1
|
Zheng F, Tian R, Lu H, Liang X, Shafiq M, Uchida S, Chen H, Ma M. Droplet Microfluidics Powered Hydrogel Microparticles for Stem Cell-Mediated Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401400. [PMID: 38881184 DOI: 10.1002/smll.202401400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/21/2024] [Indexed: 06/18/2024]
Abstract
Stem cell-related therapeutic technologies have garnered significant attention of the research community for their multi-faceted applications. To promote the therapeutic effects of stem cells, the strategies for cell microencapsulation in hydrogel microparticles have been widely explored, as the hydrogel microparticles have the potential to facilitate oxygen diffusion and nutrient transport alongside their ability to promote crucial cell-cell and cell-matrix interactions. Despite their significant promise, there is an acute shortage of automated, standardized, and reproducible platforms to further stem cell-related research. Microfluidics offers an intriguing platform to produce stem cell-laden hydrogel microparticles (SCHMs) owing to its ability to manipulate the fluids at the micrometer scale as well as precisely control the structure and composition of microparticles. In this review, the typical biomaterials and crosslinking methods for microfluidic encapsulation of stem cells as well as the progress in droplet-based microfluidics for the fabrication of SCHMs are outlined. Moreover, the important biomedical applications of SCHMs are highlighted, including regenerative medicine, tissue engineering, scale-up production of stem cells, and microenvironmental simulation for fundamental cell studies. Overall, microfluidics holds tremendous potential for enabling the production of diverse hydrogel microparticles and is worthy for various stem cell-related biomedical applications.
Collapse
Affiliation(s)
- Fangqiao Zheng
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
| | - Ruizhi Tian
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hongxu Lu
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiao Liang
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
| | - Muhammad Shafiq
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510, Japan
| | - Hangrong Chen
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ming Ma
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
2
|
Aqel S, Al-Thani N, Haider MZ, Abdelhady S, Al Thani AA, Kobeissy F, Shaito AA. Biomaterials in Traumatic Brain Injury: Perspectives and Challenges. BIOLOGY 2023; 13:21. [PMID: 38248452 PMCID: PMC10813103 DOI: 10.3390/biology13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and long-term impairment globally. TBI has a dynamic pathology, encompassing a variety of metabolic and molecular events that occur in two phases: primary and secondary. A forceful external blow to the brain initiates the primary phase, followed by a secondary phase that involves the release of calcium ions (Ca2+) and the initiation of a cascade of inflammatory processes, including mitochondrial dysfunction, a rise in oxidative stress, activation of glial cells, and damage to the blood-brain barrier (BBB), resulting in paracellular leakage. Currently, there are no FDA-approved drugs for TBI, but existing approaches rely on delivering micro- and macromolecular treatments, which are constrained by the BBB, poor retention, off-target toxicity, and the complex pathology of TBI. Therefore, there is a demand for innovative and alternative therapeutics with effective delivery tactics for the diagnosis and treatment of TBI. Tissue engineering, which includes the use of biomaterials, is one such alternative approach. Biomaterials, such as hydrogels, including self-assembling peptides and electrospun nanofibers, can be used alone or in combination with neuronal stem cells to induce neurite outgrowth, the differentiation of human neural stem cells, and nerve gap bridging in TBI. This review examines the inclusion of biomaterials as potential treatments for TBI, including their types, synthesis, and mechanisms of action. This review also discusses the challenges faced by the use of biomaterials in TBI, including the development of biodegradable, biocompatible, and mechanically flexible biomaterials and, if combined with stem cells, the survival rate of the transplanted stem cells. A better understanding of the mechanisms and drawbacks of these novel therapeutic approaches will help to guide the design of future TBI therapies.
Collapse
Affiliation(s)
- Sarah Aqel
- Medical Research Center, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
| | - Najlaa Al-Thani
- Research and Development Department, Barzan Holdings, Doha P.O. Box 7178, Qatar
| | - Mohammad Z. Haider
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria 21544, Egypt;
| | - Asmaa A. Al Thani
- Biomedical Research Center and Department of Biomedical Sciences, College of Health Science, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310, USA
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
3
|
Wang S, Wang Y, Chen B, Zhao M, Song G, Wang J, Xu J. Preparation and performance study of multichannel PLA artificial nerve conduits. Biomed Mater 2023; 18:065001. [PMID: 37582380 DOI: 10.1088/1748-605x/acf0ae] [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: 04/18/2023] [Accepted: 08/15/2023] [Indexed: 08/17/2023]
Abstract
Compared with single-channel nerve conduits, multichannel artificial nerve conduits are more beneficial for repairing damaged peripheral nerves of long-distance nerve defects. Multichannel nerve conduits can be fabricated by the mold method and the electrospinning method but with disadvantages such as low strength and large differences in batches, while the braiding method can solve this problem. In this study, polylactic acid yarns were used as the braiding yarn, and the number of spindles during braiding was varied to achieve 4, 5, 6, 7 and 8 multichannel artificial nerve conduits. A mathematical model of the number of braiding yarn spindles required to meet certain size specification parameters of the multichannel conduit was established. The cross-sectional morphology and mechanical properties of the conduits were characterized by scanning electron microscopy observation and mechanical testing; the results showed that the multichannel structure was well constructed; the tensile strength of the multichannel conduit was more than 30 times that of the rabbit tibial nerve. The biocompatibility of the conduit was tested; thein vitrocell culture results proved that the braided multichannel nerve conduits were nontoxic to Schwann cells, and the cell adhesion and proliferation were optimal in the 4-channel conduit among the multichannel conduits, which was close to the single-channel conduit.
Collapse
Affiliation(s)
- Shanlong Wang
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215021, People's Republic of China
| | - Yuyu Wang
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215021, People's Republic of China
| | - Biling Chen
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215021, People's Republic of China
| | - Mingda Zhao
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215021, People's Republic of China
| | - Gongji Song
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215021, People's Republic of China
| | - Jiannan Wang
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215021, People's Republic of China
- Key Laboratory of Textile Industry for Silk Products in Medical and Health Use, Soochow University, Suzhou 215127, People's Republic of China
| | - Jianmei Xu
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215021, People's Republic of China
- Key Laboratory of Textile Industry for Silk Products in Medical and Health Use, Soochow University, Suzhou 215127, People's Republic of China
| |
Collapse
|
4
|
Huang X, An Y, Yuan S, Chen C, Shan H, Zhang M. Silk fibroin carriers with sustained release capacity for treating neurological diseases. Front Pharmacol 2023; 14:1117542. [PMID: 37214477 PMCID: PMC10196044 DOI: 10.3389/fphar.2023.1117542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Neurological diseases such as traumatic brain injury, cerebral ischemia, Parkinson's, and Alzheimer's disease usually occur in the central and peripheral nervous system and result in nervous dysfunction, such as cognitive impairment and motor dysfunction. Long-term clinical intervention is necessary for neurological diseases where neural stem cell transplantation has made substantial progress. However, many risks remain for cell therapy, such as puncture bleeding, postoperative infection, low transplantation success rate, and tumor formation. Sustained drug delivery, which aims to maintain the desired steady-state drug concentrations in plasma or local injection sites, is considered as a feasible option to help overcome side effects and improve the therapeutic efficiency of drugs on neurological diseases. Natural polymers such as silk fibroin have excellent biocompatibility, which can be prepared for various end-use material formats, such as microsphere, gel, coating/film, scaffold/conduit, microneedle, and enables the dynamic release of loaded drugs to achieve a desired therapeutic response. Sustained-release drug delivery systems are based on the mechanism of diffusion and degradation by altering the structures of silk fibroin and drugs, factors, and cells, which can induce nerve recovery and restore the function of the nervous system in a slow and persistent manner. Based on these desirable properties of silk fibroin as a carrier with sustained-release capacity, this paper discusses the role of various forms of silk fibroin-based drug delivery materials in treating neurological diseases in recent years.
Collapse
Affiliation(s)
- Xinqi Huang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yumei An
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Shengye Yuan
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Chen Chen
- Department of Orthopedics, Dongtai People’s Hospital, Dongtai, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Mingyang Zhang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| |
Collapse
|
5
|
Du L, Qin C, Zhang H, Han F, Xue J, Wang Y, Wu J, Xiao Y, Huan Z, Wu C. Multicellular Bioprinting of Biomimetic Inks for Tendon-to-Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2301309. [PMID: 37119499 PMCID: PMC10375072 DOI: 10.1002/advs.202301309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Indexed: 06/19/2023]
Abstract
Tendon-to-bone interface has a hierarchical structure and gradient component that are conducive to distributing the stresses to achieve movement. Conventional biomaterials lack the capacity to induce synchronous repair of multiple tissues, resulting in the failure of the interface repair. Biomimetic strategies have attracted enormous attention in the field of complex structure regeneration because they can meet the different physiological requirements of multiple tissues. Herein, a biomimetic ink mimicking tendon/bone tissues is developed by combining tendon/bone-related cells and Mo-containing silicate (MS) bioceramics. Subsequently, biomimetic multicellular scaffolds are fabricated to achieve the simulation of the hierarchical structure and cellular composition of tendon-to-bone interfaces by the spatial distribution of the biomimetic inks via 3D bioprinting, which is of great significance for inducing the regeneration of complex structures in the interface region. In addition, attributed to the desirable ionic microenvironment created by MS bioceramics, the biomimetic scaffolds possess the dual function of inducing tendon/bone-related cells tenogenic and osteogenic differentiation in vitro, and promote the integrated regeneration of tendon-to-bone interfaces in vivo. The study offers a feasible strategy to construct biomimetic multicellular scaffolds with bifunction for inducing multi-lineage tissue regeneration, especially for regenerating soft-to-hard tissue interfaces.
