1
|
Zhou X, Wang P, Xie L, Chan YK, Jiao Z, Shu R, Bai D, Lai S, Deng Y. Molybdoenzymes-emulating bio-heterojunction hydrogel with rapid disinfection and macrophage reprogramming for wound regeneration. Biomaterials 2025; 320:123284. [PMID: 40121831 DOI: 10.1016/j.biomaterials.2025.123284] [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: 01/27/2025] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Developing hydrogel dressings with the capabilities to accommodate irregular wounds and provide a cascade disinfective-regenerative microenvironment for wound repair is of great importance to combating pathogenic bacteria-infected wounds but remains an ongoing challenge. To address the conundrum, we devise a molybdoenzymes-emulating bio-heterojunction (M-bioHJ) doped double network (DN) hydrogel dressing for bacterial-infected wound healing. The near-infrared (NIR) photothermal effect of the M-bioHJ facilitates the exchange of multiple dynamic crosslinking sites in the hydrogel, endowing the hydrogel with photo-remote reprocessing capabilities to completely accommodate the encountered irregular wounds and ultimately accomplish the admirable therapeutic effect. Meanwhile, the introduced M-bioHJ shows NIR light-enhanced photodynamic activity to induce a massive engendering of reactive oxygen species (ROS), allowing rapid sterilization without reliance on exogenous hydrogen peroxide. Furthermore, the Mo ions released from the M-bioHJ-encapsulated hydrogel can play a crucial role in reprogramming the macrophage phenotype and determining tissue regeneration. Both in vitro and in vivo evidences authenticate the accelerated healing potential of infected wounds through the synergistic effects of photo-reprocessing, disinfection, and macrophage-reprogramming facilitated by the hydrogel. These findings highlight the promising application prospects of such neoteric M-bioHJ-encapsulated hydrogel dressings for wound disinfection and tissue regeneration.
Collapse
Affiliation(s)
- Xiong Zhou
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Peiqi Wang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lu Xie
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, 999077, Hong Kong, China
| | - Zheng Jiao
- Swanson School of Engineering, University of Pittsburgh, Pittsburgh, 15261, USA
| | - Rui Shu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuangquan Lai
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, Guangdong 519000, China
| | - Yi Deng
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
2
|
Shen T, Wang Y, Cheng L, Bode AM, Gao Y, Zhang S, Chen X, Luo X. Oxidative complexity: The role of ROS in the tumor environment and therapeutic implications. Bioorg Med Chem 2025; 127:118241. [PMID: 40383035 DOI: 10.1016/j.bmc.2025.118241] [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: 01/27/2025] [Revised: 05/01/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Reactive oxygen species (ROS) constitutes a group of reactive molecules that play a critical role in biological processes. Varying ROS levels have been frequently observed in cancer cells and the tumor microenvironment (TME). The role of ROS displays significant complexity in cancer development and therapy. Elevated ROS levels can induce metabolic reprogramming and promote the proliferation, invasion, and metastasis of cancer cells, resulting in cancer progression. However, excessive ROS accumulation leads to the occurrence of apoptosis, pyroptosis, necroptosis, and ferroptosis in cancer cells, which restrains tumor development. In the TME, ROS frequently promotes angiogenesis and remodels the extracellular matrix (ECM) by enhancing the differentiation of cancer-associated fibroblasts (CAFs), thereby supporting tumor growth. Concurrently, high ROS levels favour immunosuppressive cells, including M2-polarized macrophages, and regulatory T cells (Tregs), while impairing the antitumor capabilities of T cells. In the aspect of cancer therapy, it is overly simplistic to merely combine chemoradiotherapy with antioxidants as a therapeutic strategy. Instead, highlighting targeted therapies that modulate ROS is essential, given their inherent complexity. Fortunately, a variety of innovative treatments have emerged, including nanodrug delivery systems (NDDS), proteolysis-targeting chimeras (PROTAC), and adoptive cell therapy (ADT), which not only exhibit synergistic effects with immune checkpoint therapy (ICT), but also enhance the antitumor capabilities of the TME. In this paper, we elucidate the mechanism of ROS production, enumerate the role of ROS in cancer development and the TME, and discuss advancements in ROS-targeted cancer therapeutics.
Collapse
Affiliation(s)
- Tingfeng Shen
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yutong Wang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Linmao Cheng
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Ya Gao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Shuntong Zhang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xue Chen
- Early Clinical Trial Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China.
| |
Collapse
|
3
|
Wang M, Xia X, Li Y, Zhang K, Cheng X, Wen X, Zhao L, Guo X, Song F, Cheng A. Mg 2+-Loaded Black Phosphorus Nanosheet Protects against Cerebral Ischemic Injury through Anti-Oxidative and Anti-Inflammatory Effects. Adv Healthc Mater 2025; 14:e2500929. [PMID: 40192293 DOI: 10.1002/adhm.202500929] [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: 02/19/2025] [Indexed: 05/17/2025]
Abstract
Ischemic stroke is a severe neurological disease, with high morbidity and mortality worldwide. To date, the treatment of ischemic stroke is limited, and its consequent ischemia-reperfusion injury is an important reason for this result. Excessive reactive oxygen species (ROS) and inflammatory storm followed by ischemia-reperfusion alter the microenvironment of cerebral ischemic penumbra, leading to the devastating damage to the brain. Herein, we design a black phosphorus nanosheets (BPNSs) loaded with magnesium ions (Mg2+) and polydopamine (PBP@Mg) to tackle the above problems. BPNSs of PBP@Mg effectively scavenge excessive ROS in neurocytes. Mg2+ plays an anti-inflammatory role in ischemic penumbra. Furthermore, polydopamine improves the stability of BPNSs. PBP@Mg is subsequently injected into the lateral ventricle of a rat model of ischemic stroke, resulting in an improvement of the ischemic microenvironment and a reduction in ischemic volume. BPNSs of PBP@Mg counteract against the excessive generation of ROS and the neuronal apoptosis in ischemic penumbra. Meanwhile, PBP@Mg dramatically suppresses inflammation by promoting the transformation of microglia from M1 to M2 in ischemic penumbra. PBP@Mg group exhibit a significantly better performance in neurofunctional behavior compared to ischemic group. Taken together, this study provides a novel therapeutic approach for cerebral ischemia-reperfusion injury via anti-oxidative and anti-inflammatory effects.
Collapse
Affiliation(s)
- Min Wang
- Jinzhou Medical University Postgraduate Training Base (Zhejiang Provincial People's Hospital), Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Nuclear Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xinyi Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yaoyao Li
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Kai Zhang
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xu Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuehua Wen
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lijun Zhao
- Jinzhou Medical University Postgraduate Training Base (Zhejiang Provincial People's Hospital), Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Nuclear Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiaopeng Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fahuan Song
- Jinzhou Medical University Postgraduate Training Base (Zhejiang Provincial People's Hospital), Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Nuclear Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Aiping Cheng
- Jinzhou Medical University Postgraduate Training Base (Zhejiang Provincial People's Hospital), Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Nuclear Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
4
|
Chang Z, Wang Z, Chen Y, Liu Y, Gao Y, Cui Y, Wang L, Liu Y, Cheng R, Liu R, Zhang L. Metabolism profiles of tannins in Phyllanthus emblica L. and its immunotherapeutic potential against hepatocellular carcinoma by re-educating tumor microenvironment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156576. [PMID: 40085988 DOI: 10.1016/j.phymed.2025.156576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide. Tumor-associated macrophages (TAMs) are key components of the immunosuppressive tumor microenvironment and represent significant obstacles to effective immunotherapy. Phyllanthus emblica L. (PE), a medicinal plant traditionally used in Tibet, has shown therapeutic promise. This study investigates the effects of the tannin fraction of PE (PE-TF) on HCC and its ability to modulate the tumor immunosuppressive microenvironment. METHODS We evaluated the antitumor efficacy of PE-TF using H22 xenografts and Hepa1-6 orthotopic mouse models. Transcriptomic analysis was performed to identify molecular targets underlying PE-TF suppression of HCC growth. Additionally, UPLC-MS/MS analysis identified the prototypic and metabolic components of PE-TF present in serum, tumor tissues, and adjacent normal liver tissues in the orthotopic HCC model. RESULTS PE-TF significantly suppressed tumor growth in both subcutaneous and orthotopic HCC models and promoted reprogramming of TAMs toward an antitumor M1 phenotype in vivo. Furthermore, PE-TF counteracted the protumoral effects mediated by bone marrow-derived macrophages (BMDMs) exposed to Hepa1-6-derived conditioned medium (HCM). Although TBH promoted macrophage M2 polarization, the reactive oxygen species (ROS)-scavenging activity of PE-TF effectively inhibited this process. Modulation of the tumor microenvironment by PE-TF-enhanced CD8+T cell infiltration and bolstered their antitumor response, as evidenced by increased transcription of perforin, IFN-γ, and IL-2. Transcriptomic analysis further revealed that T-cell receptor and cytotoxic T-cell signaling pathways are critical mediators of PE-TF' therapeutic effects. Moreover, we preliminarily characterized 79 components across serum, liver, and tumor tissues, and identified metabolic pathways for PE-TF ingredients-including methylation and glycosylation modifications of tumor-enriched constituents. Notably, seven components, such as corilagin and urolithin D, are hypothesized to possess immunomodulatory properties. CONCLUSION Our findings underscore the potential of PE-TF as an adjuvant immunotherapy for HCC. By scavenging ROS, PE-TF reverses the immunosuppressive M2-TAM phenotype and remodels the tumor microenvironment, thereby enhancing antitumor immunity. Additionally, integrating chemical and metabolic profiling offers a promising strategy for refining candidate selection in future drug discovery endeavors.
Collapse
Affiliation(s)
- Zihao Chang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Zhaohui Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yinxin Chen
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yuqi Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Ye Gao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yitong Cui
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Le Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yue Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Ruiyang Cheng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China.
| | - Runping Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China.
| | - Lanzhen Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China.
| |
Collapse
|
5
|
Zhu X, Yuan F, Sun Q, Yang C, Jiang H, Xiang X, Zhang X, Sun Z, Wei Y, Chen Q, Cai L. N-acetylcysteine remodels the tumor microenvironment of primary and recurrent mouse glioblastoma. J Neurooncol 2025; 173:131-145. [PMID: 39954037 DOI: 10.1007/s11060-025-04971-9] [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: 12/11/2024] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE Glioblastoma (GBM) exhibits a high ROS character, giving rise to an immunosuppressive microenvironment and tumor vascular abnormality. This study investigated the potential effect of N-acetylcysteine (NAC), an antioxidant, on primary and recurrent mouse brain tumors. METHODS We measured reactive oxygen species (ROS)/ glutathione (GSH) levels in human GBM. Additionally, we conducted NAC trials on primary mouse brain tumor models (GL261-Luc, CT2A-Luc) and a recurrent mouse GBM model (GL261-iCasp9-Luc). After brain tumor inoculation, mice received a daily 100 mg/kg NAC treatment, and the tumor volume was monitored via IVIS imaging. The efficacy of NAC was evaluated through survival time, tumor volume, ROS/GSH levels, M1/M2 macrophages, immune cells infiltration, and tumor vascularization. RESULTS Human GBM suffered from significant oxidative stress. With NAC treatment, mouse brain tumors exhibited a lower ROS level, more M1-like tumor-associated macrophages/microglia (TAMs), more CD8 + T cell infiltration, and a normalized vascular character. NAC inhibited tumor growth and suppressed recurrence in mouse brain tumor models. CONCLUSION NAC is a promising adjunctive drug to remodel the brain tumors microenvironment.
Collapse
Affiliation(s)
- Xiwei Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Fanen Yuan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Qian Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Chen Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Hongxiang Jiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Xi Xiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Xinyi Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Zhiqiang Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Yuxin Wei
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China.
| | - Linzhi Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
6
|
Parbin NS, Banik B. Copper peroxide incorporated BSA-NPs: a pH-responsive, self-supplying source of reactive oxygen species for cancer cell destruction via polarization of macrophages to the M1 phenotype. Chem Commun (Camb) 2025; 61:5605-5608. [PMID: 40105257 DOI: 10.1039/d5cc00216h] [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: 03/20/2025]
Abstract
Herein, we produced self-assembled CuPer-BSA nanoparticles (NPs) formed as a result of incorporation of copper peroxide into bovine serum albumin (BSA) and serving as a self-supplying source of Cu2+ and H2O2 at biologically relevant acidic pH, and in these conditions activating a Fenton-type reaction to generate hydroxyl radicals (˙OH). ROS-induced polarization of RAW 264.7 macrophages to the M1 phenotype resulted in cellular secretions that showed significant anti-cancer efficacy against HeLa cells.
Collapse
Affiliation(s)
- Nursaima Sultana Parbin
- Department of Chemistry, Cotton University, Panbazar, Guwahati-781001, Assam, India.
