1
|
Zhang N, Hu J, Liu W, Cai W, Xu Y, Wang X, Li S, Ru B. Advances in Novel Biomaterial-Based Strategies for Spinal Cord Injury Treatment. Mol Pharm 2024; 21:4764-4785. [PMID: 39235393 DOI: 10.1021/acs.molpharmaceut.3c01104] [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] [Indexed: 09/06/2024]
Abstract
Spinal cord injury (SCI) is a highly disabling neurological disorder. Its pathological process comprises an initial acute injury phase (primary injury) and a secondary injury phase (subsequent chronic injury). Although surgical, drug, and cell therapies have made some progress in treating SCI, there is no exact therapeutic strategy for treating SCI and promoting nerve regeneration due to the complexity of the pathological SCI process. The development of novel drug delivery systems to treat SCI is expected to significantly impact the individualized treatment of SCI due to its unique and excellent properties, such as active targeting and controlled release. In this review, we first describe the pathological progression of the SCI response, including primary and secondary injuries. Next, we provide a concise overview of newly developed nanoplatforms and their potential application in regulating and treating different pathological processes of SCI. Then, we introduce the existing potential problems and future clinical application perspectives of biomedical engineering-based therapies for SCI.
Collapse
Affiliation(s)
- Nannan Zhang
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Jiaqi Hu
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Wenlong Liu
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Wenjun Cai
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Yun Xu
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Xiaojuan Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shun Li
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Bin Ru
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| |
Collapse
|
2
|
Torlakcik H, Sevim S, Alves P, Mattmann M, Llacer‐Wintle J, Pinto M, Moreira R, Flouris AD, Landers FC, Chen X, Puigmartí‐Luis J, Boehler Q, Mayor TS, Kim M, Nelson BJ, Pané S. Magnetically Guided Microcatheter for Targeted Injection of Magnetic Particle Swarms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404061. [PMID: 39119930 PMCID: PMC11481240 DOI: 10.1002/advs.202404061] [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: 04/17/2024] [Revised: 07/26/2024] [Indexed: 08/10/2024]
Abstract
The initial delivery of small-scale magnetic devices such as microrobots is a key, but often overlooked, aspect for their use in clinical applications. The deployment of these devices within the dynamic environment of the human body presents significant challenges due to their dispersion caused by circulatory flows. Here, a method is introduced to effectively deliver a swarm of magnetic nanoparticles in fluidic flows. This approach integrates a magnetically navigated robotic microcatheter equipped with a reservoir for storing the magnetic nanoparticles. The microfluidic flow within the reservoir facilitates the injection of magnetic nanoparticles into the fluid stream, and a magnetic field gradient guides the swarm through the oscillatory flow to a target site. The microcatheter and reservoir are engineered to enable magnetic steering and injection of the magnetic nanoparticles. To demonstrate this approach, experiments are conducted utilizing a spinal cord phantom simulating intrathecal catheter delivery for applications in the central nervous system. These results demonstrate that the proposed microcatheter successfully concentrates nanoparticles near the desired location through the precise manipulation of magnetic field gradients, offering a promising solution for the controlled deployment of untethered magnetic micro-/nanodevices within the complex physiological circulatory systems of the human body.
Collapse
Affiliation(s)
- Harun Torlakcik
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Semih Sevim
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Pedro Alves
- Transport Phenomena Research Centre (CEFT)Engineering FacultyPorto UniversityPorto4200Portugal
- Associate Laboratory in Chemical Engineering (ALICE)Engineering FacultyPorto UniversityPorto4200Portugal
| | - Michael Mattmann
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Joaquim Llacer‐Wintle
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| | | | | | - Andreas D. Flouris
- FAME LaboratoryDepartment of Exercise ScienceUniversity of ThessalyTrikala, Karies42100Greece
| | - Fabian C. Landers
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Xiang‐Zhong Chen
- Institute of OptoelectronicsState Key Laboratory of Photovoltaic Science and TechnologyShanghai Frontiers Science Research Base of Intelligent Optoelectronics and PerceptionFudan UniversityShanghai200433P. R. China
- Yiwu Research Institute of Fudan UniversityYiwu322000P. R. China
| | - Josep Puigmartí‐Luis
- Departament de Ciència dels Materials i Química FísicaInstitut de Química Teòrica i ComputacionalUniversity of BarcelonaMartí i Franquès, 1Barcelona08028Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)Pg. Lluís Companys 23Barcelona08010Spain
| | - Quentin Boehler
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Tiago Sotto Mayor
- Transport Phenomena Research Centre (CEFT)Engineering FacultyPorto UniversityPorto4200Portugal
- Associate Laboratory in Chemical Engineering (ALICE)Engineering FacultyPorto UniversityPorto4200Portugal
| | - Minsoo Kim
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Bradley J. Nelson
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| | - Salvador Pané
- Multi‐Scale Robotics LabInstitute of Robotics and Intelligent SystemsETH ZurichTannenstrasse 3Zurich8092Switzerland
| |
Collapse
|
3
|
Zhang J, Wu P, Wen Q. Optimization strategies for mesenchymal stem cell-based analgesia therapy: a promising therapy for pain management. Stem Cell Res Ther 2024; 15:211. [PMID: 39020426 PMCID: PMC11256674 DOI: 10.1186/s13287-024-03828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
Pain is a very common and complex medical problem that has a serious impact on individuals' physical and mental health as well as society. Non-steroidal anti-inflammatory drugs and opioids are currently the main drugs used for pain management, but they are not effective in controlling all types of pain, and their long-term use can cause adverse effects that significantly impair patients' quality of life. Mesenchymal stem cells (MSCs) have shown great potential in pain treatment. However, limitations such as the low proliferation rate of MSCs in vitro and low survival rate in vivo restrict their analgesic efficacy and clinical translation. In recent years, researchers have explored various innovative approaches to improve the therapeutic effectiveness of MSCs in pain treatment. This article reviews the latest research progress of MSCs in pain treatment, with a focus on methods to enhance the analgesic efficacy of MSCs, including engineering strategies to optimize the in vitro culture environment of MSCs and to improve the in vivo delivery efficiency of MSCs. We also discuss the unresolved issues to be explored in future MSCs and pain research and the challenges faced by the clinical translation of MSC therapy, aiming to promote the optimization and clinical translation of MSC-based analgesia therapy.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Ping Wu
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| | - Qingping Wen
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| |
Collapse
|
4
|
Zhou Z, Wang J, Jiang C, Xu K, Xu T, Yu X, Fang J, Yang Y, Dai X. Advances in Hydrogels for Meniscus Tissue Engineering: A Focus on Biomaterials, Crosslinking, Therapeutic Additives. Gels 2024; 10:114. [PMID: 38391445 PMCID: PMC10887778 DOI: 10.3390/gels10020114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Meniscus tissue engineering (MTE) has emerged as a promising strategy for meniscus repair and regeneration. As versatile platforms, hydrogels have gained significant attention in this field, as they possess tunable properties that allow them to mimic native extracellular matrices and provide a suitable microenvironment. Additionally, hydrogels can be minimally invasively injected and can be adjusted to match the shape of the implant site. They can conveniently and effectively deliver bioactive additives and demonstrate good compatibility with other functional materials. These inherent qualities have made hydrogel a promising candidate for therapeutic approaches in meniscus repair and regeneration. This article provides a comprehensive review of the advancements made in the research on hydrogel application for meniscus tissue engineering. Firstly, the biomaterials and crosslinking strategies used in the formation of hydrogels are summarized and analyzed. Subsequently, the role of therapeutic additives, including cells, growth factors, and other active products, in facilitating meniscus repair and regeneration is thoroughly discussed. Furthermore, we summarize the key issues for designing hydrogels used in MTE. Finally, we conclude with the current challenges encountered by hydrogel applications and suggest potential solutions for addressing these challenges in the field of MTE. We hope this review provides a resource for researchers and practitioners interested in this field, thereby facilitating the exploration of new design possibilities.
