1
|
Qiu P, Wen M, Zhuang Z, Niu S, Tao C, Yu N, Chen Z. Biomimetic polymeric nanoreactors with photooxidation-initiated therapies and reinvigoration of antigen-dependent and antigen-free immunity. Biomaterials 2025; 314:122884. [PMID: 39405823 DOI: 10.1016/j.biomaterials.2024.122884] [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: 07/04/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/10/2024]
Abstract
Immune cell-mediated anticancer modalities usually suffer from immune cell exhaustion and limited efficacy in solid tumors. Herein, the oxygen-carrying biomimetic nanoreactors (BNR2(O2)) have been developed with photooxidation-driven therapies and antigen-dependent/antigen-free immune reinvigoration against xenograft tumors. The BNR2(O2) composes polymeric nanoreactors camouflaged with cancer cell membranes can efficiently target homotypic tumors. It continuously releases O2 to boost intracellular reactive oxygen species (ROS) to oxide diselenide bonds, which controllably releases seleninic acids and anti-folate Pemetrexed compared to hydrogen peroxide and glutathione incubation. The O2-rich microenvironment sensitizes Pemetrexed and blocks programmed cell-death ligand 1 (PD-L1) to reverse T cell immunosuppression. The ROS and Pemetrexed upregulate pro-apoptosis proteins and inhibit folate-related enzymes, which cause significant apoptosis and immunogenic cell death to stimulate dendritic cell maturation for improved secretion of cytokines, expanding antigen-dependent T cell immunity. Furthermore, by regulating the release of seleninic acids, the checkpoint receptor human leukocyte antigen E of tumor cells can be blocked to reinvigorate antigen-free natural killer cell immunity. This work offers an advanced antitumor strategy by bridging biomimetic nanoreactors and modulation of multiple immune cells.
Collapse
Affiliation(s)
- Pu Qiu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Mei Wen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Zixuan Zhuang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Shining Niu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Cheng Tao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Nuo Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China.
| | - Zhigang Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
2
|
Liu C, Ma H, Yuan S, Jin Y, Tian W. Living Cell-Mediated Self-Assembly: From Monomer Design and Morphology Regulation to Biomedical Applications. ACS NANO 2025; 19:2047-2069. [PMID: 39779487 DOI: 10.1021/acsnano.4c16669] [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: 01/11/2025]
Abstract
The self-assembly of molecules into highly ordered architectures is a ubiquitous and natural process, wherein molecules spontaneously organize into large structures to perform diverse functions. Drawing inspiration from the formation of natural nanostructures, cell-mediated self-assembly has been developed to create functional assemblies both inside and outside living cells. These techniques have been employed to regulate the cellular world by leveraging the dynamic intracellular and extracellular microenvironment. This review highlights the recent advances and future trends in living cell-mediated self-assembly, ranging from their cytocompatible monomer designs, synthetic strategies, and morphological control to functional applications. The assembly behaviors are also discussed based on the dimensionality of the self-assembled morphologies from zero to three dimensions. Finally, this review explores its promising potential for biomedical applications, clarifying the relationship between initial morphological regulation and the therapeutic effects of subsequent artificial assemblies. Through rationally designing molecular structures and precisely controlling assembly morphologies, living cell mediated self-assembly would provide an innovative platform for executing biological functions.
