1
|
Liu X, Song X. Macrophage-Centric Immunometabolic Crosstalk in Alopecia Areata Pathogenesis: Mechanisms and Therapeutic Implications. Clin Rev Allergy Immunol 2025; 68:50. [PMID: 40405030 DOI: 10.1007/s12016-025-09060-3] [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] [Accepted: 05/07/2025] [Indexed: 05/24/2025]
Abstract
Although alopecia areata (AA) is recognized as a hair loss disorder stemming from the dysregulation of cutaneous immune homeostasis, its precise pathogenesis still remains elusive. The collapse of hair follicle (HF) immune privilege (IP), leading to immune cell-mediated attack on the hair follicle, is currently the widely accepted fundamental mechanism of AA. Among the immune cells studied in this context, CD8+ T cells and regulatory T (Treg) cells are relatively well-researched, but the direct involvement of macrophages in the disease process has been less frequently demonstrated. In this review, we summarize various previous studies on macrophages and hypothesize the immune mechanisms by which macrophages contribute to the pathogenesis of AA. This exploration provides new insights for future research and potential clinical treatments.
Collapse
Affiliation(s)
- Xu Liu
- Department of Dermatology, Hangzhou Third People's Hospital, West Lake Ave 38, Hangzhou, 310009, People's Republic of China
| | - Xiuzu Song
- Department of Dermatology, Hangzhou Third People's Hospital, West Lake Ave 38, Hangzhou, 310009, People's Republic of China.
| |
Collapse
|
2
|
Xu Y, Zhang H, Nie D. Histone modifications and metabolic reprogramming in tumor-associated macrophages: a potential target of tumor immunotherapy. Front Immunol 2025; 16:1521550. [PMID: 40375990 PMCID: PMC12078272 DOI: 10.3389/fimmu.2025.1521550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 04/09/2025] [Indexed: 05/18/2025] Open
Abstract
Histone modifications, including methylation, acetylation, lactylation, phosphorylation, ubiquitination, SUMOylation, ADP-ribosylation, and crotonylation, critically regulate tumor-associated macrophages (TAMs) polarization by modulating gene expression and functional states. Reprogramming TAMs from M2 to M1 phenotypes through epigenetic targeting has emerged as a promising strategy to enhance anti-tumor immunity and improve the efficacy of cancer immunotherapy. This review explores the role of histone modifications in TAM biology, their interplay with metabolic reprogramming, and the opportunities and challenges in developing epigenetic-based therapies to advance cancer immunotherapy.
Collapse
Affiliation(s)
- Yiting Xu
- The Second Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Han Zhang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Dengyun Nie
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Parajuli N, Subedi K, Solone XK, Jiang A, Zhou L, Mi QS. Epigenetic Control of Alveolar Macrophages: Impact on Lung Health and Disease. Cells 2025; 14:640. [PMID: 40358164 PMCID: PMC12071345 DOI: 10.3390/cells14090640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alveolar macrophages (AMs) are immune cells located in the alveoli-the tiny air sacs in the lungs where gas exchange occurs. Their functions are regulated by various epigenetic mechanisms, which are essential for both healthy lung function and disease development. In the lung's microenvironment, AMs play critical roles in immune surveillance, pathogen clearance, and tissue repair. This review examines how epigenetic regulation influences AM functions and their involvement in lung diseases. Key mechanisms, such as DNA methylation, histone modifications, and non-coding RNAs, regulate gene expression in response to environmental signals. In healthy lungs, these modifications enable AMs to quickly respond to inhaled threats. However, when these processes malfunction, they could contribute to diseases such as pulmonary fibrosis, COPD, and pulmonary hypertension. By exploring how epigenetic changes affect AM polarization, plasticity, and immune responses, we can gain deeper insights into their role in lung diseases and open new avenues for treating and preventing respiratory conditions. Ultimately, understanding the epigenetic mechanisms within AMs enhances our knowledge of lung immunology and offers potential for innovative interventions to restore lung health and prevent respiratory diseases.
Collapse
Affiliation(s)
- Nirmal Parajuli
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Kalpana Subedi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Xzaviar Kaymar Solone
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Medicine, Henry Ford Health, Detroit, MI 48202, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI 48202, USA; (N.P.); (K.S.); (X.K.S.); (A.J.)
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Medicine, Henry Ford Health, Detroit, MI 48202, USA
| |
Collapse
|
4
|
Ge Y, Jiang L, Dong Q, Xu Y, Yam JWP, Zhong X. Exosome-mediated Crosstalk in the Tumor Immune Microenvironment: Critical Drivers of Hepatocellular Carcinoma Progression. J Clin Transl Hepatol 2025; 13:143-161. [PMID: 39917466 PMCID: PMC11797817 DOI: 10.14218/jcth.2024.00302] [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: 08/23/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 02/09/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant global health issue, ranking as the sixth most prevalent malignancy and the fourth leading cause of cancer-related mortality worldwide. Despite advancements in therapeutic strategies, mortality rates for HCC remain high. The tumor immune microenvironment (TIME) plays a vital role in HCC progression by influencing tumor cell survival and growth. Recent studies highlight the essential role of exosomes in mediating intercellular communication within the TIME, particularly in interactions among tumor cells, immune cells, and fibroblasts. These interactions drive critical aspects of tumor development, including immune escape, angiogenesis, drug resistance, and metastasis. A detailed understanding of the molecular mechanisms by which exosomes modulate the TIME is essential for developing targeted therapies. This review systematically evaluated the roles and regulatory mechanisms of exosomes within the TIME of HCC, examining the impact of both HCC-derived and non-HCC-derived exosomes on various cellular components within the TIME. It emphasized their regulatory effects on cell phenotypes and functions, as well as their roles in HCC progression. The review also explored the potential applications of exosome-based immunotherapies, offering new insights into improving therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Yifei Ge
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixue Jiang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingfu Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
5
|
Tan H, Cai M, Wang J, Yu T, Xia H, Zhao H, Zhang X. Harnessing Macrophages in Cancer Therapy: from Immune Modulators to Therapeutic Targets. Int J Biol Sci 2025; 21:2235-2257. [PMID: 40083710 PMCID: PMC11900799 DOI: 10.7150/ijbs.106275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
Macrophages, as the predominant phagocytes, play an essential role in pathogens defense and tissue homeostasis maintenance. In the context of cancer, tumor-associated macrophages (TAMs) have evolved into cunning actors involved in angiogenesis, cancer cell proliferation and metastasis, as well as the construction of immunosuppressive microenvironment. Once properly activated, macrophages can kill tumor cells directly through phagocytosis or attack tumor cells indirectly by stimulating innate and adaptive immunity. Thus, the prospect of targeting TAMs has sparked significant interest and emerged as a promising strategy in immunotherapy. In this review, we summarize the diverse roles and underlying mechanisms of TAMs in cancer development and immunity and highlight the TAM-based therapeutic strategies such as inhibiting macrophage recruitment, inhibiting the differentiation reprogramming of TAMs, blocking phagocytotic checkpoints, inducing trained macrophages, as well as the potential of engineered CAR-armed macrophages in cancer therapy.