Collapse
Affiliation(s)
- Zhuxing Zhou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Jiajie Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Chaoqian Jiang
- School of Materials and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Kaiwang Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Tengjing Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Xinning Yu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Jinghua Fang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Yanyu Yang
- School of Materials and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xuesong Dai
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| |
Collapse
|
5
|
Kwokdinata C, Ramanujam V, Chen J, de Oliveira PN, Nai MH, Chooi WH, Lim CT, Ng SY, David L, Chew SY. Encapsulation of Human Spinal Cord Progenitor Cells in Hyaluronan-Gelatin Hydrogel for Spinal Cord Injury Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50679-50692. [PMID: 37751213 DOI: 10.1021/acsami.3c07419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Transplanting human induced pluripotent stem cells (iPSCs)-derived spinal cord progenitor cells (SCPCs) is a promising approach to treat spinal cord injuries. However, stem cell therapies face challenges in cell survival, cell localization to the targeted site, and the control of cell differentiation. Here, we encapsulated SCPCs in thiol-modified hyaluronan-gelatin hydrogels and optimized scaffold mechanical properties and cell encapsulation density to promote cell viability and neuronal differentiation in vitro and in vivo. Different compositions of hyaluronan-gelatin hydrogels formulated by varying concentrations of poly(ethylene glycol) diacrylate were mechanically characterized by using atomic force microscopy. In vitro SCPC encapsulation study showed higher cell viability and proliferation with lower substrate Young's modulus (200 Pa vs 580 Pa) and cell density. Moreover, the soft hydrogels facilitated a higher degree of neuronal differentiation with extended filament structures in contrast to clumped cellular morphologies obtained in stiff hydrogels (p < 0.01). When transplanted in vivo, the optimized SCPC-encapsulated hydrogels resulted in higher cell survival and localization at the transplanted region as compared to cell delivery without hydrogel encapsulation at 2 weeks postimplantation within the rat spinal cord (p < 0.01). Notably, immunostaining demonstrated that the hydrogel-encapsulated SCPCs differentiated along the neuronal and oligodendroglial lineages in vivo. The lack of pluripotency and proliferation also supported the safety of the SCPC transplantation approach. Overall, the injectable hyaluronan-gelatin hydrogel shows promise in supporting the survival and neural differentiation of human SCPCs after transplantation into the spinal cord.
Collapse
Affiliation(s)
- Christy Kwokdinata
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | - Vaibavi Ramanujam
- CNRS@CREATE, Create Tower #08-01, 1 Create Way, Singapore 138602, Singapore
| | - Jiahui Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | | | - Mui Hoon Nai
- Department of Biomedical Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Wai Hon Chooi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117576, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Shi Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Laurent David
- CNRS@CREATE, Create Tower #08-01, 1 Create Way, Singapore 138602, Singapore
- Ingénierie des Matériaux Polymères IMP UMR 5223, CNRS, Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, Université de Lyon, Villeurbanne F69622, France
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
6
|
Sharma A, Fernandes DC, Reis RL, Gołubczyk D, Neumann S, Lukomska B, Janowski M, Kortylewski M, Walczak P, Oliveira JM, Maciaczyk J. Cutting-edge advances in modeling the blood-brain barrier and tools for its reversible permeabilization for enhanced drug delivery into the brain. Cell Biosci 2023; 13:137. [PMID: 37501215 PMCID: PMC10373415 DOI: 10.1186/s13578-023-01079-3] [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: 04/21/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
The blood-brain barrier (BBB) is a sophisticated structure whose full functionality is required for maintaining the executive functions of the central nervous system (CNS). Tight control of transport across the barrier means that most drugs, particularly large size, which includes powerful biologicals, cannot reach their targets in the brain. Notwithstanding the remarkable advances in characterizing the cellular nature of the BBB and consequences of BBB dysfunction in pathology (brain metastasis, neurological diseases), it remains challenging to deliver drugs to the CNS. Herein, we outline the basic architecture and key molecular constituents of the BBB. In addition, we review the current status of approaches that are being explored to temporarily open the BBB in order to allow accumulation of therapeutics in the CNS. Undoubtedly, the major concern in field is whether it is possible to open the BBB in a meaningful way without causing negative consequences. In this context, we have also listed few other important key considerations that can improve our understanding about the dynamics of the BBB.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Stereotacitc and Functional Neurosurgery, University Hospital Bonn, 53127, Bonn, Germany
| | - Diogo C Fernandes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga, Portugal
| | - Dominika Gołubczyk
- Ti-Com, Polish Limited Liability Company, 10-683, Olsztyn, Poland
- Center for Translational Medicine, Warsaw University of Life Sciences, 02-797, Warsaw, Poland
| | - Silke Neumann
- Department of Pathology, University of Otago, Dunedin, 9054, New Zealand
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Miguel Oliveira
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga, Portugal.
| | - Jarek Maciaczyk
- Department of Stereotacitc and Functional Neurosurgery, University Hospital Bonn, 53127, Bonn, Germany.
- Department of Surgical Sciences, University of Otago, Dunedin, 9054, New Zealand.
| |
Collapse
|
7
|
Sun Z, Zhu D, Zhao H, Liu J, He P, Luan X, Hu H, Zhang X, Wei G, Xi Y. Recent advance in bioactive hydrogels for repairing spinal cord injury: material design, biofunctional regulation, and applications. J Nanobiotechnology 2023; 21:238. [PMID: 37488557 PMCID: PMC10364437 DOI: 10.1186/s12951-023-01996-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
Functional hydrogels show potential application in repairing spinal cord injury (SCI) due to their unique chemical, physical, and biological properties and functions. In this comprehensive review, we present recent advance in the material design, functional regulation, and SCI repair applications of bioactive hydrogels. Different from previously released reviews on hydrogels and three-dimensional scaffolds for the SCI repair, this work focuses on the strategies for material design and biologically functional regulation of hydrogels, specifically aiming to show how these significant efforts can promoting the repairing performance of SCI. We demonstrate various methods and techniques for the fabrication of bioactive hydrogels with the biological components such as DNA, proteins, peptides, biomass polysaccharides, and biopolymers to obtain unique biological properties of hydrogels, including the cell biocompatibility, self-healing, anti-bacterial activity, injectability, bio-adhesion, bio-degradation, and other multi-functions for repairing SCI. The functional regulation of bioactive hydrogels with drugs/growth factors, polymers, nanoparticles, one-dimensional materials, and two-dimensional materials for highly effective treating SCI are introduced and discussed in detail. This work shows new viewpoints and ideas on the design and synthesis of bioactive hydrogels with the state-of-the-art knowledges of materials science and nanotechnology, and will bridge the connection of materials science and biomedicine, and further inspire clinical potential of bioactive hydrogels in biomedical fields.
Collapse
Affiliation(s)
- Zhengang Sun
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266071, People's Republic of China
- Department of Spinal Surgery, Huangdao Central Hospital, Affiliated Hospital of Qingdao University, Qingdao, 266071, China
- The Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, People's Republic of China
| | - Danzhu Zhu
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Hong Zhao
- Department of Spinal Surgery, Huangdao Central Hospital, Affiliated Hospital of Qingdao University, Qingdao, 266071, China
| | - Jia Liu
- Department of Spinal Surgery, Huangdao Central Hospital, Affiliated Hospital of Qingdao University, Qingdao, 266071, China
| | - Peng He
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xin Luan
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Huiqiang Hu
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xuanfen Zhang
- The Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, People's Republic of China.
| | - Gang Wei
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, People's Republic of China.
| | - Yongming Xi
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
8
|
Pham TLB, Nguyen DPH, Luu TTT, Nguyen LS, Binh NT, Nguyen QD, Tran PA. Encapsulation of Human Umbilical Cord Mesenchymal Stem Cells in LunaGel Photocrosslinkable Extracellular Matrix and Subcutaneous Transplantation in Mice. Biomedicines 2023; 11:biomedicines11041158. [PMID: 37189776 DOI: 10.3390/biomedicines11041158] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Stem cells have significant potential in regenerative medicines. However, a major issue with implanting stem cells in the regeneration of new tissue is the methods to implant them and cell viability and functions before and after implantation. Here we developed a simple yet effective method that used photo-crosslinkable gelatin-based hydrogel (LunaGelTM) as a scaffold for the encapsulation, expansion, and eventually, transplantation of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) into mice subcutaneously. We demonstrated the proliferation and maintenance of the original expression of mesenchymal stem cell markers as well as the ability to differentiate into mesoderm-derived cells. The hydrogel was highly stable with no signs of degradation after 20 days in PBS. The hUC-MSCs remained viable after transplantation into mice's subcutaneous pockets and migrated to integrate with the surrounding tissues. We showed a collagen-rich layer surrounding the transplanted cell-laden scaffold indicating the effects of growth factors secreted by the hUC-MSCs. A connective tissue layer was found between the implanted cell-laden scaffold and the collagen layer, and immunohistochemical staining results suggested that this tissue was derived from the MSCs which migrated from within the scaffold. The results, thus, also suggested a protective effect the scaffold has on the encapsulated cells from the antibodies and cytotoxic cells of the host immune system.
Collapse
Affiliation(s)
- Truc Le-Buu Pham
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | | | - Thao Thi-Thu Luu
- Histology-Embryology-Pathology Department, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Luong Si Nguyen
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Nguyen Trong Binh
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Quan Dang Nguyen
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Phong Anh Tran
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
| |
Collapse
|
9
|
Vieira S, Strymecka P, Stanaszek L, Silva-Correia J, Drela K, Fiedorowicz M, Malysz-Cymborska I, Janowski M, Reis RL, Łukomska B, Walczak P, Oliveira JM. Mn-Based Methacrylated Gellan Gum Hydrogels for MRI-Guided Cell Delivery and Imaging. Bioengineering (Basel) 2023; 10:bioengineering10040427. [PMID: 37106614 PMCID: PMC10135712 DOI: 10.3390/bioengineering10040427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
This work aims to engineer a new stable injectable Mn-based methacrylated gellan gum (Mn/GG-MA) hydrogel for real-time monitored cell delivery into the central nervous system. To enable the hydrogel visualization under Magnetic Resonance Imaging (MRI), GG-MA solutions were supplemented with paramagnetic Mn2+ ions before its ionic crosslink with artificial cerebrospinal fluid (aCSF). The resulting formulations were stable, detectable by T1-weighted MRI scans and also injectable. Cell-laden hydrogels were prepared using the Mn/GG-MA formulations, extruded into aCSF for crosslink, and after 7 days of culture, the encapsulated human adipose-derived stem cells remained viable, as assessed by Live/Dead assay. In vivo tests, using double mutant MBPshi/shi/rag2 immunocompromised mice, showed that the injection of Mn/GG-MA solutions resulted in a continuous and traceable hydrogel, visible on MRI scans. Summing up, the developed formulations are suitable for both non-invasive cell delivery techniques and image-guided neurointerventions, paving the way for new therapeutic procedures.
