfected into HCC cells to explore their role in immune escape. The interactions among MISP, IQGAP1, STAT3, and PD-L1 were analyzed by co-immunoprecipitation. The mechanism of M2-EVs in HCC immune escape was verified in nude mice. RESULTS MISP/IQGAP1/PD-L1 were upregulated in HCC tissues. MISP negatively-correlated with IQGAP1/PD-L1 and IQGAP1 positively-correlated with PD-L1. M2 macrophages were reduced but CD8+T cells were increased in HCC tissues with high MISP expression. M2-CM or M2-EVs inhibited the killing ability of CD8+T cells, increased HCC cell viability, impeded HCC cell apoptosis, induced CD8+T cell apoptosis, downregulated TNF-α and INF-γ, and upregulated PD-L1. M2-EVs facilitated HCC cell immune escape by potentiating IQGAP1 nuclear translocation and activating STAT3 phosphorylation through MISP downregulation. In vivo experiments further verified the action of M2-EVs through MISP. CONCLUSION M2-EVs promote HCC cell immune escape by upregulating PD-L1 through the MISP/IQGAP1/STAT3 axis.
Collapse
Affiliation(s)
- Xiaobo Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Xuxing Ye
- Department of Traditional Chinese Medicine, Jinhua Municipal Central Hospital, 351 Mingyue Street, Wucheng District, Jinhua, 321001, China
| | - Yanping Chen
- Department of Gastroenterology, Jinhua Municipal Central Hospital, 351 Mingyue Street, Wucheng District, Jinhua, 321001, China
| | - Junmei Lin
- Department of Traditional Chinese Medicine, Jinhua Municipal Central Hospital, 351 Mingyue Street, Wucheng District, Jinhua, 321001, China.
| |
Collapse
|
37
|
Ding Z, Deng Z, Li H. Single-cell transcriptome analysis reveals the key genes associated with macrophage polarization in liver cancer. Hepatol Commun 2023; 7:e0304. [PMID: 37889536 PMCID: PMC10615477 DOI: 10.1097/hc9.0000000000000304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/14/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND The aim of this study was to reveal the key genes associated with macrophage polarization in liver cancer. METHODS Data were downloaded from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas databases (TCGA). R package Seurat 4.0 was used to preprocess the downloaded single-cell sequencing data, principal component analysis, and clustering. R package SingleR was used to annotate cell types and calculate macrophage polarization scores. Spearman correlation analysis was performed to obtain key genes highly correlated with macrophage polarization in liver cancer. The Tumor IMmune Estimation Resource algorithm was used to analyze the correlation between genes and the infiltration level of macrophages. Finally, the prognostic model was constructed based on 6 macrophage polarization-related genes by multivariate Cox regression analysis. Kaplan-Meier curves and receiver operating characteristic curves validated the prognostic value of the prognostic model. RESULTS Two thousand highly variable genes were obtained after the normalization of single-cell profiles. In all, 16 principal components and 15 cell clusters were obtained. Monocytes and macrophages were the main immune cells in the microenvironment of liver cancer tissues. Macrophage polarization scores showed that cluster 5 had the highest degree of polarization. Spearman analysis yielded that a total of 6 key genes associated with macrophage polarization (CD53, TGFBI, S100A4, pyruvate kinase M, LSP1, SPP1), and Tumor IMmune Estimation Resource analysis showed that 6 key genes were significantly positively correlated with macrophage infiltration levels. The model constructed by 6 key genes could effectively evaluate the prognosis of patients with liver cancer. CONCLUSIONS The key genes associated with macrophage polarization, namely CD53, TGFBI, S100A4, pyruvate kinase M, LSP1, and SPP1, may be potential therapeutic targets for liver cancer.