Collapse
Affiliation(s)
- Lin Du
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Chen Qin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Hongjian Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Fei Han
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Jianmin Xue
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Yufeng Wang
- Nanjing First Hospital, Nanjing Medical University, 68th Changle Road, Nanjing, Jiangsu, 210006, P. R. China
| | - Jinfu Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Yin Xiao
- School of Medicine and Dentistry, Menzies Health Institute Queensland, Griffith University, Queensland, 4222, Australia
| | - Zhiguang Huan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, P. R. China
| |
Collapse
|
6
|
Pourkhodadad S, Hosseinkazemi H, Bonakdar S, Nekounam H. Biomimetic engineered approaches for neural tissue engineering: Spinal cord injury. J Biomed Mater Res B Appl Biomater 2023; 111:701-716. [PMID: 36214332 DOI: 10.1002/jbm.b.35171] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/16/2022] [Accepted: 09/03/2022] [Indexed: 01/21/2023]
Abstract
The healing process for spinal cord injuries is complex and presents many challenges. Current advances in nerve regeneration are based on promising tissue engineering techniques, However, the chances of success depend on better mimicking the extracellular matrix (ECM) of neural tissue and better supporting neurons in a three-dimensional environment. The ECM provides excellent biological conditions, including desirable morphological features, electrical conductivity, and chemical compositions for neuron attachment, proliferation and function. This review outlines the rationale for developing a construct for neuron regrowth in spinal cord injury using appropriate biomaterials and scaffolding techniques.
Collapse
Affiliation(s)
| | - Hessam Hosseinkazemi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Houra Nekounam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Lawal SK, Olojede SO, Faborode OS, Aladeyelu OS, Matshipi MN, Sulaiman SO, Naidu ECS, Rennie CO, Azu OO. Nanodelivery of antiretroviral drugs to nervous tissues. Front Pharmacol 2022; 13:1025160. [DOI: 10.3389/fphar.2022.1025160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/25/2022] [Indexed: 11/10/2022] Open
Abstract
Despite the development of effective combined antiretroviral therapy (cART), the neurocognitive impairments associated with human immunodeficiency virus (HIV) remain challenging. The presence of the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCFB) impedes the adequate penetration of certain antiretroviral drugs into the brain. In addition, reports have shown that some antiretroviral drugs cause neurotoxicity resulting from their interaction with nervous tissues due to long-term systemic exposure. Therefore, the research into the effective therapeutic modality that would cater for the HIV-associated neurocognitive disorders (HAND) and ART toxicity is now receiving broad research attention. Thus, this review explores the latest information in managing HAND using a nanoparticle drug delivery system (NDDS). We discussed the neurotoxicity profile of various approved ART. Also, we explained the applications of silver nanoparticles (AgNPs) in medicine, their different synthesis methods and their interaction with nervous tissues. Lastly, while proposing AgNPs as useful nanoparticles in properly delivering ART to enhance effectiveness and minimize neurocognitive disorders, we hypothesize that the perceived toxicity of AgNPs could be minimized by taking appropriate precautions. One such precaution is using appropriate reducing and stabilizing agents such as trisodium citrate to reduce silver ion Ag + to ground state Ag0 during the synthesis. Also, the usage of medium-sized, spherical-shaped AgNPs is encouraged in AgNPs-based drug delivery to the brain due to their ability to deliver therapeutic agents across BBB. In addition, characterization and functionalization of the synthesized AgNPs are required during the drug delivery approach. Putting all these factors in place would minimize toxicity and enhance the usage of AgNPs in delivering therapeutic agents across the BBB to the targeted brain tissue and could cater for the HIV-associated neurocognitive disorders and neurotoxic effects of antiretroviral drugs (ARDs).
Collapse
|
8
|
Liu T, Zhu W, Zhang X, He C, Liu X, Xin Q, Chen K, Wang H. Recent Advances in Cell and Functional Biomaterial Treatment for Spinal Cord Injury. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5079153. [PMID: 35978649 PMCID: PMC9377911 DOI: 10.1155/2022/5079153] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is a devastating central nervous system disease caused by accidental events, resulting in loss of sensory and motor function. Considering the multiple effects of primary and secondary injuries after spinal cord injury, including oxidative stress, tissue apoptosis, inflammatory response, and neuronal autophagy, it is crucial to understand the underlying pathophysiological mechanisms, local microenvironment changes, and neural tissue functional recovery for preparing novel treatment strategies. Treatment based on cell transplantation has become the forefront of spinal cord injury therapy. The transplanted cells provide physical and nutritional support for the damaged tissue. At the same time, the implantation of biomaterials with specific biological functions at the site of the SCI has also been proved to improve the local inhibitory microenvironment and promote axonal regeneration, etc. The combined transplantation of cells and functional biomaterials for SCI treatment can result in greater neuroprotective and regenerative effects by regulating cell differentiation, enhancing cell survival, and providing physical and directional support for axon regeneration and neural circuit remodeling. This article reviews the pathophysiology of the spinal cord, changes in the microenvironment after injury, and the mechanisms and strategies for spinal cord regeneration and repair. The article will focus on summarizing and discussing the latest intervention models based on cell and functional biomaterial transplantation and the latest progress in combinational therapies in SCI repair. Finally, we propose the future prospects and challenges of current treatment regimens for SCI repair, to provide references for scientists and clinicians to seek better SCI repair strategies in the future.
Collapse
Affiliation(s)
- Tianyi Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Wenhao Zhu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xiaoyu Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Chuan He
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xiaolong Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Qiang Xin
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Kexin Chen
- Institute of Translational Medicine, First Hospital of Jilin University, Changchun 130021, China
| | - Haifeng Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
9
|
Advances in Fibrin-Based Materials in Wound Repair: A Review. Molecules 2022; 27:molecules27144504. [PMID: 35889381 PMCID: PMC9322155 DOI: 10.3390/molecules27144504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/28/2022] [Accepted: 07/08/2022] [Indexed: 11/29/2022] Open
Abstract
The first bioprocess that occurs in response to wounding is the deterrence of local hemorrhage. This is accomplished by platelet aggregation and initiation of the hemostasis cascade. The resulting blood clot immediately enables the cessation of bleeding and then functions as a provisional matrix for wound healing, which begins a few days after injury. Here, fibrinogen and fibrin fibers are the key players, because they literally serve as scaffolds for tissue regeneration and promote the migration of cells, as well as the ingrowth of tissues. Fibrin is also an important modulator of healing and a host defense system against microbes that effectively maintains incoming leukocytes and acts as reservoir for growth factors. This review presents recent advances in the understanding and applications of fibrin and fibrin-fiber-incorporated biomedical materials applied to wound healing and subsequent tissue repair. It also discusses how fibrin-based materials function through several wound healing stages including physical barrier formation, the entrapment of bacteria, drug and cell delivery, and eventual degradation. Pure fibrin is not mechanically strong and stable enough to act as a singular wound repair material. To alleviate this problem, this paper will demonstrate recent advances in the modification of fibrin with next-generation materials exhibiting enhanced stability and medical efficacy, along with a detailed look at the mechanical properties of fibrin and fibrin-laden materials. Specifically, fibrin-based nanocomposites and their role in wound repair, sustained drug release, cell delivery to wound sites, skin reconstruction, and biomedical applications of drug-loaded fibrin-based materials will be demonstrated and discussed.
Collapse
|
10
|
Wu Y, Rakotoarisoa M, Angelov B, Deng Y, Angelova A. Self-Assembled Nanoscale Materials for Neuronal Regeneration: A Focus on BDNF Protein and Nucleic Acid Biotherapeutic Delivery. NANOMATERIALS 2022; 12:nano12132267. [PMID: 35808102 PMCID: PMC9268293 DOI: 10.3390/nano12132267] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023]
Abstract
Enabling challenging applications of nanomedicine and precision medicine in the treatment of neurodegenerative disorders requires deeper investigations of nanocarrier-mediated biomolecular delivery for neuronal targeting and recovery. The successful use of macromolecular biotherapeutics (recombinant growth factors, antibodies, enzymes, synthetic peptides, cell-penetrating peptide–drug conjugates, and RNAi sequences) in clinical developments for neuronal regeneration should benefit from the recent strategies for enhancement of their bioavailability. We highlight the advances in the development of nanoscale materials for drug delivery in neurodegenerative disorders. The emphasis is placed on nanoformulations for the delivery of brain-derived neurotrophic factor (BDNF) using different types of lipidic nanocarriers (liposomes, liquid crystalline or solid lipid nanoparticles) and polymer-based scaffolds, nanofibers and hydrogels. Self-assembled soft-matter nanoscale materials show favorable neuroprotective characteristics, safety, and efficacy profiles in drug delivery to the central and peripheral nervous systems. The advances summarized here indicate that neuroprotective biomolecule-loaded nanoparticles and injectable hydrogels can improve neuronal survival and reduce tissue injury. Certain recently reported neuronal dysfunctions in long-COVID-19 survivors represent early manifestations of neurodegenerative pathologies. Therefore, BDNF delivery systems may also help in prospective studies on recovery from long-term COVID-19 neurological complications and be considered as promising systems for personalized treatment of neuronal dysfunctions and prevention or retarding of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yu Wu
- CNRS, Institut Galien Paris-Saclay, Université Paris-Saclay, F-92290 Châtenay-Malabry, France; (Y.W.); (M.R.)
| | - Miora Rakotoarisoa
- CNRS, Institut Galien Paris-Saclay, Université Paris-Saclay, F-92290 Châtenay-Malabry, France; (Y.W.); (M.R.)
| | - Borislav Angelov
- Institute of Physics, ELI Beamlines, Academy of Sciences of the Czech Republic, Na Slovance 2, CZ-18221 Prague, Czech Republic;
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, No. 1, Jinlian Road, Longwan District, Wenzhou 325001, China;
| | - Angelina Angelova
- CNRS, Institut Galien Paris-Saclay, Université Paris-Saclay, F-92290 Châtenay-Malabry, France; (Y.W.); (M.R.)