- Department of Chemistry, Gauhati University, Gopinath Bordoloi Nagar, Guwahati-781014, Assam, India
| | - Bhabatosh Banik
- Department of Chemistry, Cotton University, Panbazar, Guwahati-781001, Assam, India.
| |
Collapse
|
7
|
Lee J, Kang Y, Lee H, Saravanakumar G, Park SA, Ahn S, Kim WJ. Amplifying glioblastoma immunotherapy: T cell shielding through Nitric oxide/reactive oxygen species scavenging nanoparticles Potentiates anti-PD-1. Biomaterials 2025; 315:122904. [PMID: 39490061 DOI: 10.1016/j.biomaterials.2024.122904] [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: 09/02/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
Despite the success of immune checkpoint blockade (ICB) therapy in various cancers, its efficacy faces challenges in glioblastoma (GBM) due to the immunosuppressive cold-tumor microenvironment. The scarcity of tumor-infiltrating T cells and the suppression of T cell activity significantly limit therapeutic outcomes in GBM. Nitric oxide (NO) and reactive oxygen species (ROS) from tumor-associated myeloid cells (TAMCs) are key contributors to T cell suppression, reducing ICB therapy effectiveness. In this study, we developed NO-ROS scavenging micelles that effectively scavenge both NO and ROS, protecting T cells from their exhausting effects. This leads to a significant increase in T cell infiltration and activation. Moreover, when combined with αPD-1, the survival rate increases to 40 % up to 120 days, enhancing therapeutic efficacy compared to αPD-1 alone. This approach not only protects T cells from the inhibitory effects of NO and ROS but also has the potential to reshape the tumor microenvironment, overcoming T cell suppression in cold tumors.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Yeoul Kang
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Hyori Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | | | - Soon A Park
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea; OmniaMed Co., Ltd, Pohang, 37666, Republic of Korea.
| |
Collapse
|
8
|
Son HH, Moon SJ. Pathogenesis of systemic sclerosis: an integrative review of recent advances. JOURNAL OF RHEUMATIC DISEASES 2025; 32:89-104. [PMID: 40134549 PMCID: PMC11931279 DOI: 10.4078/jrd.2024.0129] [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: 11/05/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 03/27/2025]
Abstract
Systemic sclerosis (SSc), or scleroderma, is a complex autoimmune connective tissue disease characterized by autoimmunity, vasculopathy, and progressive organ fibrosis, leading to severe organ dysfunction. The disease begins with a vascular injury triggered by autoimmune responses and environmental factors against a backdrop of genetic predisposition. This injury impairs angiogenesis and vasculogenesis, resulting in capillary loss and arteriolar constriction, which promotes immune cell infiltration and sustained inflammation within affected tissues. These vascular anomalies cause severe complications, including pulmonary artery hypertension, scleroderma renal crisis, and skin ulcers. Chronic inflammation fosters persistent fibroblast activation, resulting in extensive fibrosis that defines SSc. This review synthesizes the latest research on pathogenesis of SSc, highlighting the shift from fundamental research to a precision therapeutic approach. It explores the potential of technologies like flow cytometry and single-cell RNA sequencing to investigate pathogenic cell subtypes. These platforms integrate transcriptomic, genomic, proteomic, and epigenomic data to uncover insights into the underlying mechanisms of SSc pathogenesis. This review advocates for a multidisciplinary, patient-centric approach that harnesses recent scientific advances, directing future SSc research toward personalized and precise interventions.
Collapse
Affiliation(s)
- Ha-Hee Son
- Division of Rheumatology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
9
|
Rettkowski J, Romero-Mulero MC, Singh I, Wadle C, Wrobel J, Chiang D, Hoppe N, Mess J, Schönberger K, Lalioti ME, Jäcklein K, SilvaRego B, Bühler T, Karabacz N, Egg M, Demollin H, Obier N, Zhang YW, Jülicher C, Hetkamp A, Czerny M, Jones MJ, Seung H, Jain R, von Zur Mühlen C, Maier A, Lother A, Hilgendorf I, van Galen P, Kreso A, Westermann D, Rodriguez-Fraticelli AE, Heidt T, Cabezas-Wallscheid N. Modulation of bone marrow haematopoietic stem cell activity as a therapeutic strategy after myocardial infarction: a preclinical study. Nat Cell Biol 2025; 27:591-604. [PMID: 40175666 PMCID: PMC11991920 DOI: 10.1038/s41556-025-01639-4] [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: 07/09/2024] [Accepted: 02/19/2025] [Indexed: 04/04/2025]
Abstract
Myocardial infarction (MI) is a major global health concern. Although myeloid cells are crucial for tissue repair in emergency haematopoiesis after MI, excessive myelopoiesis can exacerbate scarring and impair cardiac function. Bone marrow (BM) haematopoietic stem cells (HSCs) have the unique capability to replenish the haematopoietic system, but their role in emergency haematopoiesis after MI has not yet been established. Here we collected human sternal BM samples from over 150 cardiac surgery patients, selecting 49 with preserved cardiac function. We show that MI causes detrimental transcriptional and functional changes in human BM HSCs. Lineage tracing experiments suggest that HSCs are contributors of pro-inflammatory myeloid cells infiltrating cardiac tissue after MI. Therapeutically, enforcing HSC quiescence with the vitamin A metabolite 4-oxo-retinoic acid dampens inflammatory myelopoiesis, thereby modulating tissue remodelling and preserving long-term cardiac function after MI.
Collapse
Affiliation(s)
- Jasmin Rettkowski
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Laboratory of Stem Cell Biology and Ageing, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Mari Carmen Romero-Mulero
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Indranil Singh
- Institute for Research in Biomedicine, Barcelona Institute for Science and Technology, Barcelona, Spain
- Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Carolin Wadle
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jan Wrobel
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Diana Chiang
- Spemann Graduate School of Biology and Medicine, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Natalie Hoppe
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julian Mess
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies, Freiburg, Germany
| | | | | | - Karin Jäcklein
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Beatriz SilvaRego
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Timon Bühler
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Noémie Karabacz
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
| | - Mirijam Egg
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
| | - Helen Demollin
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Nadine Obier
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Yu Wei Zhang
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Claus Jülicher
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anne Hetkamp
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Cardiovascular Surgery, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
| | - Martin Czerny
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Cardiovascular Surgery, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
| | | | - Hana Seung
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ritika Jain
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Constantin von Zur Mühlen
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Maier
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Interdisciplinary Medical Intensive Care, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter van Galen
- Division of Hematology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Antonia Kreso
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alejo E Rodriguez-Fraticelli
- Institute for Research in Biomedicine, Barcelona Institute for Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - Timo Heidt
- Department of Cardiology and Angiology, University Heart Center, Medical Center, University of Freiburg, Freiburg, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Nina Cabezas-Wallscheid
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
- Laboratory of Stem Cell Biology and Ageing, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland.
- Centre for Integrative Biological Signalling Studies, Freiburg, Germany.
| |
Collapse
|
10
|
Abulaban A, Al‐kuraishy H, Al‐Gareeb A, Ahmed E, Alruwaili M, Alexiou A, Papadakis M, El‐Saber Batiha G. The Possible Role of Metformin and Fibroblast Growth Factor-21 in Multiple Sclerosis Neuropathology: Birds of a Feather Flock Together. Eur J Neurosci 2025; 61:e70067. [PMID: 40172524 PMCID: PMC11963988 DOI: 10.1111/ejn.70067] [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: 11/05/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 04/04/2025]
Abstract
Multiple sclerosis (MS) is a progressive demyelinating disease of the CNS, characterized by inflammation, the formation of CNS plaques, and damage to the neuronal myelin sheath (Graphical abstract). Fibroblast growth factor 21 (FGF21) is involved in various metabolic disorders and neurodegenerative diseases. FGF21 and its co-receptor β-Kloth are essential in the remyelination process of MS. Metformin, an insulin-sensitizing drug that is the first-line treatment for type 2 diabetes mellitus (T2DM), may have a potential neuroprotective impact by up-regulating the production of FGF21, which may prevent the onset of neurodegenerative diseases including MS. The purpose of this review is to clarify how metformin affects MS neuropathology mechanistically via modifying FGF21. Metformin increases the expression of FGF21. Metformin also increases the expression of β-Klotho, modulates oxidative stress, reduces glutamate-induced excitotoxicity, and regulates platelet function and coagulation cascades. In conclusion, metformin can enhance the functional activity of FGF21 in counteracting the development and progression of MS. Preclinical and clinical studies are warranted in this regard.
Collapse
Affiliation(s)
- Ahmad A. Abulaban
- College of MedicineKing Saud bin Abdulaziz University for Health SciencesRiyadhSaudi Arabia
- Division of Neurology, King Abdulaziz Medical CityMinistry of the National Guard Health AffairsRiyadhSaudi Arabia
- King Abdullah International Medical Research CenterRiyadhSaudi Arabia
| | - Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBagdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBagdadIraq
| | - Eman A. Ahmed
- Department of Pharmacology, Faculty of Veterinary MedicineSuez Canal UniversityIsmailiaEgypt
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of MedicineJouf UniversitySakakaSaudi Arabia
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliPunjabIndia
- Department of Research and DevelopmentFunogenAthensGreece
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
11
|
Lee SY, Kim JH, Song Y, Kim S, Kang HJ, Kim J, Lee YJ, Seo HR. Inhibition of 11β-hydroxysteroid dehydrogenase 1 alleviates pulmonary fibrosis through inhibition of endothelial-to-mesenchymal transition and M2 macrophage polarization by upregulating heme oxygenase-1. Cell Death Dis 2025; 16:196. [PMID: 40118823 PMCID: PMC11928689 DOI: 10.1038/s41419-025-07522-2] [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: 09/27/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/24/2025]
Abstract
The intracellular enzyme 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1) catalyzes the interconversion of active glucocorticoid (cortisol) and its intrinsically inert form (cortisone) in metabolic tissues. Although 11βHSD1 is considered a promising therapeutic target in metabolic disorders such as type 2 diabetes, obesity, and nonalcoholic steatohepatitis because of its hepatic functions, its roles in other tissues have received less attention. In this study, we show that the 11βHSD1-specific inhibitor J2H-1702 facilitates the reversion of endothelial-to-mesenchymal transition in multicellular lung spheroid models encapsulating the complex crosstalk among lung cancer cells, vascular endothelial cells, and macrophages. In vascular endothelial cells, J2H-1702 not only suppressed interleukin-1α (IL-1α) expression but also attenuated reactive oxygen species-induced DNA damage by upregulating heme oxygenase-1. Additionally, in macrophages, which are key regulators of fibrogenesis, inhibition of 11βHSD1 markedly reduced IL-1β expression, thereby modulating the pro-inflammatory phenotype of activated macrophages. In mouse models of pulmonary fibrosis, including a bleomycin-induced idiopathic model and a radiation-induced model, J2H-1702 alleviated pulmonary fibrosis and markedly improved the efficacy of nintedanib. Collectively, our data suggest that J2H-1702 holds promise as a clinical candidate for the treatment of pulmonary fibrosis associated with reactive oxygen species-induced DNA damage, endothelial-to-mesenchymal transition, and inflammatory responses.
Collapse
Affiliation(s)
- Su-Yeon Lee
- Advanced Biomedical Research Lab, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Ji-Hee Kim
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, 75 nowongu nowon gil, Seoul, 139-706, Korea
| | - Yeonhwa Song
- Advanced Biomedical Research Lab, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Sanghwa Kim
- Advanced Biomedical Research Lab, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hyo Jin Kang
- R&D center, J2H Biotech Inc., Saneop-ro 156 beon-gil, Gwonseon-gu, Suwon-si, Gyeonggi-do, 16648, Republic of Korea
| | - Jason Kim
- R&D center, J2H Biotech Inc., Saneop-ro 156 beon-gil, Gwonseon-gu, Suwon-si, Gyeonggi-do, 16648, Republic of Korea
| | - Yoon-Jin Lee
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, 75 nowongu nowon gil, Seoul, 139-706, Korea
| | - Haeng Ran Seo
- Advanced Biomedical Research Lab, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
12
|
Forooshani PK, Razaviamri F, Smies A, Morath LM, Pinnaratip R, Bhuiyan MSA, Rajachar R, Goldman J, Lee BP. Accelerated dermal wound healing in diabetic mice by a H 2O 2-generating catechol-functionalized gelatin microgel. J Mater Chem B 2025; 13:3967-3979. [PMID: 40029170 DOI: 10.1039/d4tb01722f] [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: 03/05/2025]
Abstract
Physically crosslinked gelatin microgels were functionalized with a bioadhesive molecule, catechol, to study the effect of in situ generated H2O2 on full-thickness wound repair in diabetic mice. Due to the physically crosslinked nature of the microgels, they transition into a hydrogel film upon hydration. The formation of a hydrogel film was confirmed by the changes in their morphology and viscoelastic properties. Additionally, these microgels released up to 86 μM of H2O2 as a result of catechol autoxidation. The generated H2O2 completely eradicated Staphylococcus epidermidis with an initial concentration of 103 CFU mL-1. These microgels were not cytotoxic and promoted VEGF upregulation in immortalized human keratinocytes (HaCaT) in vitro. When the microgels were applied to a full-thickness dermal wound in diabetic mice, dermal wound closure was accelerated over 14 days, achieving a wound closure of 90% based on the wound area. Microgel-treated wounds also resulted in complete re-epithelialization and regeneration of new dermal tissues with morphology and structure resembling those of native tissues. These results indicate that the release of micromolar concentrations of H2O2 can accelerate wound healing in a healing-impaired animal.