Collapse
Affiliation(s)
- Sílvia Vieira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Paulina Strymecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Luiza Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Joana Silva-Correia
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Katarzyna Drela
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Michał Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Izabela Malysz-Cymborska
- Department of Neurology and Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Miroslaw Janowski
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | - Rui Luís Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Barbara Łukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Piotr Walczak
- Department of Neurology and Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | - Joaquim Miguel Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
- Correspondence: ; Tel.: +351-253510931; Fax: +351-253510909
| |
Collapse
|
10
|
Sedighi M, Shrestha N, Mahmoudi Z, Khademi Z, Ghasempour A, Dehghan H, Talebi SF, Toolabi M, Préat V, Chen B, Guo X, Shahbazi MA. Multifunctional Self-Assembled Peptide Hydrogels for Biomedical Applications. Polymers (Basel) 2023; 15:1160. [PMID: 36904404 PMCID: PMC10007692 DOI: 10.3390/polym15051160] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Self-assembly is a growth mechanism in nature to apply local interactions forming a minimum energy structure. Currently, self-assembled materials are considered for biomedical applications due to their pleasant features, including scalability, versatility, simplicity, and inexpensiveness. Self-assembled peptides can be applied to design and fabricate different structures, such as micelles, hydrogels, and vesicles, by diverse physical interactions between specific building blocks. Among them, bioactivity, biocompatibility, and biodegradability of peptide hydrogels have introduced them as versatile platforms in biomedical applications, such as drug delivery, tissue engineering, biosensing, and treating different diseases. Moreover, peptides are capable of mimicking the microenvironment of natural tissues and responding to internal and external stimuli for triggered drug release. In the current review, the unique characteristics of peptide hydrogels and recent advances in their design, fabrication, as well as chemical, physical, and biological properties are presented. Additionally, recent developments of these biomaterials are discussed with a particular focus on their biomedical applications in targeted drug delivery and gene delivery, stem cell therapy, cancer therapy and immune regulation, bioimaging, and regenerative medicine.
Collapse
Affiliation(s)
- Mahsa Sedighi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Birjand University of Medical Sciences, Birjand 9717853076, Iran
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Neha Shrestha
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Biomedicine and Translational Research, Research Institute for Bioscience and Biotechnology, Kathmandu P.O. Box 7731, Nepal
| | - Zahra Mahmoudi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan 6517838636, Iran
| | - Zahra Khademi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Alireza Ghasempour
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Hamideh Dehghan
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Seyedeh Fahimeh Talebi
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Maryam Toolabi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Bozhi Chen
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xindong Guo
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
11
|
Rybachuk O, Savytska N, Pinet É, Yaminsky Y, Medvediev V. Heterogeneous pHPMA hydrogel promotes neuronal differentiation of bone marrow derived stromal cells in vitroand in vivo. Biomed Mater 2023; 18. [PMID: 36542861 DOI: 10.1088/1748-605x/acadc3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Synthetic hydrogels composed of polymer pore frames are commonly used in medicine, from pharmacologically targeted drug delivery to the creation of bioengineering constructions used in implantation surgery. Among various possible materials, the most common are poly-[N(2-hydroxypropyl)methacrylamide] (pHPMA) derivatives. One of the pHPMA derivatives is biocompatible hydrogel, NeuroGel. Upon contact with nervous tissue, the NeuroGel's structure can support the chemical and physiological conditions of the tissue necessary for the growth of native cells. Owing to the different pore diameters in the hydrogel, not only macromolecules, but also cells can migrate. This study evaluated the differentiation of bone marrow stromal cells (BMSCs) into neurons, as well as the effectiveness of using this biofabricated system in spinal cord injuryin vivo. The hydrogel was populated with BMSCs by injection or rehydration. After cultivation, these fragments (hydrogel + BMSCs) were implanted into the injured rat spinal cord. Fragments were immunostained before implantation and seven months after implantation. During cultivation with the hydrogel, both variants (injection/rehydration) of the BMSCs culture retained their viability and demonstrated a significant number of Ki-67-positive cells, indicating the preservation of their proliferative activity. In hydrogel fragments, BMSCs also maintained their viability during the period of cocultivation and were Ki-67-positive, but in significantly fewer numbers than in the cell culture. In addition, in fragments of hydrogel with grafted BMSCs, both by the injection or rehydration versions, we observed a significant number up to 57%-63.5% of NeuN-positive cells. These results suggest that the heterogeneous pHPMA hydrogel promotes neuronal differentiation of bone marrow-derived stromal cells. Furthermore, these data demonstrate the possible use of NeuroGel implants with grafted BMSCs for implantation into damaged areas of the spinal cord, with subsequent nerve fiber germination, nerve cell regeneration, and damaged segment restoration.
Collapse
Affiliation(s)
- Oksana Rybachuk
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine.,Institute of Genetic and Regenerative Medicine, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, NAMS of Ukraine, Kyiv, Ukraine
| | - Natalia Savytska
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine.,German Center for Neurodegenerative Diseases, Tübingen, Germany
| | | | - Yurii Yaminsky
- State Institution 'Romodanov Neurosurgery Institute, NAMS of Ukraine', Kyiv, Ukraine
| | - Volodymyr Medvediev
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine.,Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
12
|
Calder D, Fathi A, Oveissi F, Maleknia S, Abrams T, Wang Y, Maitz J, Tsai KH, Maitz P, Chrzanowski W, Canoy I, Menon VA, Lee K, Ahern BJ, Lean NE, Silva DM, Young PM, Traini D, Ong HX, Mahmoud RS, Montazerian H, Khademhosseini A, Dehghani F. Thermoresponsive and Injectable Hydrogel for Tissue Agnostic Regeneration. Adv Healthc Mater 2022; 11:e2201714. [PMID: 36148581 PMCID: PMC11468498 DOI: 10.1002/adhm.202201714] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/16/2022] [Indexed: 01/28/2023]
Abstract
Injectable hydrogels can support the body's innate healing capability by providing a temporary matrix for host cell ingrowth and neovascularization. The clinical adoption of current injectable systems remains low due to their cumbersome preparation requirements, device malfunction, product dislodgment during administration, and uncontrolled biological responses at the treatment site. To address these challenges, a fully synthetic and ready-to-use injectable biomaterial is engineered that forms an adhesive hydrogel that remains at the administration site regardless of defect anatomy. The product elicits a negligible local inflammatory response and fully resorbs into nontoxic components with minimal impact on internal organs. Preclinical animal studies confirm that the engineered hydrogel upregulates the regeneration of both soft and hard tissues by providing a temporary matrix to support host cell ingrowth and neovascularization. In a pilot clinical trial, the engineered hydrogel is successfully administered to a socket site post tooth extraction and forms adhesive hydrogel that stabilizes blood clot and supports soft and hard tissue regeneration. Accordingly, this injectable hydrogel exhibits high therapeutic potential and can be adopted to address multiple unmet needs in different clinical settings.