Collapse
|
38
|
Yang D, Han F, Cai J, Sun H, Wang F, Jiang M, Zhang M, Yuan M, Zhou W, Li H, Yang L, Bai Y, Xiao L, Dong H, Cheng Q, Mao H, Zhou L, Wang R, Li Y, Nie H. N-glycosylation by N-acetylglucosaminyltransferase IVa enhances the interaction of integrin β1 with vimentin and promotes hepatocellular carcinoma cell motility. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119513. [PMID: 37295747 DOI: 10.1016/j.bbamcr.2023.119513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
N-glycosylation has been revealed to be tightly associated with cancer metastasis. As a key transferase that catalyzes the formation of β1,4 N-acetylglucosamine (β1,4GlcNAc) branches on the mannose core of N-glycans, N-acetylglucosaminyltransferase IVa (GnT-IVa) has been reported to be involved in hepatocellular carcinoma (HCC) metastasis by forming N-glycans; however, the underlying mechanisms are largely unknown. In the current study, we found that GnT-IVa was upregulated in HCC tissues and positively correlated with worse outcomes in HCC patients. We found that GnT-IVa could promote tumor growth in mice; notably, this effect was attenuated after mutating the enzymatic site (D445A) of GnT-IVa, suggesting that GnT-IVa regulated HCC progression by forming β1,4GlcNAc branches. To mechanistically investigate the role of GnT-IVa in HCC, we conducted GSEA and GO functional analysis as well as in vitro experiments. The results showed that GnT-IVa could enhance HCC cell migration, invasion and adhesion ability and increase β1,4GlcNAc branch glycans on integrin β1 (ITGB1), a tumor-associated glycoprotein that is closely involved in cell motility by interacting with vimentin. Interruption of β1,4GlcNAc branch glycan modification on ITGB1 could suppress the interaction of ITGB1 with vimentin and inhibit cell motility. These results revealed that GnT-IVa could promote HCC cell motility by affecting the biological functions of ITGB1 through N-glycosylation. In summary, our results revealed that GnT-IVa is highly expressed in HCC and can form β1,4GlcNAc branches on ITGB1, which are essential for interactions with vimentin to promote HCC cell motility. These findings not only proposed a novel mechanism for GnT-IVa in HCC progression but also revealed the significance of N-glycosylation on ITGB1 during the process, which may provide a novel target for future HCC therapy.
Collapse
Affiliation(s)
- Depeng Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Fang Han
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jialing Cai
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Handi Sun
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Fengyou Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Meiyi Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Mengmeng Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Mengfan Yuan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Wenyang Zhou
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huaxin Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lijun Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Yan Bai
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lixing Xiao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Haiyang Dong
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Qixiang Cheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Haoyu Mao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lu Zhou
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Ruonan Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Yu Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| | - Huan Nie
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
39
|
Kong P, Yang H, Tong Q, Dong X, Yi MA, Yan D. Expression of tumor-associated macrophages and PD-L1 in patients with hepatocellular carcinoma and construction of a prognostic model. J Cancer Res Clin Oncol 2023; 149:10685-10700. [PMID: 37306737 PMCID: PMC10423131 DOI: 10.1007/s00432-023-04949-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an inflammation-associated tumor involved in immune tolerance and evasion in the immune microenvironment. Immunotherapy can enhance the immune response of the body, break immune tolerance, and then recognize and kill tumor cells. The polarization homeostasis of M1 and M2 macrophages in tumor microenvironment (TME) is involved in the occurrence and development of tumors and has been considered a hot topic in tumor research. Programmed cell death ligand 1 (PD-L1) plays an important role in the polarity of TAM and affects the prognosis of HCC patients as a target of immunotherapy. To this end, efforts were hereby made to further explore the application value of PD-L1, M1 macrophages (CD86), and M2 macrophages (CD206) in the prognosis assessment of HCC, their correlation with immune cell infiltration in HCC tissues, and their bioenrichment function. METHODS The gene expression omnibus (GEO) and the Cancer Genome Atlas (TCGA) database were used to analyze the expression of PD-L1, CD86, and CD206 in different tumor tissues. The correlation between the expression of PD-L1, CD86, and CD206 and the infiltration of immune cells was analyzed using the Tumor Immune Estimation Resource (TIMER). The tissue specimens and clinicopathological data of hepatocellular carcinoma patients having undergone surgical treatment in our hospital were collected. Immunohistochemistry was used to verify the expression of PD-L1, CD86, and CD206, and analyze the relationship with clinicopathological features and prognosis of patients. Besides, nomogram was constructed to predict the overall survival (OS) of patients at 3 and 5 years. Finally, the protein-protein interaction network information was analyzed using STRING database, and GO analysis and KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis were performed to study the biological functions of PD-L1, CD86, and CD206. RESULT Bioinformatics analysis found that PD-L1, CD86, and CD206 were underexpressed in various tumor tissues including liver cancer, while the present immunohistochemical detection found that PD-L1, CD86, and CD206 were overexpressed in liver cancer tissues. Expressions of PD-L1, CD86, and CD206 were positively correlated with the infiltration level of immune cells in liver cancer, while the expression of PD-L1 was positively correlated with the degree of tumor differentiation. Meanwhile, the expression level of CD206 was positively correlated with gender and preoperative hepatitis, and patients with high expression of PD-L1 or low expression of CD86 had poor prognosis. AJCC stage, preoperative hepatitis, and the expression levels of PD-L1 and CD86 in cancer tissues were independent risk factors affecting survival of patients after radical hepatoma surgery. KEGG pathway enrichment analysis showed that PD-L1 was significantly enriched in T cell aggregation and lymphocyte aggregation, and might be involved in the formation of T cell antigen receptor CD3 complex and cell membrane. Besides, CD86 was significantly enriched in positive regulation of cell adhesion, regulation of mononuclear cell proliferation, regulation of leukocyte proliferation, and transduction of T cell receptor signaling pathway, while CD206 was significantly enriched in type 2 immune response, cellular response to LPS, cellular response to LPS, and involvement in cellular response to LPS. CONCLUSION In conclusion, these results suggest that PD-L1, CD86, and CD206 may be involved not only in the occurrence and development of HCC, but also in immune regulation, indicating the potential role of PD-L1 and CD86 as potential biomarkers and new therapeutic targets for prognosis assessment of liver cancer.