Collapse
Affiliation(s)
- Pegah Kord Forooshani
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| | - Fatemeh Razaviamri
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| | - Ariana Smies
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| | - Lea M Morath
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| | - Rattapol Pinnaratip
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| | - Md Saleh Akram Bhuiyan
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| | - Rupak Rajachar
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| | - Jeremy Goldman
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| | - Bruce P Lee
- Depart of Biomedical Engineering, Michigan Technological University, Houghton, Michigan, 49931, USA.
| |
Collapse
|
13
|
Bonacina F, Zhang X, Manel N, Yvan-Charvet L, Razani B, Norata GD. Lysosomes in the immunometabolic reprogramming of immune cells in atherosclerosis. Nat Rev Cardiol 2025; 22:149-164. [PMID: 39304748 PMCID: PMC11835540 DOI: 10.1038/s41569-024-01072-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Lysosomes have a central role in the disposal of extracellular and intracellular cargo and also function as metabolic sensors and signalling platforms in the immunometabolic reprogramming of macrophages and other immune cells in atherosclerosis. Lysosomes can rapidly sense the presence of nutrients within immune cells, thereby switching from catabolism of extracellular material to the recycling of intracellular cargo. Such a fine-tuned degradative response supports the generation of metabolic building blocks through effectors such as mTORC1 or TFEB. By coupling nutrients to downstream signalling and metabolism, lysosomes serve as a crucial hub for cellular function in innate and adaptive immune cells. Lysosomal dysfunction is now recognized to be a hallmark of atherogenesis. Perturbations in nutrient-sensing and signalling have profound effects on the capacity of immune cells to handle cholesterol, perform phagocytosis and efferocytosis, and limit the activation of the inflammasome and other inflammatory pathways. Strategies to improve lysosomal function hold promise as novel modulators of the immunoinflammatory response associated with atherosclerosis. In this Review, we describe the crosstalk between lysosomal biology and immune cell function and polarization, with a particular focus on cellular immunometabolic reprogramming in the context of atherosclerosis.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Nicolas Manel
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU), Oncoage, Nice, France
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Giuseppe D Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
14
|
Newman AAC, Dalman JM, Moore KJ. Cardiovascular Disease and Cancer: A Dangerous Liaison. Arterioscler Thromb Vasc Biol 2025; 45:359-371. [PMID: 39781742 PMCID: PMC11864891 DOI: 10.1161/atvbaha.124.319863] [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: 11/08/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
The field of cardio-oncology has traditionally focused on the impact of cancer and its therapies on cardiovascular health. Mounting clinical and preclinical evidence, however, indicates that the reverse may also be true: cardiovascular disease can itself influence tumor growth and metastasis. Numerous epidemiological studies have reported that individuals with prevalent cardiovascular disease have an increased incidence of cancer. In parallel, studies using preclinical mouse models of myocardial infarction, heart failure, and cardiac remodeling support the notion that cardiovascular disorders accelerate the growth of solid tumors and metastases. These findings have ushered in a new and burgeoning field termed reverse cardio-oncology that investigates the impact of cardiovascular disease pathophysiology on cancer emergence and progression. Recent studies have begun to illuminate the mechanisms driving this relationship, including shared risk factors, reprogramming of immune responses, changes in gene expression, and the release of cardiac factors that result in selective advantages for tumor cells or their local milieu, thus exacerbating cancer pathology. Here, we review the evidence supporting the relationship between cardiovascular disease and cancer, the mechanistic pathways enabling this connection, and the implications of these findings for patient care.
Collapse
Affiliation(s)
- Alexandra A C Newman
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jessie M Dalman
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- University of Michigan Medical School, Ann Arbor, MI 48104, USA
| | - Kathryn J Moore
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
15
|
Deng X, Huang Y, Zhang J, Chen Y, Jiang F, Zhang Z, Li T, Hou L, Tan W, Li F. Histone lactylation regulates PRKN-Mediated mitophagy to promote M2 Macrophage polarization in bladder cancer. Int Immunopharmacol 2025; 148:114119. [PMID: 39854875 DOI: 10.1016/j.intimp.2025.114119] [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: 11/04/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Bladder cancer (BCa), particularly muscle-invasive bladder cancer (MIBC), is associated with poor prognosis, partly because of immune evasion driven by M2 tumor-associated macrophages (TAMs). Understanding the regulatory mechanisms of M2 macrophage polarization via PRKN-mediated mitophagy and histone lactylation (H3K18la) is crucial for improving treatment strategies. METHODS A single-cell atlas from 46 human BCa samples was constructed to identify macrophage subpopulations. Bioinformatics analysis and experimental validation, including ChIP-seq and lactylation modulation assays, were used to investigate the role of PRKN in M2 macrophage polarization and its regulation by H3K18la. RESULTS Single-cell analysis revealed distinct macrophage subpopulations, including M1 and M2 types. PRKN was identified as a critical regulator of mitophagy in M2 macrophages, supporting their immunosuppressive function. Bulk RNA-seq and gene intersection analysis revealed a set of mitophagy-related macrophage polarization genes (Mito_Macro_RGs) enriched in mitophagy and immune pathways. Pseudotime analysis revealed that PRKN was upregulated during the M1-to-M2 transition. siRNA-mediated PRKN knockdown impaired M2 polarization, reducing the expression of CD206 and ARG1. ChIP-seq and histone lactylation modulation confirmed that H3K18la enhanced PRKN expression, promoting mitophagy and M2 polarization and thereby facilitating immune suppression and tumor progression. CONCLUSIONS Histone lactylation regulated PRKN-mediated mitophagy, promoting M2 macrophage polarization and contributing to immune evasion in BCa.
Collapse
Affiliation(s)
- Xiaolin Deng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Yuan Huang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Jinge Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Yuwen Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Feifan Jiang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Zicai Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Tanghua Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Lina Hou
- Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| | - Fei Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| |
Collapse
|
16
|
Liu X, Xu W, Feng J, Wang Y, Li K, Chen Y, Wang W, Zhao W, Ge S, Li J. Adoptive cell transfer of piezo-activated macrophage rescues immunosuppressed rodents from life-threating bacterial infections. Nat Commun 2025; 16:1363. [PMID: 39905015 PMCID: PMC11794888 DOI: 10.1038/s41467-025-56460-2] [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: 02/01/2024] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
Bacterial infections pose a significant threat to human health. Catalytic antibacterial nanoparticles that generate reactive oxygen species (ROS) are emerging, as a promising therapeutic approach in treating bacterial infection by boosting the innate immune defenses. However, the interaction between innate immune cells and these catalytic nanoparticles remains poorly understood. Here, by using rodent models of bacterial infection, we test the antimicrobial properties of ultrasound-responsive piezo-catalytic nanoparticles (piezoNP). We show that piezoNPs strongly interact with macrophages within subcutaneous abscesses caused by Staphylococcus aureus (S. aureus) infections, and demonstrate that this interaction enhances the macrophage-mediated antibacterial phagocytosis and killing activity through intracellular piezocatalysis. Moreover, we test the use of these piezo-activated macrophages (piezoMϕ) as adoptive cell therapy (ACT) for treating various immunosuppressive bacterial infections, including sepsis, pneumonia and peritonitis. Our study thus highlights the potential application of catalytic nanoparticles as a promising alternative to conventional infection treatment to effectively modulate the innate immune responses and to engineer macrophages for immunotherapy purposes.
Collapse
Affiliation(s)
- Xiaoyi Liu
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Wenxiu Xu
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Junkun Feng
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Ying Wang
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Kai Li
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Yi Chen
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Wenjun Wang
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Weiwei Zhao
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Shaohua Ge
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| | - Jianhua Li
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| |
Collapse
|
17
|
Du SL, Zhou YT, Hu HJ, Lin L, Zhang ZQ. Silica-induced ROS in alveolar macrophages and its role on the formation of pulmonary fibrosis via polarizing macrophages into M2 phenotype: a review. Toxicol Mech Methods 2025; 35:89-100. [PMID: 39223849 DOI: 10.1080/15376516.2024.2400323] [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: 06/12/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Alveolar macrophages (AMs), the first line against the invasion of foreign invaders, play a predominant role in the pathogenesis of silicosis. Studies have shown that inhaled silica dust is recognized and engulfed by AMs, resulting in the production of large amounts of silica-induced reactive oxygen species (ROS), including particle-derived ROS and macrophage-derived ROS. These ROS change the microenvironment of the AMs where the macrophage phenotype is stimulated to swift from M0 to M1 and/or M2, and ultimately emerge as the M2 phenotype to trigger silicosis. This is a complex process accompanied by various molecular biological events. Unfortunately, the detailed processes and mechanisms have not been systematically described. In this review, we first systematically introduce the process of ROS induced by silica in AMs. Then, describe the role and molecular mechanism of M2-type macrophage polarization caused by silica-induced ROS. Finally, we review the mechanism of pulmonary fibrosis induced by M2 polarized AMs. We conclude that silica-induced ROS initiate the fibrotic process of silicosis by inducing macrophage into M2 phenotype, and that targeted intervention of silica-induced ROS in AMs can reprogram the macrophage polarization and ameliorate the pathogenesis of silicosis.
Collapse
Affiliation(s)
- Shu-Ling Du
- School of Public Health, Shandong Second Medical University, Weifang, China
- School of Public Health, Jining Medical University, Jining, China
| | - Yu-Ting Zhou
- School of Public Health, Jining Medical University, Jining, China
| | - Hui-Jie Hu
- School of Public Health, Shandong Second Medical University, Weifang, China
- School of Public Health, Jining Medical University, Jining, China
| | - Li Lin
- School of Public Health, Jining Medical University, Jining, China
| | - Zhao-Qiang Zhang
- School of Public Health, Jining Medical University, Jining, China
| |
Collapse
|
18
|
Fedotova EI, Berezhnov AV, Popov DY, Shitikova EY, Vinokurov AY. The Role of mtDNA Mutations in Atherosclerosis: The Influence of Mitochondrial Dysfunction on Macrophage Polarization. Int J Mol Sci 2025; 26:1019. [PMID: 39940788 PMCID: PMC11817597 DOI: 10.3390/ijms26031019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Atherosclerosis is a complex inflammatory process associated with high-mortality cardiovascular diseases. Today, there is a growing body of evidence linking atherosclerosis to mutations of mitochondrial DNA (mtDNA). But the mechanism of this link is insufficiently studied. Atherosclerosis progression involves different cell types and macrophages are one of the most important. Due to their high plasticity, macrophages can demonstrate pro-inflammatory and pro-atherogenic (macrophage type M1) or anti-inflammatory and anti-atherogenic (macrophage type M2) effects. These two cell types, formed as a result of external stimuli, differ significantly in their metabolic profile, which suggests the central role of mitochondria in the implementation of the macrophage polarization route. According to this, we assume that mtDNA mutations causing mitochondrial disturbances can play the role of an internal trigger, leading to the formation of macrophage M1 or M2. This review provides a comparative analysis of the characteristics of mitochondrial function in different types of macrophages and their possible associations with mtDNA mutations linked with inflammation-based pathologies including atherosclerosis.
Collapse
Affiliation(s)
- Evgeniya I. Fedotova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (E.I.F.); (A.V.B.)
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Alexey V. Berezhnov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (E.I.F.); (A.V.B.)