Collapse
Affiliation(s)
- Dax Calder
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Faculty of Medicine and HealthNano InstituteThe University of SydneySydneyNSW2006Australia
- Faculty of Health and Medical SciencesSchool of Biomedical SciencesUniversity of Western AustraliaPerthWA6009Australia
| | - Ali Fathi
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- TetratherixSydneyNSW2015Australia
| | - Farshad Oveissi
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
| | | | | | - Yiwei Wang
- Burns and Reconstructive Surgery Research GroupANZAC Research InstituteConcordNSW2139Australia
| | - Joanneke Maitz
- Burns and Reconstructive Surgery Research GroupANZAC Research InstituteConcordNSW2139Australia
| | - Kevin Hung‐Yueh Tsai
- Burns and Reconstructive Surgery Research GroupANZAC Research InstituteConcordNSW2139Australia
| | - Peter Maitz
- Burns and Reconstructive Surgery Research GroupANZAC Research InstituteConcordNSW2139Australia
| | - Wojtek Chrzanowski
- Faculty of Medicine and HealthNano InstituteThe University of SydneySydneyNSW2006Australia
- Faculty of Health and Medical SciencesSchool of Biomedical SciencesUniversity of Western AustraliaPerthWA6009Australia
| | - Ivan Canoy
- Anatomical PathologyNSW Health PathologyConcord Repatriation General HospitalConcordNSW2139Australia
| | - Vivek Ashoka Menon
- Anatomical PathologyNSW Health PathologyConcord Repatriation General HospitalConcordNSW2139Australia
| | - Kenneth Lee
- Anatomical PathologyNSW Health PathologyConcord Repatriation General HospitalConcordNSW2139Australia
- School of MedicineUniversity of SydneySydneyNSW2006Australia
| | - Benjamin J. Ahern
- School of Veterinary ScienceThe University of QueenslandBrisbaneQLD4072Australia
| | - Natasha E. Lean
- School of Veterinary ScienceThe University of QueenslandBrisbaneQLD4072Australia
| | - Dina M. Silva
- Macquarie Medical SchoolFaculty of Medicine and HealthMacquarie University & Woolcock Institute of Medical ResearchThe University of SydneyGlebeNSW2037Australia
- Ab Initio PharmaCamperdownNSW2050Australia
| | - Paul M. Young
- Macquarie Medical SchoolFaculty of Medicine and HealthMacquarie University & Woolcock Institute of Medical ResearchThe University of SydneyGlebeNSW2037Australia
- Ab Initio PharmaCamperdownNSW2050Australia
| | - Daniela Traini
- Macquarie Medical SchoolFaculty of Medicine and HealthMacquarie University & Woolcock Institute of Medical ResearchThe University of SydneyGlebeNSW2037Australia
- Ab Initio PharmaCamperdownNSW2050Australia
| | - Hui Xin Ong
- Macquarie Medical SchoolFaculty of Medicine and HealthMacquarie University & Woolcock Institute of Medical ResearchThe University of SydneyGlebeNSW2037Australia
- Ab Initio PharmaCamperdownNSW2050Australia
| | | | - Hossein Montazerian
- Terasaki Institute for Biomedical InnovationLos AngelesCA90024USA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCA90095USA
- California NanoSystems Institute (CNSI)University of CaliforniaLos AngelesCA90095USA
| | | | - Fariba Dehghani
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
| |
Collapse
|
13
|
Changes in the Mechanical Properties of Alginate-Gelatin Hydrogels with the Addition of Pygeum africanum with Potential Application in Urology. Int J Mol Sci 2022; 23:ijms231810324. [PMID: 36142228 PMCID: PMC9499472 DOI: 10.3390/ijms231810324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/05/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
New hydrogel materials developed to improve soft tissue healing are an alternative for medical applications, such as tissue regeneration or enhancing the biotolerance effect in the tissue-implant–body fluid system. The biggest advantages of hydrogel materials are the presence of a large amount of water and a polymeric structure that corresponds to the extracellular matrix, which allows to create healing conditions similar to physiological ones. The present work deals with the change in mechanical properties of sodium alginate mixed with gelatin containing Pygeum africanum. The work primarily concentrates on the evaluation of the mechanical properties of the hydrogel materials produced by the sol–gel method. The antimicrobial activity of the hydrogels was investigated based on the population growth dynamics of Escherichia coli ATCC 25922 and Staphylococcus aureus ATCC 25923, as well as the degree of degradation after contact with urine using an innovative method with a urine flow simulation stand. On the basis of mechanical tests, it was found that sodium alginate-based hydrogels with gelatin showed weaker mechanical properties than without the additive. In addition, gelatin accelerates the degradation process of the produced hydrogel materials. Antimicrobial studies have shown that the presence of African plum bark extract in the hydrogel enhances the inhibitory effect on Gram-positive and Gram-negative bacteria. The research topic was considered due to the increased demand from patients for medical devices to promote healing of urethral epithelial injuries in order to prevent the formation of urethral strictures.
Collapse
|
14
|
Wilson KL, Pérez SCL, Naffaa MM, Kelly SH, Segura T. Stoichiometric Post-Modification of Hydrogel Microparticles Dictates Neural Stem Cell Fate in Microporous Annealed Particle Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201921. [PMID: 35731241 PMCID: PMC9645378 DOI: 10.1002/adma.202201921] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/07/2022] [Indexed: 05/16/2023]
Abstract
Microporous annealed particle (MAP) scaffolds are generated from assembled hydrogel microparticles (microgels). It has been previously demonstrated that MAP scaffold are porous, biocompatible, and recruit neural progenitor cells (NPCs) to the stroke cavity after injection into the stroke core. Here, the goal is to study NPC fate inside MAP scaffolds in vitro. To create plain microgels that can later be converted to contain different types of bioactivities, the inverse electron-demand Diels-Alder reaction between tetrazine and norbornene is utilized, which allows the post-modification of plain microgels stoichiometrically. As a result of adhesive peptide attachment, NPC spreading leads to contractile force generation which can be recorded by tracking microgel displacement. Alternatively, non-adhesive peptide integration results in neurosphere formation that grows within the void space of MAP scaffolds. Although the formed neurospheres do not impose a contractile force on the scaffolds, they are seen to continuously transverse the scaffolds. It is concluded that MAP scaffolds can be engineered to either promote neurogenesis or enhance stemness depending on the chosen post-modifications of the microgels, which can be key in modulating their phenotypes in various applications in vivo.
Collapse
Affiliation(s)
- Katrina L Wilson
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708-0281, USA
| | - Sasha Cai Lesher Pérez
- Department of Chemical Engineering, University of Michigan, North Campus Research Complex, Building 28, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA
| | - Moawiah M Naffaa
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Psychology and Neuroscience, Duke University, Durham, NC, 27708, USA
| | - Sean H Kelly
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708-0281, USA
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708-0281, USA
- Department of Neurology, Duke University, Durham, NC, 27708-0281, USA
- Department of Dermatology, Duke University, Durham, NC, 27708-0281, USA
| |
Collapse
|
15
|
Eco-Friendly Bio-Hydrogels Based on Antheraea Pernyi Silk Gland Protein for Cell and Drug Delivery. Gels 2022; 8:gels8070398. [PMID: 35877483 PMCID: PMC9321860 DOI: 10.3390/gels8070398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
The Antheraea Pernyi silk gland protein originates from natural organisms and synthesized by tussah silk glands and has widely potential biomaterial applications due to the superior biocompatibility. This study investigates the Antheraea Pernyi silk gland protein-based drug-loaded bio-hydrogels for bioengineered tissue fabricated by using an eco-friendly method without the harsh extracting process and the usage of toxic chemicals. The drug-loaded bio-hydrogels exhibited a porous structure and interconnected pore walls. The swelling ratio and water absorption of drug-loaded bio-hydrogels were, respectively, above 95% and 1.5 × 103%. The cumulative release of drug loaded hydrogels all reached more than 90% within 4 h, and this indicates the potential of drug-loaded hydrogels as future drug-carrying biomaterials. RSC96 Schwann cells cultured on drug-loaded hydrogels for 72 h under cell culture medium show no toxic effects and more pro-proliferative effects. The results suggest the suitability of drug-loaded bio-hydrogels as natural biopolymer for the potential in vitro RSC96 cell culture platform and other biomaterial applications.
Collapse
|
16
|
Araszkiewicz AM, Oliveira EP, Svendsen T, Drela K, Rogujski P, Malysz-Cymborska I, Fiedorowicz M, Reis RL, Oliveira JM, Walczak P, Janowski M, Lukomska B, Stanaszek L. Manganese-Labeled Alginate Hydrogels for Image-Guided Cell Transplantation. Int J Mol Sci 2022; 23:ijms23052465. [PMID: 35269609 PMCID: PMC8910205 DOI: 10.3390/ijms23052465] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 02/04/2023] Open
Abstract
Cell transplantation has been studied extensively as a therapeutic strategy for neurological disorders. However, to date, its effectiveness remains unsatisfactory due to low precision and efficacy of cell delivery; poor survival of transplanted cells; and inadequate monitoring of their fate in vivo. Fortunately, different bio-scaffolds have been proposed as cell carriers to improve the accuracy of cell delivery, survival, differentiation, and controlled release of embedded stem cells. The goal of our study was to establish hydrogel scaffolds suitable for stem cell delivery that also allow non-invasive magnetic resonance imaging (MRI). We focused on alginate-based hydrogels due to their natural origin, biocompatibility, resemblance to the extracellular matrix, and easy manipulation of gelation processes. We optimized the properties of alginate-based hydrogels, turning them into suitable carriers for transplanted cells. Human adipose-derived stem cells embedded in these hydrogels survived for at least 14 days in vitro. Alginate-based hydrogels were also modified successfully to allow their injectability via a needle. Finally, supplementing alginate hydrogels with Mn ions or Mn nanoparticles allowed for their visualization in vivo using manganese-enhanced MRI. We demonstrated that modified alginate-based hydrogels can support therapeutic cells as MRI-detectable matrices.
Collapse
Affiliation(s)
- Antonina M. Araszkiewicz
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.M.A.); (P.R.); (B.L.)
| | - Eduarda P. Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal; (E.P.O.); (R.L.R.); (J.M.O.)