Collapse
Affiliation(s)
- Panpan Kong
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Huan Yang
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Qing Tong
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Xiaogang Dong
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Mamumaimaitijiang-Abula Yi
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China
| | - Dong Yan
- The First Ward of Hepatobiliary and Pancreatic Surgery, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830001, People's Republic of China.
| |
Collapse
|
40
|
Xu K, Liu Y, Luo H, Wang T. Efferocytosis signatures as prognostic markers for revealing immune landscape and predicting immunotherapy response in hepatocellular carcinoma. Front Pharmacol 2023; 14:1218244. [PMID: 37383726 PMCID: PMC10294713 DOI: 10.3389/fphar.2023.1218244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a highly lethal liver cancer with late diagnosis; therefore, the identification of new early biomarkers could help reduce mortality. Efferocytosis, a process in which one cell engulfs another cell, including macrophages, dendritic cells, NK cells, etc., plays a complex role in tumorigenesis, sometimes promoting and sometimes inhibiting tumor development. However, the role of efferocytosis-related genes (ERGs) in HCC progression has been poorly studied, and their regulatory effects in HCC immunotherapy and drug targeting have not been reported. Methods: We downloaded efferocytosis-related genes from the Genecards database and screened for ERGs that showed significant expression changes between HCC and normal tissues and were associated with HCC prognosis. Machine learning algorithms were used to study prognostic gene features. CIBERSORT and pRRophetic R packages were used to evaluate the immune environment of HCC subtypes and predict treatment response. CCK-8 experiments conducted on HCC cells were used to assess the reliability of drug sensitivity prediction. Results: We constructed a prognostic prediction model composed of six genes, and the ROC curve showed good predictive accuracy of the risk model. In addition, two ERG-related subgroups in HCC showed significant differences in tumor immune landscape, immune response, and prognostic stratification. The CCK-8 experiment conducted on HCC cells confirmed the reliability of drug sensitivity prediction. Conclusion: Our study emphasizes the importance of efferocytosis in HCC progression. The risk model based on efferocytosis-related genes developed in our study provides a novel precision medicine approach for HCC patients, allowing clinicians to customize treatment plans based on unique patient characteristics. The results of our investigation carry noteworthy implications for the development of individualized treatment approaches involving immunotherapy and chemotherapy, thereby potentially facilitating the realization of personalized and more efficacious therapeutic interventions for HCC.
Collapse
Affiliation(s)
- Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Yu Liu
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Huiyan Luo
- Department of Oncology, Chongqing General Hospital, Chongqing, China
| | - Tengfei Wang
- Department of Equipment, Bishan Hospital of Chongqing, Chongqing, China
| |
Collapse
|
41
|
Wang Y, Yang W, Wang Q, Zhou Y. Mechanisms of esophageal cancer metastasis and treatment progress. Front Immunol 2023; 14:1206504. [PMID: 37359527 PMCID: PMC10285156 DOI: 10.3389/fimmu.2023.1206504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Esophageal cancer is a prevalent tumor of the digestive tract worldwide. The detection rate of early-stage esophageal cancer is very low, and most patients are diagnosed with metastasis. Metastasis of esophageal cancer mainly includes direct diffusion metastasis, hematogenous metastasis, and lymphatic metastasis. This article reviews the metabolic process of esophageal cancer metastasis and the mechanisms by which M2 macrophages, CAF, regulatory T cells, and their released cytokines, including chemokines, interleukins, and growth factors, form an immune barrier to the anti-tumor immune response mediated by CD8+ T cells, impeding their ability to kill tumor cells during tumor immune escape. The effect of Ferroptosis on the metastasis of esophageal cancer is briefly mentioned. Moreover, the paper also summarizes common drugs and research directions in chemotherapy, immunotherapy, and targeted therapy for advanced metastatic esophageal cancer. This review aims to serve as a foundation for further investigations into the mechanism and management of esophageal cancer metastasis.