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Daniil Y. Popov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Elena Y. Shitikova
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Andrey Y. Vinokurov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| |
Collapse
|
19
|
Wang Z, Li X, Moura AK, Hu JZ, Wang YT, Zhang Y. Lysosome Functions in Atherosclerosis: A Potential Therapeutic Target. Cells 2025; 14:183. [PMID: 39936975 PMCID: PMC11816498 DOI: 10.3390/cells14030183] [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: 12/16/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Lysosomes in mammalian cells are recognized as key digestive organelles, containing a variety of hydrolytic enzymes that enable the processing of both endogenous and exogenous substrates. These organelles digest various macromolecules and recycle them through the autophagy-lysosomal system. Recent research has expanded our understanding of lysosomes, identifying them not only as centers of degradation but also as crucial regulators of nutrient sensing, immunity, secretion, and other vital cellular functions. The lysosomal pathway plays a significant role in vascular regulation and is implicated in diseases such as atherosclerosis. During atherosclerotic plaque formation, macrophages initially engulf large quantities of lipoproteins, triggering pathogenic responses that include lysosomal dysfunction, foam cell formation, and subsequent atherosclerosis development. Lysosomal dysfunction, along with the inefficient degradation of apoptotic cells and the accumulation of modified low-density lipoproteins, negatively impacts atherosclerotic lesion progression. Recent studies have highlighted that lysosomal dysfunction contributes critically to atherosclerosis in a cell- and stage-specific manner. In this review, we discuss the mechanisms of lysosomal biogenesis and its regulatory role in atherosclerotic lesions. Based on these lysosomal functions, we propose that targeting lysosomes could offer a novel therapeutic approach for atherosclerosis, shedding light on the connection between lysosomal dysfunction and disease progression while offering new insights into potential anti-atherosclerotic strategies.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Alexandra K. Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Jenny Z. Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| |
Collapse
|
20
|
Chatzikalil E, Arvanitakis K, Kalopitas G, Florentin M, Germanidis G, Koufakis T, Solomou EE. Hepatic Iron Overload and Hepatocellular Carcinoma: New Insights into Pathophysiological Mechanisms and Therapeutic Approaches. Cancers (Basel) 2025; 17:392. [PMID: 39941760 PMCID: PMC11815926 DOI: 10.3390/cancers17030392] [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: 12/18/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Hepatocellular carcinoma (HCC), the most common form of primary liver cancer, is rising in global incidence and mortality. Metabolic dysfunction-associated steatotic liver disease (MASLD), one of the leading causes of chronic liver disease, is strongly linked to metabolic conditions that can progress to liver cirrhosis and HCC. Iron overload (IO), whether inherited or acquired, results in abnormal iron hepatic deposition, significantly impacting MASLD development and progression to HCC. While the pathophysiological connections between hepatic IO, MASLD, and HCC are not fully understood, dysregulation of glucose and lipid metabolism and IO-induced oxidative stress are being investigated as the primary drivers. Genomic analyses of inherited IO conditions reveal inconsistencies in the association of certain mutations with liver malignancies. Moreover, hepatic IO is also associated with hepcidin dysregulation and activation of ferroptosis, representing promising targets for HCC risk assessment and therapeutic intervention. Understanding the relationship between hepatic IO, MASLD, and HCC is essential for advancing clinical strategies against liver disease progression, particularly with recent IO-targeted therapies showing potential at improving liver biochemistry and insulin sensitivity. In this review, we summarize the current evidence on the pathophysiological association between hepatic IO and the progression of MASLD to HCC, underscoring the importance of early diagnosis, risk stratification, and targeted treatment for these interconnected conditions.
Collapse
Affiliation(s)
- Elena Chatzikalil
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece;
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Georgios Kalopitas
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Matilda Florentin
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece; (K.A.); (G.K.); (G.G.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Theocharis Koufakis
- Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Elena E. Solomou
- Department of Internal Medicine, University of Patras Medical School, 26500 Rion, Greece
| |
Collapse
|
21
|
Chen S, Li L, Yuan H, Gui H, Wan Q, Wang M, Lv H, Wang C, Zhu L, Nie Y, Zhang X. Intratumoral injection of R848 and poly(I:C) synergistically promoted antitumor immune responses by reprogramming macrophage polarization and activating DCs in lung cancer. Clin Exp Immunol 2025; 219:uxae110. [PMID: 40079116 PMCID: PMC12062569 DOI: 10.1093/cei/uxae110] [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: 05/13/2024] [Revised: 10/17/2024] [Accepted: 03/13/2025] [Indexed: 03/14/2025] Open
Abstract
INTRODUCTION Immunotherapy has rapidly become a primary treatment option for many lung cancer patients because of its success in treating this prevalent and deadly disease. However, the success of immunotherapy relies on overcoming the immunosuppressive tumor microenvironment, making remodeling this environment a potential strategy for lung cancer therapy. Research suggests that Toll-like receptor (TLR) agonists can impede tumor growth by promoting the conversion of tumor-associated macrophages into an M1-like state or enhancing dendritic cell development. However, there is insufficient research on the combined use of TLR agonists for treating lung cancer. METHODS In this study, we examined how TLR agonists such as resiquimod (R848) and poly(I:C) impact lung cancer treatment when used alone or in combination. In vitro, the regulatory functions and mechanisms of R848 and poly(I:C) were analyzed in primary macrophages, RAW264.7 cells, and primary dendritic cells (DCs). Tumor treatment efficacy was assessed in vivo with a Lewis lung carcinoma (LLC) mouse model. RESULTS The combination of R848 + poly(I:C) enhances the transformation of macrophages from the M2 phenotype to the M1 phenotype by increasing inflammatory cytokine levels. The percentage of mature DCs expressing MHC-II+CD11c+ and CD86+ cells was significantly higher in the R848 + poly(I:C) group compared with the other groups. Intratumoral injection of the synergistic combination of R848 + poly(I:C) suppressed tumor growth by increasing the M1:M2 ratio in TAMs, activating DCs, and attracting CD4+ and CD8+ T cells. CONCLUSION R848 + poly(I:C) synergistically induces M1-like polarization of macrophages, activates DCs, and promotes effective antitumor immunity in mice with subcutaneous LLC tumors.
Collapse
Affiliation(s)
| | - Linzhao Li
- School of Medicine, Guizhou University, Guiyang, China
| | - Haohua Yuan
- School of Medicine, Guizhou University, Guiyang, China
| | - Huan Gui
- School of Medicine, Guizhou University, Guiyang, China
| | - Quan Wan
- School of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| | - Mengjiao Wang
- School of Medicine, Guizhou University, Guiyang, China
| | - Hang Lv
- School of Medicine, Guizhou University, Guiyang, China
| | - Chenglv Wang
- School of Medicine, Guizhou University, Guiyang, China
| | - Lan Zhu
- School of Medicine, Guizhou University, Guiyang, China
| | - Yingjie Nie
- School of Medicine, Guizhou University, Guiyang, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xiangyan Zhang
- School of Medicine, Guizhou University, Guiyang, China
- School of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| |
Collapse
|
22
|
Wang H, He W, Liao J, Wang S, Dai X, Yu M, Xie Y, Chen Y. Catalytic Biomaterials-Activated In Situ Chemical Reactions: Strategic Modulation and Enhanced Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411967. [PMID: 39498674 DOI: 10.1002/adma.202411967] [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: 08/13/2024] [Revised: 10/19/2024] [Indexed: 11/07/2024]
Abstract
Chemical reactions underpin biological processes, and imbalances in critical biochemical pathways within organisms can lead to the onset of severe diseases. Within this context, the emerging field of "Nanocatalytic Medicine" leverages nanomaterials as catalysts to modulate fundamental chemical reactions specific to the microenvironments of diseases. This approach is designed to facilitate the targeted synthesis and localized accumulation of therapeutic agents, thus enhancing treatment efficacy and precision while simultaneously reducing systemic side effects. The effectiveness of these nanocatalytic strategies critically hinges on a profound understanding of chemical kinetics and the intricate interplay of reactions within particular pathological microenvironments to ensure targeted and effective catalytic actions. This review methodically explores in situ catalytic reactions and their associated biomaterials, emphasizing regulatory strategies that control therapeutic responses. Furthermore, the discussion encapsulates the crucial elements-reactants, catalysts, and reaction conditions/environments-necessary for optimizing the thermodynamics and kinetics of these reactions, while rigorously addressing both the biochemical and biophysical dimensions of the disease microenvironments to enhance therapeutic outcomes. It seeks to clarify the mechanisms underpinning catalytic biomaterials and evaluate their potential to revolutionize treatment strategies across various pathological conditions.
Collapse
Affiliation(s)
- Huijing Wang
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wenjin He
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Jing Liao
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Shuangshuang Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yu Chen
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Shanghai Institute of Materdicine, Shanghai, 200051, P. R. China
| |
Collapse
|
23
|
Ding X, Zhang J, Wan S, Wang X, Wang Z, Pu K, Wang M, Cao Y, Weng L, Zhu H, Peng F, Chao J, Pei R, Leong DT, Wang L. Non-discriminating engineered masking of immuno-evasive ligands on tumour-derived extracellular vesicles enhances tumour vaccination outcomes. NATURE NANOTECHNOLOGY 2025; 20:156-166. [PMID: 39327512 DOI: 10.1038/s41565-024-01783-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 08/08/2024] [Indexed: 09/28/2024]
Abstract
The success of personalized cancer immunotherapy depends on the initial tumour antigenic presentation to dendritic cells and macrophages. Tumour-derived extracellular vesicles (TEVs) contain abundant tumour antigenic molecules. The presence of anti-phagocytotic signals such as cluster of differentiation 47 (CD47) on the surface of the TEVs, however, leads to evasion of the same dendritic cells and macrophages. Here we show that iron oxide hydroxide nanocomposites can successfully mask TEV surfaces and unblock phagocytosis without affecting extracellular vesicles' elicited immune goals. After internalization, the mask disintegrates in the lysosome, releasing the tumour antigenic cargo. This triggers antigen presentation and promotes dendritic cell activation and maturation and macrophage reprogramming in animal models, leading to a drastic reduction of tumour volume and metastasis, and in human malignant pleural effusion clinical samples. This straightforward masking strategy eliminates the ubiquitous anti-phagocytosis block found in clinical samples and can be applied universally across all patient-specific TEVs as tumour antigenic agents for enhanced immunotherapy.
Collapse
Affiliation(s)
- Xianguang Ding
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China.
| | - JingJing Zhang
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Shuangshuang Wan
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Xu Wang
- Department of Anesthesiology and Intensive Care Unit Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiyu Wang
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Kefeng Pu
- CAS Key Laboratory of Nano-Bio Interface, Key Laboratory of Nanodevices and Applications, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Mao Wang
- CAS Key Laboratory of Nano-Bio Interface, Key Laboratory of Nanodevices and Applications, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Yi Cao
- CAS Key Laboratory of Nano-Bio Interface, Key Laboratory of Nanodevices and Applications, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Lixing Weng
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Houjuan Zhu
- Agency for Science Technology and Research, Singapore, Singapore
| | - Fei Peng
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Jie Chao
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Key Laboratory of Nanodevices and Applications, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, Singapore.
| | - Lianhui Wang
- Institute of Advanced Materials, State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts and Telecommunications, Nanjing, China.
| |
Collapse
|
24
|
Afrifa J, Ofori EG, Opoku YK, Asare KK, Sorkpor RD, Naveh-Fio IW, Armah R, Ofori S, Ephraim RKD. Oxidative Stress and Cancer Risk in Schistosomiasis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:9701021. [PMID: 39720557 PMCID: PMC11668550 DOI: 10.1155/omcl/9701021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/27/2024] [Indexed: 12/26/2024]
Abstract
Background: Schistosomiasis is considered one of the most devastating parasitic diseases globally, coming second only to malaria in terms of morbidity. The disease-causing parasite can inhabit the body for over a decade, leading to imbalances in the host's metabolic systems. The flukes and their eggs can illicit various immunological and metabolic complications resulting in the generation of reactive oxygen species (ROS). These are known to have several devastating effects on the host through increased oxidative stress, DNA mutation, and gene modifications, which can lead to fibrosis and cancer. Main Body: Here, we discuss oxidative stress and cancer risk in Schistosoma infection. The concept of ROS generation and the complex antioxidant systems that enable the parasite to evade oxidant insults and prolong its life span in the host are explored. Further, the various roles of ROS during the initiation and progression of schistosomiasis and its influence on the host are discussed. Finally, mechanisms linked to the risk of bladder cancer in Schistosoma haematobium (S. haematobium) infections are elucidated. Conclusion: Finally, we provide an opinion on how some of these mechanisms could give directions for future studies as well as provide a springboard for diagnostics and drug targeting in schistosomiasis.