- ICVS/3B’s—PT Government Associated Laboratory, 4710-057 Guimarães, Portugal
| | | | | | - Piotr Rogujski
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.M.A.); (P.R.); (B.L.)
| | - Izabela Malysz-Cymborska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland;
| | - Michal Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal; (E.P.O.); (R.L.R.); (J.M.O.)
- ICVS/3B’s—PT Government Associated Laboratory, 4710-057 Guimarães, Portugal
| | - Joaquim Miguel Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal; (E.P.O.); (R.L.R.); (J.M.O.)
- ICVS/3B’s—PT Government Associated Laboratory, 4710-057 Guimarães, Portugal
| | - Piotr Walczak
- Program for Image Guided Neurointerventions, Department of Diagnostic Radiology and Nuclear Medicine, Center for Advanced Imaging Research, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA; (P.W.); (M.J.)
| | - Miroslaw Janowski
- Program for Image Guided Neurointerventions, Department of Diagnostic Radiology and Nuclear Medicine, Center for Advanced Imaging Research, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA; (P.W.); (M.J.)
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.M.A.); (P.R.); (B.L.)
| | - Luiza Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.M.A.); (P.R.); (B.L.)
- Correspondence: ; Tel.: +48-226-086-529
| |
Collapse
|
17
|
Deng Y, Li R, Wang H, Yang B, Shi P, Zhang Y, Yang Q, Li G, Bian L. Biomaterial-Mediated Presentation of Jagged-1 Mimetic Ligand Enhances Cellular Activation of Notch Signaling and Bone Regeneration. ACS NANO 2022; 16:1051-1062. [PMID: 34967609 DOI: 10.1021/acsnano.1c08728] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The development from stem cells to adult tissues requires the delicate presentation of numerous crucial inductive cues and the activation of associated signaling pathways. The Notch signaling pathways triggered by ligands such as Jagged-1 have been demonstrated to be essential in various development processes especially in osteogenesis and ossification. However, few studies have capitalized on the osteoinductivity of the Jagged-1 mimetic ligands to enhance the osteogenesis and skeleton regeneration. In this study, we conjugate the porous hyaluronic acid hydrogels with a Jagged-1 mimetic peptide ligand (Jagged-1) and investigate the efficacy of such biomimetic functionalization to promote the mechanotransduction and osteogenesis of human mesenchymal stem cells by activating the Notch signaling pathway. Our findings indicate that the immobilized Jagged-1 mimetic ligand activates Notch signaling via the upregulation of NICD and downstream MSX2, leading to the enhanced mechanotransduction and osteogenesis of stem cells. We further demonstrate that the functionalization of the Jagged-1 ligand in the porous scaffold promotes angiogenesis, regulates macrophage recruitment and polarization, and enhances in situ regeneration of rat calvarial defects. Our findings provide valuable guidance to the design of development-inspired bioactive biomaterials for diverse biomedical applications.
Collapse
Affiliation(s)
- Yingrui Deng
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P.R. China
| | - Rui Li
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, New York 11201, United States
| | - Haixing Wang
- Department of Orthopaedics and Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, P.R China
| | - Boguang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P.R. China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yuan Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin 300211, P.R. China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, P.R China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| |
Collapse
|
18
|
Xue Y, Baig R, Dong Y. Recent advances of biomaterials in stem cell therapies. NANOTECHNOLOGY 2022; 33:10.1088/1361-6528/ac4520. [PMID: 34933291 PMCID: PMC10068913 DOI: 10.1088/1361-6528/ac4520] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
Stem cells have been utilized as 'living drugs' in clinics for decades. Their self-renewal, differentiation, and immunomodulating properties provide potential solutions for a variety of malignant diseases and disorders. However, the pathological environment may diminish the therapeutic functions and survival of the transplanted stem cells, causing failure in clinical translation. To overcome these challenges, researchers have developed biomaterial-based strategies that facilitatein vivotracking, functional engineering, and protective delivery of stem cells, paving the way for next-generation stem cell therapies. In this perspective, we briefly overview different types of stem cells and the major clinical challenges and summarize recent progress of biomaterials applied to boost stem cell therapies.
Collapse
Affiliation(s)
- Yonger Xue
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States of America
| | - Rafia Baig
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States of America
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States of America
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, United States of America
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH 43210, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States of America
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States of America
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, United States of America
| |
Collapse
|
19
|
Samal J, Segura T. Injectable biomaterial shuttles for cell therapy in stroke. Brain Res Bull 2021; 176:25-42. [PMID: 34391821 PMCID: PMC8524625 DOI: 10.1016/j.brainresbull.2021.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 07/26/2021] [Accepted: 08/06/2021] [Indexed: 01/01/2023]
Abstract
Ischemic stroke (IS) is the leading cause of disability and contributes to a significant socio-economic cost in the western world. Brain repair strategies investigated in the pre-clinical models include the delivery of drug or cell-based therapeutics; which is hindered by the complex anatomy and functional organization of the brain. Biomaterials can be instrumental in alleviating some of these challenges by providing a structural support, localization, immunomodulation and/or modulating cellular cross-talk in the brain. This review addresses the significance of and challenges associated with cell therapy in an ischemic brain. This is followed by a detailed insight into the biomaterial-based delivery systems which have been designed to provide sustained trophic factor delivery for endogenous repair and to support transplanted cell survival and integration. A biomaterial intervention uses a multifaceted approach in enhancing the survival and engraftment of cells during transplantation and this has driven them as potential candidates for the treatment of IS. The biological processes that are activated as a response to the biomaterials and how to modulate them is one of the key factors contributing to the success of the biomaterial-based therapeutic approach. Future perspectives highlight the need of a combinative approach of merging the material design with disease biology to fabricate effective biomaterial-based intervention of stroke.
Collapse
Affiliation(s)
- Juhi Samal
- Department of Biomedical Engineering, 534 Research Drive, Durham, NC 27708, United States
| | - Tatiana Segura
- Department of Biomedical Engineering, 534 Research Drive, Durham, NC 27708, United States.
| |
Collapse
|
20
|
Duan X, Li N, Chen X, Zhu N. Characterization of Tissue Scaffolds Using Synchrotron Radiation Microcomputed Tomography Imaging. Tissue Eng Part C Methods 2021; 27:573-588. [PMID: 34670397 DOI: 10.1089/ten.tec.2021.0155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Distinguishing from other traditional imaging, synchrotron radiation microcomputed tomography (SR-μCT) imaging allows for the visualization of three-dimensional objects of interest in a nondestructive and/or in situ way with better spatial resolution, deep penetration, relatively fast speed, and/or high contrast. SR-μCT has been illustrated promising for visualizing and characterizing tissue scaffolds for repairing or replacing damaged tissue or organs in tissue engineering (TE), which is of particular advance for longitudinal monitoring and tracking the success of scaffolds once implanted in animal models and/or human patients. This article presents a comprehensive review on recent studies of characterization of scaffolds based on SR-μCT and takes scaffold architectural properties, mechanical properties, degradation, swelling and wettability, and biological properties as five separate sections to introduce SR-μCT wide applications. We also discuss and highlight the unique opportunities of SR-μCT in various TE applications; conclude this article with the suggested future research directions, including the prospective applications of SR-μCT, along with its challenges and methods for improvement in the field of TE.
Collapse
Affiliation(s)
- Xiaoman Duan
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
| | - Naitao Li
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
| | - Ning Zhu
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
- Department of Chemical and Biological Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
- Canadian Light Source, Saskatoon, Canada
| |
Collapse
|
21
|
Halim A, Qu KY, Zhang XF, Huang NP. Recent Advances in the Application of Two-Dimensional Nanomaterials for Neural Tissue Engineering and Regeneration. ACS Biomater Sci Eng 2021; 7:3503-3529. [PMID: 34291638 DOI: 10.1021/acsbiomaterials.1c00490] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complexity of the nervous system structure and function, and its slow regeneration rate, makes it more difficult to treat compared to other tissues in the human body when an injury occurs. Moreover, the current therapeutic approaches including the use of autografts, allografts, and pharmacological agents have several drawbacks and can not fully restore nervous system injuries. Recently, nanotechnology and tissue engineering approaches have attracted many researchers to guide tissue regeneration in an effective manner. Owing to their remarkable physicochemical and biological properties, two-dimensional (2D) nanomaterials have been extensively studied in the tissue engineering and regenerative medicine field. The great conductivity of these materials makes them a promising candidate for the development of novel scaffolds for neural tissue engineering application. Moreover, the high loading capacity of 2D nanomaterials also has attracted many researchers to utilize them as a drug/gene delivery method to treat various devastating nervous system disorders. This review will first introduce the fundamental physicochemical properties of 2D nanomaterials used in biomedicine and the supporting biological properties of 2D nanomaterials for inducing neuroregeneration, including their biocompatibility on neural cells, the ability to promote the neural differentiation of stem cells, and their immunomodulatory properties which are beneficial for alleviating chronic inflammation at the site of the nervous system injury. It also discusses various types of 2D nanomaterials-based scaffolds for neural tissue engineering applications. Then, the latest progress on the use of 2D nanomaterials for nervous system disorder treatment is summarized. Finally, a discussion of the challenges and prospects of 2D nanomaterials-based applications in neural tissue engineering is provided.
