151
|
Bian N, Chu C, Rung S, Huangphattarakul V, Man Y, Lin J, Hu C. Immunomodulatory Biomaterials and Emerging Analytical Techniques for Probing the Immune Micro-Environment. Tissue Eng Regen Med 2023; 20:11-24. [PMID: 36241939 PMCID: PMC9852373 DOI: 10.1007/s13770-022-00491-z] [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: 08/03/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 02/01/2023] Open
Abstract
After implantation of a biomaterial, both the host immune system and properties of the material determine the local immune response. Through triggering or modulating the local immune response, materials can be designed towards a desired direction of promoting tissue repair or regeneration. High-throughput sequencing technologies such as single-cell RNA sequencing (scRNA-seq) emerging as a powerful tool for dissecting the immune micro-environment around biomaterials, have not been fully utilized in the field of soft tissue regeneration. In this review, we first discussed the procedures of foreign body reaction in brief. Then, we summarized the influences that physical and chemical modulation of biomaterials have on cell behaviors in the micro-environment. Finally, we discussed the application of scRNA-seq in probing the scaffold immune micro-environment and provided some reference to designing immunomodulatory biomaterials. The foreign body response consists of a series of biological reactions. Immunomodulatory materials regulate immune cell activation and polarization, mediate divergent local immune micro-environments and possess different tissue engineering functions. The manipulation of physical and chemical properties of scaffolds can modulate local immune responses, resulting in different outcomes of fibrosis or tissue regeneration. With the advancement of technology, emerging techniques such as scRNA-seq provide an unprecedented understanding of immune cell heterogeneity and plasticity in a scaffold-induced immune micro-environment at high resolution. The in-depth understanding of the interaction between scaffolds and the host immune system helps to provide clues for the design of biomaterials to optimize regeneration and promote a pro-regenerative local immune micro-environment.
Collapse
Affiliation(s)
- Nanyan Bian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chenyu Chu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Shengan Rung
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Vicha Huangphattarakul
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Jie Lin
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| | - Chen Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 14#, 3rd section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
152
|
Nandi D, Forster J, Ramesh A, Nguyen A, Bharadwaj H, Kulkarni A. Nanoreporter for Real-Time Monitoring of Inflammasome Activity and Targeted Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204900. [PMID: 36603165 PMCID: PMC9951342 DOI: 10.1002/advs.202204900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/18/2022] [Indexed: 06/17/2023]
Abstract
Inflammasome activation is associated with a myriad of inflammatory diseases. However, existing methods provides a limited understanding of spatiotemporal kinetics of inflammasome activation, with restricted scope for early detection of associated treatment efficacy. This limitation offers an opportunity for the development of biocompatible in-vivo inflammasome monitoring tools with translational prospects. To achieve this, they report developing a pair of lipid-based nanoparticle systems, a reporter nanoparticle consisting of a caspase-1 activatable probe alone, and a theranostic nanoparticle combining the probe with an inflammasome-inhibiting drug. This biocompatible platform enhances the probe's residence time in circulation by preventing its opsonization and allowing its sustained release over time. Their results demonstrate the specificity of reporter nanoparticles towards caspase-1 activity and provides early-on monitoring of inflammasome activation both in-vitro as well as in-vivo. Additionally, the delivery of disulfiram, an inflammasome-inhibiting drug, along with reporter probe using theranostic nanoparticles enables real-time tracking of treatment efficacy in the gouty-arthritis inflammatory model. In summary, they report an unparalleled pair of the inflammasome-associated reporter and theranostic platforms suited not only for diagnostic applications but can also detect inflammasome-targeted treatment efficiency in real-time. These findings establish two novel, sensitive nanotools for non-invasive evaluation of inflammasome-targeted immunotherapy.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Veterinary and Animal SciencesUniversity of MassachusettsAmherstMA01003USA
| | - James Forster
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Anujan Ramesh
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Biomedical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Anh Nguyen
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Hariharan Bharadwaj
- Department of PathologyUMass ChanMedical School‐BaystateSpringfieldMA01107USA
| | - Ashish Kulkarni
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Veterinary and Animal SciencesUniversity of MassachusettsAmherstMA01003USA
- Department of Biomedical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Center for Bioactive DeliveryInstitute for Applied Life SciencesUniversity of MassachusettsAmherstMA01003USA
| |
Collapse
|
153
|
Liu C, Liu Y, Xi L, He Y, Liang Y, Mak JCW, Mao S, Wang Z, Zheng Y. Interactions of Inhaled Liposome with Macrophages and Neutrophils Determine Particle Biofate and Anti-Inflammatory Effect in Acute Lung Inflammation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:479-493. [PMID: 36583377 DOI: 10.1021/acsami.2c17660] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Since most current studies have focused on exploring how phagocyte internalization of drug-loaded nanovesicles by macrophages would affect the function and therapeutic effects of infiltrated neutrophils or monocytes, research has evaluated the specificity of the inhaled nanovesicles for targeting various phagocytes subpopulations. In this study, liposomes with various charges (including neutral (L1), anionic (L2), and cationic at inflammatory sites (L3)) were constructed to investigate how particle charge determined their interactions with key phagocytes (including macrophages and neutrophils) in acute lung injury (ALI) models and to establish correlations with their biofate and overall anti-inflammatory effect. Our results clearly indicated that neutrophils were capable of rapidly sequestering L3 with a 3.2-fold increase in the cellular liposome distribution, compared to that in AMs, while 70.5% of L2 were preferentially uptaken by alveolar macrophages (AMs). Furthermore, both AMs and the infiltrated neutrophils performed as the potential vesicles for the inhaled liposomes to prolong their lung retention in ALI models, whereas AMs function as sweepers to recognize and process liposomes in the healthy lung. Finally, inhaled roflumilast-loaded macrophage or neutrophil preferential liposomes (L2 or L3) exhibited optimal anti-inflammatory effect because of the decreased AMs phagocytic capacity or the prolonged circulation times of neutrophils. Such findings will be beneficial in exploiting a potential pathway to specifically manipulate lung phagocyte functions in lung inflammatory diseases where these cells play crucial roles.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau999078, China
| | - Yihan Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau999078, China
| | - Long Xi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau999078, China
| | - Yuan He
- Department of Pharmacy, Xuzhou Medical University, Xuzhou221004, China
| | - Yingmin Liang
- School of Clinical Medicine, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
| | - Judith Choi Wo Mak
- School of Clinical Medicine, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, China
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, San Diego, California92093, United States
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau999078, China
| |
Collapse
|
154
|
Li J, Fan J, Gao Y, Huang S, Huang D, Li J, Wang X, Santos HA, Shen P, Xia B. Porous Silicon Nanocarriers Boost the Immunomodulation of Mitochondria-Targeted Bovine Serum Albumins on Macrophage Polarization. ACS NANO 2023; 17:1036-1053. [PMID: 36598186 PMCID: PMC9878978 DOI: 10.1021/acsnano.2c07439] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/29/2022] [Indexed: 05/31/2023]
Abstract
The development of nanosystems with intrinsic immunomodulatory effects on macrophage polarization is important for the macrophage-targeted immunotherapy. Here, mitochondria-targeted bovine serum albumins (BSAs) via the conjugation of fluorescent, lipophilic, and cationic rhodamine 110 molecules can efficiently enhance the gene expression of the proinflammatory phenotype of macrophages and correspondingly inhibit the gene expression of their anti-inflammatory phenotype. On this basis, porous silicon nanocarriers can further boost the immunomodulation of these mitochondria-targeted BSAs in vitro or in vivo, accompanied by the secretion of proinflammatory mediators including tumor necrosis factor α, nitric oxide, and reactive oxygen species (ROS). Meanwhile, BSA coatings can also improve the biocompatibility of porous silicon nanoparticulate cores on macrophages. Finally, the mechanism investigations demonstrate that porous silicon nanocarriers can efficiently deliver mitochondria-targeted BSA into macrophages to generate mitochondrial ROS via the interference with mitochondrial respiratory chains, which can further trigger the downstream signaling transduction pathways for the proinflammatory transition. Considering the good biosafety and versatile loading capability, this developed porous silicon@BSA nanosystem with a strong proinflmmatory regulatory effect has important potential on the combinatorial chemoimmunotherapy against cancer or viral/bacterial-related infectious diseases.
Collapse
Affiliation(s)
- Jialiang Li
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Jiqiang Fan
- State
Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive
Cancer Center, Nanjing Drum Tower Hospital, The Affiliated Hospital
of Nanjing University Medical School, Nanjing
University, Nanjing210023, China
| | - Yan Gao
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Shuodan Huang
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Di Huang
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Jiachen Li
- Department
of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AVGroningen, The Netherlands
- W.
J. Kolff Institute for Biomedical Engineering and Materials Science,
University Medical Center Groningen, University
of Groningen, Antonius
Deusinglaan 1, 9713 AVGroningen, The Netherlands
| | - Xiaoyu Wang
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| | - Hélder A. Santos
- Department
of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AVGroningen, The Netherlands
- W.
J. Kolff Institute for Biomedical Engineering and Materials Science,
University Medical Center Groningen, University
of Groningen, Antonius
Deusinglaan 1, 9713 AVGroningen, The Netherlands
| | - Pingping Shen
- Department
of Geriatric Medicine, The Second Affiliated
Hospital and Yuying Children’s Hospital of Wenzhou Medical
University, Wenzhou325027, China
- State
Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive
Cancer Center, Nanjing Drum Tower Hospital, The Affiliated Hospital
of Nanjing University Medical School, Nanjing
University, Nanjing210023, China
| | - Bing Xia
- College
of Science, Nanjing Forestry University, Nanjing210037, China
| |
Collapse
|
155
|
Li H, Luo Q, Zhang H, Ma X, Gu Z, Gong Q, Luo K. Nanomedicine embraces cancer radio-immunotherapy: mechanism, design, recent advances, and clinical translation. Chem Soc Rev 2023; 52:47-96. [PMID: 36427082 DOI: 10.1039/d2cs00437b] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cancer radio-immunotherapy, integrating external/internal radiation therapy with immuno-oncology treatments, emerges in the current management of cancer. A growing number of pre-clinical studies and clinical trials have recently validated the synergistic antitumor effect of radio-immunotherapy, far beyond the "abscopal effect", but it suffers from a low response rate and toxicity issues. To this end, nanomedicines with an optimized design have been introduced to improve cancer radio-immunotherapy. Specifically, these nanomedicines are elegantly prepared by incorporating tumor antigens, immuno- or radio-regulators, or biomarker-specific imaging agents into the corresponding optimized nanoformulations. Moreover, they contribute to inducing various biological effects, such as generating in situ vaccination, promoting immunogenic cell death, overcoming radiation resistance, reversing immunosuppression, as well as pre-stratifying patients and assessing therapeutic response or therapy-induced toxicity. Overall, this review aims to provide a comprehensive landscape of nanomedicine-assisted radio-immunotherapy. The underlying working principles and the corresponding design strategies for these nanomedicines are elaborated by following the concept of "from bench to clinic". Their state-of-the-art applications, concerns over their clinical translation, along with perspectives are covered.
Collapse
Affiliation(s)
- Haonan Li
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China.
| | - Qiang Luo
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China.
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Xuelei Ma
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China.
| | - Zhongwei Gu
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China.
| | - Qiyong Gong
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, 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
| | - Kui Luo
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, 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
|
156
|
Feng W, Chittò M, Moriarty TF, Li G, Wang X. Targeted Drug Delivery Systems for Eliminating Intracellular Bacteria. Macromol Biosci 2023; 23:e2200311. [PMID: 36189899 DOI: 10.1002/mabi.202200311] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Indexed: 01/19/2023]
Abstract
The intracellular survival of pathogenic bacteria requires a range of survival strategies and virulence factors. These infections are a significant clinical challenge, wherein treatment frequently fails because of poor antibiotic penetration, stability, and retention in host cells. Drug delivery systems (DDSs) are promising tools to overcome these shortcomings and enhance the efficacy of antibiotic therapy. In this review, the classification and the mechanisms of intracellular bacterial persistence are elaborated. Furthermore, the systematic design strategies applied to DDSs to eliminate intracellular bacteria are also described, and the strategies used for internalization, intracellular activation, bacterial targeting, and immune enhancement are highlighted. Finally, this overview provides guidance for constructing functionalized DDSs to effectively eliminate intracellular bacteria.
Collapse
Affiliation(s)
- Wenli Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.,AO Research Institute Davos, Davos, 7270, Switzerland
| | - Marco Chittò
- AO Research Institute Davos, Davos, 7270, Switzerland
| | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
157
|
Zhang X, Bai W, Hu L, Ha H, Du Y, Xiong W, Wang H, Shang P. The pleiotropic mode and molecular mechanism of macrophages in promoting tumor progression and metastasis. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:91-104. [PMID: 36071369 DOI: 10.1007/s12094-022-02932-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
Macrophages are the most abundant immune cells in primary and metastatic tumor tissues. Studies have shown that macrophages mainly exhibit a tumor-promoting phenotype and play a key role in tumor progression and metastasis. Therefore, many macrophage-targeted drugs have entered clinical trials. However, compared to preclinical studies, some clinical trial results showed that macrophage-targeted therapy did not achieve the desired effect. This may be because most of what we know about macrophages comes from in vitro experiments and animal models, while macrophages in the more complex human microenvironment are still poorly understood. With the development of technologies such as single-cell RNA sequencing, we have gained a new understanding of the origin, classification and functional mechanism of tumor-associated macrophages. Therefore, this study reviewed the recent progress of macrophages in promoting tumor progression and metastasis, aiming to provide some help for the formulation of optimal strategies for macrophage-targeted therapy.
Collapse
Affiliation(s)
- Xingxing Zhang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Wenxiu Bai
- Ultrasonic Special Examination Department, Tai An TSCM Hospital, Taian, 271000, Shandong, China
| | - Lisha Hu
- Ultrasonic Special Examination Department, Tai An TSCM Hospital, Taian, 271000, Shandong, China
| | - Hualan Ha
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Yuelin Du
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Wei Xiong
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Hongbo Wang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Panfeng Shang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
158
|
Xuan Y, Li L, Zhang C, Zhang M, Cao J, Zhang Z. The 3D-Printed Ordered Bredigite Scaffold Promotes Pro-Healing of Critical-Sized Bone Defects by Regulating Macrophage Polarization. Int J Nanomedicine 2023; 18:917-932. [PMID: 36844434 PMCID: PMC9951604 DOI: 10.2147/ijn.s393080] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/29/2023] [Indexed: 02/22/2023] Open
Abstract
Background Repairing critical-sized bone defects secondary to traumatic or tumorous damage is a complex conundrum in clinical practice; in this case, artificial scaffolds exhibited preferable outcomes. Bredigite (BRT, Ca7MgSi4O16) bioceramic possesses excellent physicochemical properties and biological activity as a promising candidate for bone tissue engineering. Methods Structurally ordered BRT (BRT-O) scaffolds were fabricated by a three-dimensional (3D) printing technique, and the random BRT (BRT-R) scaffolds and clinically available β-tricalcium phosphate (β-TCP) scaffolds were compared as control groups. Their physicochemical properties were characterized, and RAW 264.7 cells, bone marrow mesenchymal stem cells (BMSCs), and rat cranial critical-sized bone defect models were utilized for evaluating macrophage polarization and bone regeneration. Results The BRT-O scaffolds exhibited regular morphology and homogeneous porosity. In addition, the BRT-O scaffolds released higher concentrations of ionic products based on coordinated biodegradability than the β-TCP scaffolds. In vitro, the BRT-O scaffolds facilitated RWA264.7 cells polarization to pro-healing M2 macrophage phenotype, whereas the BRT-R and β-TCP scaffolds stimulated more pro-inflammatory M1-type macrophages. A conditioned medium derived from macrophages seeding on the BRT-O scaffolds notably promoted the osteogenic lineage differentiation of BMSCs in vitro. The cell migration ability of BMSCs was significantly enhanced under the BRT-O-induced immune microenvironment. Moreover, in rat cranial critical-sized bone defect models, the BRT-O scaffolds group promoted new bone formation with a higher proportion of M2-type macrophage infiltration and expression of osteogenesis-related markers. Therefore, in vivo, BRT-O scaffolds play immunomodulatory roles in promoting critical-sized bone defects by enhancing the polarization of M2 macrophages. Conclusion 3D-printed BRT-O scaffolds can be a promising option for bone tissue engineering, at least partly through macrophage polarization and osteoimmunomodulation.
Collapse
Affiliation(s)
- Yaowei Xuan
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lin Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chenping Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Min Zhang
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Junkai Cao
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhen Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| |
Collapse
|
159
|
Lou T, Chen K, Luo Q, Liu C, Yuan Y, Fan C. Periosteum-inspired in situ CaP generated nanocomposite hydrogels with strong bone adhesion and superior stretchability for accelerated distraction osteogenesis. Biomater Res 2022; 26:91. [PMID: 36581951 PMCID: PMC9801553 DOI: 10.1186/s40824-022-00330-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/27/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Distraction osteogenesis (DO) is an efficacious but lengthy procedure to reconstruct segmental bone defects under the principle of tension-stress, during which the periosteum-mediated mechanical stimulation plays a pivotal role. Inspired by the dynamic process of DO and the mechanical stimulation of periosteum, a new design of bionic periosteum was developed to simulate the mechanical transduction of natural periosteum for the application in DO procedure. METHODS In this study, an injectable organic-inorganic hybrid hydrogel was developed based on a novel combination of the PEGylated poly (glycerol sebacate) (PEGS) polymer network and in situ formed CaP nanoparticles (ICPNs). Rat bone marrow mesenchymal stem cells (rBMSCs) and human umbilical vein endothelial cells (HUVECs) were cultured and tested in vitro to evaluate biocompatibility, cell adhesion, proliferation, and pro-osteogenic and pro-angiogenic activity. In vivo experiments were conducted in the rat tibial model of distraction osteogenesis. RESULTS The developed nanocomposite hydrogels exhibited excellent injectability, robust bone adhesion, superior stretchability, and enhanced osteogenic activity. The results of in vitro and in vivo studies showed that PEGS/ICPN hydrogels could promote new bone formation and mineralization during the dynamic distraction process through the synergistic effects of angiogenesis and osteogenesis. CONCLUSIONS This periosteum-inspired nanocomposite hydrogel represents a mechanobiology approach for effectively restoring large bone defects through the dynamic DO process.
Collapse
Affiliation(s)
- Tengfei Lou
- grid.412528.80000 0004 1798 5117Orthopaedic Department, Shanghai Sixth People’s Hospital, Shanghai, 200233 People’s Republic of China
| | - Kai Chen
- grid.28056.390000 0001 2163 4895Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China ,grid.28056.390000 0001 2163 4895Frontiers Science Center for Materiobiology and Dynamic Chemistry, and Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China
| | - Qiyu Luo
- grid.412528.80000 0004 1798 5117Orthopaedic Department, Shanghai Sixth People’s Hospital, Shanghai, 200233 People’s Republic of China
| | - Changsheng Liu
- grid.28056.390000 0001 2163 4895Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China ,grid.28056.390000 0001 2163 4895Frontiers Science Center for Materiobiology and Dynamic Chemistry, and Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China
| | - Yuan Yuan
- grid.28056.390000 0001 2163 4895Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China ,grid.28056.390000 0001 2163 4895Frontiers Science Center for Materiobiology and Dynamic Chemistry, and Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237 People’s Republic of China
| | - Cunyi Fan
- grid.412528.80000 0004 1798 5117Orthopaedic Department, Shanghai Sixth People’s Hospital, Shanghai, 200233 People’s Republic of China
| |
Collapse
|
160
|
Dong J, Wang W, Zhou W, Zhang S, Li M, Li N, Pan G, Zhang X, Bai J, Zhu C. Immunomodulatory biomaterials for implant-associated infections: from conventional to advanced therapeutic strategies. Biomater Res 2022; 26:72. [PMID: 36471454 PMCID: PMC9721013 DOI: 10.1186/s40824-022-00326-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/19/2022] [Indexed: 12/11/2022] Open
Abstract
Implant-associated infection (IAI) is increasingly emerging as a serious threat with the massive application of biomaterials. Bacteria attached to the surface of implants are often difficult to remove and exhibit high resistance to bactericides. In the quest for novel antimicrobial strategies, conventional antimicrobial materials often fail to exert their function because they tend to focus on direct bactericidal activity while neglecting the modulation of immune systems. The inflammatory response induced by host immune cells was thought to be a detrimental force impeding wound healing. However, the immune system has recently received increasing attention as a vital player in the host's defense against infection. Anti-infective strategies based on the modulation of host immune defenses are emerging as a field of interest. This review explains the importance of the immune system in combating infections and describes current advanced immune-enhanced anti-infection strategies. First, the characteristics of traditional/conventional implant biomaterials and the reasons for the difficulty of bacterial clearance in IAI were reviewed. Second, the importance of immune cells in the battle against bacteria is elucidated. Then, we discuss how to design biomaterials that activate the defense function of immune cells to enhance the antimicrobial potential. Based on the key premise of restoring proper host-protective immunity, varying advanced immune-enhanced antimicrobial strategies were discussed. Finally, current issues and perspectives in this field were offered. This review will provide scientific guidance to enhance the development of advanced anti-infective biomaterials.
Collapse
Affiliation(s)
- Jiale Dong
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Wenzhi Wang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Wei Zhou
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Siming Zhang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Meng Li
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China ,grid.263761.70000 0001 0198 0694Medical College, Soochow University, 215006 Suzhou, Jiangsu P. R. China
| | - Ning Li
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Guoqing Pan
- grid.440785.a0000 0001 0743 511XInstitute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 212013 Zhenjiang, China
| | - Xianzuo Zhang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Jiaxiang Bai
- grid.263761.70000 0001 0198 0694Medical College, Soochow University, 215006 Suzhou, Jiangsu P. R. China
| | - Chen Zhu
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| |
Collapse
|
161
|
Kim Y, Thangam R, Yoo J, Heo J, Park JY, Kang N, Lee S, Yoon J, Mun KR, Kang M, Min S, Kim SY, Son S, Kim J, Hong H, Bae G, Kim K, Lee S, Yang L, Lee JY, Kim J, Park S, Kim DH, Lee KB, Jang WY, Kim BH, Paulmurugan R, Cho SW, Song HC, Kang SJ, Sun W, Zhu Y, Lee J, Kim HJ, Jang HS, Kim JS, Khademhosseini A, Kim Y, Kim S, Kang H. Photoswitchable Microgels for Dynamic Macrophage Modulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2205498. [PMID: 36268986 DOI: 10.1002/adma.202205498] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/22/2022] [Indexed: 06/16/2023]
Abstract
Dynamic manipulation of supramolecular self-assembled structures is achieved irreversibly or under non-physiological conditions, thereby limiting their biomedical, environmental, and catalysis applicability. In this study, microgels composed of azobenzene derivatives stacked via π-cation and π-π interactions are developed that are electrostatically stabilized with Arg-Gly-Asp (RGD)-bearing anionic polymers. Lateral swelling of RGD-bearing microgels occurs via cis-azobenzene formation mediated by near-infrared-light-upconverted ultraviolet light, which disrupts intermolecular interactions on the visible-light-absorbing upconversion-nanoparticle-coated materials. Real-time imaging and molecular dynamics simulations demonstrate the deswelling of RGD-bearing microgels via visible-light-mediated trans-azobenzene formation. Near-infrared light can induce in situ swelling of RGD-bearing microgels to increase RGD availability and trigger release of loaded interleukin-4, which facilitates the adhesion structure assembly linked with pro-regenerative polarization of host macrophages. In contrast, visible light can induce deswelling of RGD-bearing microgels to decrease RGD availability that suppresses macrophage adhesion that yields pro-inflammatory polarization. These microgels exhibit high stability and non-toxicity. Versatile use of ligands and protein delivery can offer cytocompatible and photoswitchable manipulability of diverse host cells.
Collapse
Affiliation(s)
- Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Institute for High Technology Materials and Devices, Korea University, Seoul, 02841, Republic of Korea
| | - Jounghyun Yoo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jeongyun Heo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jung Yeon Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jiwon Yoon
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Kwang Rok Mun
- Materials Architecturing Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Misun Kang
- Materials Architecturing Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Seong Yeol Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Subin Son
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Jihwan Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Gunhyu Bae
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Kanghyeon Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sanghyeok Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Ja Yeon Lee
- Materials Architecturing Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jinjoo Kim
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Steve Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Bong Hoon Kim
- Daegu Gyeongbuk Institute of Science and Technology (DGIST), Department of Robotics and Mechatronics Engineering, Daegu, 42988, Republic of Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
| | - Hyun-Cheol Song
- Electronic Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KIST-SKKU Carbon-Neutral Research Center, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Seok Ju Kang
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Wujin Sun
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Ho Seong Jang
- Materials Architecturing Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Nano & Information Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Yongju Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Sehoon Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
162
|
Wang P, Wang X. Mimicking the native bone regenerative microenvironment for in situ repair of large physiological and pathological bone defects. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
163
|
Zhang W, Xia S, Weng T, Yang M, Shao J, Zhang M, Wang J, Xu P, Wei J, Jin R, Yu M, Zhang Z, Han C, Wang X. Antibacterial coaxial hydro-membranes accelerate diabetic wound healing by tuning surface immunomodulatory functions. Mater Today Bio 2022; 16:100395. [PMID: 36042855 PMCID: PMC9420385 DOI: 10.1016/j.mtbio.2022.100395] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/19/2022] Open
Abstract
Diabetic foot ulcers, typical non-healing wounds, represent a severe clinical problem. Advanced glycation end-products (AGEs), which create a prolonged pro-inflammatory micro-environment in defective sites, can be responsible for refractoriness of these ulcers. Macrophages are polarized to the M2 phenotype to facilitate the transition from a pro-inflammatory microenvironment to an anti-inflammatory microenvironment, which has been demonstrated to be an effective way to accelerate diabetic wound closure. Herein, we developed coaxial hydro-membranes mimicking the extracellular matrix structure that are capable of anti-inflammatory and antibacterial functions for diabetic wound repair. These fibrous membranes maintain a moist microenvironment to support cell proliferation. Macrophages grow in an elongated shape on the surface of the fibrous membranes. The fibrous membranes effectively impaired macrophage AGE-induced M1 polarization and induced macrophage polarization towards the M2 phenotype. The effects of the fibrous membranes on the interactions between macrophages and repair cells under a diabetic condition were also investigated. Furthermore, in vivo results from a full-thickness diabetic wound model confirmed the potential of the coaxial hydro-membranes to accelerate wound healing. This study's results indicate that the developed bioactive anti-inflammatory and antibacterial wound dressing can affect AGE-induced macrophage activation and crosstalk between macrophages and fibroblasts for treating diabetic wounds.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Sizhan Xia
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Tingting Weng
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Min Yang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Jiaming Shao
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Manjia Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jialiang Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Pengqing Xu
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Jintao Wei
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
- Department of Emergency Medicine, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- Institute of Emergency Medicine, Zhejiang University, Hangzhou, 310000, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Jiefang Road 88, Hangzhou, 310009, China
| | - Ronghua Jin
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Meirong Yu
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Zhongtao Zhang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Chunmao Han
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
| | - Xingang Wang
- Department of Burns and Wound Care Center, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- The Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310000, China
- Corresponding author. Department of Burns & Wound Care Center, the Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China.
| |
Collapse
|
164
|
Li Y, Yang L, Hou Y, Zhang Z, Chen M, Wang M, Liu J, Wang J, Zhao Z, Xie C, Lu X. Polydopamine-mediated graphene oxide and nanohydroxyapatite-incorporated conductive scaffold with an immunomodulatory ability accelerates periodontal bone regeneration in diabetes. Bioact Mater 2022; 18:213-227. [PMID: 35387166 PMCID: PMC8961429 DOI: 10.1016/j.bioactmat.2022.03.021] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/22/2022] [Accepted: 03/12/2022] [Indexed: 12/11/2022] Open
Abstract
Regenerating periodontal bone tissues in the aggravated inflammatory periodontal microenvironment under diabetic conditions is a great challenge. Here, a polydopamine-mediated graphene oxide (PGO) and hydroxyapatite nanoparticle (PHA)-incorporated conductive alginate/gelatin (AG) scaffold is developed to accelerate periodontal bone regeneration by modulating the diabetic inflammatory microenvironment. PHA confers the scaffold with osteoinductivity and PGO provides a conductive pathway for the scaffold. The conductive scaffold promotes bone regeneration by transferring endogenous electrical signals to cells and activating Ca2+ channels. Moreover, the scaffold with polydopamine-mediated nanomaterials has a reactive oxygen species (ROS)-scavenging ability and anti-inflammatory activity. It also exhibits an immunomodulatory ability that suppresses M1 macrophage polarization and activates M2 macrophages to secrete osteogenesis-related cytokines by mediating glycolytic and RhoA/ROCK pathways in macrophages. The scaffold induces excellent bone regeneration in periodontal bone defects of diabetic rats because of the synergistic effects of good conductive, ROS-scavenging, anti-inflammatory, and immunomodulatory abilities. This study provides fundamental insights into the synergistical effects of conductivity, osteoinductivity, and immunomodulatory abilities on bone regeneration and offers a novel strategy to design immunomodulatory biomaterials for treatment of immune-related diseases and tissue regeneration. The conductive PGO-PHA-AG scaffold can activate Ca2+ channels. •The PGO-PHA-AG scaffold had ROS-scavenging and anti-inflammatory activities. •The scaffold exhibited an immunomodulatory ability. •The scaffold induced excellent periodontal bone regeneration in diabetes.
Collapse
Affiliation(s)
- Yazhen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lu Yang
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yue Hou
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Maoxia Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jin Liu
- Lab for Aging Research and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Corresponding author.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Corresponding author.
| | - Chaoming Xie
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Corresponding author.
| | - Xiong Lu
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- Corresponding author.
| |
Collapse
|
165
|
Lian Q, Zheng S, Shi Z, Li K, Chen R, Wang P, Liu H, Chen Y, Zhong Q, Liu Q, Pan X, Gao J, Gao C, Liu W, Wu X, Zhang Y, Zhang Y, Wang J, Cheng H. Using a degradable three-layer sandwich-type coating to prevent titanium implant infection with the combined efficient bactericidal ability and fast immune remodeling property. Acta Biomater 2022; 154:650-666. [PMID: 36306986 DOI: 10.1016/j.actbio.2022.10.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/18/2022] [Accepted: 10/14/2022] [Indexed: 01/24/2023]
Abstract
Titanium (Ti) implant-associated infections are a challenge in orthopedic surgery, for which a series of antibacterial coatings have been designed and fabricated to reduce the risk of bacterial contamination. Herein, we created a degradable three-layer sandwich-type coating to achieve long-term antibacterial effects while simultaneously reconstructing the local immune microenvironment. The vancomycin (Van)-loaded vaterite coating constitutes the outer and inner layers, whereas Interleukin-12 (IL-12)-containing liposomes embedded in sodium alginate constitutes the middle layer. Van, released from the vaterite, demonstrated a favorable and rapid bactericidal ability against the representative methicillin-sensitive S. aureus (MSSA) and methicillin-resistant S. aureus (MRSA) strains. The released IL-12 exhibited the desired immune reconstitution abilities, actively facilitating defenses against subsequent bacterial invasions. Furthermore, the biocompatibility and cell-binding feature of the multifunctional coating was beneficial for achieving solid interface intergradation. Overall, the benefits of the three-layer sandwich-type coating, including the convenient fabrication process, efficient antimicrobial activity, fast immune remodeling property, fine cell-binding feature, and biodegradability, highlight its promising translational potential in preventing implant infection. STATEMENT OF SIGNIFICANCE: To prevent titanium implant infections, researchers have designed various antibacterial coatings. However, most of these coatings focused only on killing the invading bacteria over a limited postoperative period. However, the local immune microenvironment is compromised during surgery. Local immune deflection impedes the ability of the local immune defenses to clear bacteria and limits immune memory building from active defense against long-term subsequent bacterial invasions. Furthermore, these coatings are usually nondegradable and differ substantially from bone components, thereby impairing the integration of the coating and bone interface and generating concerns about implant stability and bacterial contamination. In this work, we synthesized a degradable coating that provides sustained antibacterial activity, promotes immune reconstitution, and simultaneously achieves solid bone integration.
Collapse
Affiliation(s)
- Qiang Lian
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shaowei Zheng
- Department of Orthopedic, Huizhou First Hospital, Guangdong Medical University, Huizhou 516003, China; Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhe Shi
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kangxian Li
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Rong Chen
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Pinkai Wang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Haibing Liu
- Department of Orthopedic, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang 421001, China
| | - Yuhang Chen
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiang Zhong
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qi Liu
- South China Advanced Institute for Soft Matter Science and Technology, South China University of Technology, Guangzhou 510640, China
| | - Xin Pan
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Gao
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chenghao Gao
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 43000, China
| | - Weilu Liu
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xuanpin Wu
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yayun Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 43000, China
| | - Yang Zhang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Jian Wang
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Hao Cheng
- Department of Orthopedic, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
166
|
Zeng J, Sun Y, Sun S, Jiang M, Zhang D, Li W, Liu Z, Shang H, Guan X, Zhang W. Leveraging Nanodrug Delivery System for Simultaneously Targeting Tumor Cells and M2 Tumor-Associated Macrophages for Efficient Colon Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50475-50484. [PMID: 36327132 DOI: 10.1021/acsami.2c11534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Tumor-associated macrophages (TAMs) widely exist in the solid tumors, which participate in the entire course of tumor development and execute momentous impacts. Therefore, manipulating TAMs has been identified as an expecting strategy with immense potential for cancer therapy. Herein, a nanodrug delivery system was leveraged for simultaneously targeting tumor cells and M2-type TAMs for efficient colon cancer therapy. The broad-spectrum anticancer chemotherapeutic drug doxorubicin (DOX) was hitchhiked in a mannose-modified bovine serum albumin (MAN-BSA) carrier. The DOX@MAN-BSA nanodrug delivery system was verified to possess feasible physical performances for unhindered systemic circulation and active targeting on colon tumors. DOX@MAN-BSA nanoparticles could be preferentially swallowed by colon tumor cells and M2 TAMs through mannose receptor-mediated endocytosis. Further in vivo antitumor therapy in CT26 colon tumor-bearing mice has achieved remarkable suppression efficacy with satisfactory biosafety. Leveraging the nanodrug delivery system for simultaneously targeting tumor cells and M2 TAMs has contributed a feasible strategy to collaboratively repress the malignant tumor cells and the collusive M2 TAMs for efficient cancer therapy.
Collapse
Affiliation(s)
- Jun Zeng
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Yanju Sun
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Shuo Sun
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Mingxia Jiang
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Daijuan Zhang
- Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - Wentong Li
- Department of Pathology, Weifang Medical University, Weifang 261053, China
| | - Zhijun Liu
- Department of Medical Microbiology, Weifang Medical University, Weifang 261053, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiuwen Guan
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Weifen Zhang
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
167
|
Liu X, Li Y, Sun Y, Chen B, Du W, Li Y, Gu N. Construction of functional magnetic scaffold with temperature control switch for long-distance vascular injury. Biomaterials 2022; 290:121862. [DOI: 10.1016/j.biomaterials.2022.121862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 10/01/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022]
|
168
|
Zhong JX, Raghavan P, Desai TA. Harnessing Biomaterials for Immunomodulatory-Driven Tissue Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022; 9:224-239. [PMID: 37333620 PMCID: PMC10272262 DOI: 10.1007/s40883-022-00279-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/08/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022]
Abstract
Abstract The immune system plays a crucial role during tissue repair and wound healing processes. Biomaterials have been leveraged to assist in this in situ tissue regeneration process to dampen the foreign body response by evading or suppressing the immune system. An emerging paradigm within regenerative medicine is to use biomaterials to influence the immune system and create a pro-reparative microenvironment to instigate endogenously driven tissue repair. In this review, we discuss recent studies that focus on immunomodulation of innate and adaptive immune cells for tissue engineering applications through four biomaterial-based mechanisms of action: biophysical cues, chemical modifications, drug delivery, and sequestration. These materials enable augmented regeneration in various contexts, including vascularization, bone repair, wound healing, and autoimmune regulation. While further understanding of immune-material interactions is needed to design the next generation of immunomodulatory biomaterials, these materials have already demonstrated great promise for regenerative medicine. Lay Summary The immune system plays an important role in tissue repair. Many biomaterial strategies have been used to promote tissue repair, and recent work in this area has looked into the possibility of doing repair by tuning. Thus, we examined the literature for recent works showcasing the efficacy of these approaches in animal models of injuries. In these studies, we found that biomaterials successfully tuned the immune response and improved the repair of various tissues. This highlights the promise of immune-modulating material strategies to improve tissue repair.
Collapse
Affiliation(s)
- Justin X. Zhong
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
| | - Preethi Raghavan
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
| | - Tejal A. Desai
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
- Department of Bioengineering, University of California, Berkeley, CA 94720 USA
- School of Engineering, Brown University, Providence, RI 02912 USA
| |
Collapse
|
169
|
Lin CW, Hung CM, Chen WJ, Chen JC, Huang WY, Lu CS, Kuo ML, Chen SG. New Horizons of Macrophage Immunomodulation in the Healing of Diabetic Foot Ulcers. Pharmaceutics 2022; 14:pharmaceutics14102065. [PMID: 36297499 PMCID: PMC9606988 DOI: 10.3390/pharmaceutics14102065] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetic foot ulcers (DFUs) are one of the most costly and troublesome complications of diabetes mellitus. The wound chronicity of DFUs remains the main challenge in the current and future treatment of this condition. Persistent inflammation results in chronic wounds characterized by dysregulation of immune cells, such as M1 macrophages, and impairs the polarization of M2 macrophages and the subsequent healing process of DFUs. The interactive regulation of M1 and M2 macrophages during DFU healing is critical and seems manageable. This review details how cytokines and signalling pathways are co-ordinately regulated to control the functions of M1 and M2 macrophages in normal wound repair. DFUs are defective in the M1-to-M2 transition, which halts the whole wound-healing machinery. Many pre-clinical and clinical innovative approaches, including the application of topical insulin, CCL chemokines, micro RNAs, stem cells, stem-cell-derived exosomes, skin substitutes, antioxidants, and the most recent Phase III-approved ON101 topical cream, have been shown to modulate the activity of M1 and M2 macrophages in DFUs. ON101, the newest clinically approved product in this setting, is designed specifically to down-regulate M1 macrophages and further modulate the wound microenvironment to favour M2 emergence and expansion. Finally, the recent evolution of macrophage modulation therapies and techniques will improve the effectiveness of the treatment of diverse DFUs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ming-Liang Kuo
- Microbio Co., Ltd., Taipei 115, Taiwan
- Correspondence: (M.-L.K.); or (S.-G.C.); Tel.: +886-2-27031298 (ext. 550) (M.-L.K.); +886-2-27031098 (ext. 551) (S.-G.C.)
| | - Shyi-Gen Chen
- Oneness Biotech Co., Ltd., Taipei 106, Taiwan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: (M.-L.K.); or (S.-G.C.); Tel.: +886-2-27031298 (ext. 550) (M.-L.K.); +886-2-27031098 (ext. 551) (S.-G.C.)
| |
Collapse
|
170
|
Luo Y, Yuan P, Hu S, Wang H, Zhang H, Ma L. Inflammatory environment-adaptive patterned surface for spatiotemporal immunomodulation of macrophages. Acta Biomater 2022; 153:139-148. [PMID: 36167238 DOI: 10.1016/j.actbio.2022.09.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022]
Abstract
Designing biomaterials with precise immunomodulation can help to decipher the dynamic interactions between macrophages and biomaterials to match the tissue healing process. Although some advanced stimuli-responsive immunomodulatory biomaterials were reported for cell dynamic modulation, while most triggers need external stimuli by manual intervention, there would be the inevitable errors and uncertainties. Thus, developing immunomodulatory biomaterials with adaptive abilities, which can recognize the inflammation signals, change their properties spatiotemporally under the microenvironment triggers, and provide feedback to realize macrophages modulation in different healing stages, has become a promising strategy. In this work, we developed an inflammation-adaptive Arg-Gly-Asp (RGD) -patterned surface for spatiotemporal immunomodulation of macrophage. We fabricated a methacrylated hyaluronic acid (MA-HA) hydrogel with thiol-functionalized RGD-patterned surface by employing photolithography technology. Then, thiol-functionalized RGD contained ROS-cleavable linker was filled the remaining sites and consequently, a dynamic surface with temporary homogeneous RGD was obtained. Under the overproduction of ROS by the inflammation-activated macrophages, the linker was cleaved, and the homogeneous RGD surface was transformed to the RGD patterned surface, which triggered elongation of macrophages and consequently the upregulated expressions of arginase-1, IL-10 and TNF-β1, indicating the polarization toward to anti-inflammatory phenotype. Developing inflammatory environment-adaptive surface for spatiotemporal modulation of macrophages polarization provides a precise and smart strategy for the healing-matched immunomodulation to facilitate healing outcomes. STATEMENT OF SIGNIFICANCE: Designing biomaterials with precise immunomodulation can help to decipher the dynamic interactions between macrophages and biomaterials to match tissue repair process. Some immunomodulatory biomaterials were reported for cell dynamic modulation, while most triggers need external manual intervention. Thus, we developed an immunomodulatory biomaterial with inflammation-adaptive patterned surface, which can recognize abnormal signals and change its properties spatiotemporally under the microenvironment triggers, and provide feedback to realize macrophages modulation in different stages. The dynamic surface can adapt to the changes of microenvironment and dynamically to match the cell behavior and tissue healing process on demand without external manual intervention. Additionally, the surface achieves the balance of macrophages with pro- and anti-inflammatory phenotypes in the tissue repair process.
Collapse
Affiliation(s)
- Yilun Luo
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Peiqi Yuan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Sentao Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hanwen Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Haiqi Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
171
|
Huang D, Xu K, Huang X, Lin N, Ye Y, Lin S, Zhang J, Shao J, Chen S, Shi M, Zhou X, Lin P, Xue Y, Yu C, Yu X, Ye Z, Cheng K. Remotely Temporal Scheduled Macrophage Phenotypic Transition Enables Optimized Immunomodulatory Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203680. [PMID: 36031402 DOI: 10.1002/smll.202203680] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Indexed: 06/15/2023]
Abstract
Precise timing of macrophage polarization plays a pivotal role in immunomodulation of tissue regeneration, yet most studies mainly focus on M2 macrophages for their anti-inflammatory and regenerative effects while the essential proinflammatory role of the M1 phenotype on the early inflammation stage is largely underestimated. Herein, a superparamagnetic hydrogel capable of timely controlling macrophage polarization is constructed by grafting superparamagnetic nanoparticles on collagen nanofibers. The magnetic responsive hydrogel network enables efficient polarization of encapsulated macrophage to the M2 phenotype through the podosome/Rho/ROCK mechanical pathway in response to static magnetic field (MF) as needed. Taking advantage of remote accessibility of magnetic field together with the superparamagnetic hydrogels, a temporal engineered M1 to M2 transition course preserving the essential role of M1 at the early stage of tissue healing, as well as enhancing the prohealing effect of M2 at the middle/late stages is established via delayed MF switch. Such precise timing of macrophage polarization matching the regenerative process of injured tissue eventually leads to optimized immunomodulatory bone healing in vivo. Overall, this study offers a remotely time-scheduled approach for macrophage polarization, which enables precise manipulation of inflammation progression during tissue healing.
Collapse
Affiliation(s)
- Donghua Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Kaicheng Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Xin Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Nong Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Yuxiao Ye
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Suya Lin
- School of Material Science and Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Jiamin Zhang
- School of Material Science and Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Jiaqi Shao
- The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Mingmin Shi
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Xingzhi Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Peng Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Yucheng Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Chengcheng Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Xiaohua Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Zhaoming Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, 310000, P. R. China
| | - Kui Cheng
- School of Material Science and Engineering, University of New South Wales, Sydney, 2052, Australia
| |
Collapse
|
172
|
Xin P, Han S, Huang J, You X, Wu J. Natural Soybean Milk-Derived Bioactive Coatings for Enhanced Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2022; 14:34480-34487. [PMID: 35858126 DOI: 10.1021/acsami.2c09689] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Foodborne biomaterials, derived from diets, comprise selfassembled collections of many micro- or nanoscale units with abundant nutrients and active substances. In this study, soybean milk (SBM) was selected as a tissue engineering product for simple and feasible wound repair. SBM is a common drink prepared from soybeans and is rich in soy protein, soy isoflavones, and other bioactive components. Thus, SBM has substantial potential for antioxidation and tissue remodeling. Here, the multifunctional effect of SBM as a bioactive coating for promoting wound healing was studied. The results showed that SBM has good biocompatibility and biological activity. It efficiently scavenges intracellular reactive oxygen species, significantly enhances epithelial cell migration, and improves angiogenesis, thereby accelerating tissue remodeling. The results of animal experiments further confirmed that the SBM-bioinspired coating has promising applications for cutaneous wound regeneration.
Collapse
Affiliation(s)
- Peikun Xin
- School of Biomedical Engineering, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shuyan Han
- School of Biomedical Engineering, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510006, China
| | - Jun Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Xinru You
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Jun Wu
- School of Biomedical Engineering, State Key Laboratory of Oncology in South China, Sun Yat-Sen University, Guangzhou 510006, China
- RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| |
Collapse
|
173
|
Fan C, Xu Q, Hao R, Wang C, Que Y, Chen Y, Yang C, Chang J. Multi-functional wound dressings based on silicate bioactive materials. Biomaterials 2022; 287:121652. [PMID: 35785753 DOI: 10.1016/j.biomaterials.2022.121652] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/22/2022]
Abstract
Most traditional wound dressings passively offer a protective barrier for the wounds, which lacks the initiative in stimulating tissue regeneration. In addition, cutaneous wound healing is usually accompanied by various complicated conditions, including bacterial infection, skin cancer, and damaged skin appendages, bringing further challenges for wound management in clinic. Therefore, an ideal wound dressing should not only actively stimulate wound healing but also hold multi-functions for solving problems associated with different specific wound conditions. Recent studies have demonstrated that silicate bioceramics and bioglasses are one type of promising materials for the development of wound dressings, as they can actively accelerate wound healing by regulating endothelial cells, dermal fibroblasts, macrophages, and epidermal cells. In particular, silicate-based biomaterials can be further functionalized by specific structural design or doping with functional components, which endow materials with enhanced bioactivities or expanded physicochemical properties such as photothermal, photodynamic, chemodynamic, or imaging properties. The functionalized materials can be used to address wound healing with different demands including but not limited to antibacterial, anticancer, skin appendages regeneration, and wound monitoring. In this review, we summarized the current research on the development of silicate-based multi-functional wound dressings and prospected the development of advanced wound dressings in the future.
Collapse
Affiliation(s)
- Chen Fan
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Qing Xu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, PR China
| | - Ruiqi Hao
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Chun Wang
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Yumei Que
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Yanxin Chen
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Chen Yang
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China.
| | - Jiang Chang
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China; State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, PR China.
| |
Collapse
|
174
|
Zhang Q, Shi L, He H, Liu X, Huang Y, Xu D, Yao M, Zhang N, Guo Y, Lu Y, Li H, Zhou J, Tan J, Xing M, Luo G. Down-Regulating Scar Formation by Microneedles Directly via a Mechanical Communication Pathway. ACS NANO 2022; 16:10163-10178. [PMID: 35617518 PMCID: PMC9331171 DOI: 10.1021/acsnano.1c11016] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Excessive extracellular matrix deposition drives fibroblasts into a state of high mechanical stress, exacerbating pathological fibrosis and hypertrophic scar formation, leading to tissue dysfunction. This study reports a minimally invasive and convenient approach to obtaining scarless tissue using a silk fibroin microneedle patch (SF MNs). We found that by tuning the MN size and density only, the biocompatible MNs significantly decreased the scar elevation index in the rabbit ear hypertrophic scar model and increased ultimate tensile strength close to regular skin. To advance our understanding of this recent approach, we built a fibroblast-populated collagen lattice system and finite element model to study MN-mediated cellular behavior of fibroblasts. We found that the MNs reduced the fibroblasts generated contraction and mechanical stress, as indicated by decreased expression of the mechanical sensitive gene ANKRD1. Specifically, SF MNs attenuated the integrin-FAK signaling and consequently down-regulated the expression of TGF-β1, α-SMA, collagen I, and fibronectin. It resulted in a low-stress microenvironment that helps to reduce scar formation significantly. Microneedles' physical intervention via the mechanotherapeutic strategy is promising for scar-free wound healing.
Collapse
Affiliation(s)
- Qing Zhang
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Lin Shi
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Hong He
- Ministry
of Education & Key Disciplines Laboratory of Novel Micro-Nano
Devices and System Technology, Chongqing
University, Chongqing 400044, China
| | - Xingmou Liu
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
- Chongqing
Key Laboratory of Complex Systems and Bionic Control, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Yong Huang
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Dan Xu
- Department
of Pathology, Southwest Hospital, Third
Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mengyun Yao
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Ning Zhang
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Yicheng Guo
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Yifei Lu
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Haisheng Li
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Junyi Zhou
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Jianglin Tan
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Malcolm Xing
- Department
of Mechanical Engineering, University of
Manitoba, Winnipeg, R3T 2N2, Canada
| | - Gaoxing Luo
- Institute
of Burn Research, State Key Laboratory of Trauma, Burn and Combined
Injury, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
175
|
Chen S, Yang L, Ou X, Li JY, Zi CT, Wang H, Hu JM, Liu Y. A new polysaccharide platform constructs self-adjuvant nanovaccines to enhance immune responses. J Nanobiotechnology 2022; 20:320. [PMID: 35836236 PMCID: PMC9281129 DOI: 10.1186/s12951-022-01533-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Nanovaccines have shown the promising potential in controlling and eradicating the threat of infectious diseases worldwide. There has been a great need in developing a versatile strategy to conveniently construct diverse types of nanovaccines and induce potent immune responses. To that end, it is critical for obtaining a potent self-adjuvant platform to assemble with different types of antigens into nanovaccines. RESULTS In this study, we identified a new natural polysaccharide from the rhizomes of Bletilla striata (PRBS), and used this polysaccharide as a platform to construct diverse types of nanovaccines with potent self-adjuvant property. In the construction process of SARS-CoV-2 nanovaccine, PRBS molecules and RBD protein antigens were assembled into ~ 300 nm nanoparticles by hydrogen bond. For HIV nanovaccine, hydrophobic effect dominantly drove the co-assembly between PRBS molecules and Env expression plasmid into ~ 350 nm nanospheres. Importantly, PRBS can potently activate the behaviors and functions of multiple immune cells such as macrophages, B cells and dendritic cells. Depending on PRBS-mediated immune activation, these self-adjuvant nanovaccines can elicit significantly stronger antigen-specific antibody and cellular responses in vivo, in comparison with their corresponding traditional vaccine forms. Moreover, we also revealed the construction models of PRBS-based nanovaccines by analyzing multiple assembly parameters such as bond energy, bond length and interaction sites. CONCLUSIONS PRBS, a newly-identified natural polysaccharide which can co-assemble with different types of antigens and activate multiple critical immune cells, has presented a great potential as a versatile platform to develop potent self-adjuvant nanovaccines.
Collapse
Affiliation(s)
- Sisi Chen
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650000, Yunnan, China
| | - Liu Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Xia Ou
- School of Medicine, Kunming University of Science and Technology, Kunming, 650201, Yunnan, China
| | - Jin-Yu Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Cheng-Ting Zi
- College of Science, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, CAS Center for Excellence in Nanoscience, Beijing, 100190, China.
| | - Jiang-Miao Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
| | - Ye Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, 650000, Yunnan, China.
| |
Collapse
|
176
|
Tang X, Li Q, Huang T, Zhang H, Chen X, Ling J, Yang Y. Regenerative Role of T Cells in Nerve Repair and Functional Recovery. Front Immunol 2022; 13:923152. [PMID: 35865551 PMCID: PMC9294345 DOI: 10.3389/fimmu.2022.923152] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/06/2022] [Indexed: 12/17/2022] Open
Abstract
The immune system is essential in the process of nerve repair after injury. Successful modulation of the immune response is regarded as an effective approach to improving treatment outcomes. T cells play an important role in the immune response of the nervous system, and their beneficial roles in promoting regeneration have been increasingly recognized. However, the diversity of T-cell subsets also delivers both neuroprotective and neurodegenerative functions. Therefore, this review mainly discusses the beneficial impact of T-cell subsets in the repair of both peripheral nervous system and central nervous system injuries and introduces studies on various therapies based on T-cell regulation. Further discoveries in T-cell mechanisms and multifunctional biomaterials will provide novel strategies for nerve regeneration.
Collapse
Affiliation(s)
- Xiaoxuan Tang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Qiaoyuan Li
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Tingting Huang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Han Zhang
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Xiaoli Chen
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Jue Ling
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- *Correspondence: Jue Ling, ; Yumin Yang,
| | - Yumin Yang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- *Correspondence: Jue Ling, ; Yumin Yang,
| |
Collapse
|
177
|
Kim Y, Koo TM, Thangam R, Kim MS, Jang WY, Kang N, Min S, Kim SY, Yang L, Hong H, Jung HJ, Koh EK, Patel KD, Lee S, Fu HE, Jeon YS, Park BC, Kim SY, Park S, Lee J, Gu L, Kim DH, Kim TH, Lee KB, Jeong WK, Paulmurugan R, Kim YK, Kang H. Submolecular Ligand Size and Spacing for Cell Adhesion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110340. [PMID: 35476306 DOI: 10.1002/adma.202110340] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/27/2022] [Indexed: 06/14/2023]
Abstract
Cell adhesion occurs when integrin recognizes and binds to Arg-Gly-Asp (RGD) ligands present in fibronectin. In this work, submolecular ligand size and spacing are tuned via template-mediated in situ growth of nanoparticles for dynamic macrophage modulation. To tune liganded gold nanoparticle (GNP) size and spacing from 3 to 20 nm, in situ localized assemblies of GNP arrays on nanomagnetite templates are engineered. 3 nm-spaced ligands stimulate the binding of integrin, which mediates macrophage-adhesion-assisted pro-regenerative polarization as compared to 20 nm-spaced ligands, which can be dynamically anchored to the substrate for stabilizing integrin binding and facilitating dynamic macrophage adhesion. Increasing the ligand size from 7 to 20 nm only slightly promotes macrophage adhesion, not observed with 13 nm-sized ligands. Increasing the ligand spacing from 3 to 17 nm significantly hinders macrophage adhesion that induces inflammatory polarization. Submolecular tuning of ligand spacing can dominantly modulate host macrophages.
Collapse
Affiliation(s)
- Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Thomas Myeongseok Koo
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Institute for High Technology Materials and Devices, Korea University, Seoul, 02841, Republic of Korea
| | - Myeong Soo Kim
- Institute for High Technology Materials and Devices, Korea University, Seoul, 02841, Republic of Korea
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Seong Yeol Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hee Joon Jung
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- International Institute for Nanotechnology, Evanston, IL, 60208, USA
- NUANCE Center, Northwestern University, Evanston, IL, 60208, USA
| | - Eui Kwan Koh
- Seoul Center, Korea Basic Science Institute, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Republic of Korea
| | - Kapil D Patel
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Institute for High Technology Materials and Devices, Korea University, Seoul, 02841, Republic of Korea
| | - Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hong En Fu
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yoo Sang Jeon
- Institute of Engineering Research, Korea University, Seoul, 02841, Republic of Korea
| | - Bum Chul Park
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Soo Young Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Institute for High Technology Materials and Devices, Korea University, Seoul, 02841, Republic of Korea
| | - Steve Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Luo Gu
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Woong Kyo Jeong
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Young Keun Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Green Manufacturing Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
178
|
Zhang L, Chen X, Cai P, Sun H, Shen S, Guo B, Jiang Q. Reprogramming Mitochondrial Metabolism in Synovial Macrophages of Early Osteoarthritis by a Camouflaged Meta-Defensome. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202715. [PMID: 35671349 DOI: 10.1002/adma.202202715] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/30/2022] [Indexed: 06/15/2023]
Abstract
Osteoarthritis (OA) is a low-grade inflammatory and progressive joint disease, and its progression is closely associated with an imbalance in M1/M2 synovial macrophages. Repolarizing pro-inflammatory M1 macrophages into the anti-inflammatory M2 phenotype is emerging as a strategy to alleviate OA progression but is compromised by unsatisfactory efficiency. In this study, the reprogramming of mitochondrial dysfunction is pioneered with a camouflaged meta-Defensome, which can transform M1 synovial macrophages into the M2 phenotype with a high efficiency of 82.3%. The meta-Defensome recognizes activated macrophages via receptor-ligand interactions and accumulates in the mitochondria through electrostatic attractions. These meta-Defensomes are macrophage-membrane-coated polymeric nanoparticles decorated with dual ligands and co-loaded with S-methylisothiourea and MnO2 . Meta-Defensomes are demonstrated to successfully reprogram the mitochondrial metabolism of M1 macrophages by scavenging mitochondrial reactive oxygen species and inhibiting mitochondrial NO synthase, thereby increasing mitochondrial transcription factor A expression and restoring aerobic respiration. Furthermore, meta-Defensomes are intravenously injected into collagenase-induced osteoarthritis mice and effectively suppress synovial inflammation and progression of early OA, as evident from the Osteoarthritis Research Society International score. Therefore, reprogramming the mitochondrial metabolism can serve as a novel and practical approach to repolarize M1 synovial macrophages. The camouflaged meta-Defensomes are a promising therapeutic agent for impeding OA progression in tclinic.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Xiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Pingqiang Cai
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, P. R. China
| | - Han Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Siyu Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Baosheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| |
Collapse
|
179
|
Ma X, Li SJ, Liu Y, Zhang T, Xue P, Kang Y, Sun ZJ, Xu Z. Bioengineered nanogels for cancer immunotherapy. Chem Soc Rev 2022; 51:5136-5174. [PMID: 35666131 DOI: 10.1039/d2cs00247g] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent years have witnessed increasingly rapid advances in nanocarrier-based biomedicine aimed at improving treatment paradigms for cancer. Nanogels serve as multipurpose and constructed vectors formed via intramolecular cross-linking to generate drug delivery systems, which is attributed predominantly to their satisfactory biocompatibility, bio-responsiveness, high stability, and low toxicity. Recently, immunotherapy has experienced unprecedented growth and has become the preferred strategy for cancer treatment, and mainly involves the mobilisation of the immune system and an enhanced anti-tumour immunity of the tumour microenvironment. Despite the inspiring success, immunotherapeutic strategies are limited due to the low response rates and immune-related adverse events. Like other nanomedicines, nanogels are comparably limited by lower focal enrichment rates upon introduction into the organism via injection. Because nanogels are three-dimensional cross-linked aqueous materials that exhibit similar properties to natural tissues and are structurally stable, they can comfortably cope with shear forces and serum proteins in the bloodstream, and the longer circulation life increases the chance of nanogel accumulation in the tumour, conferring deep tumour penetration. The large specific surface area can reduce or eliminate off-target effects by introducing stimuli-responsive functional groups, allowing multiple physical and chemical modifications for specific purposes to improve targeting to specific immune cell subpopulations or immune organs, increasing the bioavailability of the drug, and conferring a low immune-related adverse events on nanogel therapies. The slow release upon reaching the tumour site facilitates long-term awakening of the host's immune system, ultimately achieving enhanced therapeutic effects. As an effective candidate for cancer immunotherapy, nanogel-based immunotherapy has been widely used. In this review, we mainly summarize the recent advances of nanogel-based immunotherapy to deliver immunomodulatory small molecule drugs, antibodies, genes and cytokines, to target antigen presenting cells, form cancer vaccines, and enable chimeric antigen receptor (CAR)-T cell therapy. Future challenges as well as expected and feasible prospects for clinical treatment are also highlighted.
Collapse
Affiliation(s)
- Xianbin Ma
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Shu-Jin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Yuantong Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Tian Zhang
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Peng Xue
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Yuejun Kang
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Zhigang Xu
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| |
Collapse
|
180
|
Lee S, Park J, Kim S, Ok J, Yoo JI, Kim YS, Ahn Y, Kim TI, Ko HC, Lee JY. High-Performance Implantable Bioelectrodes with Immunocompatible Topography for Modulation of Macrophage Responses. ACS NANO 2022; 16:7471-7485. [PMID: 35438981 DOI: 10.1021/acsnano.1c10506] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Implantable bioelectrodes enable precise recording or stimulation of electrical signals with living tissues in close contact. However, their performance is frequently compromised owing to inflammatory tissue reactions, which macrophages either induce or resolve by polarizing to an inflammatory (M1) or noninflammatory (M2) phenotype, respectively. Thus, we aimed to fabricate biocompatible and functional implantable conductive polymer bioelectrodes with optimal topography for the modulation of macrophage responses. To this end, we produced heparin-doped polypyrrole (PPy/Hep) electrodes of different surface roughness, with Ra values from 5.5 to 17.6 nm, by varying the charge densities during electrochemical synthesis. In vitro culture revealed that macrophages on rough PPy/Hep electrodes preferentially polarized to noninflammatory phenotypes. In particular, PPy/Hep-900 (Ra = 14 nm) was optimal with respect to electrochemical properties and the suppression of inflammatory M1 polarization. In vivo implantation indicated that PPy/Hep-900 significantly reduced macrophage recruitment, suppressed inflammatory polarization, and mitigated fibrotic tissue formation. In addition, the implanted PPy/Hep-900 electrodes could successfully record electrocardiographic signals for up to 10 days without substantial decreases in sensitivity, while other electrodes substantially lost their signal sensitivity during implantation. Altogether, we demonstrate that modulating the surface features of PPy/Hep can benefit the design and applications of high-performance and high-biocompatibility bioelectrodes.
Collapse
Affiliation(s)
- Sanghun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Junggeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Semin Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jehyung Ok
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jung Il Yoo
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Yong Sook Kim
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Youngkeun Ahn
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Tae-Il Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Heung Cho Ko
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| |
Collapse
|
181
|
Fang Y, Zhang Z, Liu Y, Gao T, Liang S, Chu Q, Guan L, Mu W, Fu S, Yang H, Zhang N, Liu Y. Artificial Assembled Macrophage Co-Deliver Black Phosphorus Quantum Dot and CDK4/6 Inhibitor for Colorectal Cancer Triple-Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:20628-20640. [PMID: 35477252 DOI: 10.1021/acsami.2c01305] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
In recent years, therapeutic strategies based on macrophages have been inspiringly developed, but due to the high intricacy and immunosuppression of the tumor microenvironment, the widespread use of these strategies still faces significant challenges. Herein, an artificial assembled macrophage concept (AB@LM) was presented to imitate the main antitumor abilities of macrophages of tumor targeting, promoting the antitumor immunity, and direct tumor-killing effects. The artificial assembled macrophage (AB@LM) was prepared through an extrusion method, which is to fuse the macrophage membrane with abemaciclib and black phosphorus quantum dot (BPQD)-loaded liposomes. AB@LM showed good stability and tumor targeting ability with the help of macrophage membrane. Furthermore, AB@LM reversed the immunosuppressive tumor microenvironment by inhibiting regulatory T cells (Tregs) and stimulating the maturation of antigen-presenting cells to activate the antitumor immune response through triggering an immunogenic cell death effect. More importantly, in the colorectal tumor model in vivo, a strong cooperative therapeutic effect of photo/chemo/immunotherapy was observed with high tumor inhibition rate (95.3 ± 2.05%). In conclusion, AB@LM exhibits excellent antitumor efficacy by intelligently mimicking the abilities of macrophages. A promising therapeutic strategy for tumor treatment based on imitating macrophages was provided in this study.
Collapse
Affiliation(s)
- Yuxiao Fang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Zipeng Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Yang Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Tong Gao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Shuang Liang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Qihui Chu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Li Guan
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Weiwei Mu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Shunli Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Huizhen Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Na Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Yongjun Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| |
Collapse
|
182
|
Zhou X, Zhang Z, Jiang W, Hu M, Meng Y, Li W, Zhou X, Wang C. Naringenin is a Potential Anabolic Treatment for Bone Loss by Modulating Osteogenesis, Osteoclastogenesis, and Macrophage Polarization. Front Pharmacol 2022; 13:872188. [PMID: 35586056 PMCID: PMC9108355 DOI: 10.3389/fphar.2022.872188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Bone undergoes constant remodeling of formation by osteoblasts and resorption by osteoclasts. In particular, macrophages have been reported to play an essential role in the regulation of bone homeostasis and regeneration. Naringenin, the predominant flavanone in citrus fruits, is reported to exert anti-inflammatory, anti-osteoclastic, and osteogenic effects. However, whether naringenin could modulate the crosstalk between macrophages and osteoblasts/osteoclasts remains to be investigated. In this study, we confirmed that naringenin enhanced osteogenesis and inhibited osteoclastogenesis directly. Naringenin promoted M2 transition and the secretion of osteogenic cytokines including IL-4, IL-10, BMP2, and TGF-β, while suppressing LPS-induced M1 polarization and the production of proinflammatory factors such as TNF-α and IL-1β. In addition, the coculture of primary bone mesenchymal stem cells (BMSCs)/bone marrow monocytes (BMMs) with macrophages showed that the naringenin-treated medium significantly enhanced osteogenic differentiation and impeded osteoclastic differentiation in both inflammatory and non-inflammatory environment. Moreover, in vivo experiments demonstrated that naringenin remarkably reversed LPS-induced bone loss and assisted the healing of calvarial defect. Taken together, naringenin serves as a potential anabolic treatment for pathological bone loss.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Zheng Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- College of Basic Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Weiwei Jiang
- Department of Critical Care Medicine, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Miao Hu
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- College of Basic Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yichen Meng
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Wenfang Li
- Department of Critical Care Medicine, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Wenfang Li, ; Xuhui Zhou, ; Ce Wang,
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Wenfang Li, ; Xuhui Zhou, ; Ce Wang,
| | - Ce Wang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Wenfang Li, ; Xuhui Zhou, ; Ce Wang,
| |
Collapse
|
183
|
Zarubova J, Hasani-Sadrabadi MM, Ardehali R, Li S. Immunoengineering strategies to enhance vascularization and tissue regeneration. Adv Drug Deliv Rev 2022; 184:114233. [PMID: 35304171 PMCID: PMC10726003 DOI: 10.1016/j.addr.2022.114233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022]
Abstract
Immune cells have emerged as powerful regulators of regenerative as well as pathological processes. The vast majority of regenerative immunoengineering efforts have focused on macrophages; however, growing evidence suggests that other cells of both the innate and adaptive immune system are as important for successful revascularization and tissue repair. Moreover, spatiotemporal regulation of immune cells and their signaling have a significant impact on the regeneration speed and the extent of functional recovery. In this review, we summarize the contribution of different types of immune cells to the healing process and discuss ways to manipulate and control immune cells in favor of vascularization and tissue regeneration. In addition to cell delivery and cell-free therapies using extracellular vesicles, we discuss in situ strategies and engineering approaches to attract specific types of immune cells and modulate their phenotypes. This field is making advances to uncover the extraordinary potential of immune cells and their secretome in the regulation of vascularization and tissue remodeling. Understanding the principles of immunoregulation will help us design advanced immunoengineering platforms to harness their power for tissue regeneration.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | | | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
184
|
Zhuang Z, Sun S, Chen K, Zhang Y, Han X, Zhang Y, Sun K, Cheng F, Zhang L, Wang H. Gelatin-based Colloidal vs. Monolithic Gels to Regulate Macrophage-mediated Inflammatory Response. Tissue Eng Part C Methods 2022; 28:351-362. [PMID: 35469426 DOI: 10.1089/ten.tec.2022.0044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Unlike conventional monolithic hydrogels with covalent crosslinkage that are typically elastic, colloidal gels assembled by reversibly assembled particles as building blocks have shown fascinating viscoelastic properties. They follow a gel-sol transition upon yielding and recover to the initial state upon the release of the shear force (so-called shear-thinning and self-healing behavior); this makes them an ideal candidate as injectable and moldable biomaterials for tissue regeneration. The immune response provoked by the implantation of the colloidal gels with special viscoelastic and structural features is critical for the successful integration of the implants with the host tissues, which, however, remains little explored. Since macrophages are known as the primary immune cells in determining the inflammatory response against the implants, we herein investigated in vitro macrophage polarization and in vivo inflammatory response induced by gelatin-based colloidal gels as compared to monolithic gels. Specifically, self-healing colloidal gels composed of pure gelatin nanoparticles, or methacrylate gelatin (GelMA) nanoparticles to allow secondary covalent crosslinkage were compared with GelMA bulk hydrogels. We demonstrated that hydrogel's elasticity plays a more dominant role rather than the structural feature in determining in vitro macrophage polarization evidenced by the stiffer gels inducing pro-inflammation M2 macrophage phenotype as compared to soft gels. However, subcutaneous implantation revealed a significantly alleviated immune response characterized by less fibrous capsule formation for the colloidal gels as compared to bulk gels of similar matrix elasticity. We speculated this can be related to the improved permeability of the colloidal gels for cell penetration, thereby leading to less fibrosis. In general, this study provided in-depth insight into the biophysical regulator of hydrogel materials on macrophage behavior and related inflammatory response, which can further direct future implant design and predict biomaterial-host interactions for immunotherapy and regenerative medicine.
Collapse
Affiliation(s)
- Zhumei Zhuang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Shengnan Sun
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Kaiwen Chen
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Yue Zhang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Xiaoman Han
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Yang Zhang
- Health Science Center, School of Stomatology, Shenzhen University, Shenzhen, China
| | - Kai Sun
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Fang Cheng
- Key State Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Lijun Zhang
- Optometric Center, Dalian Eye Hospital, Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, China
| | - Huanan Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian, China
| |
Collapse
|
185
|
Polymeric Coatings and Antimicrobial Peptides as Efficient Systems for Treating Implantable Medical Devices Associated-Infections. Polymers (Basel) 2022; 14:polym14081611. [PMID: 35458361 PMCID: PMC9024559 DOI: 10.3390/polym14081611] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 02/04/2023] Open
Abstract
Many infections are associated with the use of implantable medical devices. The excessive utilization of antibiotic treatment has resulted in the development of antimicrobial resistance. Consequently, scientists have recently focused on conceiving new ways for treating infections with a longer duration of action and minimum environmental toxicity. One approach in infection control is based on the development of antimicrobial coatings based on polymers and antimicrobial peptides, also termed as “natural antibiotics”.
Collapse
|
186
|
Kim Y, Jung HJ, Lee Y, Koo S, Thangam R, Jang WY, Kim SY, Park S, Lee S, Bae G, Patel KD, Wei Q, Lee KB, Paulmurugan R, Jeong WK, Hyeon T, Kim D, Kang H. Manipulating Nanoparticle Aggregates Regulates Receptor-Ligand Binding in Macrophages. J Am Chem Soc 2022; 144:5769-5783. [PMID: 35275625 DOI: 10.1021/jacs.1c08861] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The receptor-ligand interactions in cells are dynamically regulated by modulation of the ligand accessibility. In this study, we utilize size-tunable magnetic nanoparticle aggregates ordered at both nanometer and atomic scales. We flexibly anchor magnetic nanoparticle aggregates of tunable sizes over the cell-adhesive RGD ligand (Arg-Gly-Asp)-active material surface while maintaining the density of dispersed ligands accessible to macrophages at constant. Lowering the accessible ligand dispersity by increasing the aggregate size at constant accessible ligand density facilitates the binding of integrin receptors to the accessible ligands, which promotes the adhesion of macrophages. In high ligand dispersity, distant magnetic manipulation to lift the aggregates (which increases ligand accessibility) stimulates the binding of integrin receptors to the accessible ligands available under the aggregates to augment macrophage adhesion-mediated pro-healing polarization both in vitro and in vivo. In low ligand dispersity, distant control to drop the aggregates (which decreases ligand accessibility) repels integrin receptors away from the aggregates, thereby suppressing integrin receptor-ligand binding and macrophage adhesion, which promotes inflammatory polarization. Here, we present "accessible ligand dispersity" as a novel fundamental parameter that regulates receptor-ligand binding, which can be reversibly manipulated by increasing and decreasing the ligand accessibility. Limitless tuning of nanoparticle aggregate dimensions and morphology can offer further insight into the regulation of receptor-ligand binding in host cells.
Collapse
Affiliation(s)
- Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hee Joon Jung
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- NUANCE Center, Northwestern University, Evanston, Illinois 60208, United States
| | - Yunjung Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sagang Koo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul 02841, Republic of Korea
| | - Seong Yeol Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Sangwoo Park
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Gunhyu Bae
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Kapil Dev Patel
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Palo Alto, California 94304, United States
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford University, Palo Alto, California 94304, United States
| | - Woong Kyo Jeong
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul 02841, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Dokyoon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan 15588, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
187
|
Liu Y, Hu P, Zheng Z, Zhong D, Xie W, Tang Z, Pan B, Luo J, Zhang W, Wang X. Photoresponsive Vaccine-Like CAR-M System with High-Efficiency Central Immune Regulation for Inflammation-Related Depression. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108525. [PMID: 34897839 DOI: 10.1002/adma.202108525] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/25/2021] [Indexed: 06/14/2023]
Abstract
Increasing evidence suggests that activation of microglia-induced neuroinflammation plays a crucial role in the pathophysiology of depression. Consequently, targeting the central nervous system to reduce neuroinflammation holds great promise for the treatment of depression. However, few drugs can enter the brain via a circulatory route through the blood-brain barrier (BBB) to reach the central nervous system efficiently, which limits the pharmacological treatment for neuropsychiatric diseases. Herein, a light-responsive system named UZPM, consisting of blue-emitting NaYF4 :Yb, Tm@zeolitic-imidazolate framework (UCNP@ZIF-8), photoacid (PA), and melatonin (MT) is developed to address the above issues. Meanwhile, UZPM is introduced into macrophages by functional liposomes fusion and modified with hydroxylamine groups on the cell surface. Aldehyde-modified cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) is used as a chimeric antigen receptor (CAR) targeting group to modify the surface of macrophages by aldehyde/hydroxylamine condensation to precisely target central M1-type microglia (CAR-M-UZPM). Both in vitro and in vivo experiments demonstrate that the CAR-M-UZPM drug delivery system can efficiently penetrate the BBB, targeting centrally activated microglia, and thus, inhibiting the M1-type polarization of microglia, producing continuous vaccine-like anti-inflammatory effects that prevent the occurrence and development of inflammation-related depression.
Collapse
Affiliation(s)
- Yu Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, P. R. China
| | - Ping Hu
- Institute of Translational Medicine, School of Life Sciences, Nanchang University, Nanchang, 330088, P. R. China
| | - Zhiheng Zheng
- Institute of Life Science, School of Life Sciences, Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, 330088, P. R. China
| | - Da Zhong
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
| | - Weichang Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
| | - Zhibo Tang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
| | - Bingxing Pan
- Institute of Life Science, School of Life Sciences, Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, 330088, P. R. China
| | - Jun Luo
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, P. R. China
| | - Wenhua Zhang
- Institute of Life Science, School of Life Sciences, Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, 330088, P. R. China
| | - Xiaolei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
| |
Collapse
|
188
|
Li J, Dekanovsky L, Khezri B, Wu B, Zhou H, Sofer Z. Biohybrid Micro- and Nanorobots for Intelligent Drug Delivery. CYBORG AND BIONIC SYSTEMS 2022; 2022:9824057. [PMID: 36285309 PMCID: PMC9494704 DOI: 10.34133/2022/9824057] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/18/2022] [Indexed: 08/12/2023] Open
Abstract
Biohybrid micro- and nanorobots are integrated tiny machines from biological components and artificial components. They can possess the advantages of onboard actuation, sensing, control, and implementation of multiple medical tasks such as targeted drug delivery, single-cell manipulation, and cell microsurgery. This review paper is to give an overview of biohybrid micro- and nanorobots for smart drug delivery applications. First, a wide range of biohybrid micro- and nanorobots comprising different biological components are reviewed in detail. Subsequently, the applications of biohybrid micro- and nanorobots for active drug delivery are introduced to demonstrate how such biohybrid micro- and nanorobots are being exploited in the field of medicine and healthcare. Lastly, key challenges to be overcome are discussed to pave the way for the clinical translation and application of the biohybrid micro- and nanorobots.
Collapse
Affiliation(s)
- Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czech Republic
| | - Lukas Dekanovsky
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czech Republic
| | - Bahareh Khezri
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czech Republic
| | - Bing Wu
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czech Republic
| | - Huaijuan Zhou
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czech Republic
| | - Zdenek Sofer
- Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague 6, Czech Republic
| |
Collapse
|
189
|
Cai C, Zhang X, Li Y, Liu X, Wang S, Lu M, Yan X, Deng L, Liu S, Wang F, Fan C. Self-Healing Hydrogel Embodied with Macrophage-Regulation and Responsive-Gene-Silencing Properties for Synergistic Prevention of Peritendinous Adhesion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106564. [PMID: 34816470 DOI: 10.1002/adma.202106564] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/17/2021] [Indexed: 05/24/2023]
Abstract
Antiadhesion barriers such as films and hydrogels used to wrap repaired tendons are important for preventing the formation of adhesion tissue after tendon surgery. However, sliding of the tendon can compress the adjacent hydrogel barrier and cause it to rupture, which may then lead to unexpected inflammation. Here, a self-healing and deformable hyaluronic acid (HA) hydrogel is constructed as a peritendinous antiadhesion barrier. Matrix metalloproteinase-2 (MMP-2)-degradable gelatin-methacryloyl (GelMA) microspheres (MSs) encapsulated with Smad3-siRNA nanoparticles are entrapped within the HA hydrogel to inhibit fibroblast proliferation and prevent peritendinous adhesion. GelMA MSs are responsively degraded by upregulation of MMP-2, achieving on-demand release of siRNA nanoparticles. Silencing effect of Smad3-siRNA nanoparticles is around 75% toward targeted gene. Furthermore, the self-healing hydrogel shows relatively attenuated inflammation compared to non-healing hydrogel. The mean adhesion scores of composite barrier group are 1.67 ± 0.51 and 2.17 ± 0.75 by macroscopic and histological evaluation, respectively. The proposed self-healing hydrogel antiadhesion barrier with MMP-2-responsive drug release behavior is highly effective for decreasing inflammation and inhibiting tendon adhesion. Therefore, this research provides a new strategy for the development of safe and effective antiadhesion barriers.
Collapse
Affiliation(s)
- Chuandong Cai
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China
| | - Xinshu Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China
| | - Yuange Li
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China
| | - Xuanzhe Liu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China
| | - Shuo Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China
| | - Mingkuan Lu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China
| | - Xiong Yan
- Department of Orthopaedics, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shen Liu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China
| | - Fei Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China
| |
Collapse
|
190
|
Coburn PT, Li X, Li JY, Kishimoto Y, Li-Jessen NY. Progress in Vocal Fold Regenerative Biomaterials: An Immunological Perspective. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100119. [PMID: 35434718 PMCID: PMC9007544 DOI: 10.1002/anbr.202100119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Vocal folds, housed in the upper respiratory tract, are important to daily breathing, speech and swallowing functions. Irreversible changes to the vocal fold mucosae, such as scarring and atrophy, require a regenerative medicine approach to promote a controlled regrowth of the extracellular matrix (ECM)-rich mucosa. Various biomaterial systems have been engineered with an emphasis on stimulating local vocal fold fibroblasts to produce new ECM. At the same time, it is imperative to limit the foreign body reaction and associated immune components that can hinder the integration of the biomaterial into the host tissue. Modern biomaterial designs have become increasingly focused on actively harnessing the immune system to accelerate and optimize the process of tissue regeneration. An array of physical and chemical biomaterial parameters have been reported to effectively modulate local immune cells, such as macrophages, to initiate tissue repair, stimulate ECM production, promote biomaterial-tissue integration, and restore the function of the vocal folds. In this perspective paper, the unique immunological profile of the vocal folds will first be reviewed. Key physical and chemical biomaterial properties relevant to immunomodulation will then be highlighted and discussed. A further examination of the physicochemical properties of recent vocal fold biomaterials will follow to generate deeper insights into corresponding immune-related outcomes. Lastly, a perspective will be offered on the opportunity of integrating material-led immunomodulatory strategies into future vocal fold tissue engineering therapies.
Collapse
Affiliation(s)
- Patrick T. Coburn
- School of Communication Sciences and Disorders, McGill University, Canada
| | - Xuan Li
- Department of Mechanical Engineering, McGill University, Canada
| | - Jianyu. Y. Li
- Department of Mechanical Engineering, McGill University, Canada
- Department of Biomedical Engineering, McGill University, Canada
| | - Yo Kishimoto
- Department of Otolaryngology – Head & Neck Surgery, Kyoto University, Kyoto, Japan
| | - Nicole Y.K. Li-Jessen
- School of Communication Sciences and Disorders, McGill University, Canada
- Department of Biomedical Engineering, McGill University, Canada
- Department of Otolaryngology – Head & Neck Surgery, McGill University, Canada
| |
Collapse
|
191
|
He Y, Tian M, Li X, Hou J, Chen S, Yang G, Liu X, Zhou S. A Hierarchical-Structured Mineralized Nanofiber Scaffold with Osteoimmunomodulatory and Osteoinductive Functions for Enhanced Alveolar Bone Regeneration. Adv Healthc Mater 2022; 11:e2102236. [PMID: 34779582 DOI: 10.1002/adhm.202102236] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/07/2021] [Indexed: 02/05/2023]
Abstract
Alveolar bone resorption is a major cause of teeth loss and jeopardizes the osseointegration of dental implants, greatly affecting patient's quality of life and health. It is still a great challenge to completely regenerate the alveolar bone defect through traditional guided bone regeneration (GBR) membranes due to their limited bioactivity and regeneration potential. Herein, a new hierarchical-structured mineralized nanofiber (HMF) scaffold, which is combined with both anisotropic and isotropic nanofibrous surface topography and the mineralized particles, is fabricated via a simple template-assisted electrospinning technology and in situ mineralization method. This HMF scaffold can not only directly induce osteogenic differentiation of bone mesenchymal stem cells (osteoinduction), but also stimulate macrophage toward pro-healing (M2) phenotype-polarization with an elevated secretion of the pro-healing cytokines, eventually enhancing the osteogenesis (osteoimmunomodulation). The results of in vivo rat alveolar bone defect repair experiments demonstrate that as compared with the combination of commercial Bio-Gide and Bio-Oss, the single HMF scaffold shows comparable or even superior bone repair effect, with better tissue-integration and more suitable degradation time and accompanied by a simplified operation.
Collapse
Affiliation(s)
- Yang He
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu 610031 P. R. China
| | - Mi Tian
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Department of Orthodontics West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Xilin Li
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu 610031 P. R. China
| | - Jianwen Hou
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu 610031 P. R. China
| | - Song Chen
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Department of Orthodontics West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Guang Yang
- College of Medicine Southwest Jiaotong University Chengdu 610031 China
| | - Xian Liu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases Department of Orthodontics West China Hospital of Stomatology Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu 610031 P. R. China
| |
Collapse
|
192
|
Wang T, Bai J, Lu M, Huang C, Geng D, Chen G, Wang L, Qi J, Cui W, Deng L. Engineering immunomodulatory and osteoinductive implant surfaces via mussel adhesion-mediated ion coordination and molecular clicking. Nat Commun 2022; 13:160. [PMID: 35013289 PMCID: PMC8748715 DOI: 10.1038/s41467-021-27816-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 12/07/2021] [Indexed: 01/10/2023] Open
Abstract
Immune response and new tissue formation are important aspects of tissue repair. However, only a single aspect is generally considered in previous biomedical interventions, and the synergistic effect is unclear. Here, a dual-effect coating with immobilized immunomodulatory metal ions (e.g., Zn2+) and osteoinductive growth factors (e.g., BMP-2 peptide) is designed via mussel adhesion-mediated ion coordination and molecular clicking strategy. Compared to the bare TiO2 group, Zn2+ can increase M2 macrophage recruitment by up to 92.5% in vivo and upregulate the expression of M2 cytokine IL-10 by 84.5%; while the dual-effect of Zn2+ and BMP-2 peptide can increase M2 macrophages recruitment by up to 124.7% in vivo and upregulate the expression of M2 cytokine IL-10 by 171%. These benefits eventually significantly enhance bone-implant mechanical fixation (203.3 N) and new bone ingrowth (82.1%) compared to the bare TiO2 (98.6 N and 45.1%, respectively). Taken together, the dual-effect coating can be utilized to synergistically modulate the osteoimmune microenvironment at the bone-implant interface, enhancing bone regeneration for successful implantation. Immune response and new tissue formation are important aspects of tissue repair but often only one aspect is considered in biomedical interventions. Here, the authors report on the use of a mussel-like surface coating to immobilize immune modulating metal ions and growth factors and demonstrated improved in vivo outcomes.
Collapse
Affiliation(s)
- Tao Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, P. R. China.,Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, 200080, Shanghai, P. R. China.,Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, 314000, Jiaxing, P. R. China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, P. R. China
| | - Min Lu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Chenglong Huang
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, 314000, Jiaxing, P. R. China
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, P. R. China
| | - Gang Chen
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, 314000, Jiaxing, P. R. China
| | - Lei Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, P. R. China
| | - Jin Qi
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, P. R. China.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, P. R. China.
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, P. R. China.
| |
Collapse
|
193
|
Hong H, Min S, Koo S, Lee Y, Yoon J, Jang WY, Kang N, Thangam R, Choi H, Jung HJ, Han SB, Wei Q, Yu SH, Kim DH, Paulmurugan R, Jeong WK, Lee KB, Hyeon T, Kim D, Kang H. Dynamic Ligand Screening by Magnetic Nanoassembly Modulates Stem Cell Differentiation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105460. [PMID: 34655440 DOI: 10.1002/adma.202105460] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/19/2021] [Indexed: 06/13/2023]
Abstract
In native microenvironment, diverse physical barriers exist to dynamically modulate stem cell recruitment and differentiation for tissue repair. In this study, nanoassembly-based magnetic screens of various sizes are utilized, and they are elastically tethered over an RGD ligand (cell-adhesive motif)-presenting material surface to generate various nanogaps between the screens and the RGDs without modulating the RGD density. Large screens exhibiting low RGD distribution stimulate integrin clustering to facilitate focal adhesion, mechanotransduction, and differentiation of stem cells, which are not observed with small screens. Magnetic downward pulling of the large screens decreases the nanogaps, which dynamically suppress the focal adhesion, mechanotransduction, and differentiation of stem cells. Conversely, magnetic upward pulling of the small screens increases the nanogaps, which dynamically activates focal adhesion, mechanotransduction, and differentiation of stem cells. This regulation mechanism is also shown to be effective in the microenvironment in vivo. Further diversifying the geometries of the physical screens can further enable diverse modalities of multifaceted and safe unscreening of the distributed RGDs to unravel and modulate stem cell differentiation for tissue repair.
Collapse
Affiliation(s)
- Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sagang Koo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yunjung Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinho Yoon
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyojun Choi
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hee Joon Jung
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- International Institute for Nanotechnology, Evanston, IL, 60208, USA
- NUANCE Center, Northwestern University, Evanston, IL, 60208, USA
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, China
| | - Seung-Ho Yu
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Woong Kyo Jeong
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dokyoon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan, 15588, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
194
|
Lin J, Song X, Yin H, Song N, Wang Y, Li Z, Luo F, Tan H, He X, Li J. Citicoline–liposome/polyurethane composite scaffolds regulate the inflammatory response of microglia to promote nerve regeneration. JOURNAL OF MATERIALS SCIENCE 2022; 57:2073-2088. [DOI: 10.1007/s10853-021-06628-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/12/2021] [Indexed: 01/03/2025]
|
195
|
Jiang S, Zeng Q, Zhao K, Liu J, Sun Q, Huang K, He Y, Zhang X, Wang H, Shi X, Feng C, Deng X, Wei Y. Chirality Bias Tissue Homeostasis by Manipulating Immunological Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105136. [PMID: 34601779 DOI: 10.1002/adma.202105136] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/15/2021] [Indexed: 06/13/2023]
Abstract
The physiological chirality of extracellular environments is substantially affected by pathological diseases. However, how this stereochemical variation drives host immunity remains poorly understood. Here, it is reported that pathology-mimetic M-nanofibrils-but not physiology-mimetic P-nanofibrils-act as a defense mechanism that helps to restore tissue homeostasis by manipulating immunological response. Quantitative multi-omics in vivo and in vitro shows that M-nanofibrils significantly inhibit inflammation and promote tissue regeneration by upregulating M2 macrophage polarization and downstream immune signaling compared with P-nanofibrils. Molecular analysis and theoretical simulation demonstrate that M-chirality displays higher stereo-affinity to cellular binding, which induces higher cellular contractile stress and activates mechanosensitive ion channel PIEZOl to conduct Ca2+ influx. In turn, the nuclear transfer of STAT is biased by Ca2+ influx to promote M2 polarization. These findings underscore the structural mechanisms of disease, providing design basis for immunotherapy with bionic functional materials.
Collapse
Affiliation(s)
- Shengjie Jiang
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Qiang Zeng
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Kai Zhao
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Jinying Liu
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China
| | - Qiannan Sun
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Kang Huang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Ying He
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials and Dental Medical Devices Testing Center, National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Hui Wang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Xinghua Shi
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Chuanliang Feng
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiaotong University, Shanghai, 200240, P. R. China
| | - Xuliang Deng
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yan Wei
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| |
Collapse
|
196
|
The characterization, cytotoxicity, macrophage response and tissue regeneration of decellularized cartilage in costal cartilage defects. Acta Biomater 2021; 136:147-158. [PMID: 34563726 DOI: 10.1016/j.actbio.2021.09.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022]
Abstract
After harvesting multiple costal cartilages, the local defect disrupts the integrity of the chest wall and may lead to obvious thoracic complications, such as local depression and asymmetry of the bilateral thoracic height. Decellularized materials have been used for tissue reconstruction in clinical surgeries. To apply xenogenic decellularized cartilage in costal cartilage defects, porcine-derived auricular and costal cartilage was tested for characterization, cytotoxicity, macrophage response, and tissue regeneration. Most of the DNA and α-Gal were effectively removed, and the collagen was well preserved after the decellularization process. The glycosaminoglycan (GAG) content decreased significantly compared to that in untreated cartilage. The decellularized auricular cartilage had a larger pore size, more pores, and a higher degradation rate than the decellularized costal cartilage. No apparent nuclei or structural damage was observed in the extracellular matrix. The decellularized auricular cartilage had a higher cell proliferation rate and more prominent immunomodulatory effect than the other groups. Two types of decellularized cartilage, particularly decellularized auricular cartilage, promoted the tissue regeneration in the cartilage defect area, combined with noticeable cartilage morphology and increased chondrogenic gene expression. In our research, the functional components and structure of the extracellular matrix were well preserved after the decellularization process. The decellularized cartilage had better biocompatibility and suitable microenvironment for tissue regeneration in the defect area, suggesting its potential application in cartilage repair during the surgery. STATEMENT OF SIGNIFICANCE: Autologous costal cartilage has been widely used in various surgeries, while the cartilage defects after the harvesting of multiple costal cartilages may cause localized chest wall deformities. Decellularized cartilage is an ideal material that could be produced in the factory and applied in surgeries. In this study, both decellularized costal cartilage and auricular cartilage preserved original structure, functional biocompatibility, immunosuppressive effects, and promoted tissue regeneration in the cartilage defect area.
Collapse
|
197
|
Zhang P, Meng J, Li Y, Yang C, Hou Y, Tang W, McHugh KJ, Jing L. Nanotechnology-enhanced immunotherapy for metastatic cancer. Innovation (N Y) 2021; 2:100174. [PMID: 34766099 PMCID: PMC8571799 DOI: 10.1016/j.xinn.2021.100174] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
A vast majority of cancer deaths occur as a result of metastasis. Unfortunately, effective treatments for metastases are currently lacking due to the difficulty of selectively targeting these small, delocalized tumors distributed across a variety of organs. However, nanotechnology holds tremendous promise for improving immunotherapeutic outcomes in patients with metastatic cancer. In contrast to conventional cancer immunotherapies, rationally designed nanomaterials can trigger specific tumoricidal effects, thereby improving immune cell access to major sites of metastasis such as bone, lungs, and lymph nodes, optimizing antigen presentation, and inducing a persistent immune response. This paper reviews the cutting-edge trends in nano-immunoengineering for metastatic cancers with an emphasis on different nano-immunotherapeutic strategies. Specifically, it discusses directly reversing the immunological status of the primary tumor, harnessing the potential of peripheral immune cells, preventing the formation of a pre-metastatic niche, and inhibiting the tumor recurrence through postoperative immunotherapy. Finally, we describe the challenges facing the integration of nanoscale immunomodulators and provide a forward-looking perspective on the innovative nanotechnology-based tools that may ultimately prove effective at eradicating metastatic diseases.
Collapse
Affiliation(s)
- Peisen Zhang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Junli Meng
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Yingying Li
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Chen Yang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wen Tang
- South China Advanced Institute for Soft Matter Science and Technology, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main Street, MS-142, Houston, TX 77005, USA
| | - Lihong Jing
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| |
Collapse
|
198
|
Yang G, Lu SB, Li C, Chen F, Ni JS, Zha M, Li Y, Gao J, Kang T, Liu C, Li K. Type I macrophage activator photosensitizer against hypoxic tumors. Chem Sci 2021; 12:14773-14780. [PMID: 34820093 PMCID: PMC8597846 DOI: 10.1039/d1sc04124j] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022] Open
Abstract
Photodynamic immunotherapy has emerged as a promising strategy to treat cancer. However, the hypoxic nature of most solid tumors and notoriously immunosuppressive tumor microenvironment could greatly compromise the efficacy of photodynamic immunotherapy. To address this challenge, we rationally synthesized a type I photosensitizer of TPA-DCR nanoparticles (NPs) with aggregation-enhanced reactive oxygen species generation via an oxygen-independent pathway. We demonstrated that the free radicals produced by TPA-DCR NPs could reprogram M0 and M2 macrophages into an anti-tumor state, which is not restricted by the hypoxic conditions. The activated M1 macrophages could further induce the immunogenic cell death of cancer cells by secreting pro-inflammatory cytokines and phagocytosis. In addition, in vivo anti-tumor experiments revealed that the TPA-DCR NPs could further trigger tumor immune response by re-educating tumor-associated macrophages toward M1 phenotype and promoting T cell infiltration. Overall, this work demonstrates the design of type I organic photosensitizers and mechanistic investigation of their superior anti-tumor efficacy. The results will benefit the exploration of advanced strategies to regulate the tumor microenvironment for effective photodynamic immunotherapy against hypoxic tumors.
Collapse
Affiliation(s)
- Guang Yang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Song-Bo Lu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Chong Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Feng Chen
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Jen-Shyang Ni
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Menglei Zha
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Yaxi Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Ji Gao
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Tianyi Kang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Chao Liu
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Kai Li
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| |
Collapse
|
199
|
Luo Y, Zheng X, Yuan P, Ye X, Ma L. Light-induced dynamic RGD pattern for sequential modulation of macrophage phenotypes. Bioact Mater 2021; 6:4065-4072. [PMID: 33997493 PMCID: PMC8089772 DOI: 10.1016/j.bioactmat.2021.04.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 12/04/2022] Open
Abstract
Due to the critical roles of macrophage in immune response and tissue repair, harnessing macrophage phenotypes dynamically to match the tissue healing process on demand attracted many attentions. Although there have developed many advanced platforms with dynamic features for cell manipulation, few studies have designed a dynamic chemical pattern to sequentially polarize macrophage phenotypes and meet the immune requirements at various tissue repair stages. Here, we propose a novel strategy for spatiotemporal manipulation of macrophage phenotypes by a UV-induced dynamic Arg-Gly-Asp (RGD) pattern. By employing a photo-patterning technique and the specific interaction between cyclodextrin (CD) and azobenzene-RGD (Azo-RGD), we prepared a polyethylene glycol-dithiol/polyethylene glycol-norbornene (PEG-SH/PEG-Nor) hydrogel with dynamic RGD-patterned surface. After irradiation with 365-nm UV light, the homogeneous RGD surface was transformed to the RGD-patterned surface which induced morphological transformation of macrophages from round to elongated and subsequent phenotypic transition from pro-inflammation to anti-inflammation. The mechanism of phenotypic polarization induced by RGD pattern was proved to be related to Rho-associated protein kinase 2 (ROCK2). Sequential modulation of macrophage phenotypes by the dynamic RGD-patterned surface provides a remote and non-invasive strategy to manipulate immune reactions and achieve optimized healing outcomes.
Collapse
Affiliation(s)
- Yilun Luo
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiaowen Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Peiqi Yuan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xingyao Ye
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
200
|
Xu T, Yu S, Zhang J, Wu S. Dysregulated tumor-associated macrophages in carcinogenesis, progression and targeted therapy of gynecological and breast cancers. J Hematol Oncol 2021; 14:181. [PMID: 34717710 PMCID: PMC8557603 DOI: 10.1186/s13045-021-01198-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Gynecological and breast cancers are a group of heterogeneous malignant tumors. Although existing treatment strategies have ameliorated the clinical outcomes of patients, the overall survival rate of advanced diseases remains unsatisfactory. Increasing evidence has indicated that the development and prognosis of tumors are closely related to the tumor microenvironment (TME), which restricts the immune response and provokes malignant progression. Tumor-associated macrophages (TAMs) are the main component of TME and act as a key regulator in tumor metastasis, immunosuppression and therapeutic resistance. Several preclinical trials have studied potential drugs that target TAMs to achieve potent anticancer therapy. This review focuses on the various functions of TAMs and how they influence the carcinogenesis of gynecological and breast cancers through regulating cancer cell proliferation, tumor angiogenesis and tumor-related immunosuppression. Besides, we also discuss the potential application of disabling TAMs signaling as a part of cancer therapeutic strategies, as well as CAR macrophages, TAMs-based vaccines and TAMs nanobiotechnology. These research advances support that targeting TAMs combined with conventional therapy might be used as effective therapeutics for gynecological and breast cancers in the future.
Collapse
Affiliation(s)
- Tianhan Xu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Sihui Yu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China. .,Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Sufang Wu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|