Collapse
Affiliation(s)
- Huabing Tan
- Department of Infectious Diseases, Hepatology Institute, Renmin Hospital, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, Hubei Province, China
- General internal medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Meihe Cai
- Department of Traditional Chinese Medicine, Zhushan Renmin Hospital, Zhushan, 442200, China
| | | | - Tao Yu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Houjun Xia
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huanbin Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Present: Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoyu Zhang
- Department of Gastrointestinal Surgery, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
6
|
Kuhl GC, Tangney M. Bacterial-Mediated In Situ Engineering of Tumour-Associated Macrophages for Cancer Immunotherapy. Cancers (Basel) 2025; 17:723. [PMID: 40075571 PMCID: PMC11899205 DOI: 10.3390/cancers17050723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/29/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Tumour-associated macrophages (TAMs) are critical components of the tumour microenvironment (TME), significantly influencing cancer progression and treatment resistance. This review aims to explore the innovative use of engineered bacteria to reprogram TAMs, enhancing their anti-tumour functions and improving therapeutic outcomes. METHODS We conducted a systematic review following a predefined protocol. Multiple databases were searched to identify relevant studies on TAMs, their phenotypic plasticity, and the use of engineered bacteria for reprogramming. Inclusion and exclusion criteria were applied to select studies, and data were extracted using standardised forms. Data synthesis was performed to summarise the findings, focusing on the mechanisms and therapeutic benefits of using non-pathogenic bacteria to modify TAMs. RESULTS The review summarises the findings that engineered bacteria can selectively target TAMs, promoting a shift from the tumour-promoting M2 phenotype to the tumour-fighting M1 phenotype. This reprogramming enhances pro-inflammatory responses and anti-tumour activity within the TME. Evidence from various studies indicates significant tumour regression and improved immune responses following bacterial therapy. CONCLUSIONS Reprogramming TAMs using engineered bacteria presents a promising strategy for cancer therapy. This approach leverages the natural targeting abilities of bacteria to modify TAMs directly within the tumour, potentially improving patient outcomes and offering new insights into immune-based cancer treatments. Further research is needed to optimise these methods and assess their clinical applicability.
Collapse
Affiliation(s)
- Gabriela Christina Kuhl
- Cancer Research @UCC, College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
| | - Mark Tangney
- Cancer Research @UCC, College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland
| |
Collapse
|
7
|
Topham B, Hock B, Phillips E, Wiggins G, Currie M. The Role of Innate Priming in Modifying Tumor-associated Macrophage Phenotype. FRONT BIOSCI-LANDMRK 2024; 29:418. [PMID: 39735978 DOI: 10.31083/j.fbl2912418] [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: 07/08/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 12/31/2024]
Abstract
Tumor-associated macrophages (TAMs) are innate immune cells that exert far reaching influence over the tumor microenvironment (TME). Depending on cues within the local environment, TAMs may promote tumor angiogenesis, cancer cell invasion and immunosuppression, or, alternatively, inhibit tumor progression via neoantigen presentation, tumoricidal reactive oxygen species generation and pro-inflammatory cytokine secretion. Therefore, TAMs have a pivotal role in determining tumor progression and response to therapy. TAM phenotypes are driven by cytokines and physical cues produced by tumor cells, adipocytes, fibroblasts, pericytes, immune cells, and other cells within the TME. Research has shown that TAMs can be primed by environmental stimuli, adding another layer of complexity to the environmental context that determines TAM phenotype. Innate priming is a functional consequence of metabolic and epigenetic reprogramming of innate cells by a primary stimulant, resulting in altered cellular response to future secondary stimulation. Innate priming offers a novel target for development of cancer immunotherapy and improved prognosis of disease, but also raises the risk of exacerbating existing inflammatory pathologies. This review will discuss the mechanisms underlying innate priming including metabolic and epigenetic modification, its relevance to TAMs and tumor progression, and possible clinical implications for cancer treatment.