Collapse
Affiliation(s)
- Yusheng Wang
- Department of Thoracic Surgery, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Wei Yang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Qianyun Wang
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Yong Zhou
- Department of Gastroenterology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
42
|
Pan B, Chen Z, Zhang X, Wang Z, Yao Y, Wu X, Qiu J, Lin H, Yu L, Tu H, Tang N. 2,5-dimethylcelecoxib alleviated NK and T-cell exhaustion in hepatocellular carcinoma via the gastrointestinal microbiota-AMPK-mTOR axis. J Immunother Cancer 2023; 11:e006817. [PMID: 37316264 DOI: 10.1136/jitc-2023-006817] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND 2,5-dimethylcelecoxib (DMC), a derivative of celecoxib, is an inhibitor of microsomal prostaglandin E synthase-1 (mPGES-1). Our previous studies have demonstrated that DMC inhibits the expression of programmed death-ligand 1 on hepatocellular carcinoma (HCC) cells to prevent tumor progression. However, the effect and mechanism of DMC on HCC infiltrating immune cells remain unclear. METHODS In this study, single-cell-based high-dimensional mass cytometry was performed on the tumor microenvironment of HCC mice treated with DMC, celecoxib and MK-886 (a known mPGES-1 inhibitor). Moreover, 16S ribosomal RNA sequencing was employed to analyze how DMC improved the tumor microenvironment of HCC by remodeling the gastrointestinal microflora. RESULTS We found that (1) DMC significantly inhibited the growth of HCC and improved the prognosis of the mice, and this depended on the stronger antitumor activity of natural killer (NK) and T cells; (2) compared with celecoxib and MK-886, DMC significantly enhanced the cytotoxic and stem-like potential, and inhibited exhaustion of NK and T cells; (3) mechanistically, DMC inhibited the expression of programmed cell death protein-1 and upregulated interferon-γ expression of NK and T cells via the gastrointestinal microbiota (Bacteroides acidifaciens, Odoribacter laneus, and Odoribacter splanchnicus)-AMPK-mTOR axis. CONCLUSIONS Our study uncovers the role of DMC in improving the tumor microenvironment of HCC, which not only enriches the relationship between the mPGES-1/prostaglandin E2 pathway and the antitumor function of NK and T cells, but also provide an important strategic reference for multitarget or combined immunotherapy of HCC.Cite Now.
Collapse
Affiliation(s)
- Banglun Pan
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhanfei Chen
- Department of Laboratory Medicine, Affiliated Hospital of Putian University, Putian, China
- Key Laboratory of Medical Microecology (Putian University), Fujian Province University, Putian, China
| | - Xiaoxia Zhang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zengbin Wang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuxin Yao
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoxuan Wu
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiacheng Qiu
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hua Lin
- Department of Laboratory Medicine, Affiliated Hospital of Putian University, Putian, China
- Key Laboratory of Medical Microecology (Putian University), Fujian Province University, Putian, China
| | - Liumin Yu
- Department of Laboratory Medicine, Affiliated Hospital of Putian University, Putian, China
| | - Haijian Tu
- School of Basic Medical Sciences, Putian University, Putian, China
| | - Nanhong Tang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Cancer Center of Fujian Medical University, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
43
|
Zheng N, Wang T, Luo Q, Liu Y, Yang J, Zhou Y, Xie G, Ma Y, Yuan X, Shen L. M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance. Oncoimmunology 2023; 12:2210959. [PMID: 37197441 PMCID: PMC10184604 DOI: 10.1080/2162402x.2023.2210959] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023] Open
Abstract
T-cell-based immune checkpoint blockade therapy (ICB) can be undermined by local immunosuppressive M2-like tumor-associated macrophages (TAMs). However, modulating macrophages has proved difficult as the molecular and functional features of M2-TAMs on tumor growth are still uncertain. Here we reported that immunosuppressive M2 macrophages render cancer cells resistant to CD8+ T-cell-dependent tumor-killing refractory ICB efficacy by secreting exosomes. Proteomics and functional studies revealed that M2 macrophage-derived exosome (M2-exo) transmitted apolipoprotein E (ApoE) to cancer cells conferring ICB resistance by downregulated MHC-I expression curbing tumor intrinsic immunogenicity. Mechanistically, M2 exosomal ApoE diminished the tumor-intrinsic ATPase activity of binding immunoglobulin protein (BiP) to decrease tumor MHC-I expression. Sensitizing ICB efficacy can be achieved by the administration of ApoE ligand, EZ-482, enhancing ATPase activity of BiP to boost tumor-intrinsic immunogenicity. Therefore, ApoE may serve as a predictor and a potential therapeutic target for ICB resistance in M2-TAMs-enriched cancer patients. Collectively, our findings signify that the exosome-mediated transfer of functional ApoE from M2 macrophages to the tumor cells confers ICB resistance. Our findings also provide a preclinical rationale for treating M2-enriched tumors with ApoE ligand, EZ-482, to restore sensitivity to ICB immunotherapy.