Collapse
Affiliation(s)
- Alexander Halim
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Kai-Yun Qu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Xiao-Feng Zhang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Ning-Ping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| |
Collapse
|
22
|
Kalkowski L, Golubczyk D, Kwiatkowska J, Holak P, Milewska K, Janowski M, Oliveira JM, Walczak P, Malysz-Cymborska I. Two in One: Use of Divalent Manganese Ions as Both Cross-Linking and MRI Contrast Agent for Intrathecal Injection of Hydrogel-Embedded Stem Cells. Pharmaceutics 2021; 13:pharmaceutics13071076. [PMID: 34371767 PMCID: PMC8309201 DOI: 10.3390/pharmaceutics13071076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/04/2022] Open
Abstract
Cell therapy is a promising tool for treating central nervous system (CNS) disorders; though, the translational efforts are plagued by ineffective delivery methods. Due to the large contact surface with CNS and relatively easy access, the intrathecal route of administration is attractive in extensive or global diseases such as stroke or amyotrophic lateral sclerosis (ALS). However, the precision and efficacy of this approach are still a challenge. Hydrogels were introduced to minimize cell sedimentation and improve cell viability. At the same time, contrast agents were integrated to allow image-guided injection. Here, we report using manganese ions (Mn2+) as a dual agent for cross-linking alginate-based hydrogels and magnetic resonance imaging (MRI). We performed in vitro studies to test the Mn2+ alginate hydrogel formulations for biocompatibility, injectability, MRI signal retention time, and effect on cell viability. The selected formulation was injected intrathecally into pigs under MRI control. The biocompatibility test showed a lack of immune response, and cells suspended in the hydrogel showed greater viability than monolayer culture. Moreover, Mn2+-labeled hydrogel produced a strong T1 MRI signal, which enabled MRI-guided procedure. We confirmed the utility of Mn2+ alginate hydrogel as a carrier for cells in large animals and a contrast agent at the same time.
Collapse
Affiliation(s)
- Lukasz Kalkowski
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
| | - Dominika Golubczyk
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
| | - Joanna Kwiatkowska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
| | - Piotr Holak
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| | - Kamila Milewska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
| | - Miroslaw Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (M.J.); (P.W.)
| | - Joaquim Miguel Oliveira
- a3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, 4806-909 Guimarães, Portugal
| | - Piotr Walczak
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (M.J.); (P.W.)
| | - Izabela Malysz-Cymborska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
- Correspondence: ; Tel.: +48-605118887
| |
Collapse
|
23
|
Thomas HM, Ahangar P, Fitridge R, Kirby GTS, Mills SJ, Cowin AJ. Plasma-polymerized pericyte patches improve healing of murine wounds through increased angiogenesis and reduced inflammation. Regen Biomater 2021; 8:rbab024. [PMID: 34221447 PMCID: PMC8242226 DOI: 10.1093/rb/rbab024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022] Open
Abstract
Pericytes have the potential to be developed as a cell therapy for the treatment of wounds; however, the efficacy of any cell therapy relies on the successful delivery of intact and functioning cells. Here, the effect of delivering pericytes on wound repair was assessed alongside the development of a surface-functionalized pericyte patch. Plasma polymerization (PP) was used to functionalize the surface of silicone patches with heptylamine (HA) or acrylic acid (AA) monomers. Human pericytes were subsequently delivered to murine excisional wounds by intradermal injection or using the pericyte-laden patches and the comparative effects on wound healing, inflammation and revascularization determined. The AA surface provided the superior transfer of the cells to de-epidermized dermis. Excisional murine wounds treated either with pericytes injected directly into the wound or with the pericyte-laden AA patches showed improved healing with decreased neutrophil infiltration and reduced numbers of macrophages in the wounds. Pericyte delivery also enhanced angiogenesis through a mechanism independent of VEGF signalling. Pericytes, when delivered to wounds, improved healing responses by dampening inflammation and promoting angiogenesis. Delivery of pericytes using PP-AA-functionalized patches was equally as effective as direct injection of pericytes into wounds. Pericyte-functionalized dressings may therefore be a clinically relevant approach for the treatment of wounds.
Collapse
Affiliation(s)
- Hannah M Thomas
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide SA 5000, Australia
| | - Parinaz Ahangar
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide SA 5000, Australia
| | - Robert Fitridge
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide SA 5005, Australia
| | - Giles T S Kirby
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia
| | - Stuart J Mills
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide SA 5000, Australia
| | - Allison J Cowin
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide SA 5000, Australia
| |
Collapse
|
24
|
Zhang X, Gong B, Zhai J, Zhao Y, Lu Y, Zhang L, Xue J. A Perspective: Electrospun Fibers for Repairing Spinal Cord Injury. Chem Res Chin Univ 2021. [DOI: 10.1007/s40242-021-1162-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
25
|
Gebeyehu A, Surapaneni SK, Huang J, Mondal A, Wang VZ, Haruna NF, Bagde A, Arthur P, Kutlehria S, Patel N, Rishi AK, Singh M. Polysaccharide hydrogel based 3D printed tumor models for chemotherapeutic drug screening. Sci Rep 2021; 11:372. [PMID: 33431915 PMCID: PMC7801509 DOI: 10.1038/s41598-020-79325-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 12/02/2020] [Indexed: 01/29/2023] Open
Abstract
A series of stable and ready-to-use bioinks have been developed based on the xeno-free and tunable hydrogel (VitroGel) system. Cell laden scaffold fabrication with optimized polysaccharide-based inks demonstrated that Ink H4 and RGD modified Ink H4-RGD had excellent rheological properties. Both bioinks were printable with 25-40 kPa extrusion pressure, showed 90% cell viability, shear-thinning and rapid shear recovery properties making them feasible for extrusion bioprinting without UV curing or temperature adjustment. Ink H4-RGD showed printability between 20 and 37 °C and the scaffolds remained stable for 15 days at temperature of 37 °C. 3D printed non-small-cell lung cancer (NSCLC) patient derived xenograft cells (PDCs) showed rapid spheroid growth of size around 500 µm in diameter and tumor microenvironment formation within 7 days. IC50 values demonstrated higher resistance of 3D spheroids to docetaxel (DTX), doxorubicin (DOX) and erlotinib compared to 2D monolayers of NSCLC-PDX, wild type triple negative breast cancer (MDA-MB-231 WT) and lung adenocarcinoma (HCC-827) cells. Results of flow property, shape fidelity, scaffold stability and biocompatibility of H4-RGD suggest that this hydrogel could be considered for 3D cell bioprinting and also for in-vitro tumor microenvironment development for high throughput screening of various anti-cancer drugs.
Collapse
Affiliation(s)
- Aragaw Gebeyehu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Sunil Kumar Surapaneni
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - John Huang
- TheWell Bioscience, North Brunswick, New Jersey, 08902, USA
| | - Arindam Mondal
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | | | | | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Peggy Arthur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Shallu Kutlehria
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Nil Patel
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Arun K Rishi
- John D. Dingell VA Medical Center, Karmanos Cancer Institute, Department of Oncology, Wayne State University, Detroit, MI, 48201, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA.
| |
Collapse
|
26
|
Kook YM, Hwang S, Kim H, Rhee KJ, Lee K, Koh WG. Cardiovascular tissue regeneration system based on multiscale scaffolds comprising double-layered hydrogels and fibers. Sci Rep 2020; 10:20321. [PMID: 33230134 PMCID: PMC7683622 DOI: 10.1038/s41598-020-77187-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022] Open
Abstract
We report a technique to reconstruct cardiovascular tissue using multiscale scaffolds incorporating polycaprolactone fibers with double-layered hydrogels comprising fibrin hydrogel surrounded by secondary alginate hydrogel. The scaffolds compartmentalized cells into the core region of cardiac tissue and the peripheral region of blood vessels to construct cardiovascular tissue, which was accomplished by a triple culture system of adipose-derived mesenchymal stem cells (ADSCs) with C2C12 myoblasts on polycaprolactone (PCL) fibers along with human umbilical vein endothelial cells (HUVECs) in fibrin hydrogel. The secondary alginate hydrogel prevented encapsulated cells from migrating outside scaffold and maintained the scaffold structure without distortion after subcutaneous implantation. According to in vitro studies, resultant scaffolds promoted new blood vessel formation as well as cardiomyogenic phenotype expression of ADSCs. Cardiac muscle-specific genes were expressed from stem cells and peripheral blood vessels from HUVECs were also successfully developed in subcutaneously implanted cell-laden multiscale scaffolds. Furthermore, the encapsulated stem cells modulated the immune response of scaffolds by secreting anti-inflammatory cytokines for successful tissue construction. Our study reveals that multiscale scaffolds can be promising for the remodeling and transplantation of cardiovascular tissue.