Collapse
Affiliation(s)
- Ben Topham
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| | - Barry Hock
- Haematology Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| | - George Wiggins
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| | - Margaret Currie
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| |
Collapse
|
8
|
Satam S, Palekar N, Premkumar K, Shankar BS. Sirtinol, a SIRT1 inhibitor, inhibits the EMT and metastasis of 4T1 breast cancer cells and impacts the tumor microenvironment. Immunopharmacol Immunotoxicol 2024; 46:829-842. [PMID: 39373058 DOI: 10.1080/08923973.2024.2412110] [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: 08/19/2023] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
INTRODUCTION The impact of epigenetic drugs on metastasis and the immunological microenvironment is poorly understood. In this study, we looked at how sirtinol, a SIRT1 inhibitor, affected epithelial-mesenchymal transition (EMT), metastasis, and the immune cells. MATERIALS AND METHODS In vitro experiments were carried out using tumor conditioned medium (TCM). For in vivo experiments, sirtinol was administered i.p. in tumor bearing BALB/c mice at a dose of 2 mg/kg body weight either alone or in combination with cisplatin. Estimation of cytokines was carried out using ELISA or ELIspot. Estimation of different markers was done using flow cytometry or western blot. RESULTS Sirtinol, a SIRT1 inhibitor, was found to be cytotoxic to 4T1 breast cancer cells with no synergistic effects with cisplatin, both under in vitro and in vivo conditions (p < 0.05). Sirtinol significantly reduced cancer cell metastasis to the spleen which was supported by in vitro findings such as decreased vimentin expression and cell mobility in migration and wound healing assays (p < 0.01). Studies on the effects of 4T1 tumor-conditioned medium on spleen cells indicated changes in T cell proliferation as well as differentiation (p < 0.01). In tumor bearing mice, spleen cells showed elevated IFN-γ secretion, increased CD11b+ cells, and decreased T cells (p < 0.01). This was reversed by sirtinol as well as the combination treatment, which may also have contributed to metastasis inhibition (p < 0.01). CONCLUSION Sirtinol, a SIRT1 inhibitor inhibits EMT and metastasis of 4T1 breast cancer cells and also has an impact on the immune microenvironment.
Collapse
Affiliation(s)
- Sharvari Satam
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Nitya Palekar
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Kavitha Premkumar
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Bhavani S Shankar
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
9
|
Xu H, Russell SN, Steiner K, O'Neill E, Jones KI. Targeting PI3K-gamma in myeloid driven tumour immune suppression: a systematic review and meta-analysis of the preclinical literature. Cancer Immunol Immunother 2024; 73:204. [PMID: 39105848 PMCID: PMC11303654 DOI: 10.1007/s00262-024-03779-2] [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: 12/07/2023] [Accepted: 07/11/2024] [Indexed: 08/07/2024]
Abstract
The intricate interplay between immune and stromal cells within the tumour microenvironment (TME) significantly influences tumour progression. Myeloid cells, including tumour-associated macrophages (TAMs), neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs), contribute to immune suppression in the TME (Nakamura and Smyth in Cell Mol Immunol 17(1):1-12 (2020). https://doi.org/10.1038/s41423-019-0306-1 ; DeNardo and Ruffell in Nat Rev Immunol 19(6):369-382 (2019). https://doi.org/10.1038/s41577-019-0127-6 ). This poses a significant challenge for novel immunotherapeutics that rely on host immunity to exert their effect. This systematic review explores the preclinical evidence surrounding the inhibition of phosphoinositide 3-kinase gamma (PI3Kγ) as a strategy to reverse myeloid-driven immune suppression in solid tumours. EMBASE, MEDLINE, and PubMed databases were searched on 6 October 2022 using keyword and subject heading terms to capture relevant studies. The studies, focusing on PI3Kγ inhibition in animal models, were subjected to predefined inclusion and exclusion criteria. Extracted data included tumour growth kinetics, survival endpoints, and immunological responses which were meta-analysed. PRISMA and MOOSE guidelines were followed. A total of 36 studies covering 73 animal models were included in the review and meta-analysis. Tumour models covered breast, colorectal, lung, skin, pancreas, brain, liver, prostate, head and neck, soft tissue, gastric, and oral cancer. The predominant PI3Kγ inhibitors were IPI-549 and TG100-115, demonstrating favourable specificity for the gamma isoform. Combination therapies, often involving chemotherapy, radiotherapy, immune checkpoint inhibitors, biological agents, or vaccines, were explored in 81% of studies. Analysis of tumour growth kinetics revealed a statistically significant though heterogeneous response to PI3Kγ monotherapy, whereas the tumour growth in combination treated groups were more consistently reduced. Survival analysis showed a pronounced increase in median overall survival with combination therapy. This systematic review provides a comprehensive analysis of preclinical studies investigating PI3Kγ inhibition in myeloid-driven tumour immune suppression. The identified studies underscore the potential of PI3Kγ inhibition in reshaping the TME by modulating myeloid cell functions. The combination of PI3Kγ inhibition with other therapeutic modalities demonstrated enhanced antitumour effects, suggesting a synergistic approach to overcome immune suppression. These findings support the potential of PI3Kγ-targeted therapies, particularly in combination regimens, as a promising avenue for future clinical exploration in diverse solid tumour types.
Collapse
Affiliation(s)
- Haonan Xu
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Keaton Ian Jones
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Wang K, Chen X, Lin P, Wu J, Huang Q, Chen Z, Tian J, Wang H, Tian Y, Shi M, Qian M, Hui B, Zhu Y, Li L, Yao R, Bian H, Zhu P, Chen R, Chen L. CD147-K148me2-Driven Tumor Cell-Macrophage Crosstalk Provokes NSCLC Immunosuppression via the CCL5/CCR5 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400611. [PMID: 38873823 PMCID: PMC11304266 DOI: 10.1002/advs.202400611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/15/2024] [Indexed: 06/15/2024]
Abstract
Immunosuppression is a major hallmark of tumor progression in non-small cell lung cancer (NSCLC). Cluster of differentiation 147 (CD147), an important pro-tumorigenic factor, is closely linked to NSCLC immunosuppression. However, the role of CD147 di-methylation in the immunosuppressive tumor microenvironment (TME) remains unclear. Here, di-methylation of CD147 at Lys148 (CD147-K148me2) is identified as a common post-translational modification (PTM) in NSCLC that is significantly associated with unsatisfying survival outcomes among NSCLC sufferers, especially those in the advanced stages of the disease. The methyltransferase NSD2 catalyzes CD147 to generate CD147-K148me2. Further analysis demonstrates that CD147-K148me2 reestablishes the immunosuppressive TME and promotes NSCLC progression. Mechanistically, this modification promotes the interaction between cyclophilin A (CyPA) and CD147, and in turn, increases CCL5 gene transcription by activating p38-ZBTB32 signaling, leading to increased NSCLC cell-derived CCL5 secretion. Subsequently, CD147-K148me2-mediated CCL5 upregulation facilitates M2-like tumor-associated macrophage (TAM) infiltration in NSCLC tissues via CCL5/CCR5 axis-dependent intercellular crosstalk between tumor cells and macrophages, which is inhibited by blocking CD147-K148me2 with the targeted antibody 12C8. Overall, this study reveals the role of CD147-K148me2-driven intercellular crosstalk in the development of NSCLC immunosuppression, and provides a potential interventional strategy for PTM-targeted NSCLC therapy.