Collapse
Affiliation(s)
- Naisheng Zheng
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Tingting Wang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Department of Clinical Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Qin Luo
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Department of Clinical Laboratory, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, Guangdong, P.R. China
| | - Yi Liu
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Junyao Yang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Guohua Xie
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yanhui Ma
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xiangliang Yuan
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
44
|
Zhang C, Wei S, Dai S, Li X, Wang H, Zhang H, Sun G, Shan B, Zhao L. The NR_109/FUBP1/c-Myc axis regulates TAM polarization and remodels the tumor microenvironment to promote cancer development. J Immunother Cancer 2023; 11:jitc-2022-006230. [PMID: 37217247 DOI: 10.1136/jitc-2022-006230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and exert an important role in tumor progression. Due to the heterogeneity and plasticity of TAMs, modulating the polarization states of TAMs is considered as a potential therapeutic strategy for tumors. Long noncoding RNAs (lncRNAs) have been implicated in various physiological and pathological processes, yet the underlying mechanism on how lncRNAs manipulate the polarization states of TAMs is still unclear and remains to be further investigated. METHODS Microarray analyses were employed to characterize the lncRNA profile involved in THP-1-induced M0, M1 and M2-like macrophage. Among those differentially expressed lncRNAs, NR_109 was further studied, for its function in M2-like macrophage polarization and the effects of the condition medium or macrophages mediated by NR_109 on tumor proliferation, metastasis and TME remodeling both in vitro and in vivo. Moreover, we revealed how NR_109 interacted with far upstream element-binding protein 1 (FUBP1) to regulate the protein stability through hindering ubiquitination modification by competitively binding with JVT-1. Finally, we examined sections of tumor patients to probe the correlation among the expression of NR_109 and related proteins, showing the clinical significance of NR_109. RESULTS We found that lncRNA NR_109 was highly expressed in M2-like macrophages. Knockdown NR_109 impeded IL-4 induced M2-like macrophage polarization and significantly reduced the activity of M2-like macrophages to support the proliferation and metastasis of tumor cells in vitro and in vivo. Mechanistically, NR_109 competed with JVT-1 to bind FUBP1 at its C-terminus domain, impeded the ubiquitin-mediated degradation of FUBP1, activated c-Myc transcription and thus promoted M2-like macrophages polarization. Meanwhile, as a transcription factor, c-Myc could bind to the promoter of NR_109 and enhance the transcription of NR_109. Clinically, high NR_109 expression was found in CD163+ TAMs from tumor tissues and was positively correlated with poor clinical stages of patients with gastric cancer and breast cancer. CONCLUSIONS Our work revealed for the first time that NR_109 exerted a crucial role in regulating the phenotype-remodeling and function of M2-like macrophages via a NR_109/FUBP1/c-Myc positive feedback loop. Thus, NR_109 has great translational potentials in the diagnosis, prognosis and immunotherapy of cancer.