- Correspondence:
| |
Collapse
|
11
|
Moazamiyanfar R, Halabian R, Ghollasi M, Poormoghadam D, Entezari M, Endorami SE. Neural Differentiation of Human-Induced Pluripotent Stem Cells (hiPSc) on Surface-Modified Nanofibrous Scaffolds Coated with Platelet-Rich Plasma. Neurochem Res 2022; 47:1991-2001. [PMID: 35359243 DOI: 10.1007/s11064-022-03584-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 02/19/2022] [Accepted: 03/17/2022] [Indexed: 10/18/2022]
Abstract
The field of tissue engineering exploits living cells in a variety of ways to restore, maintain, or enhance tissues and organs. Between stem cells, human induced pluripotent stem cells (hiPSCs), are very important due to their wide abilities. Growth factors can support proliferation, differentiation, and migration of hiPSCs. Platelet-rich plasma (PRP) could be used as the source of growth factors for hiPSCs. In the present study, proliferation and neural differentiation of hiPSCs on surface-modified nanofibrous Poly-L-lactic acid (PLLA) coated with platelet-rich plasma was investigated. The results of in vitro analysis showed that on the surface, which was modified nanofibrous scaffolds coated with platelet-rich plasma, significantly enhanced hiPSCs proliferation and neural differentiation were observed. Whereas the MTT ([3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide]) results showed biocompatibility of surface-modified nanofibrous scaffolds coated with platelet-rich plasma and the usage of these modified nanoscaffolds in neural tissue engineering in vivo is promising for the future.
Collapse
Affiliation(s)
- Reza Moazamiyanfar
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Marzieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Delaram Poormoghadam
- Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Seyed Ehsan Endorami
- Immunogenetics Research Center, Department of Medical Biotechnology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
12
|
Mesenchymal Stromal Cells Preconditioning: A New Strategy to Improve Neuroprotective Properties. Int J Mol Sci 2022; 23:ijms23042088. [PMID: 35216215 PMCID: PMC8878691 DOI: 10.3390/ijms23042088] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Neurological diseases represent one of the main causes of disability in human life. Consequently, investigating new strategies capable of improving the quality of life in neurological patients is necessary. For decades, researchers have been working to improve the efficacy and safety of mesenchymal stromal cells (MSCs) therapy based on MSCs’ regenerative and immunomodulatory properties and multilinear differentiation potential. Therefore, strategies such as MSCs preconditioning are useful to improve their application to restore damaged neuronal circuits following neurological insults. This review is focused on preconditioning MSCs therapy as a potential application to major neurological diseases. The aim of our work is to summarize both the in vitro and in vivo studies that demonstrate the efficacy of MSC preconditioning on neuronal regeneration and cell survival as a possible application to neurological damage.
Collapse
|
13
|
Advanced approaches to regenerate spinal cord injury: The development of cell and tissue engineering therapy and combinational treatments. Biomed Pharmacother 2021; 146:112529. [PMID: 34906773 DOI: 10.1016/j.biopha.2021.112529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is a central nervous system (CNS) devastate event that is commonly caused by traumatic or non-traumatic events. The reinnervation of spinal cord axons is hampered through a myriad of devices counting on the damaged myelin, inflammation, glial scar, and defective inhibitory molecules. Unfortunately, an effective treatment to completely repair SCI and improve functional recovery has not been found. In this regard, strategies such as using cells, biomaterials, biomolecules, and drugs have been reported to be effective for SCI recovery. Furthermore, recent advances in combinatorial treatments, which address various aspects of SCI pathophysiology, provide optimistic outcomes for spinal cord regeneration. According to the global importance of SCI, the goal of this article review is to provide an overview of the pathophysiology of SCI, with an emphasis on the latest modes of intervention and current advanced approaches for the treatment of SCI, in conjunction with an assessment of combinatorial approaches in preclinical and clinical trials. So, this article can give scientists and clinicians' clues to help them better understand how to construct preclinical and clinical studies that could lead to a breakthrough in spinal cord regeneration.
Collapse
|
14
|
Siddiqui AM, Islam R, Cuellar CA, Silvernail JL, Knudsen B, Curley DE, Strickland T, Manske E, Suwan PT, Latypov T, Akhmetov N, Zhang S, Summer P, Nesbitt JJ, Chen BK, Grahn PJ, Madigan NN, Yaszemski MJ, Windebank AJ, Lavrov IA. Newly regenerated axons via scaffolds promote sub-lesional reorganization and motor recovery with epidural electrical stimulation. NPJ Regen Med 2021; 6:66. [PMID: 34671050 PMCID: PMC8528837 DOI: 10.1038/s41536-021-00176-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Here, we report the effect of newly regenerated axons via scaffolds on reorganization of spinal circuitry and restoration of motor functions with epidural electrical stimulation (EES). Motor recovery was evaluated for 7 weeks after spinal transection and following implantation with scaffolds seeded with neurotrophin producing Schwann cell and with rapamycin microspheres. Combined treatment with scaffolds and EES-enabled stepping led to functional improvement compared to groups with scaffold or EES, although, the number of axons across scaffolds was not different between groups. Re-transection through the scaffold at week 6 reduced EES-enabled stepping, still demonstrating better performance compared to the other groups. Greater synaptic reorganization in the presence of regenerated axons was found in group with combined therapy. These findings suggest that newly regenerated axons through cell-containing scaffolds with EES-enabled motor training reorganize the sub-lesional circuitry improving motor recovery, demonstrating that neuroregenerative and neuromodulatory therapies cumulatively enhancing motor function after complete SCI.
Collapse
Affiliation(s)
| | - Riazul Islam
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Carlos A Cuellar
- School of Sport Sciences, Universidad Anáhuac México, Campus Norte, Huixquilucan, State of Mexico, Mexico
| | | | - Bruce Knudsen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Dallece E Curley
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | | | - Emilee Manske
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Neuroscience, Scripps College, Claremont, CA, USA
| | - Parita T Suwan
- Paracelsus Medical Private University, Salzburg, Austria
| | - Timur Latypov
- Division of Brain, Imaging, and Behaviour - Systems Neuroscience, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nafis Akhmetov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Shuya Zhang
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Priska Summer
- Paracelsus Medical Private University, Salzburg, Austria
| | | | - Bingkun K Chen
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Peter J Grahn
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Igor A Lavrov
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
15
|
Rathnam C, Yang L, Castro-Pedrido S, Luo J, Cai L, Lee KB. Hybrid SMART spheroids to enhance stem cell therapy for CNS injuries. SCIENCE ADVANCES 2021; 7:eabj2281. [PMID: 34586845 PMCID: PMC8480929 DOI: 10.1126/sciadv.abj2281] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Although stem cell therapy holds enormous potential for treating debilitating injuries and diseases in the central nervous system, low survival and inefficient differentiation have restricted its clinical applications. Recently, 3D cell culture methods, such as stem cell–based spheroids and organoids, have demonstrated advantages by incorporating tissue-mimetic 3D cell-cell interactions. However, a lack of drug and nutrient diffusion, insufficient cell-matrix interactions, and tedious fabrication procedures have compromised their therapeutic effects in vivo. To address these issues, we developed a biodegradable nanomaterial-templated 3D cell assembly method that enables the formation of hybrid stem cell spheroids with deep drug delivery capabilities and homogeneous incorporation of 3D cell-matrix interactions. Hence, high survival rates, controlled differentiation, and functional recovery were demonstrated in a spinal cord injury animal model. Overall, our hybrid stem cell spheroids represent a substantial development of material-facilitated 3D cell culture systems and can pave the way for stem cell–based treatment of CNS injuries.
Collapse
Affiliation(s)
- Christopher Rathnam
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Letao Yang
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Sofia Castro-Pedrido
- Department of Biomedical Engineering Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jeffrey Luo
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Li Cai
- Department of Biomedical Engineering Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
16
|
Peressotti S, Koehl GE, Goding JA, Green RA. Self-Assembling Hydrogel Structures for Neural Tissue Repair. ACS Biomater Sci Eng 2021; 7:4136-4163. [PMID: 33780230 PMCID: PMC8441975 DOI: 10.1021/acsbiomaterials.1c00030] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
Hydrogel materials have been employed as biological scaffolds for tissue regeneration across a wide range of applications. Their versatility and biomimetic properties make them an optimal choice for treating the complex and delicate milieu of neural tissue damage. Aside from finely tailored hydrogel properties, which aim to mimic healthy physiological tissue, a minimally invasive delivery method is essential to prevent off-target and surgery-related complications. The specific class of injectable hydrogels termed self-assembling peptides (SAPs), provide an ideal combination of in situ polymerization combined with versatility for biofunctionlization, tunable physicochemical properties, and high cytocompatibility. This review identifies design criteria for neural scaffolds based upon key cellular interactions with the neural extracellular matrix (ECM), with emphasis on aspects that are reproducible in a biomaterial environment. Examples of the most recent SAPs and modification methods are presented, with a focus on biological, mechanical, and topographical cues. Furthermore, SAP electrical properties and methods to provide appropriate electrical and electrochemical cues are widely discussed, in light of the endogenous electrical activity of neural tissue as well as the clinical effectiveness of stimulation treatments. Recent applications of SAP materials in neural repair and electrical stimulation therapies are highlighted, identifying research gaps in the field of hydrogels for neural regeneration.