Collapse
Affiliation(s)
- Ke Wang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Xiaohong Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Peng Lin
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Jiao Wu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Qiang Huang
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- School of MedicineShanghai UniversityShanghai200444China
| | - Zhi‐Nan Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Jiale Tian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Hao Wang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ye Tian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Mingyan Shi
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Meirui Qian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Bengang Hui
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- Department of Thoracic Surgery of Tangdu HospitalFourth Military Medical UniversityXi'an710038China
| | - Yumeng Zhu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ling Li
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Rui Yao
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Huijie Bian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ping Zhu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ruo Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Liang Chen
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- School of MedicineShanghai UniversityShanghai200444China
| |
Collapse
|
11
|
Li Y, Jin H, Li Q, Shi L, Mao Y, Zhao L. The role of RNA methylation in tumor immunity and its potential in immunotherapy. Mol Cancer 2024; 23:130. [PMID: 38902779 PMCID: PMC11188252 DOI: 10.1186/s12943-024-02041-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
RNA methylation, a prevalent post-transcriptional modification, has garnered considerable attention in research circles. It exerts regulatory control over diverse biological functions by modulating RNA splicing, translation, transport, and stability. Notably, studies have illuminated the substantial impact of RNA methylation on tumor immunity. The primary types of RNA methylation encompass N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A), and N7-methylguanosine (m7G), and 3-methylcytidine (m3C). Compelling evidence underscores the involvement of RNA methylation in regulating the tumor microenvironment (TME). By affecting RNA translation and stability through the "writers", "erasers" and "readers", RNA methylation exerts influence over the dysregulation of immune cells and immune factors. Consequently, RNA methylation plays a pivotal role in modulating tumor immunity and mediating various biological behaviors, encompassing proliferation, invasion, metastasis, etc. In this review, we discussed the mechanisms and functions of several RNA methylations, providing a comprehensive overview of their biological roles and underlying mechanisms within the tumor microenvironment and among immunocytes. By exploring how these RNA modifications mediate tumor immune evasion, we also examine their potential applications in immunotherapy. This review aims to provide novel insights and strategies for identifying novel targets in RNA methylation and advancing cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Yan Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Haoer Jin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qingling Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Liangrong Shi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yitao Mao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Luqing Zhao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
12
|
Quaranta V, Ballarò C, Giannelli G. Macrophages Orchestrate the Liver Tumor Microenvironment. Cancers (Basel) 2024; 16:1772. [PMID: 38730724 PMCID: PMC11083142 DOI: 10.3390/cancers16091772] [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: 04/11/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
Liver cancer is one of the leading causes of cancer-related mortality. Hepatocellular carcinoma and cholangiocarcinoma are the most common types, and despite numerous advances, therapeutic options still remain poor for these cancer patients. Tumor development and progression strictly depend on a supportive tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are the most abundant immune cells population within a tumorigenic liver; they sustain cancer cells' growth and invasiveness, and their presence is correlated with a poor prognosis. Furthermore, TAM cross-talk with cells and components of the TME promotes immunosuppression, a desmoplastic response, and angiogenesis. In this review, we summarize the latest advances in understanding TAM heterogeneity and function, with a particular focus on TAM modulation of the TME. We also discuss the potential of targeting macrophage subpopulations and how this is now being exploited in current clinical trials for the treatment of liver cancer.
Collapse
Affiliation(s)
- Valeria Quaranta
- National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy (G.G.)
| | | | | |
Collapse
|
13
|
Kim SW, Kim CW, Moon YA, Kim HS. Reprogramming of tumor-associated macrophages by metabolites generated from tumor microenvironment. Anim Cells Syst (Seoul) 2024; 28:123-136. [PMID: 38577621 PMCID: PMC10993762 DOI: 10.1080/19768354.2024.2336249] [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: 01/25/2024] [Accepted: 03/17/2024] [Indexed: 04/06/2024] Open
Abstract
The tumor microenvironment comprises both tumor and non-tumor stromal cells, including tumor-associated macrophages (TAMs), endothelial cells, and carcinoma-associated fibroblasts. TAMs, major components of non-tumor stromal cells, play a crucial role in creating an immunosuppressive environment by releasing cytokines, chemokines, growth factors, and immune checkpoint proteins that inhibit T cell activity. During tumors develop, cancer cells release various mediators, including chemokines and metabolites, that recruit monocytes to infiltrate tumor tissues and subsequently induce an M2-like phenotype and tumor-promoting properties. Metabolites are often overlooked as metabolic waste or detoxification products but may contribute to TAM polarization. Furthermore, macrophages display a high degree of plasticity among immune cells in the tumor microenvironment, enabling them to either inhibit or facilitate cancer progression. Therefore, TAM-targeting has emerged as a promising strategy in tumor immunotherapy. This review provides an overview of multiple representative metabolites involved in TAM phenotypes, focusing on their role in pro-tumoral polarization of M2.