Collapse
Affiliation(s)
- Cong Zhang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy; Clinical Oncology Research Center, Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Sisi Wei
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy; Clinical Oncology Research Center, Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Suli Dai
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy; Clinical Oncology Research Center, Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoya Li
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy; Clinical Oncology Research Center, Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huixia Wang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy; Clinical Oncology Research Center, Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongtao Zhang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Guogui Sun
- Department of Hebei Key Laboratory of Medical-industrial Integration Precision Medicine, Affiliated Hospital, North China University of Science and Technology, Tangshan, China
| | - Baoen Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy; Clinical Oncology Research Center, Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy; Clinical Oncology Research Center, Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
45
|
Zhao H, Wei S, Zhou D, Liu Y, Guo Z, Fang C, Pang X, Li F, Hou H, Cui X. Blocking the CXCL1-CXCR2 axis enhances the effects of doxorubicin in HCC by remodelling the tumour microenvironment via the NF-κB/IL-1β/CXCL1 signalling pathway. Cell Death Discov 2023; 9:120. [PMID: 37037815 PMCID: PMC10085981 DOI: 10.1038/s41420-023-01424-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/12/2023] Open
Abstract
Inflammation is a core mechanism for oncogenesis. Chemokines act as important mediators of chronic inflammation and the tumour inflammatory response. However, there is limited information on chemokines in hepatocellular carcinoma (HCC), a disease for which almost all cases are derived from chronic liver inflammation. Here, we explored the protumor effects of CXCL1, a commonly elevated inflammatory chemokine in cirrhosis, in HCC. The protumor role was confirmed in clinical samples from HCC patients. CXCL1 enhanced tumorigenesis in the hepatic inflammatory microenvironment directly by acting on tumour cells and indirectly through promoting the recruitment of macrophages. The increase in the number of macrophages in the tumour microenvironment (TME) promoted tumour cell epithelial-mesenchymal transition (EMT) and significantly increased CXCL1 levels in the TME partly through NF-κB/IL-1β activation. To investigate the potential therapeutic value of CXCL1 in HCC with an inflammatory background, an antibody blocking CXCL1 was used alone or combined with the chemotherapy agent doxorubicin (DOX), with the goal of reshaping the TME. It has been shown that blocking CXCL1-CXCR2 inhibits tumour progression and reduces macrophage recruitment in the TME. The combination regimen has been shown to synergistically reduce the number of pro-tumour macrophages in the TME and suppress tumour progression. This provides insight into therapeutic strategies for treating HCC patients with high CXCL1 expression.
Collapse
Affiliation(s)
- Huiyong Zhao
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Sheng Wei
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Dachen Zhou
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yongfan Liu
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Zicheng Guo
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Chuibao Fang
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Xiaoxi Pang
- Department of Nuclear Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| | - Fei Li
- Department of Nuclear Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| | - Hui Hou
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China.
| | - Xiao Cui
- Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
46
|
Li J, Zhao C, Wang D, Wang S, Dong H, Wang D, Yang Y, Li J, Cui F, He X, Qin J. ZIM3 activation of CCL25 expression in pulmonary metastatic nodules of osteosarcoma recruits M2 macrophages to promote metastatic growth. Cancer Immunol Immunother 2023; 72:903-916. [PMID: 36161509 DOI: 10.1007/s00262-022-03300-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 09/20/2022] [Indexed: 10/14/2022]
Abstract
Tumor-associated macrophages (TAMs) play an important role in tumor growth and metastasis. However, the involvement of TAMs infiltration in pulmonary osteosarcoma (OS) metastasis remains poorly understood. Therefore, the effect of OS cells on macrophages migration was investigated by in vivo and in vitro experiments to evaluate the infiltration and mechanism of TAMs in pulmonary OS metastases. The results showed that the zinc finger protein ZIM3 was upregulated in OS cells than in osteoblasts and activated the expression of CCL25, which subsequently promoted the migration of M2 macrophages. CCL25 or ZIM3 silencing in OS cells inhibited the infiltration of M2 macrophages and the formation of pulmonary metastatic nodules in a mouse model of pulmonary OS metastasis and prolonged the survival of the mice. Furthermore, bioinformatics analyses revealed that CCL25 and ZIM3 expressions are negatively correlated with the prognosis of OS patients. In conclusion, this study found that a large number of M2 TAMs were recruited into pulmonary metastatic nodules of OS through the activation of the ZIM3-CCL25 axis in OS cells, thereby facilitating OS metastasis. Therefore, the suppression of ZIM3-CCL25-induced recruitment of M2 TAMs to the metastatic sites might be considered as a therapeutic approach to inhibit the growth of pulmonary OS metastases.
Collapse
Affiliation(s)
- Jing Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Chenguang Zhao
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Dong Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Shuang Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi Province, People's Republic of China
| | - Hui Dong
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Difan Wang
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi Province, People's Republic of China
| | - Yubing Yang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Jiaxi Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Feng Cui
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Xijing He
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China.
| | - Jie Qin
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China.
