1
|
Zhu J, Wang Y, Zhu K, Zhang C. Advances in understanding the role of squalene epoxidase in cancer prognosis and resistance. Mol Biol Rep 2025; 52:162. [PMID: 39869140 DOI: 10.1007/s11033-025-10276-x] [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: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Recently, there has been burgeoning interest in the involvement of cholesterol metabolism in cancer. Squalene epoxidase (SQLE), as a critical rate-limiting enzyme in the cholesterol synthesis pathway, has garnered attention due to its overexpression in various cancer types, thereby significantly impacting tumor prognosis and resistance mechanisms. Firstly, SQLE contributes to unfavorable prognosis through diverse mechanisms, encompassing modulation of the PI3K/AKT signaling pathway, manipulation of the cancer microenvironment, and participation in ferroptosis. Secondly, directing efforts towards targeting SQLE, via mechanisms such as the PI3K/AKT pathway, presents promising avenues for overcoming resistance to conventional therapies such as endocrine cancer therapy, chemotherapy, immunotherapy, or radiotherapy. Moreover, the effectiveness of SQLE protein inhibitors in impeding cancer progression may either depend directly on SQLE inhibition or function through alternative pathways separate from SQLE. This mini-review offers insights into the intricate mechanisms through which SQLE affects the prognosis and resistance profiles across diverse cancer types, while succinctly elucidating the mechanisms underpinning the anticancer effects of SQLE protein inhibitors. Furthermore, this mini-review underscores the necessity for further investigations into the interplay between SQLE and cancer, proposing potential avenues for future research, with the aim of serving as a reference for exploring the mechanisms governing the role of SQLE in cancer regulation.
Collapse
Affiliation(s)
- Jiazhuang Zhu
- Department of Orthopedic Surgery, Institute of Bone Tumor, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yongjie Wang
- Department of Orthopedic Surgery, Institute of Bone Tumor, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200092, China
| | - Kunpeng Zhu
- Department of Orthopedic Surgery, Institute of Bone Tumor, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Chunlin Zhang
- Department of Orthopedic Surgery, Institute of Bone Tumor, Shanghai Tenth People's Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
2
|
Kim WJ, Bae J, Lee EH, Kim J, Kim PJ, Ma PX, Woo KM. Long noncoding RNA MALAT1 mediates fibrous topography-driven pathologic calcification through trans-differentiation of myoblasts. Mater Today Bio 2024; 28:101182. [PMID: 39205874 PMCID: PMC11357808 DOI: 10.1016/j.mtbio.2024.101182] [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: 03/04/2024] [Revised: 07/14/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
Prosthesis-induced pathological calcification is a significant challenge in biomaterial applications and is often associated with various reconstructive medical procedures. It is uncertain whether the fibrous extracellular matrix (ECM) adjacent to biomaterials directly triggers osteogenic trans-differentiation in nearby cells. To investigate this possibility, we engineered a heterogeneous polystyrene fibrous matrix (PSF) designed to mimic the ECM. Our findings revealed that the myoblasts grown on this PSF acquired osteogenic properties, resulting in mineralization both in vitro and in vivo. Transcriptomic analyses indicated a notable upregulation in the expression of the long noncoding RNA metastsis-associated lung adenocarcinoma transcript 1 (Malat1) in the C2C12 myoblasts cultured on PSF. Intriguingly, silencing Malat1 curtailed the PSF-induced mineralization and downregulated the expression of bone morphogenetic proteins (Bmps) and osteogenic markers. Further, we found that PSF prompted the activation of Yap1 signaling and epigenetic modifications in the Malat1 promoter, crucial for the expression of Malat1. These results indicate that the fibrous matrix adjacent to biomaterials can instigate Malat1 upregulation, subsequently driving osteogenic trans-differentiation in myoblasts and ectopic calcification through its transcriptional regulation of osteogenic genes, including Bmps. Our findings point to a novel therapeutic avenue for mitigating prosthesis-induced pathological calcification, heralding new possibilities in the field of biomaterial-based therapies.