Collapse
Affiliation(s)
- Seung Woo Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Chan Woo Kim
- Cancer Immunotherapy Evaluation Team, Non-Clinical Evaluation Center, Osong Medical Innovation Foundation (KBIO Health), Cheongju, Republic of Korea
| | - Young-Ah Moon
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
14
|
Lin TT, Xiong W, Chen GH, He Y, Long L, Gao XF, Zhou JL, Lv WW, Huang YZ. Epigenetic-based combination therapy and liposomal codelivery overcomes osimertinib-resistant NSCLC via repolarizing tumor-associated macrophages. Acta Pharmacol Sin 2024; 45:867-878. [PMID: 38114644 PMCID: PMC10943229 DOI: 10.1038/s41401-023-01205-4] [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: 07/22/2023] [Accepted: 11/19/2023] [Indexed: 12/21/2023]
Abstract
Osimertinib (Osi) is widely used as a first-line treatment for non-small cell lung cancer (NSCLC) with EGFR mutations. However, the majority of patients treated with Osi eventually relapse within a year. The mechanisms of Osi resistance remain largely unexplored, and efficient strategies to reverse the resistance are urgently needed. Here, we developed a lactoferrin-modified liposomal codelivery system for the combination therapy of Osi and panobinostat (Pan), an epigenetic regulator of histone acetylation. We demonstrated that the codelivery liposomes could efficiently repolarize tumor-associated macrophages (TAM) from the M2 to M1 phenotype and reverse the epithelial-mesenchymal transition (EMT)-associated drug resistance in the tumor cells, as well as suppress glycolysis, lactic acid production, and angiogenesis. Our results suggested that the combination therapy of Osi and Pan mediated by liposomal codelivery is a promising strategy for overcoming Osi resistance in NSCLC.
Collapse
Affiliation(s)
- Ting-Ting Lin
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, 256603, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wei Xiong
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528437, China
| | - Gui-Hua Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China
| | - Yang He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li Long
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xin-Fu Gao
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Jia-Lin Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528437, China
| | - Wen-Wen Lv
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, 256603, China.
| | - Yong-Zhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528437, China.
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai, 201203, China.
| |
Collapse
|
15
|
Almeida L, Dhillon-LaBrooy A, Sparwasser T. The evolutionary tug-of-war of macrophage metabolism during bacterial infection. Trends Endocrinol Metab 2024; 35:235-248. [PMID: 38040578 DOI: 10.1016/j.tem.2023.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 12/03/2023]
Abstract
The function and phenotype of macrophages are intimately linked with pathogen detection. On sensing pathogen-derived signals and molecules, macrophages undergo a carefully orchestrated process of polarization to acquire pathogen-clearing properties. This phenotypic change must be adequately supported by metabolic reprogramming that is now known to support the acquisition of effector function, but also generates secondary metabolites with direct microbicidal activity. At the same time, bacteria themselves have adapted to both manipulate and take advantage of macrophage-specific metabolic adaptations. Here, we summarize the current knowledge on macrophage metabolism during infection, with a particular focus on understanding the 'arms race' between host immune cells and bacteria during immune responses.
Collapse
Affiliation(s)
- Luís Almeida
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany.
| | - Ayesha Dhillon-LaBrooy
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany.
| |
Collapse
|
16
|
Malavasi E, Adamo M, Zamprogno E, Vella V, Giamas G, Gagliano T. Decoding the Tumour Microenvironment: Molecular Players, Pathways, and Therapeutic Targets in Cancer Treatment. Cancers (Basel) 2024; 16:626. [PMID: 38339377 PMCID: PMC10854614 DOI: 10.3390/cancers16030626] [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: 11/23/2023] [Revised: 12/16/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
The tumour microenvironment (TME) is a complex and constantly evolving collection of cells and extracellular components. Cancer cells and the surrounding environment influence each other through different types of processes. Characteristics of the TME include abnormal vasculature, altered extracellular matrix, cancer-associated fibroblast and macrophages, immune cells, and secreted factors. Within these components, several molecules and pathways are altered and take part in the support of the tumour. Epigenetic regulation, kinases, phosphatases, metabolic regulators, and hormones are some of the players that influence and contribute to shaping the tumour and the TME. All these characteristics contribute significantly to cancer progression, metastasis, and immune escape, and may be the target for new approaches for cancer treatment.
Collapse
Affiliation(s)
- Eleonora Malavasi
- Cancer Cell Signalling Laboratory, Department of Medicine, University of Udine, 33100 Udine, Italy; (E.M.); (M.A.); (E.Z.)
| | - Manuel Adamo
- Cancer Cell Signalling Laboratory, Department of Medicine, University of Udine, 33100 Udine, Italy; (E.M.); (M.A.); (E.Z.)
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK;
| | - Elisa Zamprogno
- Cancer Cell Signalling Laboratory, Department of Medicine, University of Udine, 33100 Udine, Italy; (E.M.); (M.A.); (E.Z.)
| | - Viviana Vella
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK;
| | - Georgios Giamas
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK;
| | - Teresa Gagliano
- Cancer Cell Signalling Laboratory, Department of Medicine, University of Udine, 33100 Udine, Italy; (E.M.); (M.A.); (E.Z.)
| |
Collapse
|
17
|
Nunes SP, Morales L, Rubio C, Munera-Maravilla E, Lodewijk I, Suárez-Cabrera C, Martínez VG, Pérez-Escavy M, Pérez-Crespo M, Alonso Sánchez M, Montesinos E, San José-Enériz E, Agirre X, Prósper F, Pineda-Lucena A, Henrique R, Dueñas M, Correia MP, Jerónimo C, Paramio JM. Modulation of tumor microenvironment by targeting histone acetylation in bladder cancer. Cell Death Discov 2024; 10:1. [PMID: 38172127 PMCID: PMC10764810 DOI: 10.1038/s41420-023-01786-3] [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: 06/28/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Alterations in the epigenetic machinery in both tumor and immune cells contribute to bladder cancer (BC) development, constituting a promising target as an alternative therapeutic option. Here, we have explored the effects of a novel histone deacetylase (HDAC) inhibitor CM-1758, alone or in combination with immune checkpoint inhibitors (ICI) in BC. We determined the antitumor effects of CM-1758 in various BC cell lines together with the induction of broad transcriptional changes, with focus on the epigenetic regulation of PD-L1. Using an immunocompetent syngeneic mouse model of metastatic BC, we studied the effects of CM-1758 alone or in combination with anti-PD-L1 not only on tumor cells, but also in the tumor microenvironment. In vitro, we found that CM-1758 has cytotoxic and cytostatic effects either by inducing apoptosis or cell cycle arrest in BC cells at low micromolar levels. PD-L1 is epigenetically regulated by histone acetylation marks and is induced after treatment with CM-1758. We also observed that treatment with CM-1758 led to an important delay in tumor growth and a higher CD8 + T cell tumor infiltration. Moreover, anti-PD-L1 alone or in combination with CM-1758 reprogramed macrophage differentiation towards a M1-like polarization state and increased of pro-inflammatory cytokines systemically, yielding potential further antitumor effects. Our results suggest the possibility of combining HDAC inhibitors with immunotherapies for the management of advanced metastatic BC.