| |
Collapse
|
47
|
Wu H, Feng J, Zhong W, Zouxu X, Xiong Z, Huang W, Zhang C, Wang X, Yi J. Model for predicting immunotherapy based on M2 macrophage infiltration in TNBC. Front Immunol 2023; 14:1151800. [PMID: 36999020 PMCID: PMC10043239 DOI: 10.3389/fimmu.2023.1151800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
IntroductionCompared to other types of breast cancer, triple-negative breast cancer (TNBC) does not effectively respond to hormone therapy and HER2 targeted therapy, showing a poor prognosis. There are currently a limited number of immunotherapeutic drugs available for TNBC, a field that requires additional development.MethodsCo-expressing genes with M2 macrophages were analyzed based on the infiltration of M2 macrophages in TNBC and the sequencing data in The Cancer Genome Atlas (TCGA) database. Consequently, the influence of these genes on the prognoses of TNBC patients was analyzed. GO analysis and KEGG analysis were performed for exploring potential signal pathways. Lasso regression analysis was conducted for model construction. The TNBC patients were scored by the model, and patients were divided into high- and low-risk groups. Subsequently, the accuracy of model was further verified using GEO database and patients information from the Cancer Center of Sun Yat-sen University. On this basis, we analyzed the accuracy of prognosis prediction, correlation with immune checkpoint, and immunotherapy drug sensitivity in different groups.ResultsOur findings revealed that OLFML2B, MS4A7, SPARC, POSTN, THY1, and CD300C genes significantly influenced the prognosis of TNBC. Moreover, MS4A7, SPARC, and CD300C were finally determined for model construction, and the model showed good accuracy in prognosis prediction. And 50 immunotherapy drugs with therapeutic significance in different groups were screened, which were assessed possible immunotherapeutics that have potential application and demonstrated the high precision of our prognostic model for predictive analysis.ConclusionMS4A7, SPARC, and CD300C, the three main genes used in our prognostic model, offer good precision and clinical application potential. Fifty immune medications were assessed for their ability to predict immunotherapy drugs, providing a novel approach to immunotherapy for TNBC patients and a more reliable foundation for applying drugs in subsequent treatments.
Collapse
Affiliation(s)
- Haoming Wu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Shantou, Guangdong, China
| | - Jikun Feng
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Wenjing Zhong
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Xiazi Zouxu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Zhengchong Xiong
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Weiling Huang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Chao Zhang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Xi Wang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
- *Correspondence: Xi Wang, ; Jiarong Yi,
| | - Jiarong Yi
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, the State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
- *Correspondence: Xi Wang, ; Jiarong Yi,
| |
Collapse
|
48
|
Ma C, Wang C, Zhang Y, Li Y, Fu K, Gong L, Zhou H, Li Y. Phillygenin inhibited M1 macrophage polarization and reduced hepatic stellate cell activation by inhibiting macrophage exosomal miR-125b-5p. Biomed Pharmacother 2023; 159:114264. [PMID: 36652738 DOI: 10.1016/j.biopha.2023.114264] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/18/2023] Open
Abstract
Liver fibrosis (LF) is an important stage in chronic liver disease development, characterized by hepatic stellate cell (HSC) activation and excessive extracellular matrix deposition. Phillygenin (PHI), an active component in the traditional Chinese medicine Forsythiae Fructus with a significant anti-inflammatory effect, has been proved to inhibit HSC activation. Macrophages can polarize to pro-inflammatory M1 phenotype and anti-inflammatory M2 phenotype, participating in LF development. Currently, Forsythiae Fructus and its many components have been proved to inhibit the inflammatory activation of macrophages. However, there is no direct evidence that PHI can regulate macrophage polarization, and the relationship between macrophage polarization and the anti-LF effect of PHI has not been studied. In this study, we found that PHI inhibited the co-expression of CD80 and CD86, and inhibited the mRNA expression and protein secretion of related inflammatory cytokines in RAW264.7 cells. For mechanism, PHI was found to inhibit the JAK1/JAK2-STAT1 and Notch1 signaling pathways. Subsequently, mHSCs were co-cultured with the conditioned media or exosomes from macrophages with different treatments. It was found that the conditioned media and exosomes from PHI-treated macrophages inhibited the expression of MMP2, TIMP1, TGF-β, α-SMA, COL1 and NF-κB in mHSCs. Moreover, through bioinformatic analysis and cell transfection, we confirmed that PHI reduced HSC activation by inhibiting the overexpression of miR-125b-5p in M1 macrophage-derived exosomes and restoring Stard13 expression in mHSCs. On the whole, PHI could inhibit M1 macrophage polarization by suppressing the JAK1/JAK2-STAT1 and Notch1 signaling pathways, and reduce HSC activation by inhibiting macrophage exosomal miR-125b-5p targeting Stard13. DATA AVAILABILITY: The raw data supporting the conclusions of this study are available in the article/Supplementary figures, and can be obtained from the first or corresponding author.
Collapse
Affiliation(s)
- Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yanzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Lihong Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Honglin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
49
|
Mao X, Zhou D, Lin K, Zhang B, Gao J, Ling F, Zhu L, Yu S, Chen P, Zhang C, Zhang C, Ye G, Fong S, Chen G, Luo W. Single-cell and spatial transcriptome analyses revealed cell heterogeneity and immune environment alternations in metastatic axillary lymph nodes in breast cancer. Cancer Immunol Immunother 2023; 72:679-695. [PMID: 36040519 DOI: 10.1007/s00262-022-03278-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/12/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND Tumor heterogeneity plays essential roles in developing cancer therapies, including therapies for breast cancer (BC). In addition, it is also very important to understand the relationships between tumor microenvironments and the systematic immune environment. METHODS Here, we performed single-cell, VDJ sequencing and spatial transcriptome analyses on tumor and adjacent normal tissue as well as axillar lymph nodes (LNs) and peripheral blood mononuclear cells (PBMCs) from 8 BC patients. RESULTS We found that myeloid cells exhibited environment-dependent plasticity, where a group of macrophages with both M1 and M2 signatures possessed high tumor specificity spatially and was associated with worse patient survival. Cytotoxic T cells in tumor sites evolved in a separate path from those in the circulatory system. T cell receptor (TCR) repertoires in metastatic LNs showed significant higher consistency with TCRs in tumor than those in nonmetastatic LNs and PBMCs, suggesting the existence of common neo-antigens across metastatic LNs and primary tumor cites. In addition, the immune environment in metastatic LNs had transformed into a tumor-like status, where pro-inflammatory macrophages and exhausted T cells were upregulated, accompanied by a decrease in B cells and neutrophils. Finally, cell interactions showed that cancer-associated fibroblasts (CAFs) contributed most to shaping the immune-suppressive microenvironment, while CD8+ cells were the most signal-responsive cells. CONCLUSIONS This study revealed the cell structures of both micro- and macroenvironments, revealed how different cells diverged in related contexts as well as their prognostic capacities, and displayed a landscape of cell interactions with spatial information.
Collapse
Affiliation(s)
- Xiaofan Mao
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China.,Medical Engineering Technology Research and development center of Immune Repertoire in Foshan, The First People's Hospital of Foshan, Foshan, China
| | - Dan Zhou
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Kairong Lin
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China.,Medical Engineering Technology Research and development center of Immune Repertoire in Foshan, The First People's Hospital of Foshan, Foshan, China
| | - Beiying Zhang
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China.,Medical Engineering Technology Research and development center of Immune Repertoire in Foshan, The First People's Hospital of Foshan, Foshan, China
| | - Juntao Gao
- MOE Key Laboratory of Bioinformatics; Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist; Department of Automation, Tsinghua University, Beijing, China
| | - Fei Ling
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Lewei Zhu
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Sifei Yu
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China.,Medical Engineering Technology Research and development center of Immune Repertoire in Foshan, The First People's Hospital of Foshan, Foshan, China
| | - Peixian Chen
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Chuling Zhang
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China.,Medical Engineering Technology Research and development center of Immune Repertoire in Foshan, The First People's Hospital of Foshan, Foshan, China
| | - Chunguo Zhang
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China.,Medical Engineering Technology Research and development center of Immune Repertoire in Foshan, The First People's Hospital of Foshan, Foshan, China
| | - Guolin Ye
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Simon Fong
- Department of Computer and Information Science, University of Macau, Macau SAR, China
| | - Guoqiang Chen
- Department of Rheumatology and Immunology, The First People's Hospital of Foshan, Foshan, China.
| | - Wei Luo
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China. .,Medical Engineering Technology Research and development center of Immune Repertoire in Foshan, The First People's Hospital of Foshan, Foshan, China.
| |
Collapse
|
50
|
Yao J, Chen Y, Lin Z. Exosomes: Mediators in microenvironment of colorectal cancer. Int J Cancer 2023. [PMID: 36760212 DOI: 10.1002/ijc.34471] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 01/08/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023]
Abstract
Tumor microenvironment, the soil where tumor thrives, plays a critical role in the development and progression of colorectal cancer (CRC). Various cell signaling molecules in the environment promote tumor angiogenesis, immune tolerance and facilitate immune escape. Exosomes, as messengers between tumor and host cells, are considered key mediators involved in the tumor-accelerating environment. However, the exosome-mediated communication networks in the CRC microenvironment are still largely unclear. In this review, we summarized the relationship between TME and CRC based on recent literature. Then, we revealed the unique impacts and signal molecules of exosomes on account of their regulatory role in the flora, hypoxia, inflammatory and immunological microenvironment of CRC. Finally, we summarized the therapeutically effective of exosomes in CRC microenvironment and discussed their current status and prospects, aiming to provide new molecular targets and a theoretical basis for the CRC treatment.
Collapse
Affiliation(s)
- Jiali Yao
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yingrui Chen
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Zhijie Lin
- Department of Immunology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, China
| |
Collapse
|