Collapse
Affiliation(s)
- Yun-Min Kook
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea
| | - Soonjae Hwang
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University at Wonju, Wonju, Gangwon-do, 220-710, Republic of Korea.,Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, Republic of Korea
| | - Hyerim Kim
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University at Wonju, Wonju, Gangwon-do, 220-710, Republic of Korea
| | - Kangwon Lee
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea. .,Advanced Institutes of Convergence Technology, Suwon, Gyeonggi-do, Republic of Korea.
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-749, Republic of Korea.
| |
Collapse
|
27
|
Qu W, Chen B, Shu W, Tian H, Ou X, Zhang X, Wang Y, Wu M. Polymer-Based Scaffold Strategies for Spinal Cord Repair and Regeneration. Front Bioeng Biotechnol 2020; 8:590549. [PMID: 33117788 PMCID: PMC7576679 DOI: 10.3389/fbioe.2020.590549] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022] Open
Abstract
The injury to the spinal cord is among the most complex fields of medical development. Spinal cord injury (SCI) leads to acute loss of motor and sensory function beneath the injury level and is linked to a dismal prognosis. Currently, while a strategy that could heal the injured spinal cord remains unforeseen, the latest advancements in polymer-mediated approaches demonstrate promising treatment forms to remyelinate or regenerate the axons and to integrate new neural cells in the SCI. Moreover, they possess the capacity to locally deliver synergistic cells, growth factors (GFs) therapies and bioactive substances, which play a critical role in neuroprotection and neuroregeneration. Here, we provide an extensive overview of the SCI characteristics, the pathophysiology of SCI, and strategies and challenges for the treatment of SCI in a review. This review highlights the recent encouraging applications of polymer-based scaffolds in developing the novel SCI therapy.
Collapse
Affiliation(s)
- Wenrui Qu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Bingpeng Chen
- The Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China
| | - Wentao Shu
- Department of Biobank, Division of Clinical Research, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Heng Tian
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xiaolan Ou
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xi Zhang
- Department of Burn Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yinan Wang
- Department of Biobank, Division of Clinical Research, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Minfei Wu
- The Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Piejko M, Walczak P, Li X, Bulte JWM, Janowski M. In Vitro Assessment of Fluorine Nanoemulsion-Labeled Hyaluronan-Based Hydrogels for Precise Intrathecal Transplantation of Glial-Restricted Precursors. Mol Imaging Biol 2020; 21:1071-1078. [PMID: 30850968 DOI: 10.1007/s11307-019-01341-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE We studied the feasibility of labeling hydrogel scaffolds with a fluorine nanoemulsion for 19F- magnetic resonance imaging (MRI) to enable non-invasive visualization of their precise placement and potential degradation. PROCEDURE Hyaluronan-based hydrogels (activated hyaluronan, HA) with increasing concentrations of fluorine nanoemulsion (V-sense) were prepared to measure the gelation time and oscillatory stress at 1 h and 7 days after the beginning of gelation. All biomechanical measurements were conducted with an ARES 2 rheometer. Diffusion of fluorine from the hydrogel: Three hydrogels in various Vs to HA volumetric ratios (1:50, 1:10, and 1:5) were prepared in duplicate. Hydrogels were incubated at 37 °C. To induce diffusion, three hydrogels were agitated at 1000 rpm. 1H and 19F MRI scans were acquired at 1, 3, 7 days and 2 months after gel preparation on a Bruker Ascend 750 scanner. To quantify fluorine content, scans were analyzed using Voxel Tracker 2.0. Assessment of cell viability in vitro and in vivo: Luciferase-positive mouse glial-restricted progenitors (GRPs) were embedded in 0:1, 1:50, 1:10, and 1:5 Vs:HA mixtures (final cell concentration =1 × 107/ml). For the in vitro assay, mixtures were placed in 96-wells plate in triplicate and bioluminescence was measured after 1, 3, 7, 14, 21, and 28 days. For in vivo experiments, Vs/HA mixtures containing GRPs were injected subcutaneously in SCID mice and BLI was acquired at 1, 3, 7, and 14 days post-injection. RESULTS Mixing of V-sense at increasing ratios of 1:50, 1:10, and 1:5 v/v of fluorine/activated hyaluronan (HA) hydrogel gradually elongated the gelation time from 194 s for non-fluorinated controls to 304 s for 1:5 V-sense:HA hydrogels, while their elastic properties slightly decreased. There was no release of V-sense from hydrogels maintained in stationary conditions over 2 months. The addition of V-sense positively affected in vitro survival of scaffolded GRPs in a dose-dependent manner. CONCLUSIONS These results show that hydrogel fluorination does not impair its beneficial properties for scaffolded cells, which may be used to visualize scaffolded GRP transplants with 19F MRI.
Collapse
Affiliation(s)
- Marcin Piejko
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,3rd Department of General Surgery, Jagiellonian University Medical College, Krakow, Poland
| | - Piotr Walczak
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology and Neurosurgery, University of Warmia and Mazury, Olsztyn, Poland
| | - Xiaowei Li
- Translational Tissue Engineering Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Mary and Dick Holland Regenerative Medicine Program, Department of Neurological Sciences, The University of Nebraska Medical Center, Omaha, NE, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miroslaw Janowski
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
29
|
Le Thi P, Son JY, Lee Y, Ryu SB, Park KM, Park KD. Enzymatically Crosslinkable Hyaluronic Acid-Gelatin Hybrid Hydrogels as Potential Bioinks for Tissue Regeneration. Macromol Res 2020. [DOI: 10.1007/s13233-020-8052-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
30
|
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
|
31
|
Vieira S, Strymecka P, Stanaszek L, Silva-Correia J, Drela K, Fiedorowicz M, Malysz-Cymborska I, Rogujski P, Janowski M, Reis RL, Lukomska B, Walczak P, Oliveira JM. Methacrylated gellan gum and hyaluronic acid hydrogel blends for image-guided neurointerventions. J Mater Chem B 2020; 8:5928-5937. [DOI: 10.1039/d0tb00877j] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mn-Based gellan gum hydrogels for cell delivery and real-time tracking on image-guided neuro-procedures.
Collapse
Affiliation(s)
- Sílvia Vieira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Paulina Strymecka
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Luiza Stanaszek
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Katarzyna Drela
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Michał Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Izabela Malysz-Cymborska
- Department of Neurology and Neurosurgery, School of Medicine
- Collegium Medicum
- University of Warmia and Mazury
- Olsztyn
- Poland
| | - Piotr Rogujski
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Miroslaw Janowski
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| | - Barbara Lukomska
- NeuroRepair Department
- Mossakowski Medical Research Centre
- Polish Academy of Sciences
- Warsaw
- Poland
| | - Piotr Walczak
- Department of Neurology and Neurosurgery, School of Medicine
- Collegium Medicum
- University of Warmia and Mazury
- Olsztyn
- Poland
| | - J. Miguel Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine
- AvePark – Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- 4805-017 Barco
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
| |
Collapse
|
32
|
Nitinol thin films functionalized with CAR-T cells for the treatment of solid tumours. Nat Biomed Eng 2019; 4:195-206. [PMID: 31819155 DOI: 10.1038/s41551-019-0486-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/07/2019] [Indexed: 12/11/2022]
Abstract
Micropatterned nickel titanium (commonly known as nitinol) thin films with complex designs, high structural resolution and excellent biocompatibility can be cheaply fabricated using magnetron sputtering. Here, we show that these benefits can be leveraged to fabricate micromesh implants that are loaded with tumour-specific human chimeric antigen receptor (CAR)-T cells for the treatment of solid tumours. In a mouse model of non-resectable ovarian cancer, the cell-loaded nitinol thin films spatially conformed to the implantation site, fostered the rapid expansion of T cells, delivered a high density of T cells directly to the tumour and significantly improved animal survival. We also show that self-expandable stents that were coated with T-cell-loaded films and implanted into subcutaneous tumours in mice improved the duration of stent patency by delaying tumour ingrowth. By providing direct access to tumours, CAR-T-cell-loaded micropatterned nitinol thin films can improve the effects of cell-based therapies.