Collapse
Affiliation(s)
- Justice Afrifa
- Department of Medical Laboratory Science, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Eric Gyamerah Ofori
- Department of Biology Education, Faculty of Science Education, University of Education, Winneba, Ghana
| | - Yeboah Kwaku Opoku
- Department of Biology Education, Faculty of Science Education, University of Education, Winneba, Ghana
| | - Kwame Kumi Asare
- Infectious and Non-Communicable Diseases, Biomedical and Clinical Research Centre, University of Cape Coast, Cape Coast, Ghana
| | - Rosemary Doe Sorkpor
- Inspectorate Directorate, Food and Drugs Authority, Cape Coast P.O. Box CC13733, Ghana
| | - Ibrahim W. Naveh-Fio
- Department of Medical Laboratory Science, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Richard Armah
- Department of Medical Laboratory Science, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Sandra Ofori
- Department of Medical Laboratory Science, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Richard K. D. Ephraim
- Department of Medical Laboratory Science, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
25
|
Butucescu M, Imre M, Rus-Hrincu F, Voicu-Balasea B, Popa A, Moisa M, Ripszky A, Neculau C, Pituru SM, Pârvu S. Cell-Type-Specific ROS-AKT/mTOR-Autophagy Interplay-Should It Be Addressed in Periimplantitis? Diagnostics (Basel) 2024; 14:2784. [PMID: 39767145 PMCID: PMC11727345 DOI: 10.3390/diagnostics14242784] [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: 11/04/2024] [Accepted: 12/09/2024] [Indexed: 01/03/2025] Open
Abstract
Periimplantitis represents an inflammatory disease of the soft and hard tissues surrounding the osseointegrated dental implant, triggering progressive damage to the alveolar bone. Cumulative data have revealed that periimplantitis plays a crucial part in implant failure. Due to the strategic roles of autophagy and its upstream coordinator, the AKT/mTOR pathway, in inflammatory responses, the crosstalk between them in the context of periimplantitis should become a key research target, as it opens up an area of interesting data with clinical significance. Therefore, in this article, we aimed to briefly review the existing data concerning the complex roles played by ROS in the interplay between the AKT/mTOR signaling pathway and autophagy in periimplantitis, in each of the main cell types involved in periimplantitis pathogenesis and evolution. Knowing how to modulate specifically the autophagic machinery in each of the cellular types involved in the healing and osseointegration steps post implant surgery can help the clinician to make the most appropriate post-surgery decisions. These decisions might be crucial in order to prevent the occurrence of periimplantitis and ensure the proper conditions for effective osseointegration, depending on patients' clinical particularities.
Collapse
Affiliation(s)
- Mihai Butucescu
- Department of Organization, Professional Legislation and Management of the Dental Office, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Marina Imre
- Department of Prosthodontics, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Calea Plevnei, 010221 Bucharest, Romania;
| | - Florentina Rus-Hrincu
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Bianca Voicu-Balasea
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Alexandra Popa
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Mihai Moisa
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Alexandra Ripszky
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Cristina Neculau
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Silviu Mirel Pituru
- Department of Organization, Professional Legislation and Management of the Dental Office, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Simona Pârvu
- National Institute of Public Health, General Medicine Faculty, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
26
|
O'Reilly A, Zhao W, Wickström S, Arnér ESJ, Kiessling R. Reactive oxygen species: Janus-faced molecules in the era of modern cancer therapy. J Immunother Cancer 2024; 12:e009409. [PMID: 39645234 PMCID: PMC11629020 DOI: 10.1136/jitc-2024-009409] [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: 10/05/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024] Open
Abstract
Oxidative stress, that is, an unbalanced increase in reactive oxygen species (ROS), contributes to tumor-induced immune suppression and limits the efficacy of immunotherapy. Cancer cells have inherently increased ROS production, intracellularly through metabolic perturbations and extracellularly through activation of NADPH oxidases, which promotes cancer progression. Further increased ROS production or impaired antioxidant systems, induced, for example, by chemotherapy or radiotherapy, can preferentially kill cancer cells over healthy cells. Inflammatory cell-derived ROS mediate immunosuppressive effects of myeloid-derived suppressor cells and activated granulocytes, hampering antitumor effector cells such as T cells and natural killer (NK) cells. Cancer therapies modulating ROS levels in tumors may thus have entirely different consequences when targeting cancer cells versus immune cells. Here we discuss the possibility of developing more efficient cancer therapies based on reduction-oxidation modulation, as either monotherapies or in combination with immunotherapy. Short-term, systemic administration of antioxidants or drugs blocking ROS production can boost the immune system and act in synergy with immunotherapy. However, prolonged use of antioxidants can instead enhance tumor progression. Alternatives to systemic antioxidant administration are under development where gene-modified or activated T cells and NK cells are shielded ex vivo against the harmful effects of ROS before the infusion to patients with cancer.
Collapse
Affiliation(s)
- Aine O'Reilly
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, University College Cork, Cork, Ireland
- The Christie NHS Foundation Trust, Manchester, UK
| | - Wenchao Zhao
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Stina Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Patient area Head and Neck, Lung and Skin, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
Du Y, Wang L, Perez-Castro L, Conacci-Sorrell M, Sieber M. Non-cell autonomous regulation of cell-cell signaling and differentiation by mitochondrial ROS. J Cell Biol 2024; 223:e202401084. [PMID: 39535785 PMCID: PMC11561560 DOI: 10.1083/jcb.202401084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/14/2024] [Accepted: 08/13/2024] [Indexed: 11/16/2024] Open
Abstract
Mitochondrial reactive oxygen species (ROS) function intrinsically within cells to induce cell damage, regulate transcription, and cause genome instability. However, we know little about how mitochondrial ROS production non-cell autonomously impacts cell-cell signaling. Here, we show that mitochondrial dysfunction inhibits the plasma membrane localization of cell surface receptors that drive cell-cell communication during oogenesis. Within minutes, we found that mitochondrial ROS impairs exocyst membrane binding and leads to defective endosomal recycling. This endosomal defect impairs the trafficking of receptors, such as the Notch ligand Delta, during oogenesis. Remarkably, we found that overexpressing RAB11 restores ligand trafficking and rescues the developmental defects caused by ROS production. ROS production from adjacent cells acutely initiates a transcriptional response associated with growth and migration by suppressing Notch signaling and inducing extra cellualr matrix (ECM) remodeling. Our work reveals a conserved rapid response to ROS production that links mitochondrial dysfunction to the non-cell autonomous regulation of cell-cell signaling.
Collapse
Affiliation(s)
- Yipeng Du
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lei Wang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Matthew Sieber
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
28
|
Luo Y, Liu R, Yuan G, Pan Y. Polyphenols for stroke therapy: the role of oxidative stress regulation. Food Funct 2024; 15:11383-11399. [PMID: 39497601 DOI: 10.1039/d4fo01900h] [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: 11/26/2024]
Abstract
Stroke is associated with a high incidence and disability rate, which seriously endangers human health. Oxidative stress (OS) plays a crucial role in the underlying pathologic progression of cerebral damage in stroke. Emerging experimental studies suggest that polyphenols have antioxidant potential and express protective effects after different types of strokes, but no breakthrough has been achieved in clinical studies. Nanomaterials, due to small characteristic sizes, can be used to deliver drugs, and have shown excellent performance in the treatment of various diseases. The drug delivery capability of nanomaterials has significant implications for the clinical translation and application of polyphenols. This comprehensive review introduces the mechanism of oxidative stress in stroke, and also summarizes the antioxidant effects of polyphenols on reactive oxygen species generation and oxidative stress after stroke. Also, the application characteristics and research progress of nanomaterials in the treatment of stroke with antioxidants are presented.
Collapse
Affiliation(s)
- Yusong Luo
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Ruolan Liu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Guoqiang Yuan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yawen Pan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Academician Workstation, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
29
|
Yao M, Li M, Peng D, Wang Y, Li S, Zhang D, Yang B, Qiu HJ, Li LF. Unraveling Macrophage Polarization: Functions, Mechanisms, and "Double-Edged Sword" Roles in Host Antiviral Immune Responses. Int J Mol Sci 2024; 25:12078. [PMID: 39596148 PMCID: PMC11593441 DOI: 10.3390/ijms252212078] [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: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Numerous viruses that propagate through the respiratory tract may be initially engulfed by macrophages (Mφs) within the alveoli, where they complete their first replication cycle and subsequently infect the adjacent epithelial cells. This process can lead to significant pathological damage to tissues and organs, leading to various diseases. As essential components in host antiviral immune systems, Mφs can be polarized into pro-inflammatory M1 Mφs or anti-inflammatory M2 Mφs, a process involving multiple signaling pathways and molecular mechanisms that yield diverse phenotypic and functional features in response to various stimuli. In general, when infected by a virus, M1 macrophages secrete pro-inflammatory cytokines to play an antiviral role, while M2 macrophages play an anti-inflammatory role to promote the replication of the virus. However, recent studies have shown that some viruses may exhibit the opposite trend. Viruses have evolved various strategies to disrupt Mφ polarization for efficient replication and transmission. Notably, various factors, such as mechanical softness, the altered pH value of the endolysosomal system, and the homeostasis between M1/M2 Mφs populations, contribute to crucial events in the viral replication cycle. Here, we summarize the regulation of Mφ polarization, virus-induced alterations in Mφ polarization, and the antiviral mechanisms associated with these changes. Collectively, this review provides insights into recent advances regarding Mφ polarization in host antiviral immune responses, which will contribute to the development of precise prevention strategies as well as management approaches to disease incidence and transmission.
Collapse
Affiliation(s)
- Meng Yao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Meilin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Dingkun Peng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Yijing Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Ding Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Bo Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Lian-Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| |
Collapse
|
30
|
Li J, Xie J, Wang Y, Li X, Yang L, Zhao M, Chen C. Development of Biomaterials to Modulate the Function of Macrophages in Wound Healing. Bioengineering (Basel) 2024; 11:1017. [PMID: 39451393 PMCID: PMC11504998 DOI: 10.3390/bioengineering11101017] [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: 08/27/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Wound healing is a complex and precisely regulated process that encompasses multiple stages, including inflammation, anti-inflammation, and tissue repair. It involves various cells and signaling molecules, with macrophages demonstrating a significant degree of plasticity and playing a crucial regulatory role at different stages. In recent years, the use of biomaterials, which include both natural and synthetic polymers or macromolecules, has proliferated for the purpose of enhancing wound healing. This review summarizes how these diverse biomaterials promote wound healing by modulating macrophage behavior and examines the broader implications of these modulations. Additionally, we discuss the limitations associated with the clinical application of immunomodulatory biomaterials and propose potential solutions. Finally, we look towards future developments in the design of immunomodulatory biomaterials intended to enhance wound healing.
Collapse
Affiliation(s)
- Jiacheng Li
- Department of Plastic Surgery, The Second Affiliated Hospital, Dalian Medical University, Dalian 116041, China; (J.L.); (X.L.)
| | - Jiatong Xie
- The Second Clinical College, Dalian Medical University, Dalian 116044, China;
| | - Yaming Wang
- The First Affiliated Hospital, Dalian Medical University, Dalian 116014, China;
| | - Xixian Li
- Department of Plastic Surgery, The Second Affiliated Hospital, Dalian Medical University, Dalian 116041, China; (J.L.); (X.L.)
| | - Liqun Yang
- Research Center for Biomedical Materials, Engineering Research Center of Ministry, Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110022, China;
| | - Muxin Zhao
- Department of Plastic Surgery, The Second Affiliated Hospital, Dalian Medical University, Dalian 116041, China; (J.L.); (X.L.)
| | - Chaoxian Chen
- School of Materials Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, Peking University, Beijing 100871, China
| |
Collapse
|
31
|
Wang Q, Yin X, Huang X, Zhang L, Lu H. Impact of mitochondrial dysfunction on the antitumor effects of immune cells. Front Immunol 2024; 15:1428596. [PMID: 39464876 PMCID: PMC11502362 DOI: 10.3389/fimmu.2024.1428596] [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: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
Mitochondrial dysfunction, a hallmark of immune cell failure, affects the antitumor effects of immune cells through metabolic reprogramming, fission, fusion, biogenesis, and immune checkpoint signal transduction of mitochondria. According to researchers, restoring damaged mitochondrial function can enhance the efficacy of immune cells. Nevertheless, the mechanism of mitochondrial dysfunction in immune cells in patients with cancer is unclear. In this review, we recapitulate the impact of mitochondrial dysfunction on the antitumor effects of T cells, natural killer cells, dendritic cells, and tumor-associated macrophage and propose that targeting mitochondria can provide new strategies for antitumor therapy.
Collapse
Affiliation(s)
- Quan Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangzhi Yin
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Huang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haijun Lu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
32
|
Chen CJ, Wang HC, Hou YC, Wu YY, Shieh CC, Shan YS. Blocking M2-Like Macrophage Polarization Using Decoy Oligodeoxynucleotide-Based Gene Therapy Prevents Immune Evasion for Pancreatic Cancer Treatment. Mol Cancer Ther 2024; 23:1431-1445. [PMID: 38907533 PMCID: PMC11443249 DOI: 10.1158/1535-7163.mct-23-0767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/29/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
M2-like macrophages exhibit immunosuppressive activity and promote pancreatic cancer progression. Reactive oxygen species (ROS) affect macrophage polarization; however, the mechanism remains unclear. This study aimed to elucidate the underlying molecular basis and design a gene therapy to inhibit M2-like polarization. Microarray analysis and immunofluorescence staining were performed in M1-like and M2-like macrophages to ascertain the expression of CYBB, a major intracellular ROS source. Coculture assay and syngeneic orthotopic pancreatic cancer mice models were used to study the mechanism of M2-like skewing. Decoy oligodeoxynucleotides (ODNs) were designed to manipulate CYBB transcription to inhibit M2-like polarization and control tumor growth. Lipopolysaccharide treatment polarized U937 cells to M1-like macrophages in which CYBB expression was increased. In contrast, coculture with PANC-1 cells induced M2-like polarization in U937 cells with CYBB downregulation. High CD204 M2-like expression in combination with low CYBB expression was associated with the worst prognosis in patients with pancreatic cancer. STAT6 and HDAC2 in U937 cells were activated by cancer cell-derived IL4 after coculture and then bound to the CYBB promoter to repress CYBB expression, resulting in M2-like polarization. Diphenyleneiodonium, 8λ³-iodatricyclo[7.4.0.02,⁷]trideca-1(13),2,4,6,9,11-hexaen-8-ylium chloride that inhibits ROS production could block this action. Knockdown of STAT6 and HDAC2 also inhibited M2-like polarization and maintained the M1-like phenotype of U937 cells after coculture. Decoy ODNs interrupting the binding of STAT6 to the CYBB promoter counteracted M2-like polarization and tumor growth and triggered antitumor immunity in vivo. Gene therapy using STAT6-CYBB decoy ODNs can inhibit M2-like polarization, representing a potential therapeutic tool for pancreatic cancer.