Collapse
Affiliation(s)
- Sandra P Nunes
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Doctoral Program in Biomedical Sciences, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
| | - Lucia Morales
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Carolina Rubio
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ester Munera-Maravilla
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Iris Lodewijk
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Cristian Suárez-Cabrera
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Victor G Martínez
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Mercedes Pérez-Escavy
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
| | - Miriam Pérez-Crespo
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Miguel Alonso Sánchez
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
| | - Esther Montesinos
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
| | - Edurne San José-Enériz
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, Pamplona, Spain
| | - Xabier Agirre
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, Pamplona, Spain
| | - Felipe Prósper
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, Pamplona, Spain
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Pamplona, Spain
| | - Antonio Pineda-Lucena
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal
| | - Marta Dueñas
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Margareta P Correia
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal
| | - Jesús M Paramio
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.
- Biomedical Research Institute I+12, University Hospital "12 de Octubre", Madrid, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
18
|
Liu R, Zhao E, Yu H, Yuan C, Abbas MN, Cui H. Methylation across the central dogma in health and diseases: new therapeutic strategies. Signal Transduct Target Ther 2023; 8:310. [PMID: 37620312 PMCID: PMC10449936 DOI: 10.1038/s41392-023-01528-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 08/26/2023] Open
Abstract
The proper transfer of genetic information from DNA to RNA to protein is essential for cell-fate control, development, and health. Methylation of DNA, RNAs, histones, and non-histone proteins is a reversible post-synthesis modification that finetunes gene expression and function in diverse physiological processes. Aberrant methylation caused by genetic mutations or environmental stimuli promotes various diseases and accelerates aging, necessitating the development of therapies to correct the disease-driver methylation imbalance. In this Review, we summarize the operating system of methylation across the central dogma, which includes writers, erasers, readers, and reader-independent outputs. We then discuss how dysregulation of the system contributes to neurological disorders, cancer, and aging. Current small-molecule compounds that target the modifiers show modest success in certain cancers. The methylome-wide action and lack of specificity lead to undesirable biological effects and cytotoxicity, limiting their therapeutic application, especially for diseases with a monogenic cause or different directions of methylation changes. Emerging tools capable of site-specific methylation manipulation hold great promise to solve this dilemma. With the refinement of delivery vehicles, these new tools are well positioned to advance the basic research and clinical translation of the methylation field.
Collapse
Affiliation(s)
- Ruochen Liu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Erhu Zhao
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Huijuan Yu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Chaoyu Yuan
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
19
|
Farhana A, Alsrhani A, Khan YS, Rasheed Z. Cancer Bioenergetics and Tumor Microenvironments-Enhancing Chemotherapeutics and Targeting Resistant Niches through Nanosystems. Cancers (Basel) 2023; 15:3836. [PMID: 37568652 PMCID: PMC10416858 DOI: 10.3390/cancers15153836] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/16/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer is an impending bottleneck in the advanced scientific workflow to achieve diagnostic, prognostic, and therapeutic success. Most cancers are refractory to conventional diagnostic and chemotherapeutics due to their limited targetability, specificity, solubility, and side effects. The inherent ability of each cancer to evolve through various genetic and epigenetic transformations and metabolic reprogramming underlies therapeutic limitations. Though tumor microenvironments (TMEs) are quite well understood in some cancers, each microenvironment differs from the other in internal perturbations and metabolic skew thereby impeding the development of appropriate diagnostics, drugs, vaccines, and therapies. Cancer associated bioenergetics modulations regulate TME, angiogenesis, immune evasion, generation of resistant niches and tumor progression, and a thorough understanding is crucial to the development of metabolic therapies. However, this remains a missing element in cancer theranostics, necessitating the development of modalities that can be adapted for targetability, diagnostics and therapeutics. In this challenging scenario, nanomaterials are modular platforms for understanding TME and achieving successful theranostics. Several nanoscale particles have been successfully researched in animal models, quite a few have reached clinical trials, and some have achieved clinical success. Nanoparticles exhibit an intrinsic capability to interact with diverse biomolecules and modulate their functions. Furthermore, nanoparticles can be functionalized with receptors, modulators, and drugs to facilitate specific targeting with reduced toxicity. This review discusses the current understanding of different theranostic nanosystems, their synthesis, functionalization, and targetability for therapeutic modulation of bioenergetics, and metabolic reprogramming of the cancer microenvironment. We highlight the potential of nanosystems for enhanced chemotherapeutic success emphasizing the questions that remain unanswered.
Collapse
Affiliation(s)
- Aisha Farhana
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| | - Yusuf Saleem Khan
- Department of Anatomy, College of Medicine, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| | - Zafar Rasheed
- Department of Pathology, College of Medicine, Qassim University, P.O. Box 6655, Buraidah 51452, Qassim, Saudi Arabia
| |
Collapse
|
20
|
Yao H, Cheng L, Chen D, Zhang Q, Qiu L, Ren SH, Dou BT, Wang H, Huang J, Fan FY. Role of the bone marrow microenvironment in multiple myeloma treatment using CAR-T therapy. Expert Rev Anticancer Ther 2023; 23:807-815. [PMID: 37343305 DOI: 10.1080/14737140.2023.2229029] [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: 04/19/2023] [Accepted: 06/20/2023] [Indexed: 06/23/2023]
Abstract
INTRODUCTION Multiple myeloma (MM) is a malignant tumor caused by abnormal proliferation of bone marrow (BM) plasma cells and is the second most common hematologic malignancy. A variety of CAR-T cells targeting multiple myeloma-specific markers have shown good efficacy in clinical trials. However, CAR-T therapy still limits the insufficient duration of efficacy and recurrence of the disease. AREAS COVERED This article reviews the cell populations in the bone marrow of MM, and discusses the potential way to improve the efficiency of CAR-T cells in the treatment of MM by targeting the bone marrow microenvironment. EXPERT OPINION The limits of CAR-T therapy in MM may related to the impairment of T cell activity in the bone marrow microenvironment. This article reviews the cell populations of the immune microenvironment and nonimmune microenvironment in the bone marrow of multiple myeloma, and discusses the potential way to improve the efficiency of CAR-T cells in the treatment of MM by targeting the bone marrow. This may provides a new idea for the CAR-T therapy of multiple myeloma.
Collapse
Affiliation(s)
- Hao Yao
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Lei Cheng
- Department of Pharmacy, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Dan Chen
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Qian Zhang
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Ling Qiu
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Shi-Hui Ren
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Bai-Tao Dou
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| | - Huan Wang
- Department of Hematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, SiChuan, China
- University of Electronic Science and Technology of China, Chengdu, SiChuan, China
| | - Juan Huang
- Department of Hematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, SiChuan, China
| | - Fang-Yi Fan
- Department of Hematology and Hematopoietic Stem Cell Transplantation Center, General Hospital of the Chinese People's Liberation Army Western Theatre, Chengdu, SiChuan, China
| |
Collapse
|
21
|
Gan L, Zhao Y, Fu Y, Chen Q. The potential role of m6A modifications on immune cells and immunotherapy. Biomed Pharmacother 2023; 160:114343. [PMID: 36758318 DOI: 10.1016/j.biopha.2023.114343] [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: 12/07/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/10/2023] Open
Abstract
N6-methyladenosine (m6A), is the most prevalent and reversible post-transcriptional epigenetic modification of RNA in mammals. Dysregulation of m6A modifications impacts RNA procession, degradation, translocation, and translation, disrupting immune cell homeostasis and promoting tumor initiation and development. Here, we discuss an -up-to-date summary of the mechanisms by which m6A modifications regulate immune cell anti-tumor as well as self-homeostasis. We also present how the dysregulation of m6A modifications intrinsic to tumor cells regulates the function of immune cells in the tumor microenvironment. Meanwhile, we described some specific inhibitors targeting m6A modulators and discussed their potential use in cancer treatments.
Collapse
Affiliation(s)
- Linchuan Gan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China
| | - Yuxiang Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China
| | - Yajuan Fu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China.
| |
Collapse
|
22
|
Gorini F, Miceli M, de Antonellis P, Amente S, Zollo M, Ferrucci V. Epigenetics and immune cells in medulloblastoma. Front Genet 2023; 14:1135404. [PMID: 36968588 PMCID: PMC10036437 DOI: 10.3389/fgene.2023.1135404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Medulloblastoma (MB) is a highly malignant childhood tumor of the cerebellum. Transcriptional and epigenetic signatures have classified MB into four molecular subgroups, further stratified into biologically different subtypes with distinct somatic copy-number aberrations, driver genes, epigenetic alterations, activated pathways, and clinical outcomes. The brain tumor microenvironment (BTME) is of importance to regulate a complex network of cells, including immune cells, involved in cancer progression in brain malignancies. MB was considered with a “cold” immunophenotype due to the low influx of immune cells across the blood brain barrier (BBB). Recently, this assumption has been reconsidered because of the identification of infiltrating immune cells showing immunosuppressive phenotypes in the BTME of MB tumors. Here, we are providing a comprehensive overview of the current status of epigenetics alterations occurring during cancer progression with a description of the genomic landscape of MB by focusing on immune cells within the BTME. We further describe how new immunotherapeutic approaches could influence concurring epigenetic mechanisms of the immunosuppressive cells in BTME. In conclusion, the modulation of these molecular genetic complexes in BTME during cancer progression might enhance the therapeutic benefit, thus firing new weapons to fight MB.
Collapse
Affiliation(s)
- Francesca Gorini
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Marco Miceli
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Pasqualino de Antonellis
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Stefano Amente
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- DAI Medicina di Laboratorio e Trasfusionale, ‘AOU Federico II Policlinico, Naples, Italy
| | - Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- *Correspondence: Veronica Ferrucci,
| |
Collapse
|
23
|
Yu MY, Jia HJ, Zhang J, Ran GH, Liu Y, Yang XH. Exosomal miRNAs-mediated macrophage polarization and its potential clinical application. Int Immunopharmacol 2023; 117:109905. [PMID: 36848789 DOI: 10.1016/j.intimp.2023.109905] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 03/01/2023]
Abstract
Macrophages are highly heterogeneous and plastic immune cells that play an important role in the fight against pathogenic microorganisms and tumor cells. After different stimuli, macrophages can polarize to the M1 phenotype to show a pro-inflammatory effect and the M2 phenotype to show an anti-inflammatory effect. The balance of macrophage polarization is highly correlated with disease progression, and therapeutic approaches to reprogram macrophages by targeting macrophage polarization are feasible. There are a large number of exosomes in tissue cells, which can transmit information between cells. In particular, microRNAs (miRNAs) in the exosomes can regulate the polarization of macrophages and further affect the progression of various diseases. At the same time, exosomes are also effective "drug" carriers, laying the foundation for the clinical application of exosomes. This review describes some pathways involved in M1/M2 macrophage polarization and the effects of miRNA carried by exosomes from different sources on the polarization of macrophages. Finally, the application prospects and challenges of exosomes/exosomal miRNAs in clinical treatment are also discussed.
Collapse
Affiliation(s)
- Ming Yun Yu
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, No. 21 Bohai Road, Caofeidian Eco-city, Tangshan, 063210 Hebei, China
| | - Hui Jie Jia
- School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Jing Zhang
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, No. 21 Bohai Road, Caofeidian Eco-city, Tangshan, 063210 Hebei, China
| | - Guang He Ran
- Department of Medical Laboratory, Chang shou District Hospital of Traditional Chinese Medicine, No. 1 Xinglin Road, Peach Blossom New Town, Changshou District, 401200 Chongqing, China
| | - Yan Liu
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, No. 21 Bohai Road, Caofeidian Eco-city, Tangshan, 063210 Hebei, China.
| | - Xiu Hong Yang
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, No. 21 Bohai Road, Caofeidian Eco-city, Tangshan, 063210 Hebei, China.
| |
Collapse
|
24
|
Luo JF, Zhou H, Lio CK. Akebia Saponin D Inhibits the Inflammatory Reaction by Inhibiting the IL-6-STAT3-DNMT3b Axis and Activating the Nrf2 Pathway. Molecules 2022; 27:molecules27196236. [PMID: 36234773 PMCID: PMC9614599 DOI: 10.3390/molecules27196236] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022] Open
Abstract
Akebia saponin D (ASD) is derived from the Dipsacus asper Wall. ex Henry, which is a traditional Chinese medicine commonly used to treat rheumatic arthritis (RA). However, the in-depth mechanism of the anti-inflammatory effect of ASD is still unclear. This study aimed to preliminarily explore the anti-inflammatory effect of ASD and the underlying mechanisms from the perspective of DNA methylation and inflammation-related pathways. We found that ASD significantly reduced the production of multiple inflammatory mediators, including nitric oxide (NO) and prostaglandin E2 (PGE2), in LPS-induced RAW264.7 cells. The expression of DNA methyltransferase (DNMT) 3b and inducible nitric oxide synthase (iNOS) was also obviously inhibited by the ASD treatment. The protein and mRNA levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were also significantly inhibited by ASD. ASD inhibited the macrophage M1 phenotype, inhibited the high level of DNMT3b, and downregulated the signal transducer and activator of the transcription 3 (STAT3) pathway to exert its anti-inflammatory activity. Furthermore, DNMT3b siRNA and Nrf2 siRNA significantly promoted the anti-inflammatory effect of ASD. Our study demonstrates for the first time that ASD inhibits the IL-6-STAT3-DNMT3b axis and activates the nuclear factor-E2-related factor 2 (Nrf2) signaling pathway to achieve its inhibitory effect on inflammatory reactions.
Collapse
Affiliation(s)
- Jin-Fang Luo
- Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guian District, Guiyang 550025, China
| | - Hua Zhou
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
- Correspondence:
| | - Chon-Kit Lio
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medcine and Immune Disease Research, Guangzhou 510006, China
| |
Collapse
|
25
|
Lou X, Zhao K, Xu J, Shuai L, Niu H, Cao Z, Wang J, Zhang Y. CCL8 as a promising prognostic factor in diffuse large B-cell lymphoma via M2 macrophage interactions: A bioinformatic analysis of the tumor microenvironment. Front Immunol 2022; 13:950213. [PMID: 36072582 PMCID: PMC9441746 DOI: 10.3389/fimmu.2022.950213] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUNDS Prior investigations of the tumor microenvironment (TME) of diffuse large B-cell lymphoma (DLBCL) have shown that immune and stromal cells are key contributing factors to patients' outcome. However, challenges remain in finding reliable prognostic biomarkers based on cell infiltration. In this study, we attempted to shed some light on chemokine C-C motif chemokine ligand 8 (CCL8) in DLBCL via interaction with M2 macrophages. METHODS The Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithm was applied to evaluate immune and stromal scores from transcriptomic profiles of 443 DLBCL samples from The Cancer Genome Atlas (TCGA) and GSE10846 datasets. Immune cell infiltration (ICI) clusters were obtained based on different immune cell infiltrations of each sample, and gene clusters were derived through differentially expressed genes (DEGs) between the distinct ICI clusters. Five immune-related hub genes related to overall survival (OS) and clinical stages were obtained by COX regression analysis and protein-protein interaction (PPI) network construction then verified by quantitative real-time PCR (qPCR) and immunofluorescence staining in the FFPE tissues. The Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and TIMER websites were employed to explore the biological functions of CCL8-related DEGs. Uni- and multivariable Cox regression analyses were performed to analyze CCL8 as an independent prognostic risk factor in GSE10846 and were verified in other independent GEO cohorts. RESULTS A higher stromal score was associated with favorable prognosis in DLBCL. Patients in the ICI B cluster and gene B clusters had a better follow-up status with a higher programmed death ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen 4 (CTLA4) expression. Most of ICI-related DEGs were enriched for immune-related signaling pathways. Five hub genes with a distinct prognosis association were identified, including CD163, which is a biomarker of M2 macrophages, and CCL8. Abundant M2 macrophages were discovered in the high-CCL8 expression group. The functional analysis indicated that CCL8 is a key component of immune-related processes and secretory granule groups. Cox regression analysis and data from other GSE datasets yielded additional evidence of the prognostic value of CCL8 in DLBCL. CONCLUSIONS CCL8 has been implicated in macrophage recruitment in several solid tumors, and only a few reports have been published on the role of CCL8 in the pathogenesis of hematological malignancies. This article attempted to find out TME-related genes that associated with the survival in DLBCL patients. CCL8 was identified to be involved in immune activities. Importantly, a series of bioinformatics analysis indicated that CCL8 might become an effective target for DLBCL, which interacts with M2 macrophage and immune checkpoint. The potential related mechanisms need to be further elucidated.
Collapse
Affiliation(s)
- Xiaoli Lou
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ke Zhao
- Department of Pathology, The Affiliated Jiangyin Hospital of Nantong Universtiy, Jiangyin, China
| | - Jingze Xu
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lixiong Shuai
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hui Niu
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhifei Cao
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Juan Wang
- Department of Pathology, Suzhou Wuzhong People’s Hospital, Suzhou, China
| | - Yongsheng Zhang
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|