1
|
Sun R, Wang M, Zeng T, Chen H, Yoshitomi T, Takeguchi M, Kawazoe N, Yang Y, Chen G. Scaffolds functionalized with matrix metalloproteinase-responsive release of miRNA for synergistic magnetic hyperthermia and sensitizing chemotherapy of drug-tolerant breast cancer. Bioact Mater 2025; 44:205-219. [PMID: 39502841 PMCID: PMC11535879 DOI: 10.1016/j.bioactmat.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/19/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
Combining hyperthermia and chemotherapy for maximum anticancer efficacy remains a challenge because drug-tolerant cancer cells often evade this synergistic treatment due to drug resistance and asynchronous drug release. In this study, multifunctional scaffolds were designed to efficiently treat drug-tolerant breast cancer by improving the sensitization of breast cancer cells and synchronizing anticancer drug release with magnetic hyperthermia. The scaffolds contained microRNA-encapsulated matrix metalloproteinase-cleavable liposomes, doxorubicin-encapsulated thermoresponsive liposomes and Fe3O4 nanoparticles. The scaffolds could release microRNA specifically to improve the sensitization of breast cancer cells to anticancer drugs. The scaffolds also showed excellent hyperthermia effects under alternating magnetic field irradiation. Moreover, doxorubicin release was synchronized with magnetic hyperthermia. In vitro and in vivo studies demonstrated that the scaffolds effectively reduced drug resistance and eliminated doxorubicin-tolerant MDA-MB-231 cells through the synergistic effect of magnetic hyperthermia and sensitizing chemotherapy. Additionally, the scaffolds could support the proliferation and adipogenic differentiation of stem cells for adipose tissue regeneration after killing cancer cells at a late therapeutic stage. These composite scaffolds offer an innovative strategy for treating breast cancer, with synergistic anticancer effects and regenerative functions.
Collapse
Affiliation(s)
- Rui Sun
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Man Wang
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Tianjiao Zeng
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Huajian Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
| | - Toru Yoshitomi
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
| | - Masaki Takeguchi
- Research Center for Energy and Environmental Materials, National Institute for Materials Science, Ibaraki, 305-0047, Japan
| | - Naoki Kawazoe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, Ibaraki, 305-8572, Japan
| | - Guoping Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Ibaraki, 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| |
Collapse
|
2
|
Shi C, Chen T, Li Y, Li W, Shen Y, Cai K, Wang M, Chen Y. Encapsulation of individual mammalian cells as a cell-based drug delivery carrier for lung cancer treatment. J Control Release 2024; 378:209-220. [PMID: 39662681 DOI: 10.1016/j.jconrel.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Cell-based delivery holds great promise for advancing cell therapy, but it often faces challenges such as low cell survival rates and immunological rejection during cell injection and circulation. In this study, we develop an alternative approach aimed at engineering cell membranes to produce encapsulated individual mammalian (EIM) cells. The encapsulation shell was formed by catalyzing the reaction of H2O2 with hyaluronic acid-dopamine (HA-DA) on the cell surface using horseradish peroxidase (HRP) enzymes. This protective shell not only protects live mammalian cells from physical stress but also from biological assaults. The individual cell encapsulation system enables the loading of cells with chemotherapy drugs, enhancing their targeting ability towards tumor sites and resulting in over 5.1-fold cell enrichment at metastatic tumors, thereby improving tumor-killing efficacy and reducing metastasis. Overall, the individual cell encapsulation system demonstrates potential for targeted drug delivery to the lungs in the treatment of lung cancer by reducing side effects and enhancing treatment outcomes.
Collapse
Affiliation(s)
- Chaochen Shi
- Medical 3D Printing Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China; Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China
| | - Tao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China
| | - Yingying Li
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China
| | - Wenshuai Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China
| | - Yuan Shen
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China
| | - Kehan Cai
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China
| | - Ming Wang
- Medical Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China
| | - Yazhou Chen
- Medical 3D Printing Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, PR China; Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China.
| |
Collapse
|
3
|
Wang B, Hasturk O, Kumarasinghe U, Rudolph S, Staii C, Chen Y, Kaplan DL. Temporary Nanoencapsulation of Human Intestinal Organoids Using Silk Ionomers. Adv Healthc Mater 2024:e2403176. [PMID: 39648539 DOI: 10.1002/adhm.202403176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/03/2024] [Indexed: 12/10/2024]
Abstract
Human intestinal organoids (HIOs) are vital for modeling intestinal development, disease, and therapeutic tissue regeneration. However, their susceptibility to stress, immunological attack, and environmental fluctuations limits their utility in research and therapeutic applications. This study evaluated the effectiveness of temporary silk protein-based layer-by-layer (LbL) nanoencapsulation technique to enhance the viability and functions of HIOs against common biomedical stressors, without compromising their native functions. Cell viability and differentiation capacity are assessed, finding that nanoencapsulation significantly improved HIO survival under the various environmental perturbations studied without compromising cellular functionality. Post-stress exposures, the encapsulated HIOs still successfully differentiated into essential intestinal cell types such as enterocytes, goblet cells, enteroendocrine cells, and Paneth cells. Moreover, the silk nanocoatings effectively protected against environmental stressors such as ultraviolet (UV) light exposure, protease degradation, antibody binding, and cytokine-induced inflammation. This nanoencapsulation technique shows promise for advancing HIO applications in disease modeling, drug testing, and potential transplantation therapies.
Collapse
Affiliation(s)
- Brooke Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Onur Hasturk
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | | | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Cristian Staii
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| | - Ying Chen
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| | - David L Kaplan
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| |
Collapse
|
4
|
Ariaeenejad S, Zeinalabedini M, Sadeghi A, Gharaghani S, Mardi M. Enhancing nutritional and potential antimicrobial properties of poultry feed through encapsulation of metagenome-derived multi-enzymes. BMC Biotechnol 2024; 24:76. [PMID: 39379947 PMCID: PMC11463139 DOI: 10.1186/s12896-024-00904-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The encapsulation of metagenome-derived multi-enzymes presents a novel approach to improving poultry feed by enhancing nutrient availability and reducing anti-nutritional factors. By integrating and encapsulated enzymes such as carbohydrate-hydrolyzing enzymes, protease, lipase, and laccase into feed formulations, this method not only improves feed digestibility but also potentially contributes to animal health and productivity through antimicrobial properties. RESULTS This study investigates the encapsulation of metagenome-derived enzymes, including carbohydrate-hydrolyzing enzymes, protease, lipase, and laccase, using Arabic and Guar gums as encapsulating agents. The encapsulated multi-enzymes exhibited significant antimicrobial activity, achieving a 92.54% inhibition rate against Escherichia coli at a concentration of 6 U/mL. Fluorescence tracking with FITC-labeled enzymes confirmed efficient encapsulation and distribution, while physical characterization, including moisture content and solubility assessments, along with Atomic Force Microscopy (AFM) imaging, validated successful encapsulation. The encapsulated enzymes also effectively hydrolyzed poultry feed, leading to an increase in phenolic content and antioxidant activity, as confirmed by 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid (ABTS) and 2,2-diphenyl-1-picrylhydrazyl (DPPH) assays. CONCLUSIONS The encapsulated multi-enzymes improved the overall feed quality by increasing reducing sugars and enhancing physical properties such as solubility and water-holding capacity. The encapsulated multi-enzymes improved the overall feed quality by increasing reducing sugars, antioxidant activity and enhancing physical properties such as solubility and water-holding capacity. Scanning Electron Microscopy (SEM) and Fourier-Transform Infrared Spectroscopy (FTIR) analyses confirmed the enzymatic breakdown of the feed structure. These results suggest that supplementing poultry feed with encapsulated multi-enzymes can enhance its physical, nutritional, and functional properties, leading to improved digestibility and overall feed quality.
Collapse
Affiliation(s)
- Shohreh Ariaeenejad
- Department of Systems and Synthetic Biology, Agricultural Biotechnology Research Institute of Iran (ABRII), Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran.
| | - Mehrshad Zeinalabedini
- Department of Systems and Synthetic Biology, Agricultural Biotechnology Research Institute of Iran (ABRII), Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Akram Sadeghi
- Department of Microbial Biotechnology, Agricultural Research Education and Extension Organization (AREEO), Agricultural Biotechnology Research Institute of Iran (ABRII), Karaj, Iran
| | - Sajjad Gharaghani
- Laboratory of Bioinformatics & Drug Design (LBD), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Mohsen Mardi
- Department of Systems and Synthetic Biology, Agricultural Biotechnology Research Institute of Iran (ABRII), Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
5
|
Whitewolf J, Highley CB. Conformal encapsulation of mammalian stem cells using modified hyaluronic acid. J Mater Chem B 2024; 12:7122-7134. [PMID: 38946474 PMCID: PMC11268093 DOI: 10.1039/d4tb00223g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024]
Abstract
Micro- and nanoencapsulation of cells has been studied as a strategy to protect cells from environmental stress and promote survival during delivery. Hydrogels used in encapsulation can be modified to influence cell behaviors and direct assembly in their surroundings. Here, we report a system that conformally encapsulated stem cells using hyaluronic acid (HA). We successfully modified HA with lipid, thiol, and maleimide pendant groups to facilitate a hydrogel system in which HA was deposited onto cell plasma membranes and subsequently crosslinked through thiol-maleimide click chemistry. We demonstrated conformal encapsulation of both neural stem cells (NSCs) and mesenchymal stromal cells (MSCs), with viability of both cell types greater than 90% after encapsulation. Additional material could be added to the conformal hydrogel through alternating addition of thiol-modified and maleimide-modified HA in a layering process. After encapsulation, we tracked egress and viability of the cells over days and observed differential responses of cell types to conformal hydrogels both according to cell type and the amount of material deposited on the cell surfaces. Through the design of the conformal hydrogels, we showed that multicellular assembly could be created in suspension and that encapsulated cells could be immobilized on surfaces. In conjunction with photolithography, conformal hydrogels enabled rapid assembly of encapsulated cells on hydrogel substrates with resolution at the scale of 100 μm.
Collapse
Affiliation(s)
- Jack Whitewolf
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA.
| | - Christopher B Highley
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA.
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
6
|
Xuan L, Hou Y, Liang L, Wu J, Fan K, Lian L, Qiu J, Miao Y, Ravanbakhsh H, Xu M, Tang G. Microgels for Cell Delivery in Tissue Engineering and Regenerative Medicine. NANO-MICRO LETTERS 2024; 16:218. [PMID: 38884868 PMCID: PMC11183039 DOI: 10.1007/s40820-024-01421-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/26/2024] [Indexed: 06/18/2024]
Abstract
Microgels prepared from natural or synthetic hydrogel materials have aroused extensive attention as multifunctional cells or drug carriers, that are promising for tissue engineering and regenerative medicine. Microgels can also be aggregated into microporous scaffolds, promoting cell infiltration and proliferation for tissue repair. This review gives an overview of recent developments in the fabrication techniques and applications of microgels. A series of conventional and novel strategies including emulsification, microfluidic, lithography, electrospray, centrifugation, gas-shearing, three-dimensional bioprinting, etc. are discussed in depth. The characteristics and applications of microgels and microgel-based scaffolds for cell culture and delivery are elaborated with an emphasis on the advantages of these carriers in cell therapy. Additionally, we expound on the ongoing and foreseeable applications and current limitations of microgels and their aggregate in the field of biomedical engineering. Through stimulating innovative ideas, the present review paves new avenues for expanding the application of microgels in cell delivery techniques.
Collapse
Affiliation(s)
- Leyan Xuan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yingying Hou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Jialin Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Kai Fan
- School of Automation, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
| | - Liming Lian
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jianhua Qiu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yingling Miao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Hossein Ravanbakhsh
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA.
| | - Mingen Xu
- School of Automation, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China.
| | - Guosheng Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| |
Collapse
|
7
|
Song Y, Dong QQ, Ni YK, Xu XL, Chen CX, Chen W. Nano-Proteolysis Targeting Chimeras (Nano-PROTACs) in Cancer Therapy. Int J Nanomedicine 2024; 19:5739-5761. [PMID: 38882545 PMCID: PMC11180470 DOI: 10.2147/ijn.s448684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules that have the capability to induce specific protein degradation. While playing a revolutionary role in effectively degrading the protein of interest (POI), PROTACs encounter certain limitations that impede their clinical translation. These limitations encompass off-target effects, inadequate cell membrane permeability, and the hook effect. The advent of nanotechnology presents a promising avenue to surmount the challenges associated with conventional PROTACs. The utilization of nano-proteolysis targeting chimeras (nano-PROTACs) holds the potential to enhance specific tissue accumulation, augment membrane permeability, and enable controlled release. Consequently, this approach has the capacity to significantly enhance the controllable degradation of target proteins. Additionally, they enable a synergistic effect by combining with other therapeutic strategies. This review comprehensively summarizes the structural basis, advantages, and limitations of PROTACs. Furthermore, it highlights the latest advancements in nanosystems engineered for delivering PROTACs, as well as the development of nano-sized PROTACs employing nanocarriers as linkers. Moreover, it delves into the underlying principles of nanotechnology tailored specifically for PROTACs, alongside the current prospects of clinical research. In conclusion, the integration of nanotechnology into PROTACs harbors vast potential in enhancing the anti-tumor treatment response and expediting clinical translation.
Collapse
Affiliation(s)
- Yue Song
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, People’s Republic of China
| | - Qing-Qing Dong
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| | - Yi-Ke Ni
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Chao-Xiang Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Wei Chen
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
8
|
Mu R, Zhu D, Abdulmalik S, Wijekoon S, Wei G, Kumbar SG. Stimuli-responsive peptide assemblies: Design, self-assembly, modulation, and biomedical applications. Bioact Mater 2024; 35:181-207. [PMID: 38327824 PMCID: PMC10847779 DOI: 10.1016/j.bioactmat.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/09/2024] Open
Abstract
Peptide molecules have design flexibility, self-assembly ability, high biocompatibility, good biodegradability, and easy functionalization, which promote their applications as versatile biomaterials for tissue engineering and biomedicine. In addition, the functionalization of self-assembled peptide nanomaterials with other additive components enhances their stimuli-responsive functions, promoting function-specific applications that induced by both internal and external stimulations. In this review, we demonstrate recent advance in the peptide molecular design, self-assembly, functional tailoring, and biomedical applications of peptide-based nanomaterials. The strategies on the design and synthesis of single, dual, and multiple stimuli-responsive peptide-based nanomaterials with various dimensions are analyzed, and the functional regulation of peptide nanomaterials with active components such as metal/metal oxide, DNA/RNA, polysaccharides, photosensitizers, 2D materials, and others are discussed. In addition, the designed peptide-based nanomaterials with temperature-, pH-, ion-, light-, enzyme-, and ROS-responsive abilities for drug delivery, bioimaging, cancer therapy, gene therapy, antibacterial, as well as wound healing and dressing applications are presented and discussed. This comprehensive review provides detailed methodologies and advanced techniques on the synthesis of peptide nanomaterials from molecular biology, materials science, and nanotechnology, which will guide and inspire the molecular level design of peptides with specific and multiple functions for function-specific applications.
Collapse
Affiliation(s)
- Rongqiu Mu
- College of Chemistry and Chemical Engineering, Qingdao University, 266071, Qingdao, China
| | - Danzhu Zhu
- College of Chemistry and Chemical Engineering, Qingdao University, 266071, Qingdao, China
| | - Sama Abdulmalik
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, 06030, USA
| | - Suranji Wijekoon
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, 06030, USA
| | - Gang Wei
- College of Chemistry and Chemical Engineering, Qingdao University, 266071, Qingdao, China
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering & Department of Materials Science and Engineering, University of Connecticut, Storrs, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, 06030, USA
| |
Collapse
|
9
|
Tian F, Zhou Y, Ma Z, Tang R, Wang X. Organismal Function Enhancement through Biomaterial Intervention. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:377. [PMID: 38392750 PMCID: PMC10891834 DOI: 10.3390/nano14040377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 02/24/2024]
Abstract
Living organisms in nature, such as magnetotactic bacteria and eggs, generate various organic-inorganic hybrid materials, providing unique functionalities. Inspired by such natural hybrid materials, researchers can reasonably integrate biomaterials with living organisms either internally or externally to enhance their inherent capabilities and generate new functionalities. Currently, the approaches to enhancing organismal function through biomaterial intervention have undergone rapid development, progressing from the cellular level to the subcellular or multicellular level. In this review, we will concentrate on three key strategies related to biomaterial-guided bioenhancement, including biointerface engineering, artificial organelles, and 3D multicellular immune niches. For biointerface engineering, excess of amino acid residues on the surfaces of cells or viruses enables the assembly of materials to form versatile artificial shells, facilitating vaccine engineering and biological camouflage. Artificial organelles refer to artificial subcellular reactors made of biomaterials that persist in the cytoplasm, which imparts cells with on-demand regulatory ability. Moreover, macroscale biomaterials with spatiotemporal regulation characters enable the local recruitment and aggregation of cells, denoting multicellular niche to enhance crosstalk between cells and antigens. Collectively, harnessing the programmable chemical and biological attributes of biomaterials for organismal function enhancement shows significant potential in forthcoming biomedical applications.
Collapse
Affiliation(s)
- Fengchao Tian
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; (F.T.); (Y.Z.)
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China;
| | - Yuemin Zhou
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; (F.T.); (Y.Z.)
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China;
| | - Zaiqiang Ma
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China;
| | - Ruikang Tang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; (F.T.); (Y.Z.)
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China;
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China; (F.T.); (Y.Z.)
| |
Collapse
|
10
|
Zhao L, Chen J, Bai B, Song G, Zhang J, Yu H, Huang S, Wang Z, Lu G. Topical drug delivery strategies for enhancing drug effectiveness by skin barriers, drug delivery systems and individualized dosing. Front Pharmacol 2024; 14:1333986. [PMID: 38293666 PMCID: PMC10825035 DOI: 10.3389/fphar.2023.1333986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/27/2023] [Indexed: 02/01/2024] Open
Abstract
Topical drug delivery is widely used in various diseases because of the advantages of not passing through the gastrointestinal tract, avoiding gastrointestinal irritation and hepatic first-pass effect, and reaching the lesion directly to reduce unnecessary adverse reactions. The skin helps the organism to defend itself against a huge majority of external aggressions and is one of the most important lines of defense of the body. However, the skin's strong barrier ability is also a huge obstacle to the effectiveness of topical medications. Allowing the bioactive, composition in a drug to pass through the stratum corneum barrier as needed to reach the target site is the most essential need for the bioactive, composition to exert its therapeutic effect. The state of the skin barrier, the choice of delivery system for the bioactive, composition, and individualized disease detection and dosing planning influence the effectiveness of topical medications. Nowadays, enhancing transdermal absorption of topically applied drugs is the hottest research area. However, enhancing transdermal absorption of drugs is not the first choice to improve the effectiveness of all drugs. Excessive transdermal absorption enhances topical drug accumulation at non-target sites and the occurrence of adverse reactions. This paper introduces topical drug delivery strategies to improve drug effectiveness from three perspectives: skin barrier, drug delivery system and individualized drug delivery, describes the current status and shortcomings of topical drug research, and provides new directions and ideas for topical drug research.
Collapse
Affiliation(s)
- Lin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiamei Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bai Bai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guili Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiwei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhang Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guanghua Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Wang X, Jia B, Lee K, Davis B, Wen C, Wang Y, Zheng H, Wang Y. Biomimetic Bacterial Capsule for Enhanced Aptamer Display and Cell Recognition. J Am Chem Soc 2024; 146:868-877. [PMID: 38153404 DOI: 10.1021/jacs.3c11208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Great effort has been made to encapsulate or coat living mammalian cells for a variety of applications ranging from diabetes treatment to three-dimensional printing. However, no study has reported the synthesis of a biomimetic bacterial capsule to display high-affinity aptamers on the cell surface for enhanced cell recognition. Therefore, we synthesized an ultrathin alginate-polylysine coating to display aptamers on the surface of living cells with natural killer (NK) cells as a model. The results show that this coating-mediated aptamer display is more stable than direct cholesterol insertion into the lipid bilayer. The half-life of the aptamer on the cell surface can be increased from less than 1.5 to over 20 h. NK cells coated with the biomimetic bacterial capsule exhibit a high efficiency in recognizing and killing target cells. Therefore, this work has demonstrated a promising cell coating method for the display of aptamers for enhanced cell recognition.
Collapse
Affiliation(s)
- Xuelin Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Bei Jia
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Kyungsene Lee
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Brandon Davis
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Connie Wen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Yixun Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Hong Zheng
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
12
|
Wang K, Zhao C, Ma Y, Yang W. Yolk-Shell Encapsulation of Cells by Biomimetic Mineralization and Visible Light-Induced Surface Graft Polymerization. Biomacromolecules 2023; 24:6032-6040. [PMID: 37967289 DOI: 10.1021/acs.biomac.3c01143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
The pursuit of low-cytotoxicity modification strategies represents a prominent avenue in cell coating research, holding immense significance for the advancement of practical living cell-related technologies. Here, we presented a novel method to fabricate encapsulated yeast cells with a yolk-shell structure by biomimetic mineralization and visible-light-induced surface graft polymerization. In this approach, an amorphous calcium carbonate (ACC) shell was first deposited on the surface of a yeast cell (cell@ACC) modified with 4 layers of self-assembled poly(diallyl dimethylammonium chloride) (PDADMAC)/poly(acrylic acid) (PAA) film using a biomimetic mineralization technique. Subsequently, polyethylenimine (PEI) was absorbed on the surface of cell@ACC by electrostatic interaction. Then, a cross-linked shell was introduced by surface-initiated graft polymerization of poly(ethylene glycol) diacrylate (PEGDA) on cell@ACC under irradiation of visible light using thioxanthone catechol-O,O'-diacetic acid as the photosensitizer. After the removal of the inner ACC shell, the yolk-shell-structured yeast cells (cell@PHS) were obtained. Due to the mild conditions of the strategy, the cell@PHS could retain 98.81% of its original viability. The introduction of the shell layer significantly prolonged the lag phase of yeast cells, which could be tuned between 5 and 25 h by regulating the thickness of the shell. Moreover, the cell@PHS showed improved resistance against lyticase due to the presence of a protective shell. After 30 days of storage, the viability of cell@PHS was 81.09%, which is significantly higher than the 19.89% viability of native yeast cells.
Collapse
Affiliation(s)
- Kanglei Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Changwen Zhao
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
- Key Laboratory of Biomedical Materials of Natural Macromolecules, Ministry of Education Beijing, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yuhong Ma
- Key Laboratory of Carbon Fiber and Functional Polymers Ministry of Education, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wantai Yang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
- Key Laboratory of Biomedical Materials of Natural Macromolecules, Ministry of Education Beijing, Beijing University of Chemical Technology, Beijing 100029, China
- Key Laboratory of Carbon Fiber and Functional Polymers Ministry of Education, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
13
|
Yang H, Yao L, Wang Y, Chen G, Chen H. Advancing cell surface modification in mammalian cells with synthetic molecules. Chem Sci 2023; 14:13325-13345. [PMID: 38033886 PMCID: PMC10685406 DOI: 10.1039/d3sc04597h] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Biological cells, being the fundamental entities of life, are widely acknowledged as intricate living machines. The manipulation of cell surfaces has emerged as a progressively significant domain of investigation and advancement in recent times. Particularly, the alteration of cell surfaces using meticulously crafted and thoroughly characterized synthesized molecules has proven to be an efficacious means of introducing innovative functionalities or manipulating cells. Within this realm, a diverse array of elegant and robust strategies have been recently devised, including the bioorthogonal strategy, which enables selective modification. This review offers a comprehensive survey of recent advancements in the modification of mammalian cell surfaces through the use of synthetic molecules. It explores a range of strategies, encompassing chemical covalent modifications, physical alterations, and bioorthogonal approaches. The review concludes by addressing the present challenges and potential future opportunities in this rapidly expanding field.
Collapse
Affiliation(s)
- He Yang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
| | - Lihua Yao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
| | - Yichen Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
| | - Gaojian Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University Suzhou 215006 Jiangsu P. R. China
| | - Hong Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
| |
Collapse
|
14
|
Cai F, Ren Y, Dai J, Yang J, Shi X. Effects of Various Cell Surface Engineering Reactions on the Biological Behavior of Mammalian Cells. Macromol Biosci 2023; 23:e2200379. [PMID: 36579789 DOI: 10.1002/mabi.202200379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/15/2022] [Indexed: 12/30/2022]
Abstract
Cell surface engineering technologies can regulate cell function and behavior by modifying the cell surface. Previous studies have mainly focused on investigating the effects of cell surface engineering reactions and materials on cell activity. However, they do not comprehensively analyze other cellular processes. This study exploits covalent bonding, hydrophobic interactions, and electrostatic interactions to modify the macromolecules succinimide ester-methoxy polyethylene glycol (NHS-mPEG), distearoyl phosphoethanolamine-methoxy polyethylene glycol (DSPE-mPEG), and poly-L-lysine (PLL), respectively, on the cell surface. This work systematically investigates the effects of the three surface engineering reactions on the behavior of human umbilical vein endothelial cells (HUVECs) and human skin fibroblasts, including viability, growth, proliferation, cell cycle, adhesion, and migration. The results reveals that the PLL modification method notably affects cell viability and G2/M arrest and has a short modification duration. However, the DSPE-mPEG and NHS-mPEG modification methods have little effect on cell viability and proliferation but have a prolonged modification duration. Moreover, the DSPE-mPEG modification method highly affects cell adherence. Further, the NHS-mPEG modification method can significantly improve the migration ability of HUVECs by reducing the area of focal adhesions. The findings of this study will contribute to the application of cell surface engineering technology in the biomedical field.
Collapse
Affiliation(s)
- Fengying Cai
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yafeng Ren
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Jiajia Dai
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China.,Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China.,Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| |
Collapse
|
15
|
Mokhtarinia K, Rezvanian P, Masaeli E. Sustainable hydrogel-based cell therapy. SUSTAINABLE HYDROGELS 2023:443-470. [DOI: 10.1016/b978-0-323-91753-7.00009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
16
|
Wen L, Li G, Huang T, Geng W, Pei H, Yang J, Zhu M, Zhang P, Hou R, Tian G, Su W, Chen J, Zhang D, Zhu P, Zhang W, Zhang X, Zhang N, Zhao Y, Cao X, Peng G, Ren X, Jiang N, Tian C, Chen ZJ. Single-cell technologies: From research to application. Innovation (N Y) 2022; 3:100342. [PMID: 36353677 PMCID: PMC9637996 DOI: 10.1016/j.xinn.2022.100342] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022] Open
Abstract
In recent years, more and more single-cell technologies have been developed. A vast amount of single-cell omics data has been generated by large projects, such as the Human Cell Atlas, the Mouse Cell Atlas, the Mouse RNA Atlas, the Mouse ATAC Atlas, and the Plant Cell Atlas. Based on these single-cell big data, thousands of bioinformatics algorithms for quality control, clustering, cell-type annotation, developmental inference, cell-cell transition, cell-cell interaction, and spatial analysis are developed. With powerful experimental single-cell technology and state-of-the-art big data analysis methods based on artificial intelligence, the molecular landscape at the single-cell level can be revealed. With spatial transcriptomics and single-cell multi-omics, even the spatial dynamic multi-level regulatory mechanisms can be deciphered. Such single-cell technologies have many successful applications in oncology, assisted reproduction, embryonic development, and plant breeding. We not only review the experimental and bioinformatics methods for single-cell research, but also discuss their applications in various fields and forecast the future directions for single-cell technologies. We believe that spatial transcriptomics and single-cell multi-omics will become the next booming business for mechanism research and commercial industry.
Collapse
Affiliation(s)
- Lu Wen
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Guoqiang Li
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Geng
- School of Chemical Engineering and Technology, Sun Yat-Sen University, Zhuhai 519082, China
| | - Hao Pei
- Mozhuo Biotech (Zhejiang) Co., Ltd., Tongxiang, Jiaxing 314500, China
| | | | - Miao Zhu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Pengfei Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Rui Hou
- Geneis (Beijing) Co., Ltd., Beijing 100102, China
| | - Geng Tian
- Geneis (Beijing) Co., Ltd., Beijing 100102, China
| | - Wentao Su
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jian Chen
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
| | - Dake Zhang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100083, China
| | - Pingan Zhu
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong 999077, China
| | - Wei Zhang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Xiuxin Zhang
- Center of Peony, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Key Laboratory of Biology and Genetic Improvement of Flower Crops (North China), Key Laboratory of Biology and Genetic Improvement of Horticultural Crops, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ning Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yunlong Zhao
- Advanced Technology Institute, University of Surrey, Guildford, Surrey, GU2 7XH, UK
- National Physical Laboratory, Teddington, Middlesex TW11 0LW, UK
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guangdun Peng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Nan Jiang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Jinfeng Laboratory, Chongqing 401329, China
| | - Caihuan Tian
- Center of Peony, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Key Laboratory of Biology and Genetic Improvement of Flower Crops (North China), Key Laboratory of Biology and Genetic Improvement of Horticultural Crops, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
17
|
Liu C, Wang M, Zhang H, Li C, Zhang T, Liu H, Zhu S, Chen J. Tumor microenvironment and immunotherapy of oral cancer. Eur J Med Res 2022; 27:198. [PMID: 36209263 PMCID: PMC9547678 DOI: 10.1186/s40001-022-00835-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
Oral cancer is one of the most common malignant tumors of the head and neck, not only affects the appearance, but also affects eating and even endangers life. The clinical treatments of oral cancer mainly include surgery, radiotherapy, and chemotherapy. However, unsatisfactory therapeutic effect and toxic side effects are still the main problems in clinical treatment. Tumor microenvironment (TME) is not only closely related to the occurrence, growth, and metastasis of tumor but also works in the diagnosis, prevention, and treatment of tumor and prognosis. Future studies should continue to investigate the relationship of TME and oral cancer therapy. This purpose of this review was to analyze the characteristics of oral cancer microenvironment, summarize the traditional oral cancer therapy and immunotherapy strategies, and finally prospect the development prospects of oral cancer immunotherapy. Immunotherapy targeting tumor microenvironment is expected to provide a new strategy for clinical treatment of oral cancer.
Collapse
Affiliation(s)
- Chang Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Min Wang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Haiyang Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Chunyan Li
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Tianshou Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Hong Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Song Zhu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China.
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China.
| |
Collapse
|
18
|
Shao J, Nie P, Yang W, Guo R, Ding D, Liang R, Wei B, Wei H. An EPO-loaded multifunctional hydrogel synergizing with adipose-derived stem cells restores neurogenic erectile function via enhancing nerve regeneration and penile rehabilitation. Bioeng Transl Med 2022; 7:e10319. [PMID: 36176612 PMCID: PMC9471998 DOI: 10.1002/btm2.10319] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 12/02/2022] Open
Abstract
Neurogenic erectile dysfunction (nED) is one of the most common and intractable postoperative complications of rectal and prostate cancer surgery and sometimes accompanies patients lifelong. The transplantation of stem cells has been proved a promising way for treatment. However, the therapeutic efficacy is severely impaired by excessive cell loss and death and poor accumulation in the injury site along with the traditional implantation strategy. Herein, an EPO-loaded multifunctional hydrogel was designed. The hydrogels' adhesive property and mechanical strength were enhanced by adding catechol-catechol adducts, thus significantly improving adipose-derived stem cells (ADSC) retention and rescuing cell loss in the injury site. Meanwhile, the sustained release of EPO effectively ameliorated the viability and paracrine activity of ADSC, leading to enhanced migration of Schwann cells and differentiation of PC12 cells in vivo. On a bilateral cavernous nerve injury rat model, the present stem cell-EPO-hydrogel implanted strategy could significantly alleviate erectile dysfunction. The higher expression of Tuj1 and lower expression of GFAP in the major pelvic ganglia (MPG) indicated the acceleration of neural differentiation while the suppressing development of astrocytes. Also, the combined therapy restored the expression levels of eNOs, nNOs, and α-SMA in penile tissues, suggesting the rehabilitation of the penis. Further analysis of Masson trichrome staining and apoptosis evaluation of the corpus cavernosum showed the preservation of vascular endothelium content and the prevention of penile fibrosis after denervation. Overall, we believe that this combined strategy presents a promising way not only for restoring neurogenic erectile function but also for the clinical translation of stem cell therapy.
Collapse
Affiliation(s)
- Jun Shao
- Department of Gastrointestinal SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Pan Nie
- Department of Gastrointestinal SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Wende Yang
- Department of Gastrointestinal SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrier Development, Department of Biomedical EngineeringJinan UniversityGuangzhouChina
| | - Dongbing Ding
- Department of Gastrointestinal SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Rongpu Liang
- Department of Gastrointestinal SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Bo Wei
- Department of Gastrointestinal SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Hongbo Wei
- Department of Gastrointestinal SurgeryThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
19
|
Zhu S, Yu C, Liu N, Zhao M, Chen Z, Liu J, Li G, Huang H, Guo H, Sun T, Chen J, Zhuang J, Zhu P. Injectable conductive gelatin methacrylate / oxidized dextran hydrogel encapsulating umbilical cord mesenchymal stem cells for myocardial infarction treatment. Bioact Mater 2022; 13:119-134. [PMID: 35224296 PMCID: PMC8844712 DOI: 10.1016/j.bioactmat.2021.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 02/03/2023] Open
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) transplantation has been proposed as a promising treatment modality for myocardial infarction (MI), but the low retention rate remains a considerable challenge. Injectable natural polymer hydrogels with conductivity ability are highly desirable as cell delivery vehicles to repair infarct myocardium and restore the cardiac function. In this work, we developed a hydrogel system based on gelatin methacrylate (GelMA) and oxidized dextran (ODEX) as cell delivery vehicles for MI. And dopamine could be used as a reductant of graphene oxide (GO) to form reductive GO (rGO). By adjusting the amount of rGO, the conductivity of hydrogels with 0.5 mg/mL rGO concentration (≈10−4 S/cm) was similar to that of natural heart tissue. In vitro cell experiments showed that the prepared hydrogels had excellent biocompatibility and cell delivery ability of UCMSCs. More importantly, GelMA-O5/rGO hydrogel could promote UCMSCs growth and proliferation, improve the myocardial differentiation ability of UCMSCs, and up-regulate the expression of cTnI and Cx43. Further in vivo experiments demonstrated that GelMA-O5/rGO/UCMSCs Hydrogel could significantly improve the ejection fraction (EF) of rats and significantly reduce myocardial infarct area compared to PBS group, promote the survival of UCMSCs, enhance the expression level of cTnI and Cx43, and decrease the expression level of caspase-3. The findings of this study suggested that the injectable conductive GelMA-O5/rGO hydrogel encapsulating UCMSCs could improve damaged myocardial tissue and reconstruct myocardial function, which will be a promising therapeutic strategy for cardiac repair. Conducting interpenetrating polymer network (IPN) hydrogels were synthesized for myocardial infarction treatment. The conductivity of hydrogel with 0.5 mg/mL rGO concentration (≈10−4 S/cm) was similar to that of natural heart tissue. The hydrogel could promote the growth and proliferation of UCMSCs, and improve the myocardial differentiation ability of UCMSCs. The hydrogel could reduce infarct size and cardiac fibrosis in the infarct zone, increase ventricular ejection fraction. The hydrogel could promote the survival of UCMSCs, up-regulate the expression level of cTnI and Cx43, down-regulate the expression level of caspase-3.
Collapse
|
20
|
Marikar SN, El-Osta A, Johnston A, Such G, Al-Hasani K. Microencapsulation-based cell therapies. Cell Mol Life Sci 2022; 79:351. [PMID: 35674842 PMCID: PMC9177480 DOI: 10.1007/s00018-022-04369-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/06/2022] [Accepted: 05/10/2022] [Indexed: 11/25/2022]
Abstract
Mapping a new therapeutic route can be fraught with challenges, but recent developments in the preparation and properties of small particles combined with significant improvements to tried and tested techniques offer refined cell targeting with tremendous translational potential. Regenerating new cells through the use of compounds that regulate epigenetic pathways represents an attractive approach that is gaining increased attention for the treatment of several diseases including Type 1 Diabetes and cardiomyopathy. However, cells that have been regenerated using epigenetic agents will still encounter immunological barriers as well as limitations associated with their longevity and potency during transplantation. Strategies aimed at protecting these epigenetically regenerated cells from the host immune response include microencapsulation. Microencapsulation can provide new solutions for the treatment of many diseases. In particular, it offers an advantageous method of administering therapeutic materials and molecules that cannot be substituted by pharmacological substances. Promising clinical findings have shown the potential beneficial use of microencapsulation for islet transplantation as well as for cardiac, hepatic, and neuronal repair. For the treatment of diseases such as type I diabetes that requires insulin release regulated by the patient's metabolic needs, microencapsulation may be the most effective therapeutic strategy. However, new materials need to be developed, so that transplanted encapsulated cells are able to survive for longer periods in the host. In this article, we discuss microencapsulation strategies and chart recent progress in nanomedicine that offers new potential for this area in the future.
Collapse
Affiliation(s)
- Safiya Naina Marikar
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Angus Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Georgina Such
- School of Chemistry, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Keith Al-Hasani
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
21
|
Chong-Boon Ong, Mohamad Suffian Mohamad Annuar. Hydrogels Responsive Towards Important Biological-Based Stimuli. POLYMER SCIENCE SERIES B 2022. [DOI: 10.1134/s1560090422200015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Wang X, Shan M, Zhang S, Chen X, Liu W, Chen J, Liu X. Stimuli-Responsive Antibacterial Materials: Molecular Structures, Design Principles, and Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104843. [PMID: 35224893 PMCID: PMC9069201 DOI: 10.1002/advs.202104843] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/30/2022] [Indexed: 05/03/2023]
Abstract
Infections are regarded as the most severe complication associated with human health, which are urgent to be solved. Stimuli-responsive materials are appealing therapeutic platforms for antibacterial treatments, which provide great potential for accurate theranostics. In this review, the advantages, the response mechanisms, and the key design principles of stimuli-responsive antibacterial materials are highlighted. The biomedical applications, the current challenges, and future directions of stimuli-responsive antibacterial materials are also discussed. First, the categories of stimuli-responsive antibacterial materials are comprehensively itemized based on different sources of stimuli, including external physical environmental stimuli (e.g., temperature, light, electricity, salt, etc.) and bacterial metabolites stimuli (e.g., acid, enzyme, redox, etc.). Second, structural characteristics, design principles, and biomedical applications of the responsive materials are discussed, and the underlying interrelationships are revealed. The molecular structures and design principles are closely related to the sources of stimuli. Finally, the challenging issues of stimuli-responsive materials are proposed. This review will provide scientific guidance to promote the clinical applications of stimuli-responsive antibacterial materials.
Collapse
Affiliation(s)
- Xianghong Wang
- School of Materials Science and EngineeringThe Key Laboratory of Material Processing and Mold of Ministry of EducationHenan Key Laboratory of Advanced Nylon Materials and ApplicationZhengzhou UniversityZhengzhou450001China
| | - Mengyao Shan
- School of Materials Science and EngineeringThe Key Laboratory of Material Processing and Mold of Ministry of EducationHenan Key Laboratory of Advanced Nylon Materials and ApplicationZhengzhou UniversityZhengzhou450001China
| | - Shike Zhang
- School of Materials Science and EngineeringThe Key Laboratory of Material Processing and Mold of Ministry of EducationHenan Key Laboratory of Advanced Nylon Materials and ApplicationZhengzhou UniversityZhengzhou450001China
| | - Xin Chen
- College of Food Science and EngineeringNational Engineering Research Center for Wheat & Corn Further ProcessingHenan University of TechnologyZhengzhou450001China
| | - Wentao Liu
- School of Materials Science and EngineeringThe Key Laboratory of Material Processing and Mold of Ministry of EducationHenan Key Laboratory of Advanced Nylon Materials and ApplicationZhengzhou UniversityZhengzhou450001China
| | - Jinzhou Chen
- School of Materials Science and EngineeringThe Key Laboratory of Material Processing and Mold of Ministry of EducationHenan Key Laboratory of Advanced Nylon Materials and ApplicationZhengzhou UniversityZhengzhou450001China
| | - Xuying Liu
- School of Materials Science and EngineeringThe Key Laboratory of Material Processing and Mold of Ministry of EducationHenan Key Laboratory of Advanced Nylon Materials and ApplicationZhengzhou UniversityZhengzhou450001China
| |
Collapse
|
23
|
Wang W, Wang S. Cell-based biocomposite engineering directed by polymers. LAB ON A CHIP 2022; 22:1042-1067. [PMID: 35244136 DOI: 10.1039/d2lc00067a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Biological cells such as bacterial, fungal, and mammalian cells always exploit sophisticated chemistries and exquisite micro- and nano-structures to execute life activities, providing numerous templates for engineering bioactive and biomorphic materials, devices, and systems. To transform biological cells into functional biocomposites, polymer-directed cell surface engineering and intracellular functionalization have been developed over the past two decades. Polymeric materials can be easily adopted by various cells through polymer grafting or in situ hydrogelation and can successfully bridge cells with other functional materials as interfacial layers, thus achieving the manufacture of advanced biocomposites through bioaugmentation of living cells and transformation of cells into templated materials. This review article summarizes the recent progress in the design and construction of cell-based biocomposites by polymer-directed strategies. Furthermore, the applications of cell-based biocomposites in broad fields such as cell research, biomedicine, and bioenergy are discussed. Last, we provide personal perspectives on challenges and future trends in this interdisciplinary area.
Collapse
Affiliation(s)
- Wenshuo Wang
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
- Shandong Energy Institute, Qingdao, 266101, China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
24
|
Nanocell hybrids for green chemistry. Trends Biotechnol 2022; 40:974-986. [PMID: 35210123 DOI: 10.1016/j.tibtech.2022.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/28/2022]
Abstract
Global concerns about reducing or minimizing the costs associated with toxic waste materials have driven the continuing development of green-cell-based biosynthesis methods. Inspired by the hybridization phenomenon of living organisms, recent interest has arisen in nanocell hybrids that possess multiple new functions. They have potential to propel biosynthesis into a new generation of green chemistry. This review article discusses the development of applications for nanocell hybrids in the areas of sustainable energy, clean environment, and green catalysis. Continuing advances in these hybrids will require combining knowledge from the fields of biology, physics, chemistry, material science, and engineering.
Collapse
|
25
|
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: 90] [Impact Index Per Article: 30.0] [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
|
26
|
Combination of GNRs-PEI/cGAMP-laden macrophages-based photothermal induced in situ tumor vaccines and immune checkpoint blockade for synergistic anti-tumor immunotherapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 133:112603. [PMID: 35525760 DOI: 10.1016/j.msec.2021.112603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/27/2021] [Accepted: 12/05/2021] [Indexed: 11/22/2022]
Abstract
Immunotherapy is an effective strategy to control and eliminate primary and metastatic tumor by restarting and restoring the specific anti-tumor immune response. However, tumor immunotherapy often showed limited efficacy due to the poor T cell responses in vivo and the tumor suppressive microenvironments. Herein, we constructed polyethyleimine modified gold nanorods (GNRs-PEI) by conjugating PEI to GNRs via SAu bonds. GNRs-PEI/cGAMP nanoparticles were formed via electrostatic interaction and then loaded by macrophages. The GNRs-PEI/cGAMP-laden macrophages (GPc-RAWs) were intravenously injected into the tumor bearing mice and the in situ tumor vaccines were obtained after NIR irradiation. Besides, anti-PD-L1 antibody, an immune checkpoint inhibitor, was introduced to reverse immunosuppressive microenvironment and assisted to achieve the synergistic anti-tumor immunotherapy. GNRs-PEI/cGAMP-laden macrophages with NIR irradiation could effectively inhibit the primary tumors, while little effect for the contralateral tumors. When combined with anti-PD-L1 antibody, the combined strategy not only inhibited the growth of primary tumor, but also significantly delayed the proliferation of the contralateral tumors. More importantly, this strategy reversed immunosuppressive microenvironment without obvious side effects. Therefore, this study provided a great immunotherapy platform for the efficient treatment of primary and metastatic tumors.
Collapse
|
27
|
Liu WM, Zhou X, Chen CY, Lv DD, Huang WJ, Peng Y, Wu HP, Chen Y, Tang D, Guo LN, Wang XL, Zhang HD, Liu XH, Yang LQ, Yu WF, Yan HX. Establishment of Functional Liver Spheroids From Human Hepatocyte-Derived Liver Progenitor-Like Cells for Cell Therapy. Front Bioeng Biotechnol 2021; 9:738081. [PMID: 34858956 PMCID: PMC8630579 DOI: 10.3389/fbioe.2021.738081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/26/2021] [Indexed: 01/18/2023] Open
Abstract
Globally, about two million people die from liver diseases every year. Liver transplantation is the only reliable therapy for severe end-stage liver disease, however, the shortage of organ donors is a huge limitation. Human hepatocytes derived liver progenitor-like cells (HepLPCs) have been reported as a novel source of liver cells for development of in vitro models, cell therapies, and tissue-engineering applications, but their functionality as transplantation donors is unclear. Here, a 3-dimensional (3D) co-culture system using HepLPCs and human umbilical vein endothelial cells (HUVECs) was developed. These HepLPC spheroids mimicked the cellular interactions and architecture of mature hepatocytes, as confirmed through ultrastructure morphology, gene expression profile and functional assays. HepLPCs encapsulated in alginate beads are able to mitigate liver injury in mice treated with carbon tetrachloride (CCL4), while alginate coating protects the cells from immune attack. We confirmed these phenomena due to HUVECs producing glial cell line-derived neurotrophic factor (GDNF) to promote HepLPCs maturation and enhance HepLPCs tight junction through MET phosphorylation. Our results display the efficacy and safety of the alginate microencapsulated spheroids in animal model with acute liver injury (ALF), which may suggest a new strategy for cell therapy.
Collapse
Affiliation(s)
- Wen-Ming Liu
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Xu Zhou
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Cai-Yang Chen
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Dong-Dong Lv
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wei-Jian Huang
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Yuan Peng
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong-Ping Wu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yi Chen
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Dan Tang
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Li-Na Guo
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiu-Li Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Hong-Dan Zhang
- Shanghai Celliver Biotechnology Co. Ltd., Shanghai, China
| | - Xiao-Hua Liu
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Li-Qun Yang
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China
| | - He-Xin Yan
- Department of Anesthesiology and Critical Care Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.,Shanghai Celliver Biotechnology Co. Ltd., Shanghai, China.,Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Delivery of extracellular matrix-enriched stem cells encapsulated with enzyme-free pH-sensitive polymer for enhancing therapeutic angiogenesis. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2021.08.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
29
|
Bone Regeneration Using MMP-Cleavable Peptides-Based Hydrogels. Gels 2021; 7:gels7040199. [PMID: 34842679 PMCID: PMC8628702 DOI: 10.3390/gels7040199] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
Accumulating evidence has suggested the significant potential of chemically modified hydrogels in bone regeneration. Despite the progress of bioactive hydrogels with different materials, structures and loading cargoes, the desires from clinical applications have not been fully validated. Multiple biological behaviors are orchestrated precisely during the bone regeneration process, including bone marrow mesenchymal stem cells (BMSCs) recruitment, osteogenic differentiation, matrix calcification and well-organized remodeling. Since matrix metalloproteinases play critical roles in such bone metabolism processes as BMSC commitment, osteoblast survival, osteoclast activation matrix calcification and microstructure remodeling, matrix metalloproteinase (MMP) cleavable peptides-based hydrogels could respond to various MMP levels and, thus, accelerate bone regeneration. In this review, we focused on the MMP-cleavable peptides, polymers, functional modification and crosslinked reactions. Applications, perspectives and limitations of MMP-cleavable peptides-based hydrogels for bone regeneration were then discussed.
Collapse
|
30
|
Jiao C, Zhao C, Ma Y, Yang W. A Versatile Strategy to Coat Individual Cell with Fully/Partially Covered Shell for Preparation of Self-Propelling Living Cells. ACS NANO 2021; 15:15920-15929. [PMID: 34591443 DOI: 10.1021/acsnano.1c03896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Coating living cells with a functional shell has been regarded as an effective way to protect them against environmental stress, regulate their biological behaviors, or extend their functionalities. Here, we reported a facile method to prepare fully or partially coated shells on an individual yeast cell surface by visible light-induced graft polymerization. In this strategy, yeast cells that were surface-absorbed with polyethylenimine (PEI) were deposited on the negatively charged glass slide to form a single layer by electrostatic interaction. Then, surface-initiated graft polymerization of poly(ethylene glycol) diacrylate (PEGDA) on yeast cells under visible light irradiation was carried out to generate cross-linked shells on the cells. The process of surface modification had negligible influence on the viability of yeast cells due to the mild reaction condition. Additionally, compared to the native yeast cells, a 17.5 h of delay in division was observed when the graft polymerization was performed under 15 mW/cm2 irradiation for 30 min. Introducing artificial shell endowed yeast cells with significant resistance against lyticase, and the protection can be enhanced by increasing the thickness of shell. Moreover, the partially coated yeast cells would be prepared by simply adjusting the reaction condition such as irradiation density and time. By immobilizing urease on the functional patch, the asymmetrically modified yeast cells exhibited self-propelling capability, and the speed of directional movement reached 4 μm/s in the presence of 200 mM urea. This tunable coating individual cell strategy with varying functionality has great potential applications in fields of cell-based drug delivery, cell therapy, biocatalysis, and tissue engineering.
Collapse
Affiliation(s)
- Chong Jiao
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Changwen Zhao
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
- Key Laboratory of Biomedical Materials of Natural Macromolecules, Ministry of Education Beijing, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yuhong Ma
- Key Laboratory of Carbon Fiber and Functional Polymers Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Wantai Yang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
- Key Laboratory of Biomedical Materials of Natural Macromolecules, Ministry of Education Beijing, Beijing University of Chemical Technology, Beijing, 100029, China
- Key Laboratory of Carbon Fiber and Functional Polymers Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
31
|
Evaluation of the effect of alginate matrices combination on insulin-secreting MIN-6 cell viability. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
32
|
Hu X, Li F, Xia F, Wang Q, Lin P, Wei M, Gong L, Low LE, Lee JY, Ling D. Dynamic nanoassembly-based drug delivery system (DNDDS): Learning from nature. Adv Drug Deliv Rev 2021; 175:113830. [PMID: 34139254 DOI: 10.1016/j.addr.2021.113830] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/19/2021] [Accepted: 06/10/2021] [Indexed: 12/18/2022]
Abstract
Dynamic nanoassembly-based drug delivery system (DNDDS) has evolved from being a mere curiosity to emerging as a promising strategy for high-performance diagnosis and/or therapy of various diseases. However, dynamic nano-bio interaction between DNDDS and biological systems remains poorly understood, which can be critical for precise spatiotemporal and functional control of DNDDS in vivo. To deepen the understanding for fine control over DNDDS, we aim to explore natural systems as the root of inspiration for researchers from various fields. This review highlights ingenious designs, nano-bio interactions, and controllable functionalities of state-of-the-art DNDDS under endogenous or exogenous stimuli, by learning from nature at the molecular, subcellular, and cellular levels. Furthermore, the assembly strategies and response mechanisms of tailor-made DNDDS based on the characteristics of various diseased microenvironments are intensively discussed. Finally, the current challenges and future perspectives of DNDDS are briefly commented.
Collapse
|
33
|
Biomaterials for Cell-Surface Engineering and Their Efficacy. J Funct Biomater 2021; 12:jfb12030041. [PMID: 34287337 PMCID: PMC8293134 DOI: 10.3390/jfb12030041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/30/2022] Open
Abstract
Literature in the field of stem cell therapy indicates that, when stem cells in a state of single-cell suspension are injected systemically, they show poor in vivo survival, while such cells show robust cell survival and regeneration activity when transplanted in the state of being attached on a biomaterial surface. Although an attachment-deprived state induces anoikis, when cell-surface engineering technology was adopted for stem cells in a single-cell suspension state, cell survival and regenerative activity dramatically improved. The biochemical signal coming from ECM (extracellular matrix) molecules activates the cell survival signal transduction pathway and prevents anoikis. According to the target disease, various therapeutic cells can be engineered to improve their survival and regenerative activity, and there are several types of biomaterials available for cell-surface engineering. In this review, biomaterial types and application strategies for cell-surface engineering are presented along with their expected efficacy.
Collapse
|
34
|
Miclotte MJ, Lawrenson SB, Varlas S, Rashid B, Chapman E, O’Reilly RK. Tuning the Cloud-Point and Flocculation Temperature of Poly(2-(diethylamino)ethyl methacrylate)-Based Nanoparticles via a Postpolymerization Betainization Approach. ACS POLYMERS AU 2021; 1:47-58. [PMID: 34476421 PMCID: PMC8389998 DOI: 10.1021/acspolymersau.1c00010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Indexed: 11/28/2022]
Abstract
The ability to tune the behavior of temperature-responsive polymers and self-assembled nanostructures has attracted significant interest in recent years, particularly in regard to their use in biotechnological applications. Herein, well-defined poly(2-(diethylamino)ethyl methacrylate) (PDEAEMA)-based core-shell particles were prepared by RAFT-mediated emulsion polymerization, which displayed a lower-critical solution temperature (LCST) phase transition in aqueous media. The tertiary amine groups of PDEAEMA units were then utilized as functional handles to modify the core-forming block chemistry via a postpolymerization betainization approach for tuning both the cloud-point temperature (T CP) and flocculation temperature (T CFT) of these particles. In particular, four different sulfonate salts were explored aiming to investigate the effect of the carbon chain length and the presence of hydroxyl functionalities alongside the carbon spacer on the particle's thermoresponsiveness. In all cases, it was possible to regulate both T CP and T CFT of these nanoparticles upon varying the degree of betainization. Although T CP was found to be dependent on the type of betainization reagent utilized, it only significantly increased for particles betainized using sodium 3-chloro-2-hydroxy-1-propanesulfonate, while varying the aliphatic chain length of the sulfobetaine only provided limited temperature variation. In comparison, the onset of flocculation for betainized particles varied over a much broader temperature range when varying the degree of betainization with no real correlation identified between T CFT and the sulfobetaine structure. Moreover, experimental results were shown to partially correlate to computational oligomer hydrophobicity calculations. Overall, the innovative postpolymerization betainization approach utilizing various sulfonate salts reported herein provides a straightforward methodology for modifying the thermoresponsive behavior of soft polymeric particles with potential applications in drug delivery, sensing, and oil/lubricant viscosity modification.
Collapse
Affiliation(s)
- Matthieu
P. J. Miclotte
- School
of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Stefan B. Lawrenson
- School
of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Spyridon Varlas
- School
of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Bilal Rashid
- BP
Exploration Operating Company Ltd., Chertsey Road, Sunbury-on-Thames,
Middlesex TW16 7LN, United
Kingdom
| | - Emma Chapman
- BP
Exploration Operating Company Ltd., Chertsey Road, Sunbury-on-Thames,
Middlesex TW16 7LN, United
Kingdom
| | - Rachel K. O’Reilly
- School
of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom,
| |
Collapse
|
35
|
Lee H, Kim N, Rheem HB, Kim BJ, Park JH, Choi IS. A Decade of Advances in Single-Cell Nanocoating for Mammalian Cells. Adv Healthc Mater 2021; 10:e2100347. [PMID: 33890422 DOI: 10.1002/adhm.202100347] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/06/2021] [Indexed: 12/14/2022]
Abstract
Strategic advances in the single-cell nanocoating of mammalian cells have noticeably been made during the last decade, and many potential applications have been demonstrated. Various cell-coating strategies have been proposed via adaptation of reported methods in the surface sciences and/or materials identification that ensure the sustainability of labile mammalian cells during chemical manipulation. Here an overview of the methodological development and potential applications to the healthcare sector in the nanocoating of mammalian cells made during the last decade is provided. The materials used for the nanocoating are categorized into polymers, hydrogels, polyphenolic compounds, nanoparticles, and minerals, and the corresponding strategies are described under the given set of materials. It also suggests, as a future direction, the creation of the cytospace system that is hierarchically composed of the physically separated but mutually interacting cellular hybrids.
Collapse
Affiliation(s)
- Hojae Lee
- Center for Cell‐Encapsulation Research Department of Chemistry KAIST Daejeon 34141 Korea
| | - Nayoung Kim
- Center for Cell‐Encapsulation Research Department of Chemistry KAIST Daejeon 34141 Korea
| | - Hyeong Bin Rheem
- Center for Cell‐Encapsulation Research Department of Chemistry KAIST Daejeon 34141 Korea
| | - Beom Jin Kim
- Department of Chemistry University of Ulsan Ulsan 44610 Korea
| | - Ji Hun Park
- Department of Science Education Ewha Womans University Seoul 03760 Korea
| | - Insung S. Choi
- Center for Cell‐Encapsulation Research Department of Chemistry KAIST Daejeon 34141 Korea
| |
Collapse
|
36
|
Zhang Z, Liu Q, Tan J, Zhan X, Liu T, Wang Y, Lu G, Wu M, Zhang Y. Coating with flexible DNA network enhanced T-cell activation and tumor killing for adoptive cell therapy. Acta Pharm Sin B 2021; 11:1965-1977. [PMID: 34386331 PMCID: PMC8343197 DOI: 10.1016/j.apsb.2021.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/28/2021] [Indexed: 12/12/2022] Open
Abstract
Adoptive cell therapy (ACT) is an emerging powerful cancer immunotherapy, which includes a complex process of genetic modification, stimulation and expansion. During these in vitro or ex vivo manipulation, sensitive cells are inescapability subjected to harmful external stimuli. Although a variety of cytoprotection strategies have been developed, their application on ACT remains challenging. Herein, a DNA network is constructed on cell surface by rolling circle amplification (RCA), and T cell-targeted trivalent tetrahedral DNA nanostructure is used as a rigid scaffold to achieve high-efficient and selective coating for T cells. The cytoprotective DNA network on T-cell surface makes them aggregate over time to form cell clusters, which exhibit more resistance to external stimuli and enhanced activities in human peripheral blood mononuclear cells and liver cancer organoid killing model. Overall, this work provides a novel strategy for in vitro T cell-selective protection, which has a great potential for application in ACT.
Collapse
Affiliation(s)
- Ziyan Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Qiaojuan Liu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jizhou Tan
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoxia Zhan
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting Liu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuting Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Gen Lu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou 510080, China
| | - Minhao Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
37
|
Kim M, Kim H, Lee YS, Lee S, Kim SE, Lee UJ, Jung S, Park CG, Hong J, Doh J, Lee DY, Kim BG, Hwang NS. Novel enzymatic cross-linking-based hydrogel nanofilm caging system on pancreatic β cell spheroid for long-term blood glucose regulation. SCIENCE ADVANCES 2021; 7:eabf7832. [PMID: 34162541 PMCID: PMC8221614 DOI: 10.1126/sciadv.abf7832] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/10/2021] [Indexed: 05/17/2023]
Abstract
Pancreatic β cell therapy for type 1 diabetes is limited by low cell survival rate owing to physical stress and aggressive host immune response. In this study, we demonstrate a multilayer hydrogel nanofilm caging strategy capable of protecting cells from high shear stress and reducing immune response by interfering cell-cell interaction. Hydrogel nanofilm is fabricated by monophenol-modified glycol chitosan and hyaluronic acid that cross-link each other to form a nanothin hydrogel film on the cell surface via tyrosinase-mediated reactions. Furthermore, hydrogel nanofilm formation was conducted on mouse β cell spheroids for the islet transplantation application. The cytoprotective effect against physical stress and the immune protective effect were evaluated. Last, caged mouse β cell spheroids were transplanted into the type 1 diabetes mouse model and successfully regulated its blood glucose level. Overall, our enzymatic cross-linking-based hydrogel nanofilm caging method will provide a new platform for clinical applications of cell-based therapies.
Collapse
Affiliation(s)
- Minji Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyunbum Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Sun Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sangjun Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Seong-Eun Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Uk-Jae Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungwon Jung
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Chung-Gyu Park
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Junsang Doh
- Department of Materials Science and Engineering, Research of Advanced Materials (RIAM), Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea
- BioMAX/N-Bio Institute, Institute of Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea.
| | - Byung-Gee Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
- BioMAX/N-Bio Institute, Institute of Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
- BioMAX/N-Bio Institute, Institute of Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
38
|
|
39
|
Montoya C, Du Y, Gianforcaro AL, Orrego S, Yang M, Lelkes PI. On the road to smart biomaterials for bone research: definitions, concepts, advances, and outlook. Bone Res 2021; 9:12. [PMID: 33574225 PMCID: PMC7878740 DOI: 10.1038/s41413-020-00131-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 01/31/2023] Open
Abstract
The demand for biomaterials that promote the repair, replacement, or restoration of hard and soft tissues continues to grow as the population ages. Traditionally, smart biomaterials have been thought as those that respond to stimuli. However, the continuous evolution of the field warrants a fresh look at the concept of smartness of biomaterials. This review presents a redefinition of the term "Smart Biomaterial" and discusses recent advances in and applications of smart biomaterials for hard tissue restoration and regeneration. To clarify the use of the term "smart biomaterials", we propose four degrees of smartness according to the level of interaction of the biomaterials with the bio-environment and the biological/cellular responses they elicit, defining these materials as inert, active, responsive, and autonomous. Then, we present an up-to-date survey of applications of smart biomaterials for hard tissues, based on the materials' responses (external and internal stimuli) and their use as immune-modulatory biomaterials. Finally, we discuss the limitations and obstacles to the translation from basic research (bench) to clinical utilization that is required for the development of clinically relevant applications of these technologies.
Collapse
Affiliation(s)
- Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
| | - Yu Du
- Department of Endodontology, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
- Guangdong Provincial Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Anthony L Gianforcaro
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Maobin Yang
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
- Department of Endodontology, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Peter I Lelkes
- Department of Endodontology, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA.
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, 19122, USA.
| |
Collapse
|
40
|
Yang Y, Zeng W, Huang P, Zeng X, Mei L. Smart materials for drug delivery and cancer therapy. VIEW 2020. [DOI: 10.1002/viw.20200042] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yao Yang
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Weiwei Zeng
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Ping Huang
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Xiaowei Zeng
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Lin Mei
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
- Tianjin Key Laboratory of Biomedical Materials Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy Institute of Biomedical Engineering Chinese Academy of Medical Sciences & Peking Union Medical College Tianjin China
| |
Collapse
|
41
|
Wieszczycka K, Filipowiak K, Buchwald T, Nowicki M. Microcapsules containing task-specific ionic liquids for Zn(II) and Cu(II) recovery from dilute aqueous solutions. Sep Purif Technol 2020. [DOI: 10.1016/j.seppur.2020.117155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
42
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
43
|
Wang W, Azizyan RA, Garro A, Kajava AV, Ventura S. Multifunctional Amyloid Oligomeric Nanoparticles for Specific Cell Targeting and Drug Delivery. Biomacromolecules 2020; 21:4302-4312. [PMID: 32885960 DOI: 10.1021/acs.biomac.0c01103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Natural selection has endorsed proteins with amazing structures and functionalities that cannot be matched by synthetic means, explaining the exponential interest in developing protein-based materials. Protein self-assembly allows fabricating complex supramolecular structures from relatively simple building blocks, a bottom-up strategy naturally employed by amyloid fibrils. However, the design of amyloid-inspired materials with biological activity is inherently difficult. Here, we exploit a modular procedure to generate functional amyloid nanostructures with tight control of their mesoscopic properties. The soft amyloid core of a yeast prion was fused to dihydrofolate reductase through flexible linkers of different sizes. This enabled us to produce, for the first time, biocompatible protein-only amyloid-like oligomeric nanoparticles with defined dimensions in which the embedded enzyme remained highly active, as assessed by biophysical and enzymatic assays. The modular design allowed one to obtain multifunctional nanoparticles by incorporating the antibody-binding Z-domain to the protein fusion. We show how these assemblies can be exploited for antibody-directed targeting of specific cell types and the localized delivery of methotrexate, resulting in the intracellular uptake of the drug by cancer cells and their death. Overall, the novel protein particles we describe in this work might find applications in areas as diverse as biocatalysis, bioimaging, or targeted therapies.
Collapse
Affiliation(s)
- Weiqiang Wang
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Rafayel A Azizyan
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237 CNRS, Université Montpellier, Montpellier 34090, France.,Institut de Biologie Computationnelle, Université Montpellier, Montpellier 34090, France
| | - Adriana Garro
- Universidad Nacional de San Luis IMASL-CONICET, San Luis D5702, Argentina
| | - Andrey V Kajava
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237 CNRS, Université Montpellier, Montpellier 34090, France.,Institut de Biologie Computationnelle, Université Montpellier, Montpellier 34090, France
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
44
|
Youn W, Kim JY, Park J, Kim N, Choi H, Cho H, Choi IS. Single-Cell Nanoencapsulation: From Passive to Active Shells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1907001. [PMID: 32255241 DOI: 10.1002/adma.201907001] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/09/2019] [Accepted: 12/23/2019] [Indexed: 06/11/2023]
Abstract
Single-cell nanoencapsulation is an emerging field in cell-surface engineering, emphasizing the protection of living cells against external harmful stresses in vitro and in vivo. Inspired by the cryptobiotic state found in nature, cell-in-shell structures are formed, which are called artificial spores and which show suppression or retardation in cell growth and division and enhanced cell survival under harsh conditions. The property requirements of the shells suggested for realization of artificial spores, such as durability, permselectivity, degradability, and functionalizability, are demonstrated with various cytocompatible materials and processes. The first-generation shells in single-cell nanoencapsulation are passive in the operation mode, and do not biochemically regulate the cellular metabolism or activities. Recent advances indicate that the field has shifted further toward the formation of active shells. Such shells are intimately involved in the regulation and manipulation of biological processes. Not only endowing the cells with new properties that they do not possess in their native forms, active shells also regulate cellular metabolism and/or rewire biological pathways. Recent developments in shell formation for microbial and mammalian cells are discussed and an outlook on the field is given.
Collapse
Affiliation(s)
- Wongu Youn
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Ji Yup Kim
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Joohyouck Park
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Nayoung Kim
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Hyunwoo Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Hyeoncheol Cho
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Insung S Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| |
Collapse
|
45
|
Sun J, Ren Y, Wang W, Hao H, Tang M, Zhang Z, Yang J, Zheng Y, Shi X. Transglutaminase-Catalyzed Encapsulation of Individual Mammalian Cells with Biocompatible and Cytoprotective Gelatin Nanoshells. ACS Biomater Sci Eng 2020; 6:2336-2345. [DOI: 10.1021/acsbiomaterials.0c00044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jimin Sun
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yafeng Ren
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Weibin Wang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Huili Hao
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Mingyu Tang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Zibo Zhang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Lab of Medical Instrument and Biopharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yunquan Zheng
- Fujian Key Lab of Medical Instrument and Biopharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - XianAi Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
- Fujian Key Lab of Medical Instrument and Biopharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| |
Collapse
|
46
|
Qin X, Li Y. Strategies To Design and Synthesize Polymer‐Based Stimuli‐Responsive Drug‐Delivery Nanosystems. Chembiochem 2020; 21:1236-1253. [DOI: 10.1002/cbic.201900550] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/23/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Xing Qin
- Laboratory of Low-Dimensional Materials ChemistryKey Laboratory for Ultrafine Materials of the Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and Technology Shanghai 200237 P.R.China
| | - Yongsheng Li
- Laboratory of Low-Dimensional Materials ChemistryKey Laboratory for Ultrafine Materials of the Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and Technology Shanghai 200237 P.R.China
| |
Collapse
|
47
|
Gaspar VM, Lavrador P, Borges J, Oliveira MB, Mano JF. Advanced Bottom-Up Engineering of Living Architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903975. [PMID: 31823448 DOI: 10.1002/adma.201903975] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/30/2019] [Indexed: 05/08/2023]
Abstract
Bottom-up tissue engineering is a promising approach for designing modular biomimetic structures that aim to recapitulate the intricate hierarchy and biofunctionality of native human tissues. In recent years, this field has seen exciting progress driven by an increasing knowledge of biological systems and their rational deconstruction into key core components. Relevant advances in the bottom-up assembly of unitary living blocks toward the creation of higher order bioarchitectures based on multicellular-rich structures or multicomponent cell-biomaterial synergies are described. An up-to-date critical overview of long-term existing and rapidly emerging technologies for integrative bottom-up tissue engineering is provided, including discussion of their practical challenges and required advances. It is envisioned that a combination of cell-biomaterial constructs with bioadaptable features and biospecific 3D designs will contribute to the development of more robust and functional humanized tissues for therapies and disease models, as well as tools for fundamental biological studies.
Collapse
Affiliation(s)
- Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Pedro Lavrador
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João Borges
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
48
|
Photo-triggered capsules based on lanthanide-doped upconverting nanoparticles for medical applications. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.213013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
49
|
Yang Y, Wang X, Wang Y, Hu X, Kawazoe N, Yang Y, Chen G. Influence of Cell Spreading Area on the Osteogenic Commitment and Phenotype Maintenance of Mesenchymal Stem Cells. Sci Rep 2019; 9:6891. [PMID: 31053728 PMCID: PMC6499796 DOI: 10.1038/s41598-019-43362-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/23/2019] [Indexed: 12/17/2022] Open
Abstract
Osteogenic differentiation and commitment of mesenchymal stem cells (MSCs) is a complex process that is induced and regulated by various biological factors and biophysical cues. Although cell spreading area, as a biophysical cue, has been demonstrated to play a critical role in the regulation of osteogenic differentiation of MSCs, it is unclear how it affects the maintenance of the committed phenotype after osteogenic differentiation of MSCs. In this study, poly (vinyl alcohol) was micropatterned on a tissue culture polystyrene surface, and the micropatterns were used to culture MSCs to control their cell spreading area. The influence of cell spreading area on osteogenic differentiation and maintenance of the differentiated phenotype of MSCs was investigated. MSCs with a larger spreading area showed a higher degree of osteogenic differentiation, slower loss of differentiated phenotype and slower re-expression of stem cell markers compared with MSCs with a smaller spreading area. A large cell spreading area was beneficial for osteogenic differentiation of MSCs and maintenance of their differentiated phenotype.
Collapse
Affiliation(s)
- Yingjun Yang
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Xinlong Wang
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Yongtao Wang
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Xiaohong Hu
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8571, Japan
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8571, Japan
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|