Collapse
Affiliation(s)
- Chang-Jung Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Hao-Chen Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Medical Imaging Center, Innovation Headquarters, National Cheng Kung University, Tainan, Taiwan.
- Comparative Medicine Center, Innovation Headquarters, National Cheng Kung University, Tainan, Taiwan.
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Yi-Ying Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Chi-Chang Shieh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Comparative Medicine Center, Innovation Headquarters, National Cheng Kung University, Tainan, Taiwan.
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
33
|
Zhu W, Qiong D, Changzhi X, Meiyu J, Hui L. Macrophage polarization regulation shed lights on immunotherapy for CaOx kidney stone disease. Biomed Pharmacother 2024; 179:117336. [PMID: 39180792 DOI: 10.1016/j.biopha.2024.117336] [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: 05/27/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024] Open
Abstract
Kidney stone disease (KSD) is a major public health concern associated with high morbidity and recurrence, places a significant burden on the health care system worldwide. Calcium oxalate (CaOx) alone or a mixture of CaOx and calcium phosphate stones accounting for more than 80 % of cases. However, beyond surgical removal, the prevention and reduction of recurrence of CaOx kidney stones have always been a challenge. Given that macrophages are traditional innate immune cells that play critical roles in the clearance of pathogens and the maintenance of tissue homeostasis, which have gained more and more interests in nephrolithiasis. Several studies recently clearly demonstrated that M2-macrophage could reduce the renal calcium oxalate (CaOx) crystal acumination, and provide premise insights and therapeutic options for KSD by modulating the macrophage phenotypes. However, the mechanism of macrophage-polarization regulation and that effects on kidney stone prevention and treatments are far from clear. Here, we comprehensively reviewed the literatures related to cytokines, epigenetic modifications and metabolic reprograming of macrophage in CaOx kidney stone disease, aimed to provide better understandings on macrophage polarization regulation as well as its potential clinical applications in CaOx kidney stone disease treatments and prevention.
Collapse
Affiliation(s)
- Wang Zhu
- Department of Urology, The People's Hospital of Longhua, Shenzhen 518109, Guangdong, China.
| | - Deng Qiong
- Department of Urology, The People's Hospital of Longhua, Shenzhen 518109, Guangdong, China
| | - Xu Changzhi
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jin Meiyu
- Department of Urology, The People's Hospital of Longhua, Shenzhen 518109, Guangdong, China
| | - Liang Hui
- Department of Urology, The People's Hospital of Longhua, Shenzhen 518109, Guangdong, China.
| |
Collapse
|
34
|
Chen HR, Sun Y, Mittler G, Rumpf T, Shvedunova M, Grosschedl R, Akhtar A. MOF-mediated PRDX1 acetylation regulates inflammatory macrophage activation. Cell Rep 2024; 43:114682. [PMID: 39207899 DOI: 10.1016/j.celrep.2024.114682] [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: 10/09/2023] [Revised: 06/27/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Signaling-dependent changes in protein phosphorylation are critical to enable coordination of transcription and metabolism during macrophage activation. However, the role of acetylation in signal transduction during macrophage activation remains obscure. Here, we identify the redox signaling regulator peroxiredoxin 1 (PRDX1) as a substrate of the lysine acetyltransferase MOF. MOF acetylates PRDX1 at lysine 197, preventing hyperoxidation and thus maintaining its activity under stress. PRDX1 K197ac responds to inflammatory signals, decreasing rapidly in mouse macrophages stimulated with bacterial lipopolysaccharides (LPSs) but not with interleukin (IL)-4 or IL-10. The LPS-induced decrease of PRDX1 K197ac elevates cellular hydrogen peroxide accumulation and augments ERK1/2, but not p38 or AKT, phosphorylation. Concomitantly, diminished PRDX1 K197ac stimulates glycolysis, potentiates H3 serine 28 phosphorylation, and ultimately enhances the production of pro-inflammatory mediators such as IL-6. Our work reveals a regulatory role for redox protein acetylation in signal transduction and coordinating metabolic and transcriptional programs during inflammatory macrophage activation.
Collapse
Affiliation(s)
- Hui-Ru Chen
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany; Albert-Ludwigs-University Freiburg, Faculty of Biology, Freiburg, Baden-Württemberg, Germany
| | - Yidan Sun
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Gerhard Mittler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Tobias Rumpf
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Maria Shvedunova
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Rudolf Grosschedl
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Asifa Akhtar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany.
| |
Collapse
|
35
|
Luo Z, Jiang M, Cheng N, Zhao X, Liu H, Wang S, Lin Q, Huang J, Guo X, Liu X, Shan X, Lu Y, Shi Y, Luo L, You J. Remodeling the hepatic immune microenvironment and demolishing T cell traps to enhance immunotherapy efficacy in liver metastasis. J Control Release 2024; 373:890-904. [PMID: 39067794 DOI: 10.1016/j.jconrel.2024.07.057] [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: 12/04/2023] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Immune checkpoint inhibitors (ICIs) exhibit compromised therapeutic efficacy in many patients with advanced cancers, particularly those with liver metastases. Much of this incapability can be ascribed as an irresponsiveness resulting from the "cold" hepatic tumor microenvironment that acts as T cell "traps" for which there currently lack countermeasures. We report a novel nanomedicine that converts the hepatic immune microenvironment to a "hot" phenotype by targeting hepatic macrophage-centric T cell elimination. Using the nanomedicine, composed of KIRA6 (an endothelium reticulum stress inhibitor), α-Tocopherol nanoemulsions, and anti-PD1 antibodies, we found its potency in murine models of orthotopic colorectal tumors and hepatic metastases, restoring immune responses and enhancing anti-tumor effects. A post-treatment scrutiny of the immune microenvironment landscape in the liver reveals repolarization of immunosuppressive hepatic macrophages, upregulation of Th1-like effector CD4+ T cells, and rejuvenation of dendritic cells along with CD8+ T cells. These findings suggest adaptations of liver-centric immune milieu modulation strategies to improve the efficacy of ICIs for a variety of "cold" tumors and their liver metastases.
Collapse
Affiliation(s)
- Zhenyu Luo
- School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Mengshi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Ningtao Cheng
- School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; Hangzhou Yuhang BoYu Intelligent Health Innovation Lab, Hangzhou, Zhejiang 311121, China.
| | - Xiaoqi Zhao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Huihui Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Qing Lin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jiaxin Huang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xuemeng Guo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xinyu Shan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
36
|
Wang H, Yung MM, Xuan Y, Chen F, Chan W, Siu MK, Long R, Jia S, Liang Y, Xu D, Song Z, Tsui SK, Ngan HY, Chan KK, Chan DW. Polyunsaturated fatty acids promote M2-like TAM deposition via dampening RhoA-YAP1 signaling in the ovarian cancer microenvironment. Exp Hematol Oncol 2024; 13:90. [PMID: 39198883 PMCID: PMC11360340 DOI: 10.1186/s40164-024-00558-8] [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: 10/11/2023] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Peritoneal metastases frequently occur in epithelial ovarian cancer (EOC), resulting in poor prognosis and survival rates. Tumor-associated-macrophages (TAMs) massively infiltrate into ascites spheroids and are multi-polarized as protumoral M2-like phenotype, orchestrating the immunosuppression and promoting tumor progression. However, the impact of omental conditioned medium/ascites (OCM/AS) on TAM polarization and its function in tumor progression remains elusive. METHODS The distribution and polarization of TAMs in primary and omental metastatic EOC patients' tumors and ascites were examined by m-IHC, FACS analysis, and immunofluorescence. QPCR, immunofluorescence, FACS analysis, lipid staining assay, ROS assay, and Seahorse real-time cell metabolic assay characterized TAMs as being polarized in the ascites microenvironment. The oncogenic role of TAMs in tumor cells was demonstrated by co-cultured migration/invasion, proliferation, and spheroid formation assays. Mechanistic studies of the regulations of TAM polarization were performed by using RNA-Seq, GTPase pull-down, G-LISA activation assays, and other biochemical assays. A Yap1 macrophages (MФs) conditional knockout (cKO) mouse model demonstrated the roles of YAP1 in TAM polarization status and its pro-metastatic function. Finally, the anti-metastatic potential of targeting TAMs through restoring YAP1 by pharmacological agonist XMU MP1 was demonstrated in vitro and in vivo. RESULTS Abundant polyunsaturated fatty acids (PUFAs) in OCM/AS suppressed RhoA-GTPase activities, which, in turn, downregulated nuclear YAP1 in MФs, leading to increased protumoral TAM polarization accompanied by elevated OXPHOS metabolism. Abolishment of YAP1 in MФs further confirmed that a higher M2/M1 ratio of TAM polarization could alleviate CD8+ T cell infiltration and cytotoxicity in vivo. Consistently, the loss of YAP1 has been observed in EOC metastatic tissues, suggesting its clinical relevance. On the contrary, restoration of YAP1 expression by pharmaceutical inhibition of MST1/2 induced conversion of M2-to-M1-like polarized MФs, elevating the infiltration of CD8+ T cells and attenuating tumor growth. CONCLUSION This study revealed that PUFAs-enriched OCM/AS of EOC promotes M2-like TAM polarization through RhoA-YAP1 inhibition, where YAP1 downregulation is required for accelerating protumoral M2-like TAM polarization, thereby causing immunosuppression and enhancing tumor progression. Conversion of M2-to-M1-like polarized MФs through Yap1 activation inhibits tumor progression and contributes to developing potential TAMs-targeted immunotherapies in combating EOC peritoneal metastases.
Collapse
Affiliation(s)
- Huogang Wang
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, P.R. China
| | - Mingo Mh Yung
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Yang Xuan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Fushun Chen
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Waisun Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Michelle Ky Siu
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Runying Long
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Shuo Jia
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P.R. China
| | - Yonghao Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P.R. China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, P.R. China
| | - Stephen Kw Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Hextan Ys Ngan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Karen Kl Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China.
| | - David W Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China.
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China.
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, P.R. China.
| |
Collapse
|
37
|
Nheu D, Petratos S. How does Nogo-A signalling influence mitochondrial function during multiple sclerosis pathogenesis? Neurosci Biobehav Rev 2024; 163:105767. [PMID: 38885889 DOI: 10.1016/j.neubiorev.2024.105767] [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: 03/17/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
Multiple sclerosis (MS) is a severe neurological disorder that involves inflammation in the brain, spinal cord and optic nerve with key disabling neuropathological outcomes being axonal damage and demyelination. When degeneration of the axo-glial union occurs, a consequence of inflammatory damage to central nervous system (CNS) myelin, dystrophy and death can lead to large membranous structures from dead oligodendrocytes and degenerative myelin deposited in the extracellular milieu. For the first time, this review covers mitochondrial mechanisms that may be operative during MS-related neurodegenerative changes directly activated during accumulating extracellular deposits of myelin associated inhibitory factors (MAIFs), that include the potent inhibitor of neurite outgrowth, Nogo-A. Axonal damage may occur when Nogo-A binds to and signals through its cognate receptor, NgR1, a multimeric complex, to initially stall axonal transport and limit the delivery of important growth-dependent cargo and subcellular organelles such as mitochondria for metabolic efficiency at sites of axo-glial disintegration as a consequence of inflammation. Metabolic efficiency in axons fails during active demyelination and progressive neurodegeneration, preceded by stalled transport of functional mitochondria to fuel axo-glial integrity.
Collapse
Affiliation(s)
- Danica Nheu
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia
| | - Steven Petratos
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia.
| |
Collapse
|
38
|
Liu H, Huang M, Xin D, Wang H, Yu H, Pu W. Natural products with anti-tumorigenesis potential targeting macrophage. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155794. [PMID: 38875811 DOI: 10.1016/j.phymed.2024.155794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/06/2024] [Accepted: 05/30/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Inflammation is a risk factor for tumorigenesis. Macrophage, a subset of immune cells with high plasticity, plays a multifaceted role in this process. Natural products, which are bioactive compounds derived from traditional herbs or foods, have exhibited diverse effects on macrophages and tumorigenesis making them a valuable resource of drug discovery or optimization in tumor prevention. PURPOSE Provide a comprehensive overview of the various roles of macrophages in tumorigenesis, as well as the effects of natural products on tumorigenesis by modulating macrophage function. METHODS A thorough literature search spanning the past two decades was carried out using PubMed, Web of Science, Elsevier, and CNKI following the PRISMA guidelines. The search terms employed included "macrophage and tumorigenesis", "natural products, macrophages and tumorigenesis", "traditional Chinese medicine and tumorigenesis", "natural products and macrophage polarization", "macrophage and tumor related microenvironment", "macrophage and tumor signal pathway", "toxicity of natural products" and combinations thereof. Furthermore, certain articles are identified through the tracking of citations from other publications or by accessing the websites of relevant journals. Studies that meet the following criteria are excluded: (1) Articles not written in English or Chinese; (2) Full texts were not available; (3) Duplicate articles and irrelevant studies. The data collected was organized and summarized based on molecular mechanisms or compound structure. RESULTS This review elucidates the multifaceted effect of macrophages on tumorigenesis, encompassing process such as inflammation, angiogenesis, and tumor cell invasion by regulating metabolism, non-coding RNA, signal transduction and intercellular crosstalk. Natural products, including vitexin, ovatodiolide, ligustilide, and emodin, as well as herbal remedies, have demonstrated efficacy in modulating macrophage function, thereby attenuating tumorigenesis. These interventions mainly focus on mitigating the initial inflammatory response or modifying the inflammatory environment within the precancerous niche. CONCLUSIONS These mechanistic insights of macrophages in tumorigenesis offer valuable ideas for researchers. The identified natural products facilitate the selection of promising candidates for future cancer drug development.
Collapse
Affiliation(s)
- Hao Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Manru Huang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Dandan Xin
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Hong Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Weiling Pu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
39
|
Carter TH, Weyer-Nichols CE, Garcia-Sanchez JI, Wilson K, Nagarkatti P, Nagarkatti M. Delta-9-Tetrahydrocannabinol Blocks Bone Marrow-Derived Macrophage Differentiation through Elimination of Reactive Oxygen Species. Antioxidants (Basel) 2024; 13:887. [PMID: 39199132 PMCID: PMC11352128 DOI: 10.3390/antiox13080887] [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: 06/05/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Macrophages are vital components of the immune system and serve as the first line of defense against pathogens. Macrophage colony-stimulating factor (M-CSF) induces macrophage differentiation from bone marrow-derived cells (BMDCs). Δ9-tetrahydrocannabiol (THC), a phytocannabinoid from the Cannabis plant, has profound anti-inflammatory properties with significant effects on myeloid cells. To investigate the effect of THC on macrophage differentiation, we cultured BMDCs with M-CSF in the presence of THC. Interestingly, THC markedly blocked the differentiation of BMDCs into CD45 + CD11b + F4/80+ macrophages. The effect of THC was independent of cannabinoid receptors CB1, and CB2, as well as other potential receptors such as GPR18, GPR55, and Adenosine 2A Receptor. RNA-seq analysis revealed that the THC-treated BMDCs displayed a significant increase in the expression of NRF2-ARE-related genes. KEGG pathway analysis revealed that the expression profiles of THC-treated cells correlated with ferroptosis and glutathione metabolism pathways. Fluorescence-based labile iron assays showed that the THC-treated BMDCs had significantly increased iron levels. Finally, THC-exposed BMDCs showed decreased levels of intracellular ROS. THC has the unique molecular property to block the Fenton Reaction, thus preventing the increase in intracellular ROS that is normally induced by high iron levels. Together, these studies demonstrated that THC blocks M-CSF-induced macrophage differentiation by inhibiting ROS production through both the induction of NRF2-ARE-related gene expression and the prevention of ROS formation via the Fenton Reaction.
Collapse
Affiliation(s)
| | | | | | | | | | - Mitzi Nagarkatti
- Department of Pathology Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA; (T.H.C.); (C.E.W.-N.); (J.I.G.-S.); (K.W.); (P.N.)
| |
Collapse
|
40
|
Zheng G, Yu W, Xu Z, Yang C, Wang Y, Yue Z, Xiao Q, Zhang W, Wu X, Zang F, Wang J, Wang L, Yuan WE, Hu B, Chen H. Neuroimmune modulating and energy supporting nanozyme-mimic scaffold synergistically promotes axon regeneration after spinal cord injury. J Nanobiotechnology 2024; 22:399. [PMID: 38970101 PMCID: PMC11225227 DOI: 10.1186/s12951-024-02594-2] [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: 11/28/2023] [Accepted: 05/28/2024] [Indexed: 07/07/2024] Open
Abstract
Spinal cord injury (SCI) represents a profound central nervous system affliction, resulting in irreversibly compromised daily activities and disabilities. SCI involves excessive inflammatory responses, which are characterized by the existence of high levels of proinflammatory M1 macrophages, and neuronal mitochondrial energy deficit, exacerbating secondary damage and impeding axon regeneration. This study delves into the mechanistic intricacies of SCI, offering insights from the perspectives of neuroimmune regulation and mitochondrial function, leading to a pro-fibrotic macrophage phenotype and energy-supplying deficit. To address these challenges, we developed a smart scaffold incorporating enzyme mimicry nanoparticle-ceriumoxide (COPs) into nanofibers (NS@COP), which aims to pioneer a targeted neuroimmune repair strategy, rescuing CGRP receptor on macrophage and concurrently remodeling mitochondrial function. Our findings indicate that the integrated COPs restore the responsiveness of pro-inflammatory macrophages to calcitonin gene-related peptide (CGRP) signal by up-regulating receptor activity modifying protein 1 (RAMP1), a vital component of the CGRP receptor. This promotes macrophage fate commitment to an anti-inflammatory pro-resolution M2 phenotype, then alleviating glial scar formation. In addition, NS@COP implantation also protected neuronal mitochondrial function. Collectively, our results suggest that the strategy of integrating nanozyme COP nanoparticles into a nanofiber scaffold provides a promising therapeutic candidate for spinal cord trauma via rational regulation of neuroimmune communication and mitochondrial function.
Collapse
Affiliation(s)
- Genjiang Zheng
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Wei Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Ministry of Education, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
- Inner Mongolia Research Institute of Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zeng Xu
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Chen Yang
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Yunhao Wang
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Zhihao Yue
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Qiangqiang Xiao
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Wenyu Zhang
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Xiaodong Wu
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Fazhi Zang
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Jianxi Wang
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Lei Wang
- Division of Orthopaedics and Traumatology, Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. Guangzhou North Road, Guangzhou, 510515, China
| | - Wei-En Yuan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China.
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China.
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Ministry of Education, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China.
- Inner Mongolia Research Institute of Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Bo Hu
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China.
| | - Huajiang Chen
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
41
|
Yang Y, Li Z, Fan X, Jiang C, Wang J, Rastegar-Kashkooli Y, Wang TJ, Wang J, Wang M, Cheng N, Yuan X, Chen X, Jiang B, Wang J. Nanozymes: Potential Therapies for Reactive Oxygen Species Overproduction and Inflammation in Ischemic Stroke and Traumatic Brain Injury. ACS NANO 2024; 18:16450-16467. [PMID: 38897929 DOI: 10.1021/acsnano.4c03425] [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: 06/21/2024]
Abstract
Nanozymes, which can selectively scavenge reactive oxygen species (ROS), have recently emerged as promising candidates for treating ischemic stroke and traumatic brain injury (TBI) in preclinical models. ROS overproduction during the early phase of these diseases leads to oxidative brain damage, which has been a major cause of mortality worldwide. However, the clinical application of ROS-scavenging enzymes is limited by their short in vivo half-life and inability to cross the blood-brain barrier. Nanozymes, which mimic the catalytic function of natural enzymes, have several advantages, including cost-effectiveness, high stability, and easy storage. These advantages render them superior to natural enzymes for disease diagnosis and therapeutic interventions. This review highlights recent advancements in nanozyme applications for ischemic stroke and TBI, emphasizing their potential to mitigate the detrimental effect of ROS overproduction, oxidative brain damage, inflammation, and blood-brain barrier compromise. Therefore, nanozymes represent a promising treatment modality for ROS overproduction conditions in future medical practices.
Collapse
Affiliation(s)
- Yunfan Yang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Zixiang Li
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
- School of International Education, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Tom J Wang
- Program in Behavioral Biology, The Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Nannan Cheng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xiqian Yuan
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| |
Collapse
|
42
|
Paduch R, Klatka M, Pieniądz P, Wertel I, Pawłowska A, Klatka J. Reciprocal Interactions of Human Monocytes and Cancer Cells in Co-Cultures In Vitro. Curr Issues Mol Biol 2024; 46:6836-6852. [PMID: 39057050 PMCID: PMC11276568 DOI: 10.3390/cimb46070408] [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: 06/04/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) includes immune and stromal cells and noncellular extracellular matrix (ECM) components. Tumor-associated macrophages (TAMs) are the most important immune cells in TME and are crucial for carcinomas' progression. The purpose was to analyze direct and indirect interactions in co-culture of tumor cells with monocytes/macrophages and, additionally, to indicate which interactions are more important for cancer development. Cytokines, reactive oxygen species, nitric oxide level, tumor cell cycle and changes in tumor cell morphology after human tumor cells (Hep-2 and RK33 cell lines) with human monocyte/macrophage (THP-1 cell line) interactions were tested. Morphology and cytoskeleton organization of tumor cells did not change after co-culture with macrophages. In co-culture of tumor cells with human monocyte, changes in the percentage of tumor cells in cell cycle phases was observed. No significant changes in reactive oxygen species (ROS) were found in the co-culture as compared to the tumor cell mono-culture. Monocytes produced about three times higher ROS than tumor cells. In co-cultures, a lower nitric oxide (NOx) level was found as compared to the sum of the production by both mono-cultures. Co-culture conditions limited the production of cytokines (IL-4, IL-10 and IL-13) as compared to the sum of their level in mono-cultures. In conclusion, macrophages influence tumor cell growth and functions. Mutual (direct and paracrine) interactions between tumor cells and macrophages changed cytokine production and tumor cell cycle profile. The data obtained may allow us to initially indicate which kind of interactions may have a greater impact on cancer development processes.
Collapse
Affiliation(s)
- Roman Paduch
- Department of Virology and Immunology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland;
- Department of General and Paediatric Ophthalmology, Medical University of Lublin, Chmielna 1, 20-079 Lublin, Poland
| | - Maria Klatka
- Department of Paediatric Endocrinology and Diabetology, Medical University, Gębali 1, 20-093 Lublin, Poland;
| | - Paulina Pieniądz
- Department of Virology and Immunology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland;
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland; (I.W.); (A.P.)
| | - Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland; (I.W.); (A.P.)
| | - Janusz Klatka
- Department of Otolaryngology and Laryngological Oncology, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland;
| |
Collapse
|
43
|
Bianconi S, Leppik L, Oppermann E, Marzi I, Henrich D. Direct Current Electrical Stimulation Shifts THP-1-Derived Macrophage Polarization towards Pro-Regenerative M2 Phenotype. Int J Mol Sci 2024; 25:7272. [PMID: 39000377 PMCID: PMC11242703 DOI: 10.3390/ijms25137272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
A macrophage shift from the M1 to the M2 phenotype is relevant for promoting tissue repair and regeneration. In a previous in vivo study, we found that direct current (DC) electrical stimulation (EStim) increased the proportion of M2 macrophages in healing tissues and directed the balance of the injury response away from healing/scarring towards regeneration. These observations led us to hypothesize that DC EStim regulates macrophage polarization towards an M2 phenotype. THP-1-derived M0, M1 (IFN-γ and LPS), and M2 (IL-4 and IL-13) macrophages were exposed (or not: control group) to 100 mV/mm of DC EStim, 1 h/day for three days. Macrophage polarization was assessed through gene and surface marker expressions and cytokine secretion profiles. Following DC EStim treatment, M0 cells exhibited an upregulation of M2 marker genes IL10, CD163, and PPARG. In M1 cells, DC EStim upregulated the gene expressions of M2 markers IL10, TGM2, and CD206 and downregulated M1 marker gene CD86. EStim treatment also reduced the surface expression of CD86 and secretion of pro-inflammatory cytokines IL-1β and IL-6. Our results suggest that DC EStim differentially exerts pro-M2 effects depending on the macrophage phenotype: it upregulates typical M2 genes in M0 and M1 cells while inhibiting M1 marker CD86 at the nuclear and protein levels and the secretion of pro-inflammatory interleukins in M1 cells. Conversely, M2 cells appear to be less responsive to the EStim treatment employed in this study.
Collapse
Affiliation(s)
- Santiago Bianconi
- Department of Trauma Surgery and Orthopedics, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Liudmila Leppik
- Department of Trauma Surgery and Orthopedics, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Elsie Oppermann
- Department of General, Visceral, Transplant and Thoracic Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Ingo Marzi
- Department of Trauma Surgery and Orthopedics, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Dirk Henrich
- Department of Trauma Surgery and Orthopedics, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| |
Collapse
|
44
|
Zhu X, Wang B, Yu H, Li C, Zhao Y, Zhong Y, Tang W, Zhou Y, Huang X, Zhu H, Wu Y, Yang K, Wei Y, Gao Z, Dong J. Icariin attenuates asthmatic airway inflammation via modulating alveolar macrophage activation based on network pharmacology and in vivo experiments. J Gene Med 2024; 26:e3718. [PMID: 38979822 DOI: 10.1002/jgm.3718] [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: 09/07/2023] [Revised: 03/23/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Icariin (ICA) inhibits inflammatory response in various diseases, but the mechanism underlying ICA treating airway inflammation in asthma needs further understood. We aimed to predict and validate the potential targets of ICA against asthma-associated airway inflammation using network pharmacology and experiments. METHODS The ovalbumin-induced asthma-associated airway inflammation mice model was established. The effects of ICA were evaluated by behavioral, airway hyperresponsiveness, lung pathological changes, inflammatory cell and cytokines counts. Next, the corresponding targets of ICA were mined via the SEA, CTD, HERB, PharmMapper, Symmap database and the literature. Pubmed-Gene and GeneCards databases were used to screen asthma and airway inflammation-related targets. The overlapping targets were used to build an interaction network, analyze gene ontology and enrich pathways. Subsequently, flow cytometry, quantitative real-time PCR and western blotting were employed for validation. RESULTS ICA alleviated the airway inflammation of asthma; 402 targets of ICA, 5136 targets of asthma and 4531 targets of airway inflammation were screened; 216 overlapping targets were matched and predicted ICA possesses the potential to modulate asthmatic airway inflammation by macrophage activation/polarization. Additionally, ICA decreased M1 but elevated M2. Potential targets that were disrupted by asthma inflammation were restored by ICA treatment. CONCLUSIONS ICA alleviates airway inflammation in asthma by inhibiting the M1 polarization of alveolar macrophages, which is related to metabolic reprogramming. Jun, Jak2, Syk, Tnf, Aldh2, Aldh9a1, Nos1, Nos2 and Nos3 represent potential targets of therapeutic intervention. The present study enhances understanding of the anti-airway inflammation effects of ICA, especially in asthma.
Collapse
Affiliation(s)
- Xiaofei Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Bin Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Congcong Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yuhang Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yuanyuan Zhong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Huahe Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yueren Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Kai Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhen Gao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Wang J, Zhang Y, Tang Q, Zhang Y, Yin Y, Chen L. Application of Antioxidant Compounds in Bone Defect Repair. Antioxidants (Basel) 2024; 13:789. [PMID: 39061858 PMCID: PMC11273992 DOI: 10.3390/antiox13070789] [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: 05/07/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Bone defects caused by trauma, tumor resection, and infections are significant clinical challenges. Excessive reactive oxygen species (ROS) usually accumulate in the defect area, which may impair the function of cells involved in bone formation, posing a serious challenge for bone repair. Due to the potent ROS scavenging ability, as well as potential anti-inflammatory and immunomodulatory activities, antioxidants play an indispensable role in the maintenance and protection of bone health and have gained increasing attention in recent years. This narrative review aims to give an overview of the main research directions on the application of antioxidant compounds in bone defect repair over the past decade. In addition, the positive effects of various antioxidants and their biomaterial delivery systems in bone repair are summarized to provide new insights for exploring antioxidant-based strategies for bone defect repair.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yubing Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China
| | - Ying Yin
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
46
|
Mao X, Tang X, Ye J, Xu S, Wang Y, Liu X, Wu Q, Lin X, Zhang M, Liu J, Yang J, Sun P. Multi-omics profiling reveal cells with novel oncogenic cluster, TRAP1 low/CAMSAP3 low, emerge more aggressive behavior and poor-prognosis in early-stage endometrial cancer. Mol Cancer 2024; 23:127. [PMID: 38880903 PMCID: PMC11181528 DOI: 10.1186/s12943-024-02039-2] [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: 05/03/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
The clinical heterogeneity of early-stage endometrial cancer (EC) is worthy of further study to identify high-quality prognostic markers and their potential role in aggressive tumor behavior. Mutation of TP53 was considered as an important primary triage in modified molecular typing for EC, it still cannot precisely predict the prognosis of EC. After proteomic analysis of cancer and para-cancerous tissues from 24 early-stage endometrioid EC patients with different survival outcomes, 13 differentially expressed proteins were screen out while 2 proteins enriched in p53 signaling pathway were further identified by single-cell transcriptome (scRNA-seq). Interestingly, tumor necrosis factor type-1 receptor-associated protein (TRAP1) and calmodulin-regulated spectrin-associated protein family member 3 (CAMSAP3) were found to be significantly downregulated in the specific cell cluster. Expectedly, the signature genes of TRAP1low/CAMSAP3low cluster included classical oncogenes. Moreover, close cellular interactions were observed between myeloid cells and the TRAP1low/CAMSAP3low cluster after systematically elucidating their relationship with tumor microenvironment (TME). The expression of TRAP1 and CAMSAP3 was verified by immunohistochemistry. Thus, a novel prediction model combining TRAP1, CAMSAP3 and TP53 was construct by multi-omics. Compared with the area under the curve, it demonstrated a significantly improvemrnt in the diagnostic efficacy in EC patients from TCGA bank. In conclusion, this work improved the current knowledge regarding the prognosis of early-stage EC through proteomics and scRNA-seq. These findings may lead to improvements in precise risk stratification of early-stage EC patients.
Collapse
Affiliation(s)
- Xiaodan Mao
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Xiaoyue Tang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Jingxuan Ye
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Shuxia Xu
- Pathology Department, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Yue Wang
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Xianhua Liu
- Pathology Department, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Qibin Wu
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Xite Lin
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Maotong Zhang
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Jiangfeng Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Juntao Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China.
| |
Collapse
|
47
|
Tran N, Mills EL. Redox regulation of macrophages. Redox Biol 2024; 72:103123. [PMID: 38615489 PMCID: PMC11026845 DOI: 10.1016/j.redox.2024.103123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024] Open
Abstract
Redox signaling, a mode of signal transduction that involves the transfer of electrons from a nucleophilic to electrophilic molecule, has emerged as an essential regulator of inflammatory macrophages. Redox reactions are driven by reactive oxygen/nitrogen species (ROS and RNS) and redox-sensitive metabolites such as fumarate and itaconate, which can post-translationally modify specific cysteine residues in target proteins. In the past decade our understanding of how ROS, RNS, and redox-sensitive metabolites control macrophage function has expanded dramatically. In this review, we discuss the latest evidence of how ROS, RNS, and metabolites regulate macrophage function and how this is dysregulated with disease. We highlight the key tools to assess redox signaling and important questions that remain.
Collapse
Affiliation(s)
- Nhien Tran
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
48
|
Pinello N, Song R, Lee Q, Calonne E, Duan KL, Wong E, Tieng J, Mehravar M, Rong B, Lan F, Roediger B, Ma CJ, Yuan BF, Rasko JEJ, Larance M, Ye D, Fuks F, Wong JJL. Dynamic changes in RNA m 6A and 5 hmC influence gene expression programs during macrophage differentiation and polarisation. Cell Mol Life Sci 2024; 81:229. [PMID: 38780787 PMCID: PMC11116364 DOI: 10.1007/s00018-024-05261-9] [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: 02/06/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
RNA modifications are essential for the establishment of cellular identity. Although increasing evidence indicates that RNA modifications regulate the innate immune response, their role in monocyte-to-macrophage differentiation and polarisation is unclear. While m6A has been widely studied, other RNA modifications, including 5 hmC, remain poorly characterised. We profiled m6A and 5 hmC epitranscriptomes, transcriptomes, translatomes and proteomes of monocytes and macrophages at rest and pro- and anti-inflammatory states. Transcriptome-wide mapping of m6A and 5 hmC reveals enrichment of m6A and/or 5 hmC on specific categories of transcripts essential for macrophage differentiation. Our analyses indicate that m6A and 5 hmC modifications are present in transcripts with critical functions in pro- and anti-inflammatory macrophages. Notably, we also discover the co-occurrence of m6A and 5 hmC on alternatively-spliced isoforms and/or opposing ends of the untranslated regions (UTR) of mRNAs with key roles in macrophage biology. In specific examples, RNA 5 hmC controls the decay of transcripts independently of m6A. This study provides (i) a comprehensive dataset to interrogate the role of RNA modifications in a plastic system (ii) a resource for exploring different layers of gene expression regulation in the context of human monocyte-to-macrophage differentiation and polarisation, (iii) new insights into RNA modifications as central regulators of effector cells in innate immunity.
Collapse
Affiliation(s)
- Natalia Pinello
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, 11400, Montevideo, Uruguay
| | - Renhua Song
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
| | - Quintin Lee
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
| | - Emilie Calonne
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB Cancer Research Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Kun-Long Duan
- The Molecular and Cell Biology Lab, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Emilie Wong
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
| | - Jessica Tieng
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
| | - Majid Mehravar
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
| | - Bowen Rong
- Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Fei Lan
- Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ben Roediger
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Skin Inflammation Group, Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
- Autoimmunity, Transplantation and Inflammation (ATI) Disease Area, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Cheng-Jie Ma
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Bi-Feng Yuan
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - John E J Rasko
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Gene and Stem Cell Therapy Program, Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Camperdown, 2050, NSW, Australia
| | - Mark Larance
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
- Charles Perkins Centre, School of Medical Sciences, University of Sydney, Sydney, 2006, Australia
| | - Dan Ye
- The Molecular and Cell Biology Lab, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - François Fuks
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB Cancer Research Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Justin J-L Wong
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia.
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia.
- Charles Perkins Centre, School of Medical Sciences, University of Sydney, Sydney, 2006, Australia.
| |
Collapse
|
49
|
Li X, Gao J, Wu C, Wang C, Zhang R, He J, Xia ZJ, Joshi N, Karp JM, Kuai R. Precise modulation and use of reactive oxygen species for immunotherapy. SCIENCE ADVANCES 2024; 10:eadl0479. [PMID: 38748805 PMCID: PMC11095489 DOI: 10.1126/sciadv.adl0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Reactive oxygen species (ROS) play an important role in regulating the immune system by affecting pathogens, cancer cells, and immune cells. Recent advances in biomaterials have leveraged this mechanism to precisely modulate ROS levels in target tissues for improving the effectiveness of immunotherapies in infectious diseases, cancer, and autoimmune diseases. Moreover, ROS-responsive biomaterials can trigger the release of immunotherapeutics and provide tunable release kinetics, which can further boost their efficacy. This review will discuss the latest biomaterial-based approaches for both precise modulation of ROS levels and using ROS as a stimulus to control the release kinetics of immunotherapeutics. Finally, we will discuss the existing challenges and potential solutions for clinical translation of ROS-modulating and ROS-responsive approaches for immunotherapy, and provide an outlook for future research.
Collapse
Affiliation(s)
- Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jingjing Gao
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomedical Engineering, Material Science and Engineering Graduate Program and The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Chengcheng Wu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Chaoyu Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ruoshi Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ziting Judy Xia
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nitin Joshi
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey M. Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
50
|
Mao Y, Wu C, Wang X, Zhang F, Qi X, Li X, Li P, Tang B. Fluorescence imaging sheds light on the immune evasion mechanisms of hepatic stellate cells mediated by superoxide anion. Commun Biol 2024; 7:558. [PMID: 38730013 PMCID: PMC11087649 DOI: 10.1038/s42003-024-06245-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: 12/31/2022] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
Whether and how the reactive oxygen species generated by hepatic stellate cells (HSCs) promote immune evasion of hepatocellular carcinoma (HCC) remains mysterious. Therefore, investigating the function of superoxide anion (O2•-), the firstly generated reactive oxygen species, during the immune evasion become necessary. In this work, we establish a novel in situ imaging method for visualization of O2•- changes in HSCs based on a new two-photon fluorescence probe TPH. TPH comprises recognition group for O2•- and HSCs targeting peptides. We observe that O2•- in HSCs gradually rose, impairing the infiltration of CD8+ T cells in HCC mice. Further studies reveal that the cyclin-dependent kinase 4 is deactivated by O2•-, and then cause the up-regulation of PD-L1. Our work provides molecular insights into HSC-mediated immune evasion of HCC, which may represent potential targets for HCC immunotherapy.
Collapse
Affiliation(s)
- Yuantao Mao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Chuanchen Wu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China.
| | - Fanghui Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Xinru Qi
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Xia Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China.
- Laoshan Laboratory, 168 Wenhai Middle Rd, Aoshanwei Jimo, Qingdao, 266237, Shandong, China.
| |
Collapse
|