Collapse
Affiliation(s)
- Chengfei Liu
- Shaanxi Key Laboratory of Macromolecular Science and Technology, Xi'an Key Laboratory of Hybrid Luminescent Materials and Photonic Device, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, P. R. China
| | - Haonan Ma
- Shaanxi Key Laboratory of Macromolecular Science and Technology, Xi'an Key Laboratory of Hybrid Luminescent Materials and Photonic Device, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, P. R. China
| | - Shengzhuo Yuan
- Shaanxi Key Laboratory of Macromolecular Science and Technology, Xi'an Key Laboratory of Hybrid Luminescent Materials and Photonic Device, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, P. R. China
| | - Yifan Jin
- Shaanxi Key Laboratory of Macromolecular Science and Technology, Xi'an Key Laboratory of Hybrid Luminescent Materials and Photonic Device, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, P. R. China
| | - Wei Tian
- Shaanxi Key Laboratory of Macromolecular Science and Technology, Xi'an Key Laboratory of Hybrid Luminescent Materials and Photonic Device, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, P. R. China
| |
Collapse
|
3
|
Gao J, Zhai Y, Lu W, Jiang X, Zhou J, Wu L, Du L, Ou C, Zhang X, He H, Zhu J, Zhang Z, Li M, Wu Y, Pan X. ROS-sensitive PD-L1 siRNA cationic selenide nanogels for self-inhibition of autophagy and prevention of immune escape. Bioact Mater 2024; 41:597-610. [PMID: 39280899 PMCID: PMC11393550 DOI: 10.1016/j.bioactmat.2024.08.013] [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: 02/22/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
In the field of cancer therapy, inhibiting autophagy has emerged as a promising strategy. However, pharmacological disruption of autophagy can lead to the upregulation of programmed death-ligand 1 (PD-L1), enabling tumor immune evasion. To address this issue, we developed innovative ROS-responsive cationic poly(ethylene imine) (PEI) nanogels using selenol chemistry-mediated multicomponent reaction (MCR) technology. This procedure involved simple mixing of low-molecular-weight PEI (LMW PEI), γ-selenobutylacetone (γ-SBL), and poly(ethylene glycol) methacrylate (PEGMA). Through high-throughput screening, we constructed a library of AxSeyOz nanogels and identified the optimized A1.8Se3O0.5/siPD-L1 nanogels, which exhibited a size of approximately 200 nm, excellent colloidal stability, and the most effective PD-L1 silencing efficacy. These nanogels demonstrated enhanced uptake by tumor cells, excellent oxidative degradation ability, and inhibited autophagy by alkalinizing lysosomes. The A1.8Se3O0.5/siPD-L1 nanogels significantly downregulated PD-L1 expression and increased the expression of major histocompatibility complex class I (MHC-I), resulting in robust proliferation of specific CD8+ T cells and a decrease in MC38 tumor growth. As a result, the A1.8Se3O0.5/siPD-L1 nanogels inhibited tumor growth through self-inhibition of autophagy, upregulation of MHC-I, and downregulation of PD-L1. Designed with dynamic diselenide bonds, the A1.8Se3O0.5/siPD-L1 nanogels showed synergistic antitumor efficacy through self-inhibition of autophagy and prevention of immune escape.
Collapse
Affiliation(s)
- Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Yonghua Zhai
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Weihong Lu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Xianghe Jiang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Jingsheng Zhou
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Longhai Du
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Chunqing Ou
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Xinyi Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Hanliang He
- The Department of Orthopedic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215028, China
| | - Jian Zhu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Meiyun Li
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xiangqiang Pan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| |
Collapse
|
4
|
Shang Y, Han D, Deng K, Zhou H, Wu M. Quercetin Boosts Pulsatile Gonadotropin-Releasing Hormone Release to Improve Luteal Function via Inhibiting NF-κB/NLRP3-Mediated Neuron Pyroptosis. Mol Nutr Food Res 2024; 68:e2400649. [PMID: 39491793 DOI: 10.1002/mnfr.202400649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/13/2024] [Indexed: 11/05/2024]
Abstract
SCOPE Luteal phase deficiency (LPD) is the main cause of infertility without an effective cure. Quercetin (QUE) is a bioactive flavonoid with antioxidant properties, while its role in treating LPD remains unclear. This study aims to investigate the therapeutic effects of QUE on infertility and menstrual disorders induced by LPD, thus further exploring the underlying mechanism. METHODS AND RESULTS Mifepristone-induced rats are used to explore the protective effects of QUE against LPD. QUE stimulates the spontaneous secretion of progesterone to improve luteal function and endometrial receptivity in LPD rats by activating the kisspeptin/GPR54 system to facilitate the gonadotropin-releasing hormone (GnRH) pulsatility. Bioinformatics analysis reveals that the core mechanism of QUE in treating LPD is to attenuate the GnRH neuron pyroptosis by inhibiting the NF-κB pathway, which is further verified in LPD rats and lipopolysaccharide (LPS)-treated GT1-7, as QUE significantly reduces the expression of key factors concerning NF-κB pathway and NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. CONCLUSION This study first proposes that neuron pyroptosis-induced GnRH pulsatility disruption accounts for the pathogenesis of LPD, and QUE facilitates the pulse secretion of GnRH to boost the spontaneous progesterone secretion by inhibiting NF-κB/NLRP3-mediated neuron pyroptosis, which provides a new therapeutic target and strategy for LPD.