Collapse
Affiliation(s)
- Woo-Jin Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jieun Bae
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun-Hye Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaehyung Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Pil-Jong Kim
- Biomedical Knowledge Engineering Laboratory, Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Peter X. Ma
- Department of Biologic and Material Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kyung Mi Woo
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Pharmacology & Dental Therapeutics, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
3
|
Liu J, Li B, Li L, Ming X, Xu ZP. Advances in Nanomaterials for Immunotherapeutic Improvement of Cancer Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403024. [PMID: 38773882 DOI: 10.1002/smll.202403024] [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: 04/15/2024] [Revised: 05/10/2024] [Indexed: 05/24/2024]
Abstract
Immuno-stimulative effect of chemotherapy (ISECT) is recognized as a potential alternative to conventional immunotherapies, however, the clinical application is constrained by its inefficiency. Metronomic chemotherapy, though designed to overcome these limitations, offers inconsistent results, with effectiveness varying based on cancer types, stages, and patient-specific factors. In parallel, a wealth of preclinical nanomaterials holds considerable promise for ISECT improvement by modulating the cancer-immunity cycle. In the area of biomedical nanomaterials, current literature reviews mainly concentrate on a specific category of nanomaterials and nanotechnological perspectives, while two essential issues are still lacking, i.e., a comprehensive analysis addressing the causes for ISECT inefficiency and a thorough summary elaborating the nanomaterials for ISECT improvement. This review thus aims to fill these gaps and catalyze further development in this field. For the first time, this review comprehensively discusses the causes of ISECT inefficiency. It then meticulously categorizes six types of nanomaterials for improving ISECT. Subsequently, practical strategies are further proposed for addressing inefficient ISECT, along with a detailed discussion on exemplary nanomedicines. Finally, this review provides insights into the challenges and perspectives for improving chemo-immunotherapy by innovations in nanomaterials.
Collapse
Affiliation(s)
- Jie Liu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 000000, China
- GoodMedX Tech Limited Company, Hong Kong SAR, 000000, China
| | - Bei Li
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
| | - Xin Ming
- Departments of Cancer Biology and Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27157, USA
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD, 4072, Australia
- Institute of Biomedical Health Technology and Engineering, and Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, 518107, China
| |
Collapse
|
4
|
Beitl KN, Reimhult E. Effect of Solvent Properties on the Critical Solution Temperature of Thermoresponsive Polymers. Int J Mol Sci 2024; 25:7734. [PMID: 39062977 PMCID: PMC11277098 DOI: 10.3390/ijms25147734] [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: 05/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The ability of thermoresponsive polymers to respond to temperature with a reversible conformational change makes them promising 'smart' materials for solutions in medical and biotechnological applications. In this work, two such polymers and structural isomers were studied: poly(N-isopropyl acrylamide) (PNiPAm) and poly(2-isopropyl-2-oxazoline) (PiPOx). We compare the critical solution temperatures (CST) of these polymers in D2O and H2O in the presence of Hofmeister series salts, as results obtained under these different solvent conditions are often compared. D2O has a higher dipole moment and electronegativity than H2O, which could significantly alter the CST transition. We used two complementary methods to measure the CST, dynamic light scattering (DLS) and differential scanning calorimetry (DSC) and found that the CST decreased significantly in D2O compared to H2O. In the presence of highly concentrated kosmotropes, the CST of both polymers decreased in both solvents. The influence of the kosmotropic anions was smaller than the water isotope effect at low ionic strengths but considerably higher at physiological ionic strengths. However, the Hofmeister anion effect was quantitatively different in H2O than in D2O, with the largest relative differences observed for Cl-, where the CSTs in D2O decreased more than in H2O measured by DLS but less by DSC. PiPOx was more sensitive than PNiPAm to the presence of chaotropes. It exhibited much higher transition enthalpies and multistep transitions, especially in aqueous solutions. Our results highlight that measurements of thermoresponsive polymer properties in D2O cannot be compared directly or quantitatively to application conditions or even measurements performed in H2O.
Collapse
Affiliation(s)
| | - Erik Reimhult
- Institute of Colloid and Biointerface Science, Department of Bionanosciences, BOKU University, Muthgasse 11, A-1190 Vienna, Austria
| |
Collapse
|
5
|
Qian Y, Chen W, Wang M, Xie Y, Qiao L, Sun Q, Gao M, Li C. Tumor Microenvironment-Specific Driven Nanoagents for Synergistic Mitochondria Damage-Related Immunogenic Cell Death and Alleviated Immunosuppression. SMALL METHODS 2024; 8:e2301231. [PMID: 38126694 DOI: 10.1002/smtd.202301231] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/06/2023] [Indexed: 12/23/2023]
Abstract
Despite significant breakthroughs in immunotherapy, the limitations of inadequate immune stimulation and stubborn immune resistance continue to present opportunities and challenges. Therefore, a two-pronged approach, encompassing the activation of immunogenic cell death (ICD) and blocking the indoleamine 2,3-dioxygenase (IDO)-mediated pathway, is devised to elicit systemic anti-tumor immunity and alleviate immunosuppression. Herein, a tumor microenvironment (TME)-specific driven nanoagent is composed of a tetrasulfide bond-bridged mesoporous silica layer (MON) coated up-conversion nanoparticles as a nano-carrier, combines Fe2+, curcumin, and indoximod for operating chemodynamic therapy/chemotherapy/immunotherapy. The consumption of glutathione (GSH) caused by MON degradation, the Fenton reaction of Fe2+, and curcumin triggering mitochondrial damage collectively exacerbate the oxidative stress, leading to a violent immunoreaction and reversal of the immunosuppressive TME through a combination of IDO-inhibitors. Meanwhile, upconversion luminescence (UCL) imaging serves as a significant guiding tool for drug delivery and the treatment of nanoagents. In vivo and in vitro experiment results demonstrate that the nanosystem not only effectively inhibits the growth of primary tumors but also induces immune priming and memory effects to reject re-challenged tumors. The strategy as a complementary approach displays great potential for future immunotherapy along with other multimodal treatment modes.
Collapse
Affiliation(s)
- Yanrong Qian
- Shenzhen Research Institute, Shandong University, Shenzhen, 518057, P. R. China
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, P. R. China
| | - Weilin Chen
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, P. R. China
| | - Man Wang
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, P. R. China
| | - Yulin Xie
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, P. R. China
| | - Luying Qiao
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, P. R. China
| | - Qianqian Sun
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, P. R. China
| | - Minghong Gao
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, P. R. China
| | - Chunxia Li
- Shenzhen Research Institute, Shandong University, Shenzhen, 518057, P. R. China
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, P. R. China
| |
Collapse
|
6
|
Zhou Y, Xu M, Shen W, Xu Y, Shao A, Xu P, Yao K, Han H, Ye J. Recent Advances in Nanomedicine for Ocular Fundus Neovascularization Disease Management. Adv Healthc Mater 2024; 13:e2304626. [PMID: 38406994 PMCID: PMC11468720 DOI: 10.1002/adhm.202304626] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/22/2024] [Indexed: 02/27/2024]
Abstract
As an indispensable part of the human sensory system, visual acuity may be impaired and even develop into irreversible blindness due to various ocular pathologies. Among ocular diseases, fundus neovascularization diseases (FNDs) are prominent etiologies of visual impairment worldwide. Intravitreal injection of anti-vascular endothelial growth factor drugs remains the primary therapy but is hurdled by common complications and incomplete potency. To renovate the current therapeutic modalities, nanomedicine emerged as the times required, which is endowed with advanced capabilities, able to fulfill the effective ocular fundus drug delivery and achieve precise drug release control, thus further improving the therapeutic effect. This review provides a comprehensive summary of advances in nanomedicine for FND management from state-of-the-art studies. First, the current therapeutic modalities for FNDs are thoroughly introduced, focusing on the key challenges of ocular fundus drug delivery. Second, nanocarriers are comprehensively reviewed for ocular posterior drug delivery based on the nanostructures: polymer-based nanocarriers, lipid-based nanocarriers, and inorganic nanoparticles. Thirdly, the characteristics of the fundus microenvironment, their pathological changes during FNDs, and corresponding strategies for constructing smart nanocarriers are elaborated. Furthermore, the challenges and prospects of nanomedicine for FND management are thoroughly discussed.
Collapse
Affiliation(s)
- Yifan Zhou
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Mingyu Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Wenyue Shen
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Yufeng Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - An Shao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Peifang Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Haijie Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, 88 Jiefang Road, Hangzhou, 310009, P. R. China
| |
Collapse
|
7
|
Ma S, Tian Z, Liu L, Zhu J, Wang J, Zhao S, Zhu Y, Zhu J, Wang W, Jiang R, Qu Y, Lei J, Zhao J, Jiang T. Cold to Hot: Tumor Immunotherapy by Promoting Vascular Normalization Based on PDGFB Nanocomposites. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308638. [PMID: 38018295 DOI: 10.1002/smll.202308638] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/04/2023] [Indexed: 11/30/2023]
Abstract
Immunotherapy is a promising cancer therapeutic strategy. However, the "cold" tumor immune microenvironment (TIME), characterized by insufficient immune cell infiltration and immunosuppressive status, limits the efficacy of immunotherapy. Tumor vascular abnormalities due to defective pericyte coverage are gradually recognized as a profound determinant in "cold" TIME establishment by hindering immune cell trafficking. Recently, several vascular normalization strategies by improving pericyte coverage have been reported, whereas have unsatisfactory efficacy and high rates of resistance. Herein, a combinatorial strategy to induce tumor vasculature-targeted pericyte recruitment and zinc ion-mediated immune activation with a platelet-derived growth factor B (PDGFB)-loaded, cyclo (Arg-Gly-Asp-D-Phe-Lys)-modified zeolitic imidazolate framework 8 (PDGFB@ZIF8-RGD) nanoplatform is proposed. PDGFB@ZIF8-RGD effectively induced tumor vascular normalization, which facilitated trafficking and infiltration of immune effector cells, including natural killer (NK) cells, M1-like macrophages and CD8+ T cells, into tumor microenvironment. Simultaneously, vascular normalization promoted the accumulation of zinc ions inside tumors to trigger effector cell immune activation and effector molecule production. The synergy between these two effects endowed PDGFB@ZIF8-RGD with superior capabilities in reprogramming the "cold" TIME to a "hot" TIME, thereby initiating robust antitumor immunity and suppressing tumor growth. This combinatorial strategy for improving immune effector cell infiltration and activation is a promising paradigm for solid tumor immunotherapy.
Collapse
Affiliation(s)
- Shouzheng Ma
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Zhimin Tian
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Lei Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, China
| | - Jun Zhu
- The Southern Theater Air Force Hospital, Guangzhou, 510000, China
| | - Jing Wang
- Department of Immunology, Air Force Medical University, Xi'an, 710032, China
| | - Shoujie Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yejing Zhu
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Wenchen Wang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Runmin Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yongquan Qu
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Junlong Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Medical Genetics and Development Biology, Air Force Medical University, Xi'an, 710032, China
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, 710000, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| |
Collapse
|
8
|
Xie C, Xu J, Wang X, Jiang S, Zheng Y, Liu Z, Jia Z, Jia Z, Lu X. Smart Hydrogels for Tissue Regeneration. Macromol Biosci 2024; 24:e2300339. [PMID: 37848181 DOI: 10.1002/mabi.202300339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/06/2023] [Indexed: 10/19/2023]
Abstract
The rapid growth in the portion of the aging population has led to a consequent increase in demand for biomedical hydrogels, together with an assortment of challenges that need to be overcome in this field. Smart hydrogels can autonomously sense and respond to the physiological/pathological changes of the tissue microenvironment and continuously adapt the response according to the dynamic spatiotemporal shifts in conditions. This along with other favorable properties, make smart hydrogels excellent materials for employing toward improving the precision of treatment for age-related diseases. The key factor during the smart hydrogel design is on accurately identifying the characteristics of natural tissues and faithfully replicating the composition, structure, and biological functions of these tissues at the molecular level. Such hydrogels can accurately sense distinct physiological and external factors such as temperature and biologically active molecules, so they may in turn actively and promptly adjust their response, by regulating their own biological effects, thereby promoting damaged tissue repair. This review summarizes the design strategies employed in the creation of smart hydrogels, their response mechanisms, as well as their applications in field of tissue engineering; and concludes by briefly discussing the relevant challenges and future prospects.
Collapse
Affiliation(s)
- Chaoming Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Jie Xu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xinyi Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Shengxi Jiang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yujia Zheng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zexin Liu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zhuo Jia
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zhanrong Jia
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523000, China
| | - Xiong Lu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
9
|
Yong J, Shu H, Zhang X, Yang K, Luo G, Yu L, Li J, Huang H. Natural Products-Based Inhaled Formulations for Treating Pulmonary Diseases. Int J Nanomedicine 2024; 19:1723-1748. [PMID: 38414528 PMCID: PMC10898359 DOI: 10.2147/ijn.s451206] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
Given the unique physiological and pathological characteristics of the lung, the direct, inhalable route is more conducive to pulmonary drug delivery and disease control than traditional systemic drug delivery, significantly circumventing drug loss, off-target effects, systemic and organ toxicity, etc., and is widely regarded as the preferred regimen for pulmonary drug delivery. However, very few lung diseases are currently treated with the preferred inhaled formulations, such as asthma, chronic obstructive pulmonary disease and pulmonary hypertension. And there is a lack of appropriate inhaled formulations for other critical lung diseases, such as lung cancer and pulmonary fibrosis, due to the fact that the physicochemical properties of the drugs and their pharmacokinetic profiles do not match the physiology of the lung, and conventional inhalation devices are unable to deliver them to the specific parts of the lung. Phytochemicals of natural origin, due to their wide availability and clear safety profile, hold great promise for the preparation of inhalable formulations to improve the current dilemma in the treatment of lung diseases. In particular, the preparation of inhalable formulations based on nano- and microparticulate carriers for drug delivery to deep lung tissues, which overcome the shortcomings of conventional inhalation therapies while targeting the drug activity directly to a specific part of the lung, may be the best approach to change the current dilemma of lung disease treatment. In this review, we discuss recent advances in nano- and micron-carrier-based inhalation formulations for the delivery of natural products for the treatment of pulmonary diseases, which may represent an opportunity for practical clinical translation of natural products.
Collapse
Affiliation(s)
- Jiangyan Yong
- Department of Clinical Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, People’s Republic of China
| | - Hongli Shu
- Department of Clinical Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, People’s Republic of China
| | - Xiao Zhang
- Department of Clinical Laboratory, Chengdu Children Special Hospital, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Kun Yang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Guining Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Lu Yu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Jiaqi Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Hong Huang
- Department of Clinical Laboratory, the People’s Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, People’s Republic of China
| |
Collapse
|
10
|
Sun D, Chang Q, Lu F. Immunomodulation in diabetic wounds healing: The intersection of macrophage reprogramming and immunotherapeutic hydrogels. J Tissue Eng 2024; 15:20417314241265202. [PMID: 39071896 PMCID: PMC11283672 DOI: 10.1177/20417314241265202] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Diabetic wound healing presents a significant clinical challenge due to the interplay of systemic metabolic disturbances and local inflammation, which hinder the healing process. Macrophages undergo a phenotypic shift from M1 to M2 during wound healing, a transition pivotal for effective tissue repair. However, in diabetic wounds, the microenvironment disrupts this phenotypic polarization, perpetuating inflammation, and impeding healing. Reprograming macrophages to restore their M2 phenotype offers a potential avenue for modulating the wound immune microenvironment and promoting healing. This review elucidates the mechanisms underlying impaired macrophage polarization toward the M2 phenotype in diabetic wounds and discusses novel strategies, including epigenetic and metabolic interventions, to promote macrophage conversion to M2. Hydrogels, with their hydrated 3D cross-linked structure, closely resemble the physiological extracellular matrix and offer advantageous properties such as biocompatibility, tunability, and versatility. These characteristics make hydrogels promising candidates for developing immunomodulatory materials aimed at addressing diabetic wounds. Understanding the role of hydrogels in immunotherapy, particularly in the context of macrophage reprograming, is essential for the development of advanced wound care solutions. This review also highlights recent advancements in immunotherapeutic hydrogels as a step toward precise and effective treatments for diabetic wounds.
Collapse
Affiliation(s)
- Dan Sun
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Chang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Zhang Z, He C, Chen X. Designing Hydrogels for Immunomodulation in Cancer Therapy and Regenerative Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308894. [PMID: 37909463 DOI: 10.1002/adma.202308894] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/26/2023] [Indexed: 11/03/2023]
Abstract
The immune system not only acts as a defense against pathogen and cancer cells, but also plays an important role in homeostasis and tissue regeneration. Targeting immune systems is a promising strategy for efficient cancer treatment and regenerative medicine. Current systemic immunomodulation therapies are usually associated with low persistence time, poor targeting to action sites, and severe side effects. Due to their extracellular matrix-mimetic nature, tunable properties and diverse bioactivities, hydrogels are intriguing platforms to locally deliver immunomodulatory agents and cells, as well as provide an immunomodulatory microenvironment to recruit, activate, and expand host immune cells. In this review, the design considerations, including polymer backbones, crosslinking mechanisms, physicochemical nature, and immunomodulation-related components, of the hydrogel platforms, are focused on. The immunomodulatory effects and therapeutic outcomes in cancer therapy and tissue regeneration of different hydrogel systems are emphasized, including hydrogel depots for delivery of immunomodulatory agents, hydrogel scaffolds for cell delivery, and immunomodulatory hydrogels depending on the intrinsic properties of materials. Finally, the remained challenges in current systems and future development of immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Zhen Zhang
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
12
|
Tian R, Li Y, Xu Z, Xu J, Liu J. Current Advances of Atomically Dispersed Metal-Centered Nanozymes for Tumor Diagnosis and Therapy. Int J Mol Sci 2023; 24:15712. [PMID: 37958697 PMCID: PMC10648793 DOI: 10.3390/ijms242115712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Nanozymes, which combine enzyme-like catalytic activity and the biological properties of nanomaterials, have been widely used in biomedical fields. Single-atom nanozymes (SANs) with atomically dispersed metal centers exhibit excellent biological catalytic activity due to the maximization of atomic utilization efficiency, unique metal coordination structures, and metal-support interaction, and their structure-activity relationship can also be clearly investigated. Therefore, they have become an emerging alternative to natural enzymes. This review summarizes the examples of nanocatalytic therapy based on SANs in tumor diagnosis and treatment in recent years, providing an overview of material classification, activity modulation, and therapeutic means. Next, we will delve into the therapeutic mechanism of SNAs in the tumor microenvironment and the advantages of synergistic multiple therapeutic modalities (e.g., chemodynamic therapy, sonodynamic therapy, photothermal therapy, chemotherapy, photodynamic therapy, sonothermal therapy, and gas therapy). Finally, this review proposes the main challenges and prospects for the future development of SANs in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Ruizhen Tian
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China; (R.T.); (Y.L.)
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (Z.X.); (J.X.)
| | - Yijia Li
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China; (R.T.); (Y.L.)
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (Z.X.); (J.X.)
| | - Zhengwei Xu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (Z.X.); (J.X.)
| | - Jiayun Xu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (Z.X.); (J.X.)
| | - Junqiu Liu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (Z.X.); (J.X.)
| |
Collapse
|
13
|
Ma X, Mao M, He J, Liang C, Xie HY. Nanoprobe-based molecular imaging for tumor stratification. Chem Soc Rev 2023; 52:6447-6496. [PMID: 37615588 DOI: 10.1039/d3cs00063j] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
The responses of patients to tumor therapies vary due to tumor heterogeneity. Tumor stratification has been attracting increasing attention for accurately distinguishing between responders to treatment and non-responders. Nanoprobes with unique physical and chemical properties have great potential for patient stratification. This review begins by describing the features and design principles of nanoprobes that can visualize specific cell types and biomarkers and release inflammatory factors during or before tumor treatment. Then, we focus on the recent advancements in using nanoprobes to stratify various therapeutic modalities, including chemotherapy, radiotherapy (RT), photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), ferroptosis, and immunotherapy. The main challenges and perspectives of nanoprobes in cancer stratification are also discussed to facilitate probe development and clinical applications.
Collapse
Affiliation(s)
- Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, P. R. China.
| |
Collapse
|