Collapse
Affiliation(s)
- Sofia Peressotti
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Gillian E. Koehl
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Josef A. Goding
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Rylie A. Green
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| |
Collapse
|
17
|
Ali MA, Bhuiyan MH. Types of biomaterials useful in brain repair. Neurochem Int 2021; 146:105034. [PMID: 33789130 DOI: 10.1016/j.neuint.2021.105034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/28/2021] [Accepted: 03/22/2021] [Indexed: 01/21/2023]
Abstract
Biomaterials is an emerging field in the study of brain tissue engineering and repair or neurogenesis. The fabrication of biomaterials that can replicate the mechanical and viscoelastic features required by the brain, including the poroviscoelastic responses, force dissipation, and solute diffusivity are essential to be mapped from the macro to the nanoscale level under physiological conditions in order for us to gain an effective treatment for neurodegenerative diseases. This research topic has identified a critical study gap that must be addressed, and that is to source suitable biomaterials and/or create reliable brain-tissue-like biomaterials. This chapter will define and discuss the various types of biomaterials, their structures, and their function-properties features which would enable the development of next-generation biomaterials useful in brain repair.
Collapse
Affiliation(s)
- M Azam Ali
- Center for Bioengineering and Nanomedicine, Faculty of Dentistry, Division of Health Sciences, University of Otago, Dunedin, New Zealand.
| | - Mozammel Haque Bhuiyan
- Center for Bioengineering and Nanomedicine, Faculty of Dentistry, Division of Health Sciences, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
18
|
Chowdhury S, Ghosh S. Nanoparticles and Stem Cells. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Abstract
Spinal cord injury results in significant loss of motor, sensory, and autonomic functions. Although a wide range of therapeutic agents have been shown to attenuate secondary injury or promote regeneration/repair in animal models of spinal cord injury, clinical translation of these strategies has been limited, in part due to difficulty in safely and effectively achieving therapeutic concentrations in the injured spinal cord tissue. Hydrogel-based drug delivery systems offer unique opportunities to locally deliver drugs to the injured spinal cord with sufficient dose and duration, while avoiding deleterious side effects associated with systemic drug administration. Such local drug delivery systems can be readily fabricated from biocompatible and biodegradable materials. In this review, hydrogel-based strategies for local drug delivery to the injured spinal cord are extensively reviewed, and recommendations are made for implementation.
Collapse
Affiliation(s)
- Robert B Shultz
- School of Biomedical Engineering, Science and Health Systems, Drexel University; Department of Neurosurgery; Department of Bioengineering, University of Pennsylvania; New Jersey Center for Biomaterials, Rutgers - The State University of New Jersey, Piscataway, NJ; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Yinghui Zhong
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
20
|
OʼShea TM, Wollenberg AL, Kim JH, Ao Y, Deming TJ, Sofroniew MV. Foreign body responses in mouse central nervous system mimic natural wound responses and alter biomaterial functions. Nat Commun 2020; 11:6203. [PMID: 33277474 PMCID: PMC7718896 DOI: 10.1038/s41467-020-19906-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/22/2020] [Indexed: 01/30/2023] Open
Abstract
Biomaterials hold promise for therapeutic applications in the central nervous system (CNS). Little is known about molecular factors that determine CNS foreign body responses (FBRs) in vivo, or about how such responses influence biomaterial function. Here, we probed these factors in mice using a platform of injectable hydrogels readily modified to present interfaces with different physiochemical properties to host cells. We found that biomaterial FBRs mimic specialized multicellular CNS wound responses not present in peripheral tissues, which serve to isolate damaged neural tissue and restore barrier functions. We show that the nature and intensity of CNS FBRs are determined by definable properties that significantly influence hydrogel functions, including resorption and molecular delivery when injected into healthy brain or stroke injuries. Cationic interfaces elicit stromal cell infiltration, peripherally derived inflammation, neural damage and amyloid production. Nonionic and anionic formulations show minimal levels of these responses, which contributes to superior bioactive molecular delivery. Our results identify specific molecular mechanisms that drive FBRs in the CNS and have important implications for developing effective biomaterials for CNS applications.
Collapse
Affiliation(s)
- Timothy M OʼShea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA
| | - Alexander L Wollenberg
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, 90095-1600, USA
| | - Jae H Kim
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA
| | - Yan Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA
| | - Timothy J Deming
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, 90095-1600, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095-1600, USA
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA.
| |
Collapse
|
21
|
Huang F, Chen T, Chang J, Zhang C, Liao F, Wu L, Wang W, Yin Z. A conductive dual-network hydrogel composed of oxidized dextran and hyaluronic-hydrazide as BDNF delivery systems for potential spinal cord injury repair. Int J Biol Macromol 2020; 167:434-445. [PMID: 33278434 DOI: 10.1016/j.ijbiomac.2020.11.206] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) often causes neuronal death and axonal degeneration. In this study, we report a new strategy for preparing injectable and conductive polysaccharides-based hydrogels that could sustainably deliver brain-derived neurotrophic factor (BDNF) for SCI repair. We used poly(lactic-co-glycolic acid) (PLGA) as a carrier to encapsulate BDNF. The resulting microspheres were then modified with tannic acid (TA). The polysaccharides-based hydrogel composed of oxidized dextran (Dex) and hyaluronic acid-hydrazide (HA) was mixed with TA-modified microspheres to form the ultimate BDNF@TA-PLGA/Dex-HA hydrogel. Our results showed that the hydrogel had properties similar to natural spinal cords. Specifically, the hydrogel had soft mechanical properties and high electrical conductivity. The cross-sectional morphology of the hydrogel exhibited a continuous and porous structure. The swelling and degradation behaviors of the Dex-HA hydrogel in vitro indicated the incorporation of TA into hydrogel matrix could improve the stability of the hydrogel matrix as well as extend the release time of BDNF from the matrix. Furthermore, results from immunostaining and real-time PCR demonstrated that BDNF@TA-PLGA/Dex-HA hydrogel could promote the differentiation of neural stem cells (NSCs) into neurons and inhibit astrocyte differentiation in vitro. These results show the great potential of this hydrogel as a biomimetic material in SCI regeneration.
Collapse
Affiliation(s)
- Fei Huang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Taiying Chen
- Department of Liver Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jun Chang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chi Zhang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Faxue Liao
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Linwei Wu
- Department of Liver Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| | - Wenbin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Zongsheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
22
|
Memanishvili T, Monni E, Tatarishivili J, Lindvall O, Tsiskaridze A, Kokaia Z, Tornero D. Poly(ester amide) microspheres are efficient vehicles for long-term intracerebral growth factor delivery and improve functional recovery after stroke. Biomed Mater 2020; 15:065020. [DOI: 10.1088/1748-605x/aba4f6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
23
|
Yang L, Conley BM, Cerqueira SR, Pongkulapa T, Wang S, Lee JK, Lee KB. Effective Modulation of CNS Inhibitory Microenvironment using Bioinspired Hybrid-Nanoscaffold-Based Therapeutic Interventions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002578. [PMID: 32893402 PMCID: PMC7606660 DOI: 10.1002/adma.202002578] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/04/2020] [Indexed: 05/11/2023]
Abstract
Central nervous system (CNS) injuries are often debilitating, and most currently have no cure. This is due to the formation of a neuroinhibitory microenvironment at injury sites, which includes neuroinflammatory signaling and non-permissive extracellular matrix (ECM) components. To address this challenge, a viscous interfacial self-assembly approach, to generate a bioinspired hybrid 3D porous nanoscaffold platform for delivering anti-inflammatory molecules and establish a favorable 3D-ECM environment for the effective suppression of the neuroinhibitory microenvironment, is developed. By tailoring the structural and biochemical properties of the 3D porous nanoscaffold, enhanced axonal growth from the dual-targeting therapeutic strategy in a human induced pluripotent stem cell (hiPSC)-based in vitro model of neuroinflammation is demonstrated. Moreover, nanoscaffold-based approaches promote significant axonal growth and functional recovery in vivo in a spinal cord injury model through a unique mechanism of anti-inflammation-based fibrotic scar reduction. Given the critical role of neuroinflammation and ECM microenvironments in neuroinhibitory signaling, the developed nanobiomaterial-based therapeutic intervention may pave a new road for treating CNS injuries.
Collapse
Affiliation(s)
- Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Brian M Conley
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Susana R Cerqueira
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, 1095 NW 14th Terrace, LPLC 4-19, Miami, FL, 33136, USA
| | - Thanapat Pongkulapa
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Shenqiang Wang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, 1095 NW 14th Terrace, LPLC 4-19, Miami, FL, 33136, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
24
|
Enhanced proliferation and differentiation of neural stem cells by peptide-containing temperature-sensitive hydrogel scaffold. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111258. [PMID: 32806302 DOI: 10.1016/j.msec.2020.111258] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 01/03/2023]
Abstract
Hydrogel has attracted great attention in the past few years as a widely used material for repairing central nerve damage. However, conventional hydrogel bio-scaffold, such as chitosan, gelatin, and sodium alginate, lack sufficient biological activity and have limited nerve repair capabilities. Therefore, to explore biologically active and intelligent hydrogel materials is particularly important and necessary for central nerve repair. Herein, we developed a temperature-sensitive hydrogel grafted with a bioactive peptide IKVAV (Ile-Lys-Val-Ala-Val, IKVAV). The hydrogel was prepared by copolymerization of N-propan-2-ylprop-2-enamide (NIPAM) and AC-PEG-IKVAV copolymers via reversible addition-fracture chain transfer (RAFT) polymerization, using polyethylene glycol (PEGDA) and N, N'-Methylenebisacrylamide (BISAM) as cross-linking agents. The prepared hydrogel scaffold demonstrates a series of excellent properties such as rapid (de)swelling performance, good biocompatibility, regular three-dimensional porous structure, and in particular good biological activity, which can guide cell fate and mediate neuron's differentiation. Therefore, the developed peptide hydrogel scaffold provides a new strategy for designing biomaterials that are widely used in tissue engineering for central nervous system injury.
Collapse
|
25
|
Rey F, Barzaghini B, Nardini A, Bordoni M, Zuccotti GV, Cereda C, Raimondi MT, Carelli S. Advances in Tissue Engineering and Innovative Fabrication Techniques for 3-D-Structures: Translational Applications in Neurodegenerative Diseases. Cells 2020; 9:cells9071636. [PMID: 32646008 PMCID: PMC7407518 DOI: 10.3390/cells9071636] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
In the field of regenerative medicine applied to neurodegenerative diseases, one of the most important challenges is the obtainment of innovative scaffolds aimed at improving the development of new frontiers in stem-cell therapy. In recent years, additive manufacturing techniques have gained more and more relevance proving the great potential of the fabrication of precision 3-D scaffolds. In this review, recent advances in additive manufacturing techniques are presented and discussed, with an overview on stimulus-triggered approaches, such as 3-D Printing and laser-based techniques, and deposition-based approaches. Innovative 3-D bioprinting techniques, which allow the production of cell/molecule-laden scaffolds, are becoming a promising frontier in disease modelling and therapy. In this context, the specific biomaterial, stiffness, precise geometrical patterns, and structural properties are to be considered of great relevance for their subsequent translational applications. Moreover, this work reports numerous recent advances in neural diseases modelling and specifically focuses on pre-clinical and clinical translation for scaffolding technology in multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
| | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
| | - Alessandra Nardini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
| | - Matteo Bordoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy;
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
| | - Cristina Cereda
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy;
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
- Correspondence: (M.T.R.); (S.C.); Tel.: +390-223-994-306 (M.T.R.); +390-250-319-825 (S.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
- Correspondence: (M.T.R.); (S.C.); Tel.: +390-223-994-306 (M.T.R.); +390-250-319-825 (S.C.)
| |
Collapse
|
26
|
Prager J, Adams CF, Delaney AM, Chanoit G, Tarlton JF, Wong LF, Chari DM, Granger N. Stiffness-matched biomaterial implants for cell delivery: clinical, intraoperative ultrasound elastography provides a 'target' stiffness for hydrogel synthesis in spinal cord injury. J Tissue Eng 2020; 11:2041731420934806. [PMID: 32670538 PMCID: PMC7336822 DOI: 10.1177/2041731420934806] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/21/2020] [Indexed: 12/14/2022] Open
Abstract
Safe hydrogel delivery requires stiffness-matching with host tissues to avoid
iatrogenic damage and reduce inflammatory reactions. Hydrogel-encapsulated cell
delivery is a promising combinatorial approach to spinal cord injury therapy,
but a lack of in vivo clinical spinal cord injury stiffness
measurements is a barrier to their use in clinics. We demonstrate that
ultrasound elastography – a non-invasive, clinically established tool – can be
used to measure spinal cord stiffness intraoperatively in canines with
spontaneous spinal cord injury. In line with recent experimental reports, our
data show that injured spinal cord has lower stiffness than uninjured cord. We
show that the stiffness of hydrogels encapsulating a clinically relevant
transplant population (olfactory ensheathing cells) can also be measured by
ultrasound elastography, enabling synthesis of hydrogels with comparable
stiffness to canine spinal cord injury. We therefore demonstrate
proof-of-principle of a novel approach to stiffness-matching hydrogel-olfactory
ensheathing cell implants to ‘real-life’ spinal cord injury values; an approach
applicable to multiple biomaterial implants for regenerative therapies.
Collapse
Affiliation(s)
- Jon Prager
- Bristol Veterinary School, University of Bristol, Bristol, UK.,The Royal Veterinary College, University of London, Hatfield, UK
| | - Christopher F Adams
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, UK
| | - Alexander M Delaney
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, UK
| | | | - John F Tarlton
- Bristol Veterinary School, University of Bristol, Bristol, UK
| | | | - Divya M Chari
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, UK
| | - Nicolas Granger
- The Royal Veterinary College, University of London, Hatfield, UK
| |
Collapse
|
27
|
Sun X, Zhang C, Xu J, Zhai H, Liu S, Xu Y, Hu Y, Long H, Bai Y, Quan D. Neurotrophin-3-Loaded Multichannel Nanofibrous Scaffolds Promoted Anti-Inflammation, Neuronal Differentiation, and Functional Recovery after Spinal Cord Injury. ACS Biomater Sci Eng 2020; 6:1228-1238. [PMID: 33464858 DOI: 10.1021/acsbiomaterials.0c00023] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The clinical therapeutics for nerve tissue regeneration and functional recovery after spinal cord injury (SCI) are very limited because of the complex biological processes and inhibitory microenvironment. Advanced biomaterials are highly desired to avoid severe secondary damage and provide guidance for axonal regrowth. Multichannel nanofibrous scaffolds were modified with gelatin and cross-linked by genipin. The gelatin-coated nanofibers exhibited strong binding affinity with neurotrophin-3, which underwent a well-controlled release and highly promoted neuronal differentiation and synapse formation of the seeded neural stem cells. The nanofibrous scaffolds fabricated by combinatorial biomaterials were implanted into complete transected spinal cords in rats. Not only were the inflammatory responses and collagen/astrocytic scar formation limited, but the functional neurons and remyelination were facilitated postsurgery, leading to highly improved functional restoration. This nanofibrous scaffold with high specific surface area can be easily modified with biomolecules, which was proven to be effective for nerve regeneration after transected SCI, and provided a springboard for advanced scaffold design in clinical applications.
Collapse
Affiliation(s)
- Xiumin Sun
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China.,School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Chi Zhang
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Jinghui Xu
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Hong Zhai
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Sheng Liu
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Yiwei Xu
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Yong Hu
- Department of Orthopaedics and Traumatology, Li Kai Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Houqing Long
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Ying Bai
- GD Functional Biomaterials Engineering Technology Research Center, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Daping Quan
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China.,GD Functional Biomaterials Engineering Technology Research Center, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
28
|
Cembran A, Bruggeman KF, Williams RJ, Parish CL, Nisbet DR. Biomimetic Materials and Their Utility in Modeling the 3-Dimensional Neural Environment. iScience 2020; 23:100788. [PMID: 31954980 PMCID: PMC6970178 DOI: 10.1016/j.isci.2019.100788] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/30/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
The brain is a complex 3-dimensional structure, the organization of which provides a local environment that directly influences the survival, proliferation, differentiation, migration, and plasticity of neurons. To probe the effects of damage and disease on these cells, a synthetic environment is needed. Three-dimensional culturing of stem cells, neural progenitors, and neurons within fabricated biomaterials has demonstrated superior biomimetic properties over conventional 2-dimensional cultureware, offering direct recapitulation of both cell-cell and cell-extracellular matrix interactions. Within this review we address the benefits of deploying biomaterials as advanced cell culture tools capable of influencing neuronal fate and as in vitro models of the native in vivo microenvironment. We highlight recent and promising biomaterials approaches toward understanding neural network and their function relevant to neurodevelopment and provide our perspective on how these materials can be engineered and programmed to study both the healthy and diseased nervous system.
Collapse
Affiliation(s)
- Arianna Cembran
- Laboratory of Advanced Biomaterials, Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT 2600, Australia
| | - Kiara F Bruggeman
- Laboratory of Advanced Biomaterials, Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT 2600, Australia
| | | | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia.
| | - David R Nisbet
- Laboratory of Advanced Biomaterials, Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT 2600, Australia.
| |
Collapse
|
29
|
Ghane N, Beigi MH, Labbaf S, Nasr-Esfahani MH, Kiani A. Design of hydrogel-based scaffolds for the treatment of spinal cord injuries. J Mater Chem B 2020; 8:10712-10738. [DOI: 10.1039/d0tb01842b] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hydrogel-based scaffold design approaches for the treatment of spinal cord injuries.
Collapse
Affiliation(s)
- Nazanin Ghane
- Department of Cellular Biotechnology Cell Science Research Center
- Royan Institute for Biotechnology
- ACECR
- Isfahan
- Iran
| | - Mohammad-Hossein Beigi
- Department of Cellular Biotechnology Cell Science Research Center
- Royan Institute for Biotechnology
- ACECR
- Isfahan
- Iran
| | - Sheyda Labbaf
- Biomaterials Research Group
- Department of Materials Engineering
- Isfahan University of Technology
- Isfahan
- Iran
| | | | - Amirkianoosh Kiani
- Silicon Hall: Micro/Nano Manufacturing Facility
- Faculty of Engineering and Applied Science
- Ontario Tech University
- Ontario
- Canada
| |
Collapse
|
30
|
He Q, Zhang J, Liao Y, Alakpa EV, Bunpetch V, Zhang J, Ouyang H. Current advances in microsphere based cell culture and tissue engineering. Biotechnol Adv 2019; 39:107459. [PMID: 31682922 DOI: 10.1016/j.biotechadv.2019.107459] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 09/12/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Qiulin He
- Department of Orthopaedic Surgery, Second Affiliated Hospital & Zhejiang University-University of Edinburgh Institute & School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingwei Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Youguo Liao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China.; Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning 530021, China
| | - Enateri Verissarah Alakpa
- Department of Orthopaedic Surgery, Second Affiliated Hospital & Zhejiang University-University of Edinburgh Institute & School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiayan Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hongwei Ouyang
- Department of Orthopaedic Surgery, Second Affiliated Hospital & Zhejiang University-University of Edinburgh Institute & School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China.; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.; China Orthopedic Regenerative Medicine Group (CORMed), China..
| |
Collapse
|
31
|
Sellers DL, Tan JKY, Pineda JMB, Peeler DJ, Porubsky VL, Olden BR, Salipante SJ, Pun SH. Targeting Ligands Deliver Model Drug Cargo into the Central Nervous System along Autonomic Neurons. ACS NANO 2019; 13:10961-10971. [PMID: 31589023 PMCID: PMC7651855 DOI: 10.1021/acsnano.9b01515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
While biologic drugs such as proteins, peptides, or nucleic acids have shown promise in the treatment of neurodegenerative diseases, the blood-brain barrier (BBB) severely limits drug delivery to the central nervous system (CNS) after systemic administration. Consequently, drug delivery challenges preclude biological drug candidates from the clinical armamentarium. In order to target drug delivery and uptake into to the CNS, we used an in vivo phage display screen to identify peptides able to target drug-uptake by the vast array of neurons of the autonomic nervous system (ANS). Using next-generation sequencing, we identified 21 candidate targeted ANS-to-CNS uptake ligands (TACL) that enriched bacteriophage accumulation and delivered protein-cargo into the CNS after intraperitoneal (IP) administration. The series of TACL peptides were synthesized and tested for their ability to deliver a model enzyme (NeutrAvidin-horseradish peroxidase fusion) to the brain and spinal cord. Three TACL-peptides facilitated significant active enzyme delivery into the CNS, with limited accumulation in off-target organs. Peptide structure and serum stability is increased when internal cysteine residues are cyclized by perfluoroarylation with decafluorobiphenyl, which increased delivery to the CNS further. TACL-peptide was demonstrated to localize in parasympathetic ganglia neurons in addition to neuronal structures in the hindbrain and spinal cord. By targeting uptake into ANS neurons, we demonstrate the potential for TACL-peptides to bypass the blood-brain barrier and deliver a model drug into the brain and spinal cord.
Collapse
Affiliation(s)
- Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - James-Kevin Y. Tan
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Julio Marco B. Pineda
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - David J. Peeler
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Veronica L. Porubsky
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Brynn R. Olden
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, Washington, 98195, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington, 98195, USA
| |
Collapse
|
32
|
Song YH, Agrawal NK, Griffin JM, Schmidt CE. Recent advances in nanotherapeutic strategies for spinal cord injury repair. Adv Drug Deliv Rev 2019; 148:38-59. [PMID: 30582938 PMCID: PMC6959132 DOI: 10.1016/j.addr.2018.12.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/12/2018] [Accepted: 12/17/2018] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a devastating and complicated condition with no cure available. The initial mechanical trauma is followed by a secondary injury characterized by inflammatory cell infiltration and inhibitory glial scar formation. Due to the limitations posed by the blood-spinal cord barrier, systemic delivery of therapeutics is challenging. Recent development of various nanoscale strategies provides exciting and promising new means of treating SCI by crossing the blood-spinal cord barrier and delivering therapeutics. As such, we discuss different nanomaterial fabrication methods and provide an overview of recent studies where nanomaterials were developed to modulate inflammatory signals, target inhibitory factors in the lesion, and promote axonal regeneration after SCI. We also review emerging areas of research such as optogenetics, immunotherapy and CRISPR-mediated genome editing where nanomaterials can provide synergistic effects in developing novel SCI therapy regimens, as well as current efforts and barriers to clinical translation of nanomaterials.
Collapse
Affiliation(s)
- Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Nikunj K Agrawal
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jonathan M Griffin
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
33
|
de la Vega L, Lee C, Sharma R, Amereh M, Willerth SM. 3D bioprinting models of neural tissues: The current state of the field and future directions. Brain Res Bull 2019; 150:240-249. [DOI: 10.1016/j.brainresbull.2019.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/30/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023]
|
34
|
Caplan H, Olson SD, Kumar A, George M, Prabhakara KS, Wenzel P, Bedi S, Toledano-Furman NE, Triolo F, Kamhieh-Milz J, Moll G, Cox CS. Mesenchymal Stromal Cell Therapeutic Delivery: Translational Challenges to Clinical Application. Front Immunol 2019; 10:1645. [PMID: 31417542 PMCID: PMC6685059 DOI: 10.3389/fimmu.2019.01645] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
For several decades, multipotent mesenchymal stromal cells (MSCs) have been extensively studied for their therapeutic potential across a wide range of diseases. In the preclinical setting, MSCs demonstrate consistent ability to promote tissue healing, down-regulate excessive inflammation and improve outcomes in animal models. Several proposed mechanisms of action have been posited and demonstrated across an array of in vitro models. However, translation into clinical practice has proven considerably more difficult. A number of prominent well-funded late-phase clinical trials have failed, thus calling out for new efforts to optimize product delivery in the clinical setting. In this review, we discuss novel topics critical to the successful translation of MSCs from pre-clinical to clinical applications. In particular, we focus on the major routes of cell delivery, aspects related to hemocompatibility, and potential safety concerns associated with MSC therapy in the different settings.
Collapse
Affiliation(s)
- Henry Caplan
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Akshita Kumar
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Mitchell George
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karthik S. Prabhakara
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Pamela Wenzel
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Supinder Bedi
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Naama E. Toledano-Furman
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Fabio Triolo
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
35
|
Gharieh A, Khoee S, Mahdavian AR. Emulsion and miniemulsion techniques in preparation of polymer nanoparticles with versatile characteristics. Adv Colloid Interface Sci 2019; 269:152-186. [PMID: 31082544 DOI: 10.1016/j.cis.2019.04.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 04/13/2019] [Accepted: 04/24/2019] [Indexed: 11/29/2022]
Abstract
In recent years, polymer nanoparticles (PNPs) have found their ways into numerous applications extending from electronics to photonics, conducting materials to sensors and medicine to biotechnology. Physical properties and surface morphology of PNPs are the most important parameters that significantly affect on their exploitations and can be controlled through the synthesis process. Emulsion and miniemulsion techniques are among the most efficient and wide-spread methods for preparation of PNPs. The objective of this review is to present and highlight the recent developments in the advanced PNPs with specific properties that are produced through emulsion and miniemulsion processes.
Collapse
Affiliation(s)
- Ali Gharieh
- Polymer Science Department, Iran Polymer & Petrochemical Institute, P.O. Box: 14965/115, Tehran, Iran
| | - Sepideh Khoee
- Polymer Laboratory, School of Chemistry, College of Science, University of Tehran, PO Box 14155 6455, Tehran, Iran
| | - Ali Reza Mahdavian
- Polymer Science Department, Iran Polymer & Petrochemical Institute, P.O. Box: 14965/115, Tehran, Iran.
| |
Collapse
|
36
|
Teleanu RI, Gherasim O, Gherasim TG, Grumezescu V, Grumezescu AM, Teleanu DM. Nanomaterial-Based Approaches for Neural Regeneration. Pharmaceutics 2019; 11:E266. [PMID: 31181719 PMCID: PMC6630326 DOI: 10.3390/pharmaceutics11060266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
Mechanical, thermal, chemical, or ischemic injury of the central or peripheral nervous system results in neuron loss, neurite damage, and/or neuronal dysfunction, almost always accompanied by sensorimotor impairment which alters the patient's life quality. The regenerative strategies for the injured nervous system are currently limited and mainly allow partial functional recovery, so it is necessary to develop new and effective approaches for nervous tissue regenerative therapy. Nanomaterials based on inorganic or organic and composite or hybrid compounds with tunable physicochemical properties and functionality proved beneficial for the transport and delivery/release of various neuroregenerative-relevant biomolecules or cells. Within the following paragraphs, we will emphasize that nanomaterial-based strategies (including nanosized and nanostructured biomaterials) represent a promising alternative towards repairing and regenerating the injured nervous system.
Collapse
Affiliation(s)
- Raluca Ioana Teleanu
- "Victor Gomoiu" Clinical Children's Hospital, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | - Oana Gherasim
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania.
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 077125 Magurele, Romania.
| | - Tudor George Gherasim
- National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania.
| | - Valentina Grumezescu
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 077125 Magurele, Romania.
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania.
| | - Daniel Mihai Teleanu
- Emergency University Hospital, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| |
Collapse
|
37
|
Katoh H, Yokota K, Fehlings MG. Regeneration of Spinal Cord Connectivity Through Stem Cell Transplantation and Biomaterial Scaffolds. Front Cell Neurosci 2019; 13:248. [PMID: 31244609 PMCID: PMC6563678 DOI: 10.3389/fncel.2019.00248] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/17/2019] [Indexed: 12/20/2022] Open
Abstract
Significant progress has been made in the treatment of spinal cord injury (SCI). Advances in post-trauma management and intensive rehabilitation have significantly improved the prognosis of SCI and converted what was once an “ailment not to be treated” into a survivable injury, but the cold hard fact is that we still do not have a validated method to improve the paralysis of SCI. The irreversible functional impairment of the injured spinal cord is caused by the disruption of neuronal transduction across the injury lesion, which is brought about by demyelination, axonal degeneration, and loss of synapses. Furthermore, refractory substrates generated in the injured spinal cord inhibit spontaneous recovery. The discovery of the regenerative capability of central nervous system neurons in the proper environment and the verification of neural stem cells in the spinal cord once incited hope that a cure for SCI was on the horizon. That hope was gradually replaced with mounting frustration when neuroprotective drugs, cell transplantation, and strategies to enhance remyelination, axonal regeneration, and neuronal plasticity demonstrated significant improvement in animal models of SCI but did not translate into a cure in human patients. However, recent advances in SCI research have greatly increased our understanding of the fundamental processes underlying SCI and fostered increasing optimism that these multiple treatment strategies are finally coming together to bring about a new era in which we will be able to propose encouraging therapies that will lead to appreciable improvements in SCI patients. In this review, we outline the pathophysiology of SCI that makes the spinal cord refractory to regeneration and discuss the research that has been done with cell replacement and biomaterial implantation strategies, both by itself and as a combined treatment. We will focus on the capacity of these strategies to facilitate the regeneration of neural connectivity necessary to achieve meaningful functional recovery after SCI.
Collapse
Affiliation(s)
- Hiroyuki Katoh
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Department of Orthopaedic Surgery - Surgical Sciences, School of Medicine, Tokai University, Tokyo, Japan
| | - Kazuya Yokota
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Division of Neurosurgery, University of Toronto, Toronto, ON, Canada.,Spine Program, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| |
Collapse
|
38
|
Abstract
Biomaterials as we know them today had their origins in the late 1940s with off-the-shelf commercial polymers and metals. The evolution of materials for medical applications from these simple origins has been rapid and impactful. This review relates some of the early history; addresses concerns after two decades of development in the twenty-first century; and discusses how advanced technologies in both materials science and biology will address concerns, advance materials used at the biointerface, and improve outcomes for patients.
Collapse
Affiliation(s)
- Buddy D. Ratner
- Departments of Bioengineering and Chemical Engineering, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
39
|
Jooybar E, Abdekhodaie MJ, Mousavi A, Zoetebier B, Dijkstra PJ. Enzymatically crosslinked hyaluronic acid microgels as a vehicle for sustained delivery of cationic proteins. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.03.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
40
|
Courtine G, Sofroniew MV. Spinal cord repair: advances in biology and technology. Nat Med 2019; 25:898-908. [PMID: 31160817 DOI: 10.1038/s41591-019-0475-6] [Citation(s) in RCA: 272] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
Individuals with spinal cord injury (SCI) can face decades with permanent disabilities. Advances in clinical management have decreased morbidity and improved outcomes, but no randomized clinical trial has demonstrated the efficacy of a repair strategy for improving recovery from SCI. Here, we summarize recent advances in biological and engineering strategies to augment neuroplasticity and/or functional recovery in animal models of SCI that are pushing toward clinical translation.
Collapse
Affiliation(s)
- Grégoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland. .,Department of Neurosurgery, University Hospital Lausanne (CHUV), Lausanne, Switzerland.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
41
|
Willerth SM, Sakiyama-Elbert SE. Combining Stem Cells and Biomaterial Scaffolds for Constructing Tissues and Cell Delivery. ACTA ACUST UNITED AC 2019. [DOI: 10.3233/stj-180001] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Combining stem cells with biomaterial scaffolds serves as a promising strategy for engineering tissues for both in vitro and in vivo applications. This updated review details commonly used biomaterial scaffolds for engineering tissues from stem cells. We first define the different types of stem cells and their relevant properties and commonly used scaffold formulations. Next, we discuss natural and synthetic scaffold materials typically used when engineering tissues, along with their associated advantages and drawbacks and gives examples of target applications. New approaches to engineering tissues, such as 3D bioprinting, are described as they provide exciting opportunities for future work along with current challenges that must be addressed. Thus, this review provides an overview of the available biomaterials for directing stem cell differentiation as a means of producing replacements for diseased or damaged tissues.
Collapse
Affiliation(s)
- Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, VIC, Canada
- Division of Medical Sciences, University of Victoria, VIC, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
42
|
Ashammakhi N, Ahadian S, Darabi MA, El Tahchi M, Lee J, Suthiwanich K, Sheikhi A, Dokmeci MR, Oklu R, Khademhosseini A. Minimally Invasive and Regenerative Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1804041. [PMID: 30565732 PMCID: PMC6709364 DOI: 10.1002/adma.201804041] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/20/2018] [Indexed: 05/03/2023]
Abstract
Advances in biomaterial synthesis and fabrication, stem cell biology, bioimaging, microsurgery procedures, and microscale technologies have made minimally invasive therapeutics a viable tool in regenerative medicine. Therapeutics, herein defined as cells, biomaterials, biomolecules, and their combinations, can be delivered in a minimally invasive way to regenerate different tissues in the body, such as bone, cartilage, pancreas, cardiac, skeletal muscle, liver, skin, and neural tissues. Sophisticated methods of tracking, sensing, and stimulation of therapeutics in vivo using nano-biomaterials and soft bioelectronic devices provide great opportunities to further develop minimally invasive and regenerative therapeutics (MIRET). In general, minimally invasive delivery methods offer high yield with low risk of complications and reduced costs compared to conventional delivery methods. Here, minimally invasive approaches for delivering regenerative therapeutics into the body are reviewed. The use of MIRET to treat different tissues and organs is described. Although some clinical trials have been performed using MIRET, it is hoped that such therapeutics find wider applications to treat patients. Finally, some future perspective and challenges for this emerging field are highlighted.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mario El Tahchi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- LBMI, Department of Physics, Lebanese University - Faculty of Sciences 2, PO Box 90656, Jdeidet, Lebanon
| | - Junmin Lee
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Kasinan Suthiwanich
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Amir Sheikhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mehmet R. Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Rahmi Oklu
- Division of Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Radiological Sciences, University of California - Los Angeles, Los Angeles, California, USA
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, California, USA
- Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Edri R, Gal I, Noor N, Harel T, Fleischer S, Adadi N, Green O, Shabat D, Heller L, Shapira A, Gat-Viks I, Peer D, Dvir T. Personalized Hydrogels for Engineering Diverse Fully Autologous Tissue Implants. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1803895. [PMID: 30406960 DOI: 10.1002/adma.201803895] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/27/2018] [Indexed: 05/22/2023]
Abstract
Despite incremental improvements in the field of tissue engineering, no technology is currently available for producing completely autologous implants where both the cells and the scaffolding material are generated from the patient, and thus do not provoke an immune response that may lead to implant rejection. Here, a new approach is introduced to efficiently engineer any tissue type, which its differentiation cues are known, from one small tissue biopsy. Pieces of omental tissues are extracted from patients and, while the cells are reprogrammed to become induced pluripotent stem cells, the extracellular matrix is processed into an immunologically matching, thermoresponsive hydrogel. Efficient cell differentiation within a large 3D hydrogel is reported, and, as a proof of concept, the generation of functional cardiac, cortical, spinal cord, and adipogenic tissue implants is demonstrated. This versatile bioengineering approach may assist to regenerate any tissue and organ with a minimal risk for immune rejection.
Collapse
Affiliation(s)
- Reuven Edri
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Idan Gal
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Nadav Noor
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tom Harel
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Sharon Fleischer
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Nofar Adadi
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ori Green
- School of Chemistry, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Doron Shabat
- School of Chemistry, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Lior Heller
- Department of Plastic Surgery, Assaf Harofeh MC, Beer Ya'akov, Zerifin, 70300, Israel
| | - Assaf Shapira
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Irit Gat-Viks
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Dan Peer
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tal Dvir
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
- Sagol Center for Regenerative Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| |
Collapse
|
44
|
Zamproni LN, Grinet MAVM, Mundim MTVV, Reis MBC, Galindo LT, Marciano FR, Lobo AO, Porcionatto M. Rotary jet-spun porous microfibers as scaffolds for stem cells delivery to central nervous system injury. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 15:98-107. [PMID: 30244084 DOI: 10.1016/j.nano.2018.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022]
Abstract
Stem cell transplantation is a promising strategy to treat brain injuries. However, cell-based therapies are limited because poor local cell engraftment. Here, we present a polylactic acid (PLA) scaffold to support mesenchymal stem cells (MSCs) delivery in stroke. We isolated bone marrow MSCs from adult C57/Bl6 mice, cultured them on PLA polymeric rough microfibrous (PRM) scaffolds obtained by rotary jet spinning, and transplanted over the brains of adult C57/Bl6 mice, carrying thermocoagulation-induced cortical stroke. No inflammatory response to PRM was found. MSCs transplantation significantly reduced the area of the lesion and PRM delivery increased MSCs retention at the injury site. In addition, PRM upregulated α6-integrin and CXCL12 production, which may be the cause for greater cell retention at the lesion site and may provide additional benefit to MSCs transplantation procedures. We conclude that PRM scaffolds offer a promising new system to deliver stem cells to injured areas of the brain.
Collapse
Affiliation(s)
- Laura N Zamproni
- Neurobiology Lab, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Mayara T V V Mundim
- Neurobiology Lab, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Marcella B C Reis
- Neurobiology Lab, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Layla T Galindo
- Neurobiology Lab, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Fernanda R Marciano
- Instituto de Ciência e Tecnologia, Universidade Brasil, São Paulo, SP, Brazil
| | - Anderson O Lobo
- Instituto de Ciência e Tecnologia, Universidade Brasil, São Paulo, SP, Brazil; Programa de Pós-graduação em Ciência e Engenharia de Materiais, Universidade Federal do Piaui, Teresina, PI, Brazil.
| | - Marimelia Porcionatto
- Neurobiology Lab, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
45
|
Jensen G, Morrill C, Huang Y. 3D tissue engineering, an emerging technique for pharmaceutical research. Acta Pharm Sin B 2018; 8:756-766. [PMID: 30258764 PMCID: PMC6148716 DOI: 10.1016/j.apsb.2018.03.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
Tissue engineering and the tissue engineering model have shown promise in improving methods of drug delivery, drug action, and drug discovery in pharmaceutical research for the attenuation of the central nervous system inflammatory response. Such inflammation contributes to the lack of regenerative ability of neural cells, as well as the temporary and permanent loss of function associated with neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and traumatic brain injury. This review is focused specifically on the recent advances in the tissue engineering model made by altering scaffold biophysical and biochemical properties for use in the treatment of neurodegenerative diseases. A portion of this article will also be spent on the review of recent progress made in extracellular matrix decellularization as a new and innovative scaffold for disease treatment.
Collapse
Affiliation(s)
| | | | - Yu Huang
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| |
Collapse
|
46
|
Nisbet DR, Wang TY, Bruggeman KF, Niclis JC, Somaa FA, Penna V, Hunt CPJ, Wang Y, Kauhausen JA, Williams RJ, Thompson LH, Parish CL. Shear Containment of BDNF within Molecular Hydrogels Promotes Human Stem Cell Engraftment and Postinfarction Remodeling in Stroke. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800113] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- D. R. Nisbet
- Laboratory of Advanced Materials Research School of Engineering The Australian National University Canberra 2601 Australia
- Biofab3D Aikenhead Center for Medical Discovery St. Vincent's Hospital Melbourne Victoria 3065 Australia
| | - T. Y. Wang
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria 3010 Australia
| | - K. F. Bruggeman
- Laboratory of Advanced Materials Research School of Engineering The Australian National University Canberra 2601 Australia
| | - J. C. Niclis
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria 3010 Australia
| | - F. A. Somaa
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria 3010 Australia
| | - V. Penna
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria 3010 Australia
| | - C. P. J. Hunt
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria 3010 Australia
| | - Y. Wang
- Laboratory of Advanced Materials Research School of Engineering The Australian National University Canberra 2601 Australia
| | - J. A. Kauhausen
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria 3010 Australia
| | - R. J. Williams
- Biofab3D Aikenhead Center for Medical Discovery St. Vincent's Hospital Melbourne Victoria 3065 Australia
- School of Engineering RMIT University Melbourne 3001 Australia
| | - L. H. Thompson
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria 3010 Australia
| | - C. L. Parish
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
47
|
Niemczyk B, Sajkiewicz P, Kolbuk D. Injectable hydrogels as novel materials for central nervous system regeneration. J Neural Eng 2018; 15:051002. [DOI: 10.1088/1741-2552/aacbab] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
48
|
Zhou H, Ma Q, Zhu P, Ren J, Reiter RJ, Chen Y. Protective role of melatonin in cardiac ischemia-reperfusion injury: From pathogenesis to targeted therapy. J Pineal Res 2018; 64. [PMID: 29363153 DOI: 10.1111/jpi.12471] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/16/2018] [Indexed: 02/06/2023]
Abstract
Acute myocardial infarction (MI) is a major cause of mortality and disability worldwide. In patients with MI, the treatment option for reducing acute myocardial ischemic injury and limiting MI size is timely and effective myocardial reperfusion using either thombolytic therapy or primary percutaneous coronary intervention (PCI). However, the procedure of reperfusion itself induces cardiomyocyte death, known as myocardial reperfusion injury, for which there is still no effective therapy. Recent evidence has depicted a promising role of melatonin, which possesses powerful antioxidative and anti-inflammatory properties, in the prevention of ischemia-reperfusion (IR) injury and the protection against cardiomyocyte death. A number of reports explored the mechanism of action behind melatonin-induced beneficial effects against myocardial IR injury. In this review, we summarize the research progress related to IR injury and discuss the unique actions of melatonin as a protective agent. Furthermore, the possible mechanisms responsible for the myocardial benefits of melatonin against reperfusion injury are listed with the prospect of the use of melatonin in clinical application.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qiang Ma
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX, USA
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
49
|
Chen YS, Harn HJ, Chiou TW. The Role of Biomaterials in Implantation for Central Nervous System Injury. Cell Transplant 2018; 27:407-422. [PMID: 29741115 PMCID: PMC6038039 DOI: 10.1177/0963689717732991] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Permanent deficits that occur in memory, sensation, and cognition can result from central nervous system (CNS) trauma that causes dysfunction and/or unregulated CNS regeneration. Some therapeutic approaches are preferentially applied to the human body. Therefore, cell transplantation, one of the therapeutic strategies, may be used to benefit people. However, poor cell viability and low efficacy are the limitations to cell transplantation strategies. Biomaterials have been widely used in several fields (e.g., triggering cell differentiation, guiding cell migration, improving wound healing, and increasing tissue regeneration) by modulating their characteristics in chemistry, topography, and softness/stiffness for highly flexible application. We reviewed implanted biomaterials to investigate the roles and influences of physical/chemical properties on cell behaviors and applications. With their unique molecular features, biomaterials are delivered in several methods and mixed with transplanted cells, which assists in increasing postimplanted biological substance efficiency on cell survival, host responses, and functional recovery of animal models. Moreover, tracking the routes of these transplanted cells using biomaterials as labeling agents is crucial for addressing their location, distribution, activity, and viability. Here, we provide comprehensive comments and up-to-date research of the application of biomaterials.
Collapse
Affiliation(s)
- Yu-Shuan Chen
- Bioinnovation Center, Tzu Chi Foundation, Hualien, Taiwan, No. 707, Sec. 3, Chung Yang Rd., Hualien 970, Taiwan, R.O.C.,Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan, No. 707, Section 3, Chung-Yang Road, Hualien 970, Taiwan, R.O.C
| | - Horng-Jyh Harn
- Bioinnovation Center, Tzu Chi foundation, Department of Pathology, Buddhist Tzu Chi General Hospital, Tzu Chi University, 707, Sec. 3, Chung Yang Rd., Hualien 970, Taiwan, R.O.C.,Horng-Jyh Harn, MD, PhD, Bioinnovation Center, Tzu Chi foundation, Department of Pathology, Buddhist Tzu Chi General Hospital, Tzu Chi University, 707, Sec. 3, Chung Yang Rd., Hualien 970, Taiwan, R.O.C.
| | - Tzyy-Wen Chiou
- Department of Life Science, Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien 97401, Taiwan, R.O.C.,Tzyy-Wen Chiou, PhD, Department of Life Science, Graduate Institute of Biotechnology, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Road, Hualien 97401, Taiwan, R.O.C.
| |
Collapse
|
50
|
Zhou CL, Zhao L, Shi HY, Liu JW, Shi JW, Kan BH, Li Z, Yu JC, Han JX. Combined acupuncture and HuangDiSan treatment affects behavior and synaptophysin levels in the hippocampus of senescence-accelerated mouse prone 8 after neural stem cell transplantation. Neural Regen Res 2018; 13:541-548. [PMID: 29623942 PMCID: PMC5900520 DOI: 10.4103/1673-5374.228760] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sanjiao acupuncture and HuangDiSan can promote the proliferation, migration and differentiation of exogenous neural stem cells in senescence-accelerated mouse prone 8 (SAMP8) mice and can improve learning and memory impairment and behavioral function in dementia-model mice. Thus, we sought to determine whether Sanjiao acupuncture and HuangDiSan can elevate the effect of neural stem cell transplantation in Alzheimer’s disease model mice. Sanjiao acupuncture was used to stimulate Danzhong (CV17), Zhongwan (CV12), Qihai (CV6), bilateral Xuehai (SP10) and bilateral Zusanli (ST36) 15 days before and after implantation of neural stem cells (5 × 105) into the hippocampal dentate gyrus of SAMP8 mice. Simultaneously, 0.2 mL HuangDiSan, containing Rehmannia Root and Chinese Angelica, was intragastrically administered. Our results demonstrated that compared with mice undergoing neural stem cell transplantation alone, learning ability was significantly improved and synaptophysin mRNA and protein levels were greatly increased in the hippocampus of mice undergoing both Sanjiao acupuncture and intragastric administration of HuangDiSan. We conclude that the combination of Sanjiao acupuncture and HuangDiSan can effectively improve dementia symptoms in mice, and the mechanism of this action might be related to the regulation of synaptophysin expression.
Collapse
Affiliation(s)
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine; Tianjin Key Laboratory of Acupuncture and Moxibustion, Tianjin, China
| | - Hui-Yan Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine; Tianjin Key Laboratory of Acupuncture and Moxibustion, Tianjin, China
| | - Jian-Wei Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiang-Wei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bo-Hong Kan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine; Tianjin Key Laboratory of Acupuncture and Moxibustion, Tianjin, China
| | - Zhen Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine; Tianjin Key Laboratory of Acupuncture and Moxibustion, Tianjin, China
| | - Jian-Chun Yu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing-Xian Han
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|