Collapse
Affiliation(s)
- Yujie Shang
- School of Basic Medical Sciences, Central South University, Changsha, China
- School of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Di Han
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Kun Deng
- School of Basic Medical Sciences, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, China
| | - Huifang Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Minghua Wu
- School of Basic Medical Sciences, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Liu X, Ren B, Ren J, Gu M, You L, Zhao Y. The significant role of amino acid metabolic reprogramming in cancer. Cell Commun Signal 2024; 22:380. [PMID: 39069612 DOI: 10.1186/s12964-024-01760-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024] Open
Abstract
Amino acid metabolism plays a pivotal role in tumor microenvironment, influencing various aspects of cancer progression. The metabolic reprogramming of amino acids in tumor cells is intricately linked to protein synthesis, nucleotide synthesis, modulation of signaling pathways, regulation of tumor cell metabolism, maintenance of oxidative stress homeostasis, and epigenetic modifications. Furthermore, the dysregulation of amino acid metabolism also impacts tumor microenvironment and tumor immunity. Amino acids can act as signaling molecules that modulate immune cell function and immune tolerance within the tumor microenvironment, reshaping the anti-tumor immune response and promoting immune evasion by cancer cells. Moreover, amino acid metabolism can influence the behavior of stromal cells, such as cancer-associated fibroblasts, regulate ECM remodeling and promote angiogenesis, thereby facilitating tumor growth and metastasis. Understanding the intricate interplay between amino acid metabolism and the tumor microenvironment is of crucial significance. Expanding our knowledge of the multifaceted roles of amino acid metabolism in tumor microenvironment holds significant promise for the development of more effective cancer therapies aimed at disrupting the metabolic dependencies of cancer cells and modulating the tumor microenvironment to enhance anti-tumor immune responses and inhibit tumor progression.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Jie Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Minzhi Gu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R, 100023, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, P.R, China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, P.R, China.
| |
Collapse
|
6
|
Zou B, Xiong Z, Yu Y, Shi S, Li X, Chen T. Rapid Selenoprotein Activation by Selenium Nanoparticles to Suppresses Osteoclastogenesis and Pathological Bone Loss. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401620. [PMID: 38621414 DOI: 10.1002/adma.202401620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/28/2024] [Indexed: 04/17/2024]
Abstract
Osteoclast hyperactivation stands as a significant pathological factor contributing to the emergence of bone disorders driven by heightened oxidative stress levels. The modulation of the redox balance to scavenge reactive oxygen species emerges as a viable approach to addressing this concern. Selenoproteins, characterized by selenocysteine (SeCys2) as the active center, are crucial for selenium-based antioxidative stress therapy for inflammatory diseases. This study reveals that surface-active elemental selenium (Se) nanoparticles, particularly lentinan-Se (LNT-Se), exhibit enhanced cellular accumulation and accelerated metabolism to SeCys2, the primary active Se form in biological systems. Consequently, LNT-Se demonstrates significant inhibition of osteoclastogenesis. Furthermore, in vivo studies underscore the superior therapeutic efficacy of LNT-Se over SeCys2, potentially attributable to the enhanced stability and safety profile of LNT-Se. Specifically, LNT-Se effectively modulates the expression of the selenoprotein GPx1, thereby exerting regulatory control over osteoclastogenesis inhibition, and the prevention of osteolysis. In summary, these results suggest that the prompt activation of selenoproteins by Se nanoparticles serves to suppress osteoclastogenesis and pathological bone loss by upregulating GPx1. Moreover, the utilization of bioactive Se species presents a promising avenue for effectively managing bone disorders.
Collapse
Affiliation(s)
- Binhua Zou
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Zushuang Xiong
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yanzi Yu
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Sujiang Shi
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Xiaoling Li
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Laboratory of Viral Pathogenesis & Infection Prevention and Control of Ministry of Education, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
7
|
Sabag B, Puthenveetil A, Levy M, Joseph N, Doniger T, Yaron O, Karako-Lampert S, Lazar I, Awwad F, Ashkenazi S, Barda-Saad M. Dysfunctional natural killer cells can be reprogrammed to regain anti-tumor activity. EMBO J 2024; 43:2552-2581. [PMID: 38637625 PMCID: PMC11217363 DOI: 10.1038/s44318-024-00094-5] [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: 10/01/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/20/2024] Open
Abstract
Natural killer (NK) cells are critical to the innate immune system, as they recognize antigens without prior sensitization, and contribute to the control and clearance of viral infections and cancer. However, a significant proportion of NK cells in mice and humans do not express classical inhibitory receptors during their education process and are rendered naturally "anergic", i.e., exhibiting reduced effector functions. The molecular events leading to NK cell anergy as well as their relation to those underlying NK cell exhaustion that arises from overstimulation in chronic conditions, remain unknown. Here, we characterize the "anergic" phenotype and demonstrate functional, transcriptional, and phenotypic similarities to the "exhausted" state in tumor-infiltrating NK cells. Furthermore, we identify zinc finger transcription factor Egr2 and diacylglycerol kinase DGKα as common negative regulators controlling NK cell dysfunction. Finally, experiments in a 3D organotypic spheroid culture model and an in vivo tumor model suggest that a nanoparticle-based delivery platform can reprogram these dysfunctional natural killer cell populations in their native microenvironment. This approach may become clinically relevant for the development of novel anti-tumor immunotherapeutic strategies.
Collapse
Affiliation(s)
- Batel Sabag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Abhishek Puthenveetil
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Moria Levy
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Noah Joseph
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Tirtza Doniger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Orly Yaron
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Sarit Karako-Lampert
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Itay Lazar
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Fatima Awwad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shahar Ashkenazi
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Mira Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel.
| |
Collapse
|
8
|
Meng X, Zhu G, Yang YG, Sun T. Targeted delivery strategies: The interactions and applications of nanoparticles in liver diseases. Biomed Pharmacother 2024; 175:116702. [PMID: 38729052 DOI: 10.1016/j.biopha.2024.116702] [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: 02/27/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
In recent years, nanoparticles have been broadly utilized in various drugs delivery formulations. Nanodelivery systems have shown promise in solving problems associated with the distribution of hydrophobic drugs and have promoted the accumulation of nanomedicines in the circulation or in organs. However, the injection dose of nanoparticles (NPs) is much greater than that needed by diseased tissues or organs. In other words, most of the NPs are localized off-target and do not reach the desired tissue or organs. With the rapid development of biodegradable and biosafety nanomaterials, the nanovectors represent assurance of safety. However, the off-target effects also induce concerns about the application of NPs, especially in the delivery of gene editing tools. Therefore, a complete understanding of the biological responses to NPs in the body will clearly guide the design of targeted delivery of NPs. The different properties of various nanodelivery systems may induce diverse interactions between carriers and organs. In this review, we describe the relationship between the liver, the most influenced organ of systemic administration of NPs, and targeted delivery nanoplatforms. Various transport vehicles have adopted multiple delivery strategies for the targeted delivery to the cells in the homeostasis liver and in diseased liver. Additionally, nanodelivery systems provide a novel strategy for treating incurable diseases. The appearance of a targeted delivery has profoundly improved the application of NPs to liver diseases.
Collapse
Affiliation(s)
- Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
9
|
Xianyu B, Pan S, Gao S, Xu H, Li T. Selenium-Containing Nanocomplexes Achieve Dual Immune Checkpoint Blockade for NK Cell Reinvigoration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306225. [PMID: 38072799 DOI: 10.1002/smll.202306225] [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: 07/23/2023] [Revised: 11/06/2023] [Indexed: 05/12/2024]
Abstract
The blockade of immune checkpoints has emerged as a promising strategy for cancer immunotherapy. However, most of the current approaches focus on T cells, leaving natural killer (NK) cell-mediated therapeutic strategies rarely explored. Here, a selenium-containing nanocomplex is developed that acts as a dual immune checkpoint inhibitor to reinvigorate NK cell-based cancer immunotherapy. The Se nanocomplex can deliver and release siRNA that targets programmed death ligand-1 (PD-L1) in tumor cells, thereby silencing the checkpoint receptor PD-L1. The intracellular reactive oxygen species generated by porphyrin derivatives in the nanocomplexes can oxidize the diselenide bond into seleninic acid, which blocks the expression of another checkpoint receptor, human leukocyte antigen E. The blockade of dual immune checkpoints shows synergistic effects on promoting NK cell-mediated antitumoral activity. This study provides a new strategy to reinvigorate NK cell immunity for the development of combined cancer immunotherapy.
Collapse
Affiliation(s)
- Banruo Xianyu
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Shuojiong Pan
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Shiqian Gao
- Key Laboratory of Polyoxometalate Science of the Ministry of Education, Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Huaping Xu
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Tianyu Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
10
|
Liu H, Chen L, Shen Y, Fan L, Zhang J, Zhu H, Shi Y, Yan S. Advances in selenium from materials to applications. NANOTECHNOLOGY 2024; 35:242003. [PMID: 38471145 DOI: 10.1088/1361-6528/ad32d3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 03/11/2024] [Indexed: 03/14/2024]
Abstract
Over the past few decades, single-element semiconductors have received a great deal of attention due to their unique light-sensitive and heat-sensitive properties, which are of great application and research significance. As one promising material, selenium, being a typical semiconductor, has attracted significant attention from researchers due to its unique properties including high optical conductivity, anisotropic, thermal conductivity, and so on. To promote the application of selenium nanomaterials in various fields, numerous studies over the past few decades have successfully synthesized selenium nanomaterials in various morphologies using a wide range of physical and chemical methods. In this paper, we review and summarise the different methods of synthesis of various morphologies of selenium nanomaterials and discuss the applications of different nanostructures of selenium nanomaterials in optoelectronic devices, chemical sensors, and biomedical applications. Finally, we discuss possible challenges for selenium nanodevices and provide an outlook on the future applications of selenium nanomaterials.
Collapse
Affiliation(s)
- Hao Liu
- School of Integrated Circuit Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing 210023, People's Republic of China
| | - Liping Chen
- School of Integrated Circuit Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing 210023, People's Republic of China
| | - Yunkun Shen
- College of Automation & College of Artificial Intelligence, Nanjing University of Posts and Telecommunications, Nanjing 210023, People's Republic of China
| | - Li Fan
- School of Materials Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing 210023, People's Republic of China
| | - Jiawei Zhang
- School of Materials Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing 210023, People's Republic of China
| | - Hongliang Zhu
- School of Materials Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing 210023, People's Republic of China
| | - Yi Shi
- National Laboratory of Solid State Microstructures School of Electronic Science and Engineering, Nanjing University, Nanjing 210093, People's Republic of China
| | - Shancheng Yan
- School of Integrated Circuit Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing 210023, People's Republic of China
| |
Collapse
|
11
|
He C, Liu C, Pan S, Tan Y, Guan J, Xu H. Polyurethane with β-Selenocarbonyl Structure Enabling the Combination of Plastic Degradation and Waste Upcycling. Angew Chem Int Ed Engl 2024; 63:e202317558. [PMID: 38156718 DOI: 10.1002/anie.202317558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/16/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
Degradable polymers offer a promising solution to mitigate global plastic pollution, but the degraded products often suffer from diminished value. Upcycling is a more sustainable approach to upgrade polymer waste into value-added products. Herein, we report a β-selenocarbonyl-containing polyurethane (SePU), which can be directly degraded under mild conditions into valuable selenium fertilizers for selenium-rich vegetable cultivation globally, enabling both plastic degradation and waste upcycling. Under oxidation condition, this polymer can be easily and selectively degraded via selenoxide elimination reaction from mixed plastic waste. The degraded product can serve as effective selenium fertilizers to increase selenium content in radish and pak choi. The SePU exhibits excellent mechanical properties. Additionally, we observed the formation of spherulites-like selenium particles within the materials during degradation for the first time. Our research offers a successful application of selenoxide elimination reaction in the field of plastic degradation for the first time, endowing plastics with both degradability and high reusable value. This strategy provides a promising solution to reduce pollution and improve economy and sustainability of plastics.
Collapse
Affiliation(s)
- Chaowei He
- Key Lab of Organic Optoelectronics & Molecular Engineering and Laboratory of Flexible Electronics Technology, Department of Chemistry, Tsinghua University, 100084, Beijing, China
| | - Cheng Liu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, 310027, Hangzhou, Zhejiang, China
| | - Shuojiong Pan
- Key Lab of Organic Optoelectronics & Molecular Engineering and Laboratory of Flexible Electronics Technology, Department of Chemistry, Tsinghua University, 100084, Beijing, China
| | - Yizheng Tan
- Key Lab of Organic Optoelectronics & Molecular Engineering and Laboratory of Flexible Electronics Technology, Department of Chemistry, Tsinghua University, 100084, Beijing, China
| | - Jun Guan
- Key Lab of Organic Optoelectronics & Molecular Engineering and Laboratory of Flexible Electronics Technology, Department of Chemistry, Tsinghua University, 100084, Beijing, China
| | - Huaping Xu
- Key Lab of Organic Optoelectronics & Molecular Engineering and Laboratory of Flexible Electronics Technology, Department of Chemistry, Tsinghua University, 100084, Beijing, China
| |
Collapse
|
12
|
Han X, Gong C, Yang Q, Zheng K, Wang Z, Zhang W. Biomimetic Nano-Drug Delivery System: An Emerging Platform for Promoting Tumor Treatment. Int J Nanomedicine 2024; 19:571-608. [PMID: 38260239 PMCID: PMC10802790 DOI: 10.2147/ijn.s442877] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
With the development of nanotechnology, nanoparticles (NPs) have shown broad prospects as drug delivery vehicles. However, they exhibit certain limitations, including low biocompatibility, poor physiological stability, rapid clearance from the body, and nonspecific targeting, which have hampered their clinical application. Therefore, the development of novel drug delivery systems with improved biocompatibility and high target specificity remains a major challenge. In recent years, biofilm mediated biomimetic nano-drug delivery system (BNDDS) has become a research hotspot focus in the field of life sciences. This new biomimetic platform uses bio-nanotechnology to encapsulate synthetic NPswithin biomimetic membrane, organically integrating the low immunogenicity, low toxicity, high tumor targeting, good biocompatibility of the biofilm with the adjustability and versatility of the nanocarrier, and shows promising applications in the field of precision tumor therapy. In this review, we systematically summarize the new progress in BNDDS used for optimizing drug delivery, providing a theoretical reference for optimizing drug delivery and designing safe and efficient treatment strategies to improve tumor treatment outcomes.
Collapse
Affiliation(s)
- Xiujuan Han
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Chunai Gong
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, People’s Republic of China
| | - Qingru Yang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Kaile Zheng
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
| | - Zhuo Wang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Wei Zhang
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
13
|
Zhang H, Yang L, Wang T, Li Z. NK cell-based tumor immunotherapy. Bioact Mater 2024; 31:63-86. [PMID: 37601277 PMCID: PMC10432724 DOI: 10.1016/j.bioactmat.2023.08.001] [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: 05/26/2023] [Revised: 07/16/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Abstract
Natural killer (NK) cells display a unique inherent ability to identify and eliminate virus-infected cells and tumor cells. They are particularly powerful for elimination of hematological cancers, and have attracted considerable interests for therapy of solid tumors. However, the treatment of solid tumors with NK cells are less effective, which can be attributed to the very complicated immunosuppressive microenvironment that may lead to the inactivation, insufficient expansion, short life, and the poor tumor infiltration of NK cells. Fortunately, the development of advanced nanotechnology has provided potential solutions to these issues, and could improve the immunotherapy efficacy of NK cells. In this review, we summarize the activation and inhibition mechanisms of NK cells in solid tumors, and the recent advances in NK cell-based tumor immunotherapy boosted by diverse nanomaterials. We also propose the challenges and opportunities for the clinical application of NK cell-based tumor immunotherapy.
Collapse
Affiliation(s)
- Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Li Yang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Tingting Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| |
Collapse
|
14
|
Li Y, Wu Y, Fang Z, Zhang Y, Ding H, Ren L, Zhang L, Gong Q, Gu Z, Luo K. Dendritic Nanomedicine with Boronate Bonds for Augmented Chemo-Immunotherapy via Synergistic Modulation of Tumor Immune Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307263. [PMID: 37743633 DOI: 10.1002/adma.202307263] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/02/2023] [Indexed: 09/26/2023]
Abstract
Unsatisfied tumor accumulation of chemotherapeutic drugs and a complicated immunosuppressive microenvironment diminish the immune response rate and the therapeutic effect. Surface modification of these drugs with target ligands can promote their cellular internalization, but the modified drugs may be subjected to unexpected immune recognition and clearance. Herein, a phenylboronic acid (PBA) group-shieldable dendritic nanomedicine that integrates an immunogenic cell death (ICD)-inducing agent (epirubicin, Epi) and an indoleamine 2,3-dioxgenase 1 (IDO1) inhibitor (NLG919) is reported for tumor chemo-immunotherapy. This NLG919-loaded Epi-conjugated PEGylated dendrimers bridged with boronate bonds (NLG919@Epi-DBP) maintains a stable nanostructure during circulation. Under a moderate acidic condition, the PBA group exposes to the sialic acid residue on the tumor cell membrane to enhance the internalization and penetration of NLG919@Epi-DBP. At pH 5.0, NLG919@Epi-DBP rapidly disassembles to release the incorporated Epi and NLG919. Epi triggers robust ICD of tumor cells that evokes strong immune response. In addition, inhibition of the IDO1 activity downregulates the metabolism of L-tryptophan to kynurenine, leading to a reduction in the recruitment of immunosuppressive cells and modulation of the tumor immune microenvironment. Collectively, this promising strategy has been demonstrated to evoke robust immune response as well as remodel the immunosuppressive microenvironment for an enhanced chemo-immunotherapeutic effect.
Collapse
Affiliation(s)
- Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yahui Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zaixiang Fang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuxin Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haitao Ding
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Long Ren
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lu Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Zhongwei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Metabolomics and Proteomics Technology Platform, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|