Collapse
|
33
|
Li S, Jiang D, Rosenkrans ZT, Barnhart TE, Ehlerding EB, Ni D, Engle JW, Cai W. Aptamer-Conjugated Framework Nucleic Acids for the Repair of Cerebral Ischemia-Reperfusion Injury. NANO LETTERS 2019; 19:7334-7341. [PMID: 31518140 PMCID: PMC6876547 DOI: 10.1021/acs.nanolett.9b02958] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Effective therapy for protecting dying neurons against cerebral ischemia-reperfusion injury (IRI) represents a substantial challenge in the treatment of ischemic strokes. Oxidative stress coupled with excessive inflammation is the main culprit for brain IRI that results in neuronal damage and disability. Specifically, complement component 5a (C5a) exacerbates the vicious cycle between oxidative stress and inflammatory responses. Herein, we propose that a framework nucleic acid (FNA) conjugated with anti-C5a aptamers (aC5a) can selectively reduce C5a-mediated neurotoxicity and effectively alleviate oxidative stress in the brain. Intrathecal injection of the aC5a-conjugated FNA (aC5a-FNA) was applied for the treatment of rats with ischemic strokes. Positron emission tomography (PET) imaging was performed to investigate the accumulation of aC5a-FNA in the penumbra and its therapeutic efficacy. Results demonstrated that aC5a-FNA could rapidly penetrate different brain regions after brain IRI. Furthermore, aC5a-FNA effectively protected neurons from brain IRI, as verified by serum tests, tissue staining, biomarker detection, and functional assessment. The protective effect of aC5a-FNA against cerebral IRI in living animals may pave the way for the translation of FNA from bench to bedside and broaden the horizon of FNA in the field of biomedicine.
Collapse
Affiliation(s)
- Shiyong Li
- Department of Rehabilitation, Second Affiliated Hospital of Nanchang University, Jiangxi 330006, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Dawei Jiang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Zachary T. Rosenkrans
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Todd E. Barnhart
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Emily B. Ehlerding
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Dalong Ni
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jonathan W. Engle
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
34
|
Li R, Lin S, Zhu M, Deng Y, Chen X, Wei K, Xu J, Li G, Bian L. Synthetic presentation of noncanonical Wnt5a motif promotes mechanosensing-dependent differentiation of stem cells and regeneration. SCIENCE ADVANCES 2019; 5:eaaw3896. [PMID: 31663014 PMCID: PMC6795506 DOI: 10.1126/sciadv.aaw3896] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 09/25/2019] [Indexed: 05/30/2023]
Abstract
Noncanonical Wnt signaling in stem cells is essential to numerous developmental events. However, no prior studies have capitalized on the osteoinductive potential of noncanonical Wnt ligands to functionalize biomaterials in enhancing the osteogenesis and associated skeleton formation. Here, we investigated the efficacy of the functionalization of biomaterials with a synthetic Wnt5a mimetic ligand (Foxy5 peptide) to promote the mechanosensing and osteogenesis of human mesenchymal stem cells by activating noncanonical Wnt signaling. Our findings showed that the immobilized Wnt5a mimetic ligand activated noncanonical Wnt signaling via the up-regulation of Disheveled 2 and downstream RhoA-ROCK signaling, leading to enhanced intracellular calcium level, F-actin stability, actomyosin contractility, and cell adhesion structure development. This enhanced mechanotransduction in stem cells promoted the in vitro osteogenic lineage commitment and the in vivo healing of rat calvarial defects. Our work provides valuable guidance for the developmentally inspired design of biomaterials for a wide array of therapeutic applications.
Collapse
Affiliation(s)
- Rui Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Sien Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Meiling Zhu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Yingrui Deng
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Xiaoyu Chen
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Kongchang Wei
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biomimetic Membranes and Textiles, Lerchenfeldstrasse 5, CH-9014 St. Gallen, Switzerland
| | - Jianbin Xu
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P. R. China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
| | - Liming Bian
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P. R. China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, P. R. China
- Center of Novel Biomaterials, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077 Hong Kong, P.R. China
| |
Collapse
|
35
|
Oliveira EP, Malysz-Cymborska I, Golubczyk D, Kalkowski L, Kwiatkowska J, Reis RL, Oliveira JM, Walczak P. Advances in bioinks and in vivo imaging of biomaterials for CNS applications. Acta Biomater 2019; 95:60-72. [PMID: 31075514 DOI: 10.1016/j.actbio.2019.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 04/25/2019] [Accepted: 05/03/2019] [Indexed: 01/03/2023]
Abstract
Due to increasing life expectancy incidence of neurological disorders is rapidly rising, thus adding urgency to develop effective strategies for treatment. Stem cell-based therapies were considered highly promising and while progress in this field is evident, outcomes of clinical trials are rather disappointing. Suboptimal engraftment, poor cell survival and uncontrolled differentiation may be the reasons behind dismal results. Clearly, new direction is needed and we postulate that with recent progress in biomaterials and bioprinting, regenerative approaches for neurological applications may be finally successful. The use of biomaterials aids engraftment of stem cells, protects them from harmful microenvironment and importantly, it facilitates the incorporation of cell-supporting molecules. The biomaterials used in bioprinting (the bioinks) form a scaffold for embedding the cells/biomolecules of interest, but also could be exploited as a source of endogenous contrast or supplemented with contrast agents for imaging. Additionally, bioprinting enables patient-specific customization with shape/size tailored for actual needs. In stroke or traumatic brain injury for example lesions are localized and focal, and usually progress with significant loss of tissue volume creating space that could be filled with artificial tissue using bioprinting modalities. The value of imaging for bioprinting technology is advantageous on many levels including design of custom shapes scaffolds based on anatomical 3D scans, assessment of performance and integration after scaffold implantation, or to learn about the degradation over time. In this review, we focus on bioprinting technology describing different printing techniques and properties of biomaterials in the context of requirements for neurological applications. We also discuss the need for in vivo imaging of implanted materials and tissue constructs reviewing applicable imaging modalities and type of information they can provide. STATEMENT OF SIGNIFICANCE: Current stem cell-based regenerative strategies for neurological diseases are ineffective due to inaccurate engraftment, low cell viability and suboptimal differentiation. Bioprinting and embedding stem cells within biomaterials at high precision, including building complex multi-material and multi-cell type composites may bring a breakthrough in this field. We provide here comprehensive review of bioinks, bioprinting techniques applicable to application for neurological disorders. Appreciating importance of longitudinal monitoring of implanted scaffolds, we discuss advantages of various imaging modalities available and suitable for imaging biomaterials in the central nervous system. Our goal is to inspire new experimental approaches combining imaging, biomaterials/bioinks, advanced manufacturing and tissue engineering approaches, and stimulate interest in image-guided therapies based on bioprinting.
Collapse
Affiliation(s)
- Eduarda P Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal
| | | | - Dominika Golubczyk
- Dept. of Neurosurgery, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Lukasz Kalkowski
- Dept. of Neurosurgery, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Joanna Kwiatkowska
- Dept. of Neurosurgery, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal
| | - J Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal
| | - Piotr Walczak
- Dept. of Neurosurgery, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland; Russell H. Morgan Dept. of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
36
|
Costa A, Adamo S, Gossetti F, D'Amore L, Ceci F, Negro P, Bruzzone P. Biological Scaffolds for Abdominal Wall Repair: Future in Clinical Application? MATERIALS 2019; 12:ma12152375. [PMID: 31349716 PMCID: PMC6695954 DOI: 10.3390/ma12152375] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Abstract
Millions of abdominal wall repair procedures are performed each year for primary and incisional hernias both in the European Union and in the United States with extremely high costs. Synthetic meshes approved for augmenting abdominal wall repair provide adequate mechanical support but have significant drawbacks (seroma formation, adhesion to viscera, stiffness of abdominal wall, and infection). Biologic scaffolds (i.e., derived from naturally occurring materials) represent an alternative to synthetic surgical meshes and are less sensitive to infection. Among biologic scaffolds, extracellular matrix scaffolds promote stem/progenitor cell recruitment in models of tissue remodeling and, in the specific application of abdominal wall repair, have enough mechanical strength to support the repair. However, many concerns remain about the use of these scaffolds in the clinic due to their higher cost of production compared with synthetic meshes, despite having the same recurrence rate. The present review aims to highlight the pros and cons of using biologic scaffolds as surgical devices for abdominal wall repair and present possible improvements to widen their use in clinical practice.
Collapse
Affiliation(s)
- Alessandra Costa
- Sezione di Istologia ed Embriologia Medica, Dipartimento SAIMLAL, Sapienza Università di Roma, Via A. Scarpa 16, 00161 Rome, Italy
| | - Sergio Adamo
- Sezione di Istologia ed Embriologia Medica, Dipartimento SAIMLAL, Sapienza Università di Roma, Via A. Scarpa 16, 00161 Rome, Italy
| | - Francesco Gossetti
- Dipartimento Assistenziale Integrato Cardio Toraco-Vascolare, Chirurgia e Trapianti d'Organo, Azienda Ospedaliera Universitaria Policlinico Umberto I. Dipartimento Universitario Chirurgia Generale e Specialistica "Paride Stefanini", Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Linda D'Amore
- Dipartimento Assistenziale Integrato Cardio Toraco-Vascolare, Chirurgia e Trapianti d'Organo, Azienda Ospedaliera Universitaria Policlinico Umberto I. Dipartimento Universitario Chirurgia Generale e Specialistica "Paride Stefanini", Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Francesca Ceci
- Dipartimento Assistenziale Integrato Cardio Toraco-Vascolare, Chirurgia e Trapianti d'Organo, Azienda Ospedaliera Universitaria Policlinico Umberto I. Dipartimento Universitario Chirurgia Generale e Specialistica "Paride Stefanini", Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Paolo Negro
- Dipartimento Assistