1
|
Luo J, Cui Y, Xu L, Zhang J, Chen J, Li X, Zeng B, Deng Z, Shao L. Layered double hydroxides for regenerative nanomedicine and tissue engineering: recent advances and future perspectives. J Nanobiotechnology 2025; 23:370. [PMID: 40405242 PMCID: PMC12096525 DOI: 10.1186/s12951-025-03448-1] [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: 03/04/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
With the rapid development of nanotechnology, layered double hydroxides (LDHs) have attracted considerable attention in the biomedical field due to their highly tunable composition and structure, superior biocompatibility, multifunctional bioactivity, and exceptional drug delivery performance. However, a focused and comprehensive review addressing the role of LDHs specifically in tissue regeneration has been lacking. This review aims to fill that gap by providing a systematic and in-depth overview of recent advances in the application of LDHs across various regenerative domains, including bone repair, cartilage reconstruction, angiogenesis, wound healing, and nerve regeneration. Beyond presenting emerging applications, the review places particular emphasis on elucidating the underlying mechanisms through which LDHs exert their therapeutic effects. Although LDHs demonstrate considerable promise in regenerative medicine, their clinical translation remains in its infancy. To address this, we not only provided our insights into the personalized problems that arise in the application of various tissues, but also focused on discussing and prospecting the common challenges in the clinical translation of LDHs. These challenges include optimizing synthesis techniques, enhancing biosafety and stability, improving drug-loading efficiency, designing multifunctional composite materials, and establishing pathways that facilitate the transition from laboratory research to clinical practice.
Collapse
Affiliation(s)
- Junsi Luo
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Yiteng Cui
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Laijun Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- School of Stomatology, Changsha Medical University, Changsha, 410219, China
| | - Junyi Zhang
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Jinhong Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Xumin Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Bin Zeng
- School of Stomatology, Changsha Medical University, Changsha, 410219, China
| | - Zhiyuan Deng
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- School of Stomatology, Changsha Medical University, Changsha, 410219, China.
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Huang C, Zhong G, Xiao J, Wang X, Huang W, Chen L, Zhang Y, Cheng S. An Injectable Kartogenin-Incorporated Hydrogel Supports Mesenchymal Stem Cells for Cartilage Tissue Engineering. Bioengineering (Basel) 2025; 12:434. [PMID: 40428053 DOI: 10.3390/bioengineering12050434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Cartilage defects and injuries often lead to osteoarthritis, posing significant challenges for cartilage repair. Traditional treatments have limited efficacy, necessitating innovative therapeutic strategies. This study aimed to develop an injectable hydrogel-based tissue engineering construct to enhance cartilage regeneration by combining mesenchymal stem cells (MSCs) and the small molecule drug kartogenin (KGN). METHODS An injectable hydrogel was synthesized by crosslinking carboxymethyl chitosan (CMC) with aldehyde-modified cellulose nanocrystals (DACNCs). KGN was incorporated into the hydrogel during crosslinking to achieve sustained drug release. Three hydrogels with varying CMC/DACNC molar ratios (MR = 0.11, 0.22, and 0.33) were developed and characterized for their structural, mechanical, and biocompatible properties. The hydrogel with the optimal ratio (MR = 0.33) was further evaluated for its ability to support MSC viability and differentiation in vitro. Additionally, signaling pathways (TGF-β, FOXO, and PI3K-AKT) were investigated to elucidate the underlying mechanisms. In vivo efficacy was assessed using a rabbit femoral trochlear cartilage defect model. RESULTS The hydrogel with a higher CMC/DACNC molar ratio (MR = 0.33) exhibited increased compressive modulus, a reduced swelling rate, and superior biocompatibility, effectively promoting MSC differentiation in vitro. Signaling pathway analysis revealed activation of the TGF-β, FOXO, and PI3K-AKT pathways, suggesting enhanced chondrogenic potential. In vivo experiments demonstrated that the KGN-MSC-encapsulated hydrogel significantly improved cartilage repair. CONCLUSIONS The injectable CMC/DACNC hydrogel, combined with KGN and MSCs, synergistically enhanced cartilage regeneration both in vitro and in vivo. This study highlights the potential of this hydrogel as a promising scaffold for cartilage tissue engineering, offering a novel therapeutic approach for cartilage defects and injuries.
Collapse
Affiliation(s)
- Chongquan Huang
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Guoqing Zhong
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangzhou 510080, China
| | - Jin Xiao
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiaolan Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangzhou 510080, China
| | - Weijuan Huang
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Lingyun Chen
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangzhou 510080, China
| | - Shi Cheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
3
|
Frączek W, Kotela A, Kotela I, Grodzik M. Nanostructures in Orthopedics: Advancing Diagnostics, Targeted Therapies, and Tissue Regeneration. MATERIALS (BASEL, SWITZERLAND) 2024; 17:6162. [PMID: 39769763 PMCID: PMC11677186 DOI: 10.3390/ma17246162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Nanotechnology, delving into the realm of nanometric structures, stands as a transformative force in orthopedics, reshaping diagnostics, and numerous regenerative interventions. Commencing with diagnostics, this scientific discipline empowers accurate analyses of various diseases and implant stability, heralding an era of unparalleled precision. Acting as carriers for medications, nanomaterials introduce novel therapeutic possibilities, propelling the field towards more targeted and effective treatments. In arthroplasty, nanostructural modifications to implant surfaces not only enhance mechanical properties but also promote superior osteointegration and durability. Simultaneously, nanotechnology propels tissue regeneration, with nanostructured dressings emerging as pivotal elements in accelerating wound healing. As we navigate the frontiers of nanotechnology, ongoing research illuminates promising avenues for further advancements, assuring a future where orthopedic practices are not only personalized but also highly efficient, promising a captivating journey through groundbreaking innovations and tailored patient care.
Collapse
Affiliation(s)
- Wiktoria Frączek
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland
| | - Andrzej Kotela
- Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszyński University, 01-938 Warsaw, Poland
| | - Ireneusz Kotela
- National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-369 Kielce, Poland
| | - Marta Grodzik
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland
| |
Collapse
|
4
|
Liu X, Liu P, Li H, Cen Y, Jiang G, Zhang W, Tian K, Wang X. Application of kartogenin for the treatment of cartilage defects: current practice and future directions. RSC Adv 2024; 14:33206-33222. [PMID: 39434994 PMCID: PMC11492430 DOI: 10.1039/d4ra06558a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
Osteoarthritis and sports injuries often lead to cartilage defects. How to promote its repair and rebuild the smooth cartilage surface has been a hot spot of research in recent years. Kartogenin (KGN), a small molecule discovered in recent years, has been shown to promote the proliferation and chondrogenic differentiation of mesenchymal stem cells (MSCs). As more and more studies have been conducted on KGN, its mechanism of action has been gradually revealed. However, KGN is insoluble in water and therefore easily removed by body fluids. In order to address such issues, a number of systems for efficient intra-articular delivery of KGN have been developed. In addition, due to the complex pathology of cartilage repair, KGN is often used in combination with other drugs to target different stages. In addition, with the rapid development of tissue engineering, scholars have combined KGN with various scaffolds by physical or chemical methods. In this paper, we firstly introduce the general properties of KGN followed by a review of the latest advances in the intra-articular delivery modes of KGN. Finally, we discuss the prospects for the application of KGN in cartilage regeneration, which is aimed at providing a new idea and target for the treatment of cartilage defects.
Collapse
Affiliation(s)
- Xuemiao Liu
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Polymer Physics and Chemistry Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Pengfei Liu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University Beijing 100191 China
| | - Han Li
- Xiongan Xuanwu Hospital Hebei 071700 China
| | - Ying Cen
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Guichun Jiang
- Liaoning Cancer Hospital & Institute, Clinical Skills Training Center Shenyang 110042 China
| | - Weiguo Zhang
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Kang Tian
- First Affiliated Hospital of Dalian Medical University Dalian 116001 China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Polymer Physics and Chemistry Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| |
Collapse
|
5
|
Cometa S, Busto F, Scalia AC, Castellaneta A, Gentile P, Cochis A, Manfredi M, Borrini V, Rimondini L, De Giglio E. Effectiveness of gellan gum scaffolds loaded with Boswellia serrata extract for in-situ modulation of pro-inflammatory pathways affecting cartilage healing. Int J Biol Macromol 2024; 277:134079. [PMID: 39038574 DOI: 10.1016/j.ijbiomac.2024.134079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/09/2024] [Accepted: 07/19/2024] [Indexed: 07/24/2024]
Abstract
In this study, we developed a composite hydrogel based on Gellan gum containing Boswellia serrata extract (BSE). BSE was either incorporated directly or loaded into an MgAl-layered double hydroxide (LDH) clay to create a multifunctional cartilage substitute. This composite was designed to provide anti-inflammatory properties while enhancing chondrogenesis. Additionally, LDH was exploited to facilitate the loading of hydrophobic BSE components and to improve the hydrogel's mechanical properties. A calcination process was also adopted on LDH to increase BSE loading. Physicochemical and mechanical characterizations were performed by spectroscopic (XPS and FTIR), thermogravimetric, rheological, compression test, weight loss and morphological (SEM) investigations. RPLC-ESI-FTMS was employed to investigate the boswellic acids release in simulated synovial fluid. The composites were cytocompatible and capable of supporting the mesenchymal stem cells (hMSC) growth in a 3D-conformation. Loading BSE resulted in the modulation of the pro-inflammatory cascade by down-regulating COX2, PGE2 and IL1β. Chondrogenesis studies demonstrated an enhanced differentiation, leading to the up-regulation of COL 2 and ACAN. This effect was attributed to the efficacy of BSE in reducing the inflammation through PGE2 down-regulation and IL10 up-regulation. Proteomics studies confirmed gene expression findings by revealing an anti-inflammatory protein signature during chondrogenesis of the cells cultivated onto loaded specimens. Concluding, BSE-loaded composites hold promise as a tool for the in-situ modulation of the inflammatory cascade while preserving cartilage healing.
Collapse
Affiliation(s)
| | - Francesco Busto
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy; INSTM, National Consortium of Materials Science and Technology, Via G. Giusti 9, 50121 Florence, Italy.
| | - Alessandro C Scalia
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Andrea Castellaneta
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy.
| | - Piergiorgio Gentile
- Newcastle University, School of Engineering, Claremont Road, NE1 7RU Newcastle upon Tyne, United Kingdom.
| | - Andrea Cochis
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Marcello Manfredi
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Vittoria Borrini
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Lia Rimondini
- Center for Translational Research on Autoimmune and Allergic Disease, CAAD, Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy.
| | - Elvira De Giglio
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy; INSTM, National Consortium of Materials Science and Technology, Via G. Giusti 9, 50121 Florence, Italy.
| |
Collapse
|
6
|
Son B, Park S, Cho S, Kim JA, Baek SH, Yoo KH, Han D, Joo J, Park HH, Park TH. Improved Neural Inductivity of Size-Controlled 3D Human Embryonic Stem Cells Using Magnetic Nanoparticles. Biomater Res 2024; 28:0011. [PMID: 38500782 PMCID: PMC10944702 DOI: 10.34133/bmr.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/12/2024] [Indexed: 03/20/2024] Open
Abstract
Background: To improve the efficiency of neural development from human embryonic stem cells, human embryoid body (hEB) generation is vital through 3-dimensional formation. However, conventional approaches still have limitations: long-term cultivation and laborious steps for lineage determination. Methods: In this study, we controlled the size of hEBs for ectodermal lineage specification using cell-penetrating magnetic nanoparticles (MNPs), which resulted in reduced time required for initial neural induction. The magnetized cells were applied to concentrated magnetic force for magnet-derived multicellular organization. The uniformly sized hEBs were differentiated in neural induction medium (NIM) and suspended condition. This neurally induced MNP-hEBs were compared with other groups. Results: As a result, the uniformly sized MNP-hEBs in NIM showed significantly improved neural inductivity through morphological analysis and expression of neural markers. Signaling pathways of the accelerated neural induction were detected via expression of representative proteins; Wnt signaling, dopaminergic neuronal pathway, intercellular communications, and mechanotransduction. Consequently, we could shorten the time necessary for early neurogenesis, thereby enhancing the neural induction efficiency. Conclusion: Overall, this study suggests not only the importance of size regulation of hEBs at initial differentiation stage but also the efficacy of MNP-based neural induction method and stimulations for enhanced neural tissue regeneration.
Collapse
Affiliation(s)
- Boram Son
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Sora Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Sungwoo Cho
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Cheongju, Chungbuk 28119, Republic of Korea
| | - Seung-Ho Baek
- Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), Ulsan 44429, Korea
| | - Ki Hyun Yoo
- SIMPLE Planet Inc., 48 Achasan-ro 17-gil, Seongdong-gu, Seoul 04799, Korea
| | - Dongoh Han
- SIMPLE Planet Inc., 48 Achasan-ro 17-gil, Seongdong-gu, Seoul 04799, Korea
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seodaemun-gu, Seoul 03760, Republic of Korea
| |
Collapse
|
7
|
Majood M, Agrawal O, Garg P, Selvam A, Yadav SK, Singh S, Kalyansundaram D, Verma YK, Nayak R, Mohanty S, Mukherjee M. Carbon quantum dot-nanocomposite hydrogel as Denovo Nexus in rapid chondrogenesis. BIOMATERIALS ADVANCES 2024; 157:213730. [PMID: 38101066 DOI: 10.1016/j.bioadv.2023.213730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/15/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
The incapability of cartilage to naturally regenerate and repair chronic muscular injuries urges the development of competent bionic rostrums. There is a need to explore faster strategies for chondrogenic engineering using mesenchymal stem cells (MSCs). Along these lines, rapid chondrocyte differentiation would benefit the transplantation demand affecting osteoarthritis (OA) and rheumatoid arthritis (RA) patients. In this report, a de novo nanocomposite was constructed by integrating biogenic carbon quantum dot (CQD) filler into synthetic hydrogel prepared from dimethylaminoethyl methacrylate (DMAEMA) and acrylic acid (AAc). The dominant structural integrity of synthetic hydrogel along with the chondrogenic differentiation potential of garlic peel derived CQDs led to faster chondrogenesis within 14 days. By means of extensive chemical and morphological characterization techniques, we illustrate that the hydrogel nanocomposite possesses lucrative features to influence rapid chondrogenesis. These results were further corroborated by bright field imaging, Alcian blue staining and Masson trichome staining. Thus, this stratagem of chondrogenic engineering conceptualizes to be a paragon in clinical wound care for the rapid manufacturing of chondrocytes.
Collapse
Affiliation(s)
- Misba Majood
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida 201313, India
| | - Omnarayan Agrawal
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida 201313, India
| | - Piyush Garg
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida 201313, India
| | - Abhyavartin Selvam
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida 201313, India; Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida 201313, India
| | - Sunil Kumar Yadav
- Center of Biomedical Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Sonu Singh
- Center of Biomedical Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Dinesh Kalyansundaram
- Center of Biomedical Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Yogesh Kumar Verma
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences, Delhi 110054, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida 201313, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT center of Excellence, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Monalisa Mukherjee
- Amity Institute of Click Chemistry Research and Studies, Amity University Uttar Pradesh, Noida 201313, India.
| |
Collapse
|
8
|
Kang J, Li Y, Qin Y, Huang Z, Wu Y, Sun L, Wang C, Wang W, Feng G, Qi Y. In Situ Deposition of Drug and Gene Nanoparticles on a Patterned Supramolecular Hydrogel to Construct a Directionally Osteochondral Plug. NANO-MICRO LETTERS 2023; 16:18. [PMID: 37975889 PMCID: PMC10656386 DOI: 10.1007/s40820-023-01228-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/20/2023] [Indexed: 11/19/2023]
Abstract
The integrated repair of bone and cartilage boasts advantages for osteochondral restoration such as a long-term repair effect and less deterioration compared to repairing cartilage alone. Constructing multifactorial, spatially oriented scaffolds to stimulate osteochondral regeneration, has immense significance. Herein, targeted drugs, namely kartogenin@polydopamine (KGN@PDA) nanoparticles for cartilage repair and miRNA@calcium phosphate (miRNA@CaP) NPs for bone regeneration, were in situ deposited on a patterned supramolecular-assembled 2-ureido-4 [lH]-pyrimidinone (UPy) modified gelation hydrogel film, facilitated by the dynamic and responsive coordination and complexation of metal ions and their ligands. This hydrogel film can be rolled into a cylindrical plug, mimicking the Haversian canal structure of natural bone. The resultant hydrogel demonstrates stable mechanical properties, a self-healing ability, a high capability for reactive oxygen species capture, and controlled release of KGN and miR-26a. In vitro, KGN@PDA and miRNA@CaP promote chondrogenic and osteogenic differentiation of mesenchymal stem cells via the JNK/RUNX1 and GSK-3β/β-catenin pathways, respectively. In vivo, the osteochondral plug exhibits optimal subchondral bone and cartilage regeneration, evidenced by a significant increase in glycosaminoglycan and collagen accumulation in specific zones, along with the successful integration of neocartilage with subchondral bone. This biomaterial delivery approach represents a significant toward improved osteochondral repair.
Collapse
Affiliation(s)
- Jiawei Kang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China
| | - Yaping Li
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, Zhejiang, People's Republic of China
| | - Yating Qin
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, Zhejiang, People's Republic of China
| | - Zhongming Huang
- The Affiliated Nanhua Hospital, Orthopedic Research Centre, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Yifan Wu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China
| | - Long Sun
- Department of Radiology, Jining No. 1 People's Hospital, Jining Medical University, Jining, 272000, Shandong, People's Republic of China
| | - Cong Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China
| | - Wei Wang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China.
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, Zhejiang, People's Republic of China.
| | - Gang Feng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China.
| | - Yiying Qi
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, 310009, Zhejiang Province, People's Republic of China.
| |
Collapse
|
9
|
Bian Y, Cai X, Lv Z, Xu Y, Wang H, Tan C, Liang R, Weng X. Layered Double Hydroxides: A Novel Promising 2D Nanomaterial for Bone Diseases Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301806. [PMID: 37329200 PMCID: PMC10460877 DOI: 10.1002/advs.202301806] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/07/2023] [Indexed: 06/18/2023]
Abstract
Bone diseases including bone defects, bone infections, osteoarthritis, and bone tumors seriously affect life quality of the patient and bring serious economic burdens to social health management, for which the current clinical treatments bear dissatisfactory therapeutic effects. Biomaterial-based strategies have been widely applied in the treatment of orthopedic diseases but are still plagued by deficient bioreactivity. With the development of nanotechnology, layered double hydroxides (LDHs) with adjustable metal ion composition and alterable interlayer structure possessing charming physicochemical characteristics, versatile bioactive properties, and excellent drug loading and delivery capabilities arise widespread attention and have achieved considerable achievements for bone disease treatment in the last decade. However, to the authors' best knowledge, no review has comprehensively summarized the advances of LDHs in treating bone disease so far. Herein, the advantages of LDHs for orthopedic disorders treatment are outlined and the corresponding state-of-the-art achievements are summarized for the first time. The potential of LDHs-based nanocomposites for extended therapeutics for bone diseases is highlighted and perspectives for LDHs-based scaffold design are proposed for facilitated clinical translation.
Collapse
Affiliation(s)
- Yixin Bian
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Xuejie Cai
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Zehui Lv
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Yiming Xu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Han Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Chaoliang Tan
- Department of Chemistry and Center of Super‐Diamond and Advanced Films (COSDAF)City University of Hong KongKowloonHong KongP. R. China
- Shenzhen Research InstituteCity University of Hong KongShenzhen518057P. R. China
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Xisheng Weng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| |
Collapse
|
10
|
Zhao D, Rong Y, Li D, He C, Chen X. Thermo-induced physically crosslinked polypeptide-based block copolymer hydrogels for biomedical applications. Regen Biomater 2023; 10:rbad039. [PMID: 37265604 PMCID: PMC10229375 DOI: 10.1093/rb/rbad039] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 06/03/2023] Open
Abstract
Stimuli-responsive synthetic polypeptide-containing block copolymers have received considerable attention in recent years. Especially, unique thermo-induced sol-gel phase transitions were observed for elaborately-designed amphiphilic diblock copolypeptides and a range of poly(ethylene glycol) (PEG)-polypeptide block copolymers. The thermo-induced gelation mechanisms involve the evolution of secondary conformation, enhanced intramolecular interactions, as well as reduced hydration and increased chain entanglement of PEG blocks. The physical parameters, including polymer concentrations, sol-gel transition temperatures and storage moduli, were investigated. The polypeptide hydrogels exhibited good biocompatibility in vitro and in vivo, and displayed biodegradation periods ranging from 1 to 5 weeks. The unique thermo-induced sol-gel phase transitions offer the feasibility of minimal-invasive injection of the precursor aqueous solutions into body, followed by in situ hydrogel formation driven by physiological temperature. These advantages make polypeptide hydrogels interesting candidates for diverse biomedical applications, especially as injectable scaffolds for 3D cell culture and tissue regeneration as well as depots for local drug delivery. This review focuses on recent advances in the design and preparation of injectable, thermo-induced physically crosslinked polypeptide hydrogels. The influence of composition, secondary structure and chirality of polypeptide segments on the physical properties and biodegradation of the hydrogels are emphasized. Moreover, the studies on biomedical applications of the hydrogels are intensively discussed. Finally, the major challenges in the further development of polypeptide hydrogels for practical applications are proposed.
Collapse
Affiliation(s)
- Dan Zhao
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- College of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yan Rong
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Dong Li
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- College of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | | | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- College of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
11
|
Guan P, Ji Y, Kang X, Liu W, Yang Q, Liu S, Lin Y, Zhang Z, Li J, Zhang Y, Liu C, Fan L, Sun Y. Biodegradable Dual-Cross-Linked Hydrogels with Stem Cell Differentiation Regulatory Properties Promote Growth Plate Injury Repair via Controllable Three-Dimensional Mechanics and a Cartilage-like Extracellular Matrix. ACS APPLIED MATERIALS & INTERFACES 2023; 15:8986-8998. [PMID: 36752284 DOI: 10.1021/acsami.2c20722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Recent breakthroughs in cell transplantation therapy have revealed the promising potential of bone marrow mesenchymal stem cells (BMSCs) for promoting the regeneration of growth plate cartilage injury. However, the high apoptosis rate and the uncertainty of the differentiation direction of cells often lead to poor therapeutic effects. Cells are often grown under three-dimensional (3D) conditions in vivo, and the stiffness and components of the extracellular matrix (ECM) are important regulators of stem cell differentiation. To this end, a 3D cartilage-like ECM hydrogel with tunable mechanical properties was designed and synthesized mainly from gelatin methacrylate (GM) and oxidized chondroitin sulfate (OCS) via dynamic Schiff base bonding under UV. The effects of scaffold stiffness and composition on the survival and differentiation of BMSCs in vitro were investigated. A rat model of growth plate injury was developed to validate the effect of the GMOCS hydrogels encapsulated with BMSCs on the repair of growth plate injury. The results showed that 3D GMOCS hydrogels with an appropriate modulus significantly promoted chondrogenic differentiation of BMSCs, and GMOCS/BMSC transplantation could effectively inhibit bone bridge formation and promote the repair of damaged growth plates. Accordingly, GMOCS/BMSC therapy can be engineered as a promising therapeutic candidate for growth plate injury.
Collapse
Affiliation(s)
- Pengfei Guan
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| | - Yuelun Ji
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| | - Xinchang Kang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Weilu Liu
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qinfeng Yang
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shencai Liu
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yeying Lin
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Zuyu Zhang
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| | - Junji Li
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| | - Yue Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Can Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lei Fan
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yongjian Sun
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
12
|
Chen C, Huang S, Chen Z, Liu Q, Cai Y, Mei Y, Xu Y, Guo R, Yan C. Kartogenin (KGN)/synthetic melanin nanoparticles (SMNP) loaded theranostic hydrogel scaffold system for multiparametric magnetic resonance imaging guided cartilage regeneration. Bioeng Transl Med 2023; 8:e10364. [PMID: 36684070 PMCID: PMC9842022 DOI: 10.1002/btm2.10364] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/06/2022] [Accepted: 06/12/2022] [Indexed: 01/25/2023] Open
Abstract
Cartilage regeneration after injury is still a great challenge in clinics, which suffers from its avascularity and poor proliferative ability. Herein we designed a novel biocompatible cellulose nanocrystal/GelMA (gelatin-methacrylate anhydride)/HAMA (hyaluronic acid-methacrylate anhydride)-blended hydrogel scaffold system, loaded with synthetic melanin nanoparticles (SMNP) and a bioactive drug kartogenin (KGN) for theranostic purpose. We found that the SMNP-KGN/Gel showed favorable mechanical property, thermal stability, and distinct magnetic resonance imaging (MRI) contrast enhancement. Meanwhile, the sustained release of KGN could recruit bone-derived mesenchymal stem cells to proliferate and differentiate into chondrocytes, which promoted cartilage regeneration in vitro and in vivo. The hydrogel degradation and cartilage restoration were simultaneously monitored by multiparametric MRI for 12 weeks, and further confirmed by histological analysis. Together, these results validated the multifunctional hydrogel as a promising tissue engineering platform for noninvasive imaging-guided precision therapy in cartilage regenerative medicine.
Collapse
Affiliation(s)
- Chuyao Chen
- Department of Medical Imaging Center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shaoshan Huang
- 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
| | - Zelong Chen
- Department of Medical Imaging Center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Qin Liu
- Department of Medical Imaging Center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yu Cai
- Clinical Research CenterZhujiang Hospital, Southern Medical UniversityGuangzhouGuangdongChina
- Center of Orthopedics, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yingjie Mei
- School of Biomedical EngineeringSouthern Medical UniversityGuangzhouChina
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang HospitalSouthern Medical 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
| | - Chenggong Yan
- Department of Medical Imaging Center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
13
|
Tipa C, Cidade MT, Borges JP, Costa LC, Silva JC, Soares PIP. Clay-Based Nanocomposite Hydrogels for Biomedical Applications: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3308. [PMID: 36234440 PMCID: PMC9565291 DOI: 10.3390/nano12193308] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
In recent decades, new and improved materials have been developed with a significant interest in three-dimensional (3D) scaffolds that can cope with the diverse needs of the expanding biomedical field and promote the required biological response in multiple applications. Due to their biocompatibility, ability to encapsulate and deliver drugs, and capacity to mimic the extracellular matrix (ECM), typical hydrogels have been extensively investigated in the biomedical and biotechnological fields. The major limitations of hydrogels include poor mechanical integrity and limited cell interaction, restricting their broad applicability. To overcome these limitations, an emerging approach, aimed at the generation of hybrid materials with synergistic effects, is focused on incorporating nanoparticles (NPs) within polymeric gels to achieve nanocomposites with tailored functionality and improved properties. This review focuses on the unique contributions of clay nanoparticles, regarding the recent developments of clay-based nanocomposite hydrogels, with an emphasis on biomedical applications.
Collapse
Affiliation(s)
- Cezar Tipa
- CENIMAT|i3N, Department of Materials Science, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Maria T. Cidade
- CENIMAT|i3N, Department of Materials Science, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - João P. Borges
- CENIMAT|i3N, Department of Materials Science, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Luis C. Costa
- I3N and Physics Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Jorge C. Silva
- CENIMAT|i3N, Department of Physics, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Paula I. P. Soares
- CENIMAT|i3N, Department of Materials Science, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| |
Collapse
|
14
|
Soh WWM, Teoh RYP, Zhu J, Xun Y, Wee CY, Ding J, Thian ES, Li J. Facile Construction of a Two-in-One Injectable Micelleplex-Loaded Thermogel System for the Prolonged Delivery of Plasmid DNA. Biomacromolecules 2022; 23:3477-3492. [PMID: 35878156 DOI: 10.1021/acs.biomac.2c00648] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nanoparticle-hydrogel systems have recently emerged as a class of interesting hybrid materials with immense potential for several biomedical applications. Remarkably, the incorporation of nanoparticles into a hydrogel may yield synergistic benefits lacking in a singular system. However, most synthetic strategies require laborious steps to achieve the system, severely restricting the process of translational research. Herein, a facile strategy to access a two-in-one system comprising two distinct polyurethane (PU)-based micellar systems is demonstrated and applied as a novel sustained gene delivery platform, where the two PUs are synthesized similarly but with slightly different compositions. One PU forms cationic micelles that complex with plasmid DNA (pDNA), which are loaded into a thermogel formed by another PU micellar system for the prolonged release of pDNA micelleplexes. Specifically, a thermogelling multiblock PU copolymer (denoted as EPH) was synthesized via the step-growth polymerization of poly(ethylene glycol), poly(propylene glycol), and poly(3-hydroxybutyrate). By further introducing a cationic extender, 3-(dimethylamino)-1,2-propanediol, into the reaction feed, a series of cationic PUs (denoted as EPHD) with varying compositions were obtained. The EPHDs formed positively charged micelles in aqueous solutions, efficiently condensed pDNA into nano-sized micelleplexes (<200 nm) at optimized w/w ratios, and mediated transient green fluorescence protein expression in HEK293T cells at 48 h post-transfection. On the other hand, aqueous EPH solution (4 wt %) was injectable at 4 °C and rapidly gelled upon heating to 37 °C to form a stable hydrogel depot. EPHD/pDNA micelleplexes were easily loaded into EPH by mixing the solutions at 4 °C, before heating to 37 °C, leading to the resultant hydrogel system. The in vitro release study revealed that while free pDNA loaded in the thermogel was completely released in 2 weeks, the release of EPHD/pDNA micelleplexes was prolonged to at least 28 days, suggesting substantial micelleplex-hydrogel interactions. Intact, bioactive, and noncytotoxic EPHD/pDNA micelleplexes in the release media were proved by gel retardation, in vitro gene transfection, and CCK-8 cytotoxicity assay results, respectively. Collectively, this work presents a simple approach to achieving and optimizing a novel two-in-one nanoparticle-hydrogel system for the prolonged delivery of pDNA and may be promising for long-term gene delivery applications.
Collapse
Affiliation(s)
- Wilson Wee Mia Soh
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Rachel Yun Pei Teoh
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore.,NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Yanran Xun
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Chien Yi Wee
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Jun Ding
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Eng San Thian
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Jun Li
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore.,NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
15
|
Jing G, Yang L, Wang H, Niu J, Li Y, Wang S. Interference of layered double hydroxide nanoparticles with pathways for biomedical applications. Adv Drug Deliv Rev 2022; 188:114451. [PMID: 35843506 DOI: 10.1016/j.addr.2022.114451] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/18/2022] [Accepted: 07/09/2022] [Indexed: 11/01/2022]
Abstract
Recent decades have witnessed a surge of explorations into the application of multifarious materials, especially biomedical applications. Among them, layered double hydroxides (LDHs) have been widely developed as typical inorganic layer materials to achieve remarkable advancements. Multiple physicochemical properties endow LDHs with excellent merits in biomedical applications. Moreover, LDH nanoplatforms could serve as "molecular switches", which are capable of the controlled release of payloads under specific physiological pH conditions but are stable during circulation in the bloodstream. In addition, LDHs themselves are composed of several specific cations and possess favorable biological effects or regulatory roles in various cellular functions. These advantages have caused LDHs to become increasingly of interest in the area of nanomedicine. Recent efforts have been devoted to revealing the potential factors that interfere with the biological pathways of LDH-based nanoparticles, such as their applications in shaping the functions of immune cells and in determining the fate of stem cells and tumor treatments, which are comprehensively described herein. In addition, several intracellular signaling pathways interfering with by LDHs in the above applications were also systematically expatiated. Finally, the future development and challenges of LDH-based nanomedicine are discussed in the context of the ultimate goal of practical clinical application.
Collapse
Affiliation(s)
- Guoxin Jing
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Linnan Yang
- Central Laboratory, First Affiliated Hospital, Anhui Medical University, Hefei, PR China
| | - Hong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Jintong Niu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Youyuan Li
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China
| | - Shilong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China.
| |
Collapse
|
16
|
Ao Y, Zhang E, Liu Y, Yang L, Li J, Wang F. Advanced Hydrogels With Nanoparticle Inclusion for Cartilage Tissue Engineering. Front Bioeng Biotechnol 2022; 10:951513. [PMID: 35845428 PMCID: PMC9277358 DOI: 10.3389/fbioe.2022.951513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Cartilage dysfunctions caused by congenital disease, trauma and osteoarthritis are still a serious threat to joint activity and quality of life, potentially leading to disability. The relatively well-established tissue engineering technology based on hydrogel is a promising strategy for cartilage defect repairing. However, several unmet challenges remain to be resolved before its wide application and clinical translation, such as weak mechanical property and compromised bioactivity. The development of nanomedicine has brought a new dawn to cartilage tissue engineering, and composite hydrogel containing nanoparticles can substantially mimic natural cartilage components with good histocompatibility, demonstrating unique biological effects. In this review, we summarize the different advanced nanoparticle hydrogels currently adopted in cartilage tissue engineering. In addition, we also discuss the various application scenarios including injection and fabrication strategies of nanocomposite hydrogel in the field of cartilage repair. Finally, the future application prospects and challenges of nanocomposite hydrogel are also highlighted.
Collapse
Affiliation(s)
- Yunong Ao
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - En Zhang
- Chongqing Institute for Food and Drug Control, Chongqing, China
| | - Yangxi Liu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Liu Yang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jun Li
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Institute for Food and Drug Control, Chongqing, China
- *Correspondence: Jun Li, ; Fuyou Wang,
| | - Fuyou Wang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- *Correspondence: Jun Li, ; Fuyou Wang,
| |
Collapse
|
17
|
Dehghan-Baniani D, Mehrjou B, Wang D, Bagheri R, Solouk A, Chu PK, Wu H. A dual functional chondro-inductive chitosan thermogel with high shear modulus and sustained drug release for cartilage tissue engineering. Int J Biol Macromol 2022; 205:638-650. [PMID: 35217083 DOI: 10.1016/j.ijbiomac.2022.02.115] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/13/2022] [Accepted: 02/18/2022] [Indexed: 12/17/2022]
Abstract
We report a chitosan-based nanocomposite thermogel with superior shear modulus resembling that of cartilage and dual pro-chondrogenic and anti-inflammatory functions. Two therapeutic agents, kartogenin (KGN) and diclofenac sodium (DS), are employed to promote chondrogenesis of stem cells and suppress inflammation, respectively. To extend the release time in a controlled manner, KGN is encapsulated in the uniform-sized starch microspheres and DS is loaded into the halloysite nanotubes. Both drug carriers are doped into the maleimide-modified chitosan hydrogel to produce a shear modulus of 167 ± 5 kPa that is comparable to that of articular cartilage (50-250 kPa). Owing to the hydrogel injectability and relatively suitable gelation time (5 ± 0.5 min) at 37 °C, this system potentially constitutes a manageable platform for clinical practice. Moreover, sustained linear drug release for over a month boosts chondro-differentiation of stem cells to eliminate the necessity for multiple administrations. Considering virtues such as thermogel strength and ability to co-deliver anti-inflammatory and chondro-inductive biomolecules continuously, the materials and strategy have promising potential in functional cartilage tissue engineering.
Collapse
Affiliation(s)
- Dorsa Dehghan-Baniani
- Department of Chemical and Biological Engineering, Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China; Polymeric Materials Research Group, Department of Materials Science and Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9466, Iran
| | - Babak Mehrjou
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Dong Wang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Reza Bagheri
- Polymeric Materials Research Group, Department of Materials Science and Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9466, Iran
| | - Atefeh Solouk
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Hongkai Wu
- Department of Chemical and Biological Engineering, Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China; Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
18
|
Huang J, Liu F, Su H, Xiong J, Yang L, Xia J, Liang Y. Advanced Nanocomposite Hydrogels for Cartilage Tissue Engineering. Gels 2022; 8:138. [PMID: 35200519 PMCID: PMC8871651 DOI: 10.3390/gels8020138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Tissue engineering is becoming an effective strategy for repairing cartilage damage. Synthesized nanocomposite hydrogels mimic the structure of natural cartilage extracellular matrices (ECMs), are biocompatible, and exhibit nano-bio effects in response to external stimuli. These inherent characteristics make nanocomposite hydrogels promising scaffold materials for cartilage tissue engineering. This review summarizes the advances made in the field of nanocomposite hydrogels for artificial cartilage. We discuss, in detail, their preparation methods and scope of application. The challenges involved for the application of hydrogel nanocomposites for cartilage repair are also highlighted.
Collapse
Affiliation(s)
- Jianghong Huang
- Department of Spine Surgery and Orthopedics, Shenzhen Second People’s Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, China; (J.H.); (J.X.); (L.Y.)
- Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Fei Liu
- Department of Biochemistry, Texas A&M University School of Medicine, Bryan, TX 77807, USA;
| | - Haijing Su
- Technology R&D Department, Shenzhen Lechuang Medical Research Institute Co., Ltd., Shenzhen 518129, China;
| | - Jianyi Xiong
- Department of Spine Surgery and Orthopedics, Shenzhen Second People’s Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, China; (J.H.); (J.X.); (L.Y.)
| | - Lei Yang
- Department of Spine Surgery and Orthopedics, Shenzhen Second People’s Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, China; (J.H.); (J.X.); (L.Y.)
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China;
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen 518020, China
| |
Collapse
|
19
|
Gonzalez-Fernandez P, Rodríguez-Nogales C, Jordan O, Allémann E. Combination of mesenchymal stem cells and bioactive molecules in hydrogels for osteoarthritis treatment. Eur J Pharm Biopharm 2022; 172:41-52. [PMID: 35114357 DOI: 10.1016/j.ejpb.2022.01.003] [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: 09/02/2021] [Revised: 12/13/2021] [Accepted: 01/17/2022] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) is a chronic and inflammatory disease with no effective regenerative treatments to date. The therapeutic potential of mesenchymal stem cells (MSCs) remains to be fully explored. Intra-articular injection of these cells promotes cartilage protection and regeneration by paracrine signaling and differentiation into chondrocytes. However, joints display a harsh avascular environment for these cells upon injection. This phenomenon prompted researchers to develop suitable injectable materials or systems for MSCs to enhance their function and survival. Among them, hydrogels can absorb a large amount of water and maintain their 3D structure but also allow incorporation of bioactive agents or small molecules in their matrix that maximize the action of MSCs. These materials possess advantageous cartilage-like features such as collagen or hyaluronic acid moieties that interact with MSC receptors, thereby promoting cell adhesion. This review provides an up-to-date overview of the progress and opportunities of MSCs entrapped into hydrogels, combined with bioactive/small molecules to improve the therapeutic effects in OA treatment.
Collapse
Affiliation(s)
- P Gonzalez-Fernandez
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - C Rodríguez-Nogales
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - O Jordan
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - E Allémann
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland.
| |
Collapse
|
20
|
Moon Y, Patel M, Um S, Lee HJ, Park S, Park SB, Cha SS, Jeong B. Folic acid pretreatment and its sustained delivery for chondrogenic differentiation of MSCs. J Control Release 2022; 343:118-130. [PMID: 35051494 DOI: 10.1016/j.jconrel.2022.01.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 10/19/2022]
Abstract
Dietary uptake of folic acid (FA) improves cartilage regeneration. In this work, we discovered that three days of FA treatment is highly effective for promoting chondrogenic differentiation of tonsil-derived mesenchymal stem cells (TMSCs). In a three-dimensional pellet culture, the levels of typical chondrogenic biomarkers, sulfated glycosaminoglycan, proteoglycan, type II collagen (COL II), SRY box transcription factor 9 (SOX 9), cartilage oligomeric matrix protein (COMP), and aggrecan (ACAN) increased significantly in proportion to FA concentration up to 30 μM. At the mRNA expression level, COL II, SOX 9, COMP, and ACAN increased 3.6-6.0-fold with FA treatment at 30 μM compared with the control system that did not receive FA treatment, and the levels with FA treatment were 1.6-2.5 times greater than those in the kartogenin-treated positive control system. FA treatment did not increase type I collagen α1 (COL I α1), an osteogenic biomarker which is a concern with most chondrogenic promoters. At the high FA concentration of 100 μM, significant decreases in chondrogenic biomarkers were observed, which might be related to DNA methylation. A thermogel system incorporating TMSCs and FA provided sustained release of FA over several days, similar to the FA treatment. The thermogel system confirmed the efficacy of FA in promoting chondrogenic promotion of TMSCs. The increased nuclear translocation of core-binding factor β subunit (CBFβ) and the runt-related transcription factor 1 (RUNX1) expression after FA treatment, together with molecular docking studies, suggest that the chondrogenic enhancement mechanism of FA is mediated by CBFβ and RUNX1.
Collapse
Affiliation(s)
- Yuna Moon
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Soyoun Um
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Sohee Park
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Soo-Bong Park
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea.
| |
Collapse
|
21
|
Hu X, Xia Z, Cai K. Recent advances of 3D hydrogel culture systems for mesenchymal stem cell-based therapy and cell behavior regulation. J Mater Chem B 2022; 10:1486-1507. [DOI: 10.1039/d1tb02537f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesenchymal stem cells (MSCs) have been increasingly recognized as resources for disease treatments and regenerative medicine. Meanwhile, the unique chemical and physical properties of hydrogels provide innate advantages to achieve...
Collapse
|
22
|
Peters JT, Wechsler ME, Peppas NA. Advanced biomedical hydrogels: molecular architecture and its impact on medical applications. Regen Biomater 2021; 8:rbab060. [PMID: 34925879 PMCID: PMC8678442 DOI: 10.1093/rb/rbab060] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
Hydrogels are cross-linked polymeric networks swollen in water, physiological aqueous solutions or biological fluids. They are synthesized by a wide range of polymerization methods that allow for the introduction of linear and branched units with specific molecular characteristics. In addition, they can be tuned to exhibit desirable chemical characteristics including hydrophilicity or hydrophobicity. The synthesized hydrogels can be anionic, cationic, or amphiphilic and can contain multifunctional cross-links, junctions or tie points. Beyond these characteristics, hydrogels exhibit compatibility with biological systems, and can be synthesized to render systems that swell or collapse in response to external stimuli. This versatility and compatibility have led to better understanding of how the hydrogel's molecular architecture will affect their physicochemical, mechanical and biological properties. We present a critical summary of the main methods to synthesize hydrogels, which define their architecture, and advanced structural characteristics for macromolecular/biological applications.
Collapse
Affiliation(s)
- Jonathan T Peters
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, 200 E. Dean Keeton, Austin, TX 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
| | - Marissa E Wechsler
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, 200 E. Dean Keeton, Austin, TX 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
- Department of Surgery and Perioperative Care, and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, 1601 Trinity St., Bldg. B, Austin, TX 78712, USA
| |
Collapse
|
23
|
Wei W, Dai H. Articular cartilage and osteochondral tissue engineering techniques: Recent advances and challenges. Bioact Mater 2021; 6:4830-4855. [PMID: 34136726 PMCID: PMC8175243 DOI: 10.1016/j.bioactmat.2021.05.011] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
In spite of the considerable achievements in the field of regenerative medicine in the past several decades, osteochondral defect regeneration remains a challenging issue among diseases in the musculoskeletal system because of the spatial complexity of osteochondral units in composition, structure and functions. In order to repair the hierarchical tissue involving different layers of articular cartilage, cartilage-bone interface and subchondral bone, traditional clinical treatments including palliative and reparative methods have showed certain improvement in pain relief and defect filling. It is the development of tissue engineering that has provided more promising results in regenerating neo-tissues with comparable compositional, structural and functional characteristics to the native osteochondral tissues. Here in this review, some basic knowledge of the osteochondral units including the anatomical structure and composition, the defect classification and clinical treatments will be first introduced. Then we will highlight the recent progress in osteochondral tissue engineering from perspectives of scaffold design, cell encapsulation and signaling factor incorporation including bioreactor application. Clinical products for osteochondral defect repair will be analyzed and summarized later. Moreover, we will discuss the current obstacles and future directions to regenerate the damaged osteochondral tissues.
Collapse
Affiliation(s)
- Wenying Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan, 528200, China
| |
Collapse
|
24
|
Murali A, Lokhande G, Deo KA, Brokesh A, Gaharwar AK. Emerging 2D Nanomaterials for Biomedical Applications. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2021; 50:276-302. [PMID: 34970073 PMCID: PMC8713997 DOI: 10.1016/j.mattod.2021.04.020] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Two-dimensional (2D) nanomaterials are an emerging class of biomaterials with remarkable potential for biomedical applications. The planar topography of these nanomaterials confers unique physical, chemical, electronic and optical properties, making them attractive candidates for therapeutic delivery, biosensing, bioimaging, regenerative medicine, and additive manufacturing strategies. The high surface-to-volume ratio of 2D nanomaterials promotes enhanced interactions with biomolecules and cells. A range of 2D nanomaterials, including transition metal dichalcogenides (TMDs), layered double hydroxides (LDHs), layered silicates (nanoclays), 2D metal carbides and nitrides (MXenes), metal-organic framework (MOFs), covalent organic frameworks (COFs) and polymer nanosheets have been investigated for their potential in biomedical applications. Here, we will critically evaluate recent advances of 2D nanomaterial strategies in biomedical engineering and discuss emerging approaches and current limitations associated with these nanomaterials. Due to their unique physical, chemical, and biological properties, this new class of nanomaterials has the potential to become a platform technology in regenerative medicine and other biomedical applications.
Collapse
Affiliation(s)
- Aparna Murali
- Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Giriraj Lokhande
- Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Kaivalya A. Deo
- Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Anna Brokesh
- Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Akhilesh K. Gaharwar
- Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Material Science and Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX 77843, USA
- Interdisciplinary Graduate Program in Genetics, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
25
|
Izbudak B, Cecen B, Anaya I, Miri AK, Bal-Ozturk A, Karaoz E. Layered double hydroxide-based nanocomposite scaffolds in tissue engineering applications. RSC Adv 2021; 11:30237-30252. [PMID: 35480250 PMCID: PMC9041101 DOI: 10.1039/d1ra03978d] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
Layered double hydroxides (LDHs), when incorporated into biomaterials, provide a tunable composition, controllable particle size, anion exchange capacity, pH-sensitive solubility, high-drug loading efficiency, efficient gene and drug delivery, controlled release and effective intracellular uptake, natural biodegradability in an acidic medium, and negligible toxicity. In this review, we study potential applications of LDH-based nanocomposite scaffolds for tissue engineering. We address how LDHs provide new solutions for nanostructure stability and enhance in vivo studies' success.
Collapse
Affiliation(s)
- Burcin Izbudak
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University Istanbul Turkey
| | - Berivan Cecen
- Biofabrication Lab, Department of Mechanical Engineering, Rowan University Glassboro NJ 08028 USA.,School of Medical Engineering, Science and Health, Rowan University Camden NJ 08103 USA.,Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istinye University 34010 Zeytinburnu Istanbul Turkey
| | - Ingrid Anaya
- Department of Bioengineering, Tecnológico de Monterrey, Campus Monterrey CP 64849 Monterrey Nuevo León México
| | - Amir K Miri
- Biofabrication Lab, Department of Mechanical Engineering, Rowan University Glassboro NJ 08028 USA.,School of Medical Engineering, Science and Health, Rowan University Camden NJ 08103 USA
| | - Ayca Bal-Ozturk
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University Istanbul Turkey .,Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University Istanbul Turkey
| | - Erdal Karaoz
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University Istanbul Turkey .,Department of Histology and Embryology, Faculty of Medicine, Istinye University Istanbul Turkey.,Center for Regenerative Medicine and Stem Cell Research and Manufacturing (LivMedCell) Istanbul Turkey
| |
Collapse
|
26
|
Zhang K, Xue K, Loh XJ. Thermo-Responsive Hydrogels: From Recent Progress to Biomedical Applications. Gels 2021; 7:77. [PMID: 34202514 PMCID: PMC8293033 DOI: 10.3390/gels7030077] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022] Open
Abstract
Thermogels are also known as thermo-sensitive or thermo-responsive hydrogels and can undergo a sol-gel transition as the temperature increases. This thermogelling behavior is the result of combined action from multiscale thermo-responsive mechanisms. From micro to macro, these mechanisms can be attributed to LCST behavior, micellization, and micelle aggregation of thermogelling polymers. Due to its facile phase conversion properties, thermogels are injectable yet can form an in situ gel in the human body. Thermogels act as a useful platform biomaterial that operates at physiological body temperatures. The purpose of this review is to summarize the recent progress in thermogel research, including investigations on the thermogel gelation mechanism and its applications in drug delivery, 3D cell culture, and tissue engineering. The review also discusses emerging directions in the study of thermogels.
Collapse
Affiliation(s)
- Kaiwen Zhang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology (SUSTech), 1088 Xueyuan Avenue, Shenzhen 518055, China;
| | - Kun Xue
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Singapore 138634, Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Singapore 138634, Singapore
- Department of Materials Science and Engineering, National University of Singapore (NUS), 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
27
|
Walker M, Luo J, Pringle EW, Cantini M. ChondroGELesis: Hydrogels to harness the chondrogenic potential of stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111822. [PMID: 33579465 DOI: 10.1016/j.msec.2020.111822] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 01/01/2023]
Abstract
The extracellular matrix is a highly complex microenvironment, whose various components converge to regulate cell fate. Hydrogels, as water-swollen polymer networks composed by synthetic or natural materials, are ideal candidates to create biologically active substrates that mimic these matrices and target cell behaviour for a desired tissue engineering application. Indeed, the ability to tune their mechanical, structural, and biochemical properties provides a framework to recapitulate native tissues. This review explores how hydrogels have been engineered to harness the chondrogenic response of stem cells for the repair of damaged cartilage tissue. The signalling processes involved in hydrogel-driven chondrogenesis are also discussed, identifying critical pathways that should be taken into account during hydrogel design.
Collapse
Affiliation(s)
- Matthew Walker
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Jiajun Luo
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Eonan William Pringle
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Marco Cantini
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK.
| |
Collapse
|
28
|
Dehghan‐Baniani D, Mehrjou B, Chu PK, Wu H. A Biomimetic Nano-Engineered Platform for Functional Tissue Engineering of Cartilage Superficial Zone. Adv Healthc Mater 2021; 10:e2001018. [PMID: 32803848 DOI: 10.1002/adhm.202001018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/15/2020] [Indexed: 12/22/2022]
Abstract
Articular cartilage has limited regeneration capacity because of its acellular and avascular nature. Although tissue engineering has been shown to be life-saving, reforming cartilage zones required by the appropriate tissue functions are challenging. Herein, the need is addressed by designing and producing a nano-engineered structure mimicking the superficial zone (SZ) of articular cartilage. The substrate is based on silk with good mechanical properties in conjunction with nano-topographical and biochemical cues. Nanopillar arrays are produced on the silk surface to regulate the stem cell morphology rendering them with a flattened ellipsoidal shape that is similar to that of chondrocytes in the SZ of natural cartilage. The cell interactions are enhanced by nitrogen ion implantation and the biomolecule, kartogenin (KGN), is loaded to promote chondrogenesis of the stem cells and furthermore, a thermosensitive chitosan hydrogel is formed on the nanopatterned silk to produce rheological properties similar to those of a synovial fluid. Based on the in vitro results and mechanical properties, it is a desirable implantable smart structure mimicking the cartilage SZ with the ability of continuous drug release for cartilage regeneration.
Collapse
Affiliation(s)
- Dorsa Dehghan‐Baniani
- Department of Chemical and Biological Engineering Division of Biomedical Engineering The Hong Kong University of Science and Technology Hong Kong China
| | - Babak Mehrjou
- Department of Physics Department of Materials Science and Engineering Department of Biomedical Engineering City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong China
| | - Paul K. Chu
- Department of Physics Department of Materials Science and Engineering Department of Biomedical Engineering City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong China
| | - Hongkai Wu
- Department of Chemical and Biological Engineering Division of Biomedical Engineering The Hong Kong University of Science and Technology Hong Kong China
- Department of Chemistry The Hong Kong University of Science and Technology Hong Kong China
- Guangzhou First People's Hospital 1 Panfu Rd, Yuexiu District Guangzhou Guangdong Province China
| |
Collapse
|
29
|
Mohammed M, Lai TS, Lin HC. Substrate stiffness and sequence dependent bioactive peptide hydrogels influence the chondrogenic differentiation of human mesenchymal stem cells. J Mater Chem B 2021; 9:1676-1685. [DOI: 10.1039/d0tb02008g] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
N-Cadherin mimetic nanofibrous biocompatible peptide hydrogels with enhanced mechanical properties for differentiation of mesenchymal stem cells into chondrocytes.
Collapse
Affiliation(s)
- Mohiuddin Mohammed
- Department of Materials Science and Engineering, National Chiao Tung University
- Hsinchu 30010
- Taiwan
| | - Tsung-Sheng Lai
- Department of Materials Science and Engineering, National Chiao Tung University
- Hsinchu 30010
- Taiwan
| | - Hsin-Chieh Lin
- Department of Materials Science and Engineering, National Chiao Tung University
- Hsinchu 30010
- Taiwan
| |
Collapse
|
30
|
Yan L, Gonca S, Zhu G, Zhang W, Chen X. Layered double hydroxide nanostructures and nanocomposites for biomedical applications. J Mater Chem B 2020; 7:5583-5601. [PMID: 31508652 DOI: 10.1039/c9tb01312a] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Layered double hydroxide (LDH) nanostructures and related nanocomposites have attracted significant interest in biomedical applications including cancer therapy, bioimaging and antibacterial treatment. These materials hold great advantages including low cost and facile preparation, convenient drug loading, high drug incorporation capacity, good biocompatibility, efficient intracellular uptake and endosome/lysosome escape, and natural biodegradability in an acidic environment. In this review, we summarize the development of three types of LDH nanostructures including pristine LDH, surface modified LDH, and LDH nanocomposites for a range of biomedical applications. The advantages and disadvantages of LDH nanostructures and insights into the future development are also discussed.
Collapse
Affiliation(s)
- Li Yan
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | | | | | | |
Collapse
|
31
|
Lim DG, Kang E, Jeong SH. pH-dependent nanodiamonds enhance the mechanical properties of 3D-printed hyaluronic acid nanocomposite hydrogels. J Nanobiotechnology 2020; 18:88. [PMID: 32522274 PMCID: PMC7288416 DOI: 10.1186/s12951-020-00647-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/06/2020] [Indexed: 01/08/2023] Open
Abstract
Nanocomposite hydrogels capable of undergoing manufacturing process have recently attracted attention in biomedical applications due to their desired mechanical properties and high functionality. 3D printing nanocomposite hydrogels of hyaluronic acid (HA)/nanodiamond (ND) revealed that the addition of ND with the low weight ratio of 0.02 wt% resulted in higher compressive force and gel breaking point, compared with HA only nanocomposites. These HA nanocomposite hydrogels loaded with surface functionalized ND allowed for the enforced compressive stress to be tuned in a pH-dependent manner. HA nanocomposite hydrogels with ND-OH at pH 8 showed an increase of 1.40-fold (0.02%: 236.18 kPa) and 1.37-fold (0.04%: 616.72 kPa) the compressive stress at the composition of 0.02 wt% and 0.04 wt, respectively, compared to those of ND-COOH (0.02%: 168.31 kPa, 0.04%: 449.59 kPa) at the same pH. Moreover, the compressive stress of HA/ND-OH (0.04 wt%) at pH 8 was mechanically enhanced 1.29-fold, compared to that of HA/ND-OH (0.04 wt%) at pH 7. These results indicate that the tunable buffering environment and interaction with the long chains of HA at the molecular level have a critical role in the dependency of the mechanical properties on pH. Due to the pH stability of the ND-OH nanophase, filament-based processing and layer-based deposition at microscale attained enforced mechanical properties of hydrogel. Fine surface tuning of the inorganic ND nanophase and controlled 3D printing leads to improved control over the pH-dependent mechanical properties of the nanocomposite hydrogels reported herein.![]()
Collapse
Affiliation(s)
- Dae Gon Lim
- College of Pharmacy, Dongguk University-Seoul, Gyeonggi, 10326, Republic of Korea
| | - Eunah Kang
- School of Chemical Engineering and Material Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Seong Hoon Jeong
- College of Pharmacy, Dongguk University-Seoul, Gyeonggi, 10326, Republic of Korea.
| |
Collapse
|
32
|
Dehghan-Baniani D, Chen Y, Wang D, Bagheri R, Solouk A, Wu H. Injectable in situ forming kartogenin-loaded chitosan hydrogel with tunable rheological properties for cartilage tissue engineering. Colloids Surf B Biointerfaces 2020; 192:111059. [PMID: 32380404 DOI: 10.1016/j.colsurfb.2020.111059] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/16/2020] [Accepted: 04/12/2020] [Indexed: 11/22/2022]
Abstract
Limited regeneration capacity of cartilage can be addressed by tissue engineering approaches including localized delivery of bioactive agents using biomaterials. Although chitosan hydrogels have been considered as appropriate candidates for these purposes, however, their poor mechanical properties limit their real applications. Here, we develop in situ forming chitosan hydrogels with enhanced shear modulus by chemical modification of chitosan using N-(β-maleimidopropyloxy) succinimide ester (BMPS). Moreover, we utilize β-Glycerophosphate (β-GP) in the hydrogels for achieving thermosensitivity. We investigate the effects of BMPS, β-GP and chitosan concentration on rheological and swelling properties of the hydrogels. Accordingly, we generate significant statistical models by response surface method to predict these properties. These models provide us beneficial tools to tune the hydrogel properties depending on the cartilage defect location and properties. Finally, we incorporate a recently discovered small biomolecule, kartogenin (KGN), for promoting chondrogenesis of stem cells into the optimized hydrogel. The hydrogel's shear modulus is 78 ± 5 kPa which covers a wide range of human articular cartilage shear modulus (50-250 kPa). It can be injected to the defects non-invasively at room temperature which gels at 37 °C within minutes. Additionally, it provides a sustained KGN release for ∼40 days that may eliminate the need of multiple injections. In vitro chondrogenic results confirm enhanced chondrogenic differentiation of human adipose mesenchymal stem cells (hAMSCs) treated with KGN-loaded hydrogel, compared to pure KGN. Based on the enhanced hydrogel shear modulus, injectability, gelation behavior, long-term drug release and in vitro results, this thermosensitive hydrogel looks promising for cartilage tissue engineering.
Collapse
Affiliation(s)
- Dorsa Dehghan-Baniani
- Department of Chemical and Biological Engineering, Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China; Polymeric Materials Research Group, Department of Materials Science and Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9466, Iran
| | - Yin Chen
- Department of Chemical and Biological Engineering, Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Dong Wang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Reza Bagheri
- Polymeric Materials Research Group, Department of Materials Science and Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9466, Iran
| | - Atefeh Solouk
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Hongkai Wu
- Department of Chemical and Biological Engineering, Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China; Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
33
|
Chen Y, Shi J, Zhang Y, Miao J, Zhao Z, Jin X, Liu L, Yu L, Shen C, Ding J. An injectable thermosensitive hydrogel loaded with an ancient natural drug colchicine for myocardial repair after infarction. J Mater Chem B 2020; 8:980-992. [PMID: 31930242 DOI: 10.1039/c9tb02523e] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Localized administration of anti-inflammatory agents benefits patients after myocardial infarction (MI) by repressing/modulating inflammatory response of the MI region and thus accelerating repair of the impaired tissues. Colchicine (Col), an ancient natural drug, has excellent anti-inflammatory effects; however, its utilization is strictly limited due to its severe systemic toxicity and narrow therapeutic window. In this study, we developed an intramyocardial delivery system of Col using an injectable, thermosensitive poly(lactide-co-glycolide)-poly(ethylene glycol)-poly(lactide-co-glycolide) (PLGA-PEG-PLGA) polymer hydrogel as the vehicle for the treatment of MI while minimizing its systemic toxicity. The aqueous PLGA-PEG-PLGA solution loaded with Col (Col@Gel) underwent a sol-gel transition at 35 °C and maintained a gel state at body temperature. Col was released from the Col@Gel in an initial burst followed by a sustained release manner for over 8 days. The in vitro cell tests showed that the Col@Gel system significantly inhibited macrophage proliferation and migration. In a mouse model of MI, a single intramyocardial administration of the Col@Gel effectively alleviated cardiac inflammation, inhibited myocardial apoptosis and fibrosis, improved cardiac function and structure, and increased mouse survival without inducing severe systemic toxicity, which was observed following intraperitoneal administration of Col solution. These results suggested that the Col@Gel system is a reliable drug delivery system for the sustained local release of Col and has great potential as an anti-inflammatory therapy for the treat of MI.
Collapse
Affiliation(s)
- Yu Chen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Jiayue Shi
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China.
| | - Yaping Zhang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Jiajun Miao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China.
| | - Zhe Zhao
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Xian Jin
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Liang Liu
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China. and Zhuhai Fudan Innovation Institute, Zhuhai, Guangdong 51900, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China. and Zhuhai Fudan Innovation Institute, Zhuhai, Guangdong 51900, China
| |
Collapse
|
34
|
Zhao Y, Ding X, Dong Y, Sun X, Wang L, Ma X, Zhu M, Xu B, Yang Q. Role of the Calcified Cartilage Layer of an Integrated Trilayered Silk Fibroin Scaffold Used to Regenerate Osteochondral Defects in Rabbit Knees. ACS Biomater Sci Eng 2020; 6:1208-1216. [PMID: 33464868 DOI: 10.1021/acsbiomaterials.9b01661] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The repair of osteochondral defects remains challenging, given the complexity of native osteochondral tissue and the limited self-repair capacity of cartilage. Osteochondral tissue engineering is a promising strategy. Here, we fabricated a biomimetic osteochondral scaffold using silk fibroin and hydroxyapatite, including a calcified cartilage layer (CCL). We studied the role played by the CCL in terms of cell viability in vivo. We established osteochondral defects in rabbit knees to investigate the effects of CCL-containing scaffolds with or without adipose tissue-derived stem cells (ADSCs). We evaluated osteochondral tissue regeneration by calculating gross observational scores, via histological and immunohistochemical assessments, by performing quantitative biochemical and biomechanical analyses of new osteochondral tissue, and via microcomputed tomography of new bone at 4, 8, and 12 weeks after surgery. In terms of surface roughness and integrity, the CCL + ADSCs group was better than the CCL and the non-CCL + ADSCs groups at all time points tested; the glycosaminoglycan and collagen type II levels of the CCL + ADSCs group were highest, reflecting the important role played by the CCL in cartilage tissue repair. Subchondral bone smoothness was better in the CCL + ADSCs group than in the non-CCL + ADSCs and CCL groups. The CCL promoted smooth subchondral bone regeneration but did not obviously affect bone strength or quality. In conclusion, a biomimetic osteochondral scaffold with a CCL, combined with autologous ADSCs, satisfactorily regenerated a rabbit osteochondral defect. The CCL enhances cartilage and subchondral bone regeneration.
Collapse
Affiliation(s)
- Yanhong Zhao
- Stomatological Hospital of Tianjin Medical University, 12 Qixiangtai Road, Heping District, Tianjin 300070, People's Republic of China
| | - Xiaoming Ding
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China.,Department of Orthopedics, Rizhao Traditional Chinese Medicine Hospital, 35 Haiwang Road, Donggang District, Rizhao, Shandong 276800, People's Republic of China
| | - Yunsheng Dong
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Lianyong Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Xinlong Ma
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Meifeng Zhu
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Baoshan Xu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| |
Collapse
|
35
|
Rasines Mazo A, Allison-Logan S, Karimi F, Chan NJA, Qiu W, Duan W, O’Brien-Simpson NM, Qiao GG. Ring opening polymerization of α-amino acids: advances in synthesis, architecture and applications of polypeptides and their hybrids. Chem Soc Rev 2020; 49:4737-4834. [DOI: 10.1039/c9cs00738e] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review provides a comprehensive overview of the latest advances in the synthesis, architectural design and biomedical applications of polypeptides and their hybrids.
Collapse
Affiliation(s)
- Alicia Rasines Mazo
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Stephanie Allison-Logan
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Fatemeh Karimi
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Nicholas Jun-An Chan
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Wenlian Qiu
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| | - Wei Duan
- School of Medicine
- Deakin University
- Geelong
- Australia
| | - Neil M. O’Brien-Simpson
- Centre for Oral Health Research
- Melbourne Dental School and the Bio21 Institute of Molecular Science and Biotechnology
- University of Melbourne
- Parkville
- Australia
| | - Greg G. Qiao
- Polymer Science Group
- Department of Chemical Engineering
- University of Melbourne
- Parkville
- Australia
| |
Collapse
|
36
|
Hu T, Mei X, Wang Y, Weng X, Liang R, Wei M. Two-dimensional nanomaterials: fascinating materials in biomedical field. Sci Bull (Beijing) 2019; 64:1707-1727. [PMID: 36659785 DOI: 10.1016/j.scib.2019.09.021] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/22/2019] [Accepted: 09/12/2019] [Indexed: 01/21/2023]
Abstract
Due to their high anisotropy and chemical functions, two-dimensional (2D) nanomaterials have attracted increasing interest and attention from various scientific fields, including functional electronics, catalysis, supercapacitors, batteries and energy materials. In the biomedical field, 2D nanomaterials have made significant contributions to the field of nanomedicine, especially in drug/gene delivery systems, multimodal imaging, biosensing, antimicrobial agents and tissue engineering. 2D nanomaterials such as graphene/graphene oxide (GO)/reduced graphene oxide (rGO), silicate clays, layered double hydroxides (LDHs), transition metal dichalcogenides (TMDs), transition metal oxides (TMOs), black phosphorus (BP), graphitic carbon nitride (g-C3N4), hexagonal boron nitride (h-BN), antimonene (AM), boron nanosheets (B NSs) and tin telluride nanosheets (SnTe NSs) possess excellent physical, chemical, optical and biological properties due to their uniform shapes, high surface-to-volume ratios and surface charge. In this review, we first introduce the properties, structures and synthetic strategies of different configurations of 2D nanomaterials. Recent advances and paradigms of 2D nanomaterials in a variety of biomedical applications, ranging from drug delivery, cancer treatment, bioimaging and tissue engineering to biosensing are discussed afterwards. In the final part, we foresee the development prospects and challenges of 2D nanomaterials after summarizing the research status of ultrathin 2D nanomaterials.
Collapse
Affiliation(s)
- Tingting Hu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xuan Mei
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yingjie Wang
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xisheng Weng
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Min Wei
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
37
|
Gan D, Wang Z, Xie C, Wang X, Xing W, Ge X, Yuan H, Wang K, Tan H, Lu X. Mussel-Inspired Tough Hydrogel with In Situ Nanohydroxyapatite Mineralization for Osteochondral Defect Repair. Adv Healthc Mater 2019; 8:e1901103. [PMID: 31609095 DOI: 10.1002/adhm.201901103] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/20/2019] [Indexed: 01/15/2023]
Abstract
Repairing osteochondral defects is a considerable challenge because it involves the breakdown of articular cartilage and underlying bone. Traditional hydrogels with a homogenized single-layer structure cannot fully restore the function of osteochondral cartilage tissue. In this study, a mussel-inspired hydrogel with a bilayer structure is developed to repair osteochondral defects. The hydrogel is synthesized by simultaneously polymerizing two layers using a one-pot method. The resulting upper and lower gelatin methacryloyl-polydopamine hydrogel layers are used as cartilage and subchondral bone repair layers, respectively. Polydopamine-induced hydroxyapatite in situ mineralization takes place in the lower layer to mimic the structure of subchondral bone. The bilayer hydrogel exhibits good mechanical properties for the synergistic effect of covalent and noncovalent bonds, as well as nanoreinforcement of mineralized hydroxyapatite. To improve the tissue-inducibility of hydrogels, transforming growth factor β3 is immobilized in the upper layer to induce cartilage regeneration, while bone morphogenetic protein 2 is immobilized in the lower layer to induce bone regeneration. Bone and cartilage repair performance of the hydrogel is examined by implantation into a full-thickness cartilage defect of a rabbit knee joint. The bilayer-structure hydrogel promotes regeneration of osteochondral tissue, thus providing a new option for repair of osteochondral defects.
Collapse
Affiliation(s)
- Donglin Gan
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Zhixiong Wang
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Chaoming Xie
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xiao Wang
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Wensi Xing
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xiang Ge
- Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, School of Mechanical Engineering, Tianjin University, Tianjin, 300354, China
| | - Huipin Yuan
- College of Physical Science and Technology, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Kefeng Wang
- National Engineering Research Center for Biomaterials, Genome Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Hui Tan
- Health Science Center, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, No. 3002, Sungang West Road, Futian District, Shenzhen, 518035, China
| | - Xiong Lu
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
38
|
Liao Y, He Q, Zhou F, Zhang J, Liang R, Yao X, Bunpetch V, Li J, Zhang S, Ouyang H. Current Intelligent Injectable Hydrogels for In Situ Articular Cartilage Regeneration. POLYM REV 2019. [DOI: 10.1080/15583724.2019.1683028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Youguo Liao
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiulin He
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Feifei Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingwei Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xudong Yao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute & School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajin Li
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Hongwei Ouyang
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute & School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
39
|
Patel M, Lee HJ, Son S, Kim H, Kim J, Jeong B. Iron Ion-Releasing Polypeptide Thermogel for Neuronal Differentiation of Mesenchymal Stem Cells. Biomacromolecules 2019; 21:143-151. [DOI: 10.1021/acs.biomac.9b01096] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Seungyi Son
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Heeju Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Jinheung Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| |
Collapse
|
40
|
Zhuang Y, Yang X, Li Y, Chen Y, Peng X, Yu L, Ding J. Sustained Release Strategy Designed for Lixisenatide Delivery to Synchronously Treat Diabetes and Associated Complications. ACS APPLIED MATERIALS & INTERFACES 2019; 11:29604-29618. [PMID: 31361112 DOI: 10.1021/acsami.9b10346] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Diabetes and its complications have become a global challenge of public health. Herein, we aimed to develop a long-acting delivery system of lixisenatide (Lixi), a glucose-dependent antidiabetic peptide, based on an injectable hydrogel for the synchronous treatment of type 2 diabetes mellitus (T2DM) and associated complications. Two triblock copolymers, poly(ε-caprolactone-co-glycolic acid)-poly(ethylene glycol)-poly(ε-caprolactone-co-glycolic acid) and poly(d,l-lactic acid-co-glycolic acid)-poly(ethylene glycol)-poly(d,l-lactic acid-co-glycolic acid) possessing temperature-induced sol-gel transitions, were synthesized by us. Compared to the two single-component hydrogels, their 1/1 mixture hydrogel not only maintained the temperature-induced gelation but also exhibited a steadier degradation profile in vivo. Both in vitro and in vivo release studies demonstrated that the mixture hydrogel provided the sustained release of Lixi for up to 9 days, which was attributed to balanced electrostatic interactions between the positive charges in the peptide and the negative charges in the polymer carrier. The hypoglycemic efficacy of Lixi delivered from the mixture hydrogel after a single subcutaneous injection into diabetic db/db mice was comparable to that of twice-daily administrations of Lixi solution for up to 9 days. Furthermore, three successive administrations of the abovementioned gel system within a month significantly increased the plasma insulin level, lowered glycosylated hemoglobin, and improved the pancreatic function of the animals. These results were superior or equivalent to those of twice-daily injections of Lixi solution for 30 days, but the number of injections was markedly reduced from 60 to 3. Finally, an improvement in hyperlipidemia, augmentation of nerve fiber density, and enhancement of motor nerve conduction velocity in the gel formulation-treated db/db mice indicated that the sustained delivery of Lixi arrested and even ameliorated diabetic complications. These findings suggested that the Lixi-loaded mixture hydrogel has great potential for the treatment of T2DM with significant improvements in the health and quality of life of patients.
Collapse
Affiliation(s)
- Yaping Zhuang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Xiaowei Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Yamin Li
- Department of Orthopaedic Surgery , Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai 200233 , China
| | - Yipei Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Xiaochun Peng
- Department of Orthopaedic Surgery , Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai 200233 , China
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| |
Collapse
|
41
|
Oh SY, Choi YM, Kim HY, Park YS, Jung SC, Park JW, Woo SY, Ryu KH, Kim HS, Jo I. Application of Tonsil-Derived Mesenchymal Stem Cells in Tissue Regeneration: Concise Review. Stem Cells 2019; 37:1252-1260. [PMID: 31287931 PMCID: PMC6852396 DOI: 10.1002/stem.3058] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/23/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022]
Abstract
Since the discovery of stem cells and multipotency characteristics of mesenchymal stem cells (MSCs), there has been tremendous development in regenerative medicine. MSCs derived from bone marrow have been widely used in various research applications, yet there are limitations such as invasiveness of obtaining samples, low yield and proliferation rate, and questions regarding their practicality in clinical applications. Some have suggested that MSCs from other sources, specifically those derived from palatine tonsil tissues, that is, tonsil‐derived MSCs (TMSCs), could be considered as a new potential therapeutic tool in regenerative medicine due to their superior proliferation rate and differentiation capabilities with low immunogenicity and ease of obtaining. Several studies have determined that TMSCs have differentiation potential not only into the mesodermal lineage but also into the endodermal as well as ectodermal lineages, expanding their potential usage and placing them as an appealing option to consider for future studies in regenerative medicine. In this review, the differentiation capacities of TMSCs and their therapeutic competencies from past studies are addressed. stem cells2019;37:1252–1260
Collapse
Affiliation(s)
- Se-Young Oh
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Young Min Choi
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ha Yeong Kim
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Yoon Shin Park
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Sung-Chul Jung
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Joo-Won Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Han Su Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
42
|
Abstract
Layered double hydroxides (LDHs) are an emergent class of biocompatible inorganic lamellar nanomaterials that have attracted significant research interest owing to their high surface-to-volume ratio, the capability to accumulate specific molecules, and the timely release to targets. Their unique properties have been employed for applications in organic catalysis, photocatalysis, sensors, drug delivery, and cell biology. Given the widespread contemporary interest in these topics, time-to-time it urges to review the recent progresses. This review aims to summarize the most recent cutting-edge reports appearing in the last years. It firstly focuses on the application of LDHs as catalysts in relevant chemical reactions and as photocatalysts for organic molecule degradation, water splitting reaction, CO2 conversion, and reduction. Subsequently, the emerging role of these materials in biological applications is discussed, specifically focusing on their use as biosensors, DNA, RNA, and drug delivery, finally elucidating their suitability as contrast agents and for cellular differentiation. Concluding remarks and future prospects deal with future applications of LDHs, encouraging researches in better understanding the fundamental mechanisms involved in catalytic and photocatalytic processes, and the molecular pathways that are activated by the interaction of LDHs with cells in terms of both uptake mechanisms and nanotoxicology effects.
Collapse
|
43
|
Jr da Costa Fernandes C, Pinto TS, Kang HR, de Magalhães Padilha P, Koh IHJ, Constantino VRL, Zambuzzi WF. Layered Double Hydroxides Are Promising Nanomaterials for Tissue Bioengineering Application. ACTA ACUST UNITED AC 2019; 3:e1800238. [PMID: 32648675 DOI: 10.1002/adbi.201800238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 05/01/2019] [Indexed: 01/07/2023]
Abstract
Layered double hydroxides (LDHs) have emerged as promising nanomaterials for human health and although it has achieved some progress on this matter, their application within bioengineering is not fully addressed. This prompted to subject fibroblasts to two compositions of LDHs (Mg2 Al-Cl and Zn2 Al-Cl), considering an acute response. First, LDH particles are addressed by scanning electron microscopy, and no significant effect of the cell culture medium on the shape of LDHs particles is reported although it seems to adsorb some soluble proteins as proposed by energy-dispersive X-ray analysis. These LDHs release magnesium, zinc, and aluminum, but there is no cytotoxic or biocompatibility effects. The data show interference to fibroblast adhesion by driving the reorganization of actin-based cytoskeleton, preliminarily to cell cycle progression. Additionally, these molecular findings are validated by performing a functional wound-healing assay, which is accompanied by a dynamic extracellular matrix remodeling in response to the LDHs. Altogether, the results show that LDHs nanomaterials modulate cell adhesion, proliferation, and migration, delineating new advances on the biomaterial field applied in the context of soft tissue bioengineering, which must be explored in health disorders, such as wound healing in burn injuries.
Collapse
Affiliation(s)
- Célio Jr da Costa Fernandes
- Lab. de Bioensaios e Dinâmica Celular, Departamento de Química e Bioquímica, Instituto de Biociências, Universidade Estadual Paulista - UNESP, Campus Botucatu, São Paulo, CEP 18618-970, Brazil
| | - Thaís Silva Pinto
- Lab. de Bioensaios e Dinâmica Celular, Departamento de Química e Bioquímica, Instituto de Biociências, Universidade Estadual Paulista - UNESP, Campus Botucatu, São Paulo, CEP 18618-970, Brazil
| | - Ha Ram Kang
- Lab. de Bioensaios e Dinâmica Celular, Departamento de Química e Bioquímica, Instituto de Biociências, Universidade Estadual Paulista - UNESP, Campus Botucatu, São Paulo, CEP 18618-970, Brazil
| | - Pedro de Magalhães Padilha
- Lab. de Bioensaios e Dinâmica Celular, Departamento de Química e Bioquímica, Instituto de Biociências, Universidade Estadual Paulista - UNESP, Campus Botucatu, São Paulo, CEP 18618-970, Brazil
| | - Ivan Hong Jun Koh
- Departamento de Cirurgia, Universidade Federal de São Paulo-UNIFESP, Rua Botucatu 740, CEP 04023-900, São Paulo, SP, Brazil
| | - Vera Regina Leopoldo Constantino
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo-USP, Av. Prof. Lineu Prestes 748, CEP 05508-000, São Paulo, SP, Brazil
| | - Willian F Zambuzzi
- Lab. de Bioensaios e Dinâmica Celular, Departamento de Química e Bioquímica, Instituto de Biociências, Universidade Estadual Paulista - UNESP, Campus Botucatu, São Paulo, CEP 18618-970, Brazil
| |
Collapse
|
44
|
Fu J, Wu B, Wei M, Huang Y, Zhou Y, Zhang Q, Du L. Prussian blue nanosphere-embedded in situ hydrogel for photothermal therapy by peritumoral administration. Acta Pharm Sin B 2019; 9:604-614. [PMID: 31193840 PMCID: PMC6543023 DOI: 10.1016/j.apsb.2018.12.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/23/2018] [Accepted: 10/30/2018] [Indexed: 12/03/2022] Open
Abstract
To establish an injectable hydrogel containing Prussian blue (PB) nanospheres for photothermal therapy against cancer, PB nanospheres were prepared by one-pot synthesis and the thermosensitive Pluronic F127 was used as the hydrogel matrix. The PB nanospheres and the hydrogel were characterized by shape, particle size, serum stability, photothermal performance upon repeated 808 nm laser irradiation, as well as the rheological features. The effect of the PB nanospheres and the hydrogel were evaluated qualitatively and quantitatively in 4T1 mouse breast cancer cells. The retention, photothermal efficacy, therapeutic effects and systemic toxicity of the hydrogel were assessed in a tumor-bearing mouse model. The PB nanospheres had a diameter of about 150 nm and exhibited satisfactory serum stability, photo-heat convert ability and repeated laser exposure stability. The hydrogel encapsulation did not negatively influence the above features of the photothermal agent. The nanosphere-containing hydrogel showed a phase transition at body temperature and, as a result, a long retention time in vivo. The photothermal agent-embedded hydrogel displayed promising photothermal therapeutic effects in the tumor-bearing mouse model with little-to-no systemic toxicity after peritumoral administration.
Collapse
Affiliation(s)
- Jijun Fu
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Bo Wu
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
- Center of Pharmaceutical Research and Development, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Minyan Wei
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yugang Huang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yi Zhou
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510700, China
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Qiang Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Lingran Du
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
45
|
Tang C, Holt BD, Wright ZM, Arnold AM, Moy AC, Sydlik SA. Injectable amine functionalized graphene and chondroitin sulfate hydrogel with potential for cartilage regeneration. J Mater Chem B 2019; 7:2442-2453. [PMID: 32255121 DOI: 10.1039/c8tb02967a] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Damaged cartilage does not readily heal and often requires surgical intervention that only modestly improves outcomes. A synthetic material that could be injected and covalently crosslinked in situ to form a bioactive, mechanically robust scaffold that promotes stem cell chondrogenic differentiation holds promise for next-generation treatment of cartilage lesions. Here, Johnson-Claisen rearrangement chemistry was performed on graphene oxide (GO) to enable functionalization with a primary amine covalently bound to the graphenic backbone through a chemically stable linker. The primary amines are used to form covalent crosslinks with chondroitin sulfate, an important component of cartilage that promotes regeneration, to form a hydrogel (EDAG-CS). The EDAG-CS system gels in situ within 10 min, and the graphenic component imparts improved mechanical properties, including stiffness (320% increase) and toughness (70% increase). EDAG-CS hydrogels are highly porous, resistant to degradation, and enable the growth of human mesenchymal stem cells and their deposition of collagen matrix. This system has potential to improve clinical outcomes of patients with cartilage damage.
Collapse
Affiliation(s)
- Caoxin Tang
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Patel M, Lee HJ, Kwon OH, Jeong B. Polypeptide Thermogel-Filled Silk Tube as a Bioactive Nerve Conduit. ACS APPLIED BIO MATERIALS 2019; 2:1967-1974. [DOI: 10.1021/acsabm.9b00026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Oh Hyeong Kwon
- Department of Polymer Science and Engineering, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi, Gyeongbuk 39177, Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| |
Collapse
|
47
|
Zhang Y, Yu J, Ren K, Zuo J, Ding J, Chen X. Thermosensitive Hydrogels as Scaffolds for Cartilage Tissue Engineering. Biomacromolecules 2019; 20:1478-1492. [PMID: 30843390 DOI: 10.1021/acs.biomac.9b00043] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Yanbo Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, P. R. China
| | - Jiakuo Yu
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, 49 Huayuanbei Road, Beijing 100191, P. R. China
| | - Kaixuan Ren
- Mork Family Department of Chemical Engineering & Materials Science, University of Southern California, 925 West 34th Street, Los Angeles, California 90089, United States of America
| | - Jianlin Zuo
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, P. R. China
| | - Jianxun Ding
- Key Laboratory
of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, 5625 Renmin Street, Changchun 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory
of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, 5625 Renmin Street, Changchun 130022, P. R. China
| |
Collapse
|
48
|
Piao H, Kim MH, Cui M, Choi G, Choy JH. Alendronate-Anionic Clay Nanohybrid for Enhanced Osteogenic Proliferation and Differentiation. J Korean Med Sci 2019; 34:e37. [PMID: 30718990 PMCID: PMC6356027 DOI: 10.3346/jkms.2019.34.e37] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/13/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Alendronate (AL), a drug for inhibiting osteoclast-mediated bone-resorption, was intercalated into an inorganic drug delivery nanovehicle, layered double hydroxide (LDH), to form a new nanohybrid, AL-LDH, with 1:1 heterostructure along the crystallographic C-axis. Based on the intercalation reaction strategy, the present AL-LDH drug delivery system (DDS) was realized with an enhanced drug efficacy of AL, which was confirmed by the improved proliferation and osteogenic differentiation of osteoblast-like cells (MG63). METHODS The AL-LDH nanohybrid was synthesized by conventional ion-exchange reaction and characterized by powder X-ray diffraction (PXRD), high-resolution transmission electron microscopy (HR-TEM), and Fourier transform infrared (FT-IR) spectroscopy. Additionally, in vitro efficacy tests, such as cell proliferation and alkaline phosphatase (ALP) activity, were analyzed. RESULTS The AL was successfully intercalated into LDH via ion-exchange reaction, and thus prepared AL-LDH DDS was X-ray single phasic and chemically well defined. The accumulated AL content in MG63 cells treated with the AL-LDH DDS nanoparticles was determined to be 10.6-fold higher than that within those treated with the intact AL after incubation for 1 hour, suggesting that intercellular permeation of AL was facilitated thanks to the hybridization with drug delivery vehicle, LDH. Furthermore, both in vitro proliferation level and ALP activity of MG63 treated with the present hybrid drug, AL-LDH, were found to be much more enhanced than those treated with the intact AL. This is surely due to the fact that LDH could deliver AL drug very efficiently, although LDH itself does not show any effect on proliferation and osteogenic differentiation of MG63 cells. CONCLUSION The present AL-LDH could be considered as a promising DDS for improving efficacy of AL.
Collapse
Affiliation(s)
- Huiyan Piao
- Department of Chemistry and Nanoscience, Center for Intelligent Nano-Bio Materials, Ewha Womans University, Seoul, Korea
| | - Myung Hun Kim
- Department of Chemistry and Nanoscience, Center for Intelligent Nano-Bio Materials, Ewha Womans University, Seoul, Korea
| | - Meiling Cui
- Department of Chemistry and Nanoscience, Center for Intelligent Nano-Bio Materials, Ewha Womans University, Seoul, Korea
| | - Goeun Choi
- Department of Chemistry and Nanoscience, Center for Intelligent Nano-Bio Materials, Ewha Womans University, Seoul, Korea
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Korea
| | - Jin-Ho Choy
- Department of Chemistry and Nanoscience, Center for Intelligent Nano-Bio Materials, Ewha Womans University, Seoul, Korea
| |
Collapse
|
49
|
Teng L, Chen Y, Jia YG, Ren L. Supramolecular and dynamic covalent hydrogel scaffolds: from gelation chemistry to enhanced cell retention and cartilage regeneration. J Mater Chem B 2019; 7:6705-6736. [DOI: 10.1039/c9tb01698h] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review highlights the most recent progress in gelation strategies of biomedical supramolecular and dynamic covalent crosslinking hydrogels and their applications for enhancing cell retention and cartilage regeneration.
Collapse
Affiliation(s)
- Lijing Teng
- School of Medicine
- South China University of Technology
- Guangzhou 510006
- China
- National Engineering Research Center for Tissue Restoration and Reconstruction
| | - Yunhua Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou 510006
- China
- School of Materials Science and Engineering
| | - Yong-Guang Jia
- National Engineering Research Center for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou 510006
- China
- School of Materials Science and Engineering
| | - Li Ren
- National Engineering Research Center for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou 510006
- China
- School of Materials Science and Engineering
| |
Collapse
|
50
|
Patel M, Park S, Lee HJ, Jeong B. Polypeptide Thermogels as Three-Dimensional Scaffolds for Cells. Tissue Eng Regen Med 2018; 15:521-530. [PMID: 30603576 PMCID: PMC6171707 DOI: 10.1007/s13770-018-0148-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Thermogel is an aqueous solution that exhibits a sol-to-gel transition as the temperature increases. Stem cells, growth factors, and differentiating factors can be incorporated in situ in the matrix during the sol-to-gel transition, leading to the formation of a three-dimensional (3D) cell-culture scaffold. METHODS The uses of thermogelling polypeptides, such as collagen, Matrigel™, elastin-like polypeptides, and synthetic polypeptides, as 3D scaffolds of cells, are summarized in this paper. RESULTS The timely supply of growth factors to the cells, cell survival, and metabolite removal is to be insured in the cell culture matrix. Various growth factors were incorporated in the matrix during the sol-to-gel transition of the thermogelling polypeptide aqueous solutions, and preferential differentiation of the incorporated stem cells into specific target cells were investigated. In addition, modulus of the matrix was controlled by post-crosslinking reactions of thermogels or employing composite systems. Chemical functional groups as well as biological factors were selected appropriately for targeted differentiation of the incorporated stem cells. CONCLUSION In addition to all the advantages of thermogels including mild conditions for cell-incorporation and controlled supplies of the growth factors, polypeptide thermogels provide neutral pH environments to the cells during the degradation of the gel. Polypeptide thermogels as an injectable scaffold can be a promising system for their eventual in vivo applications in stem cell therapy.
Collapse
Affiliation(s)
- Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760 Korea
| | - Sohee Park
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760 Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760 Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760 Korea
| |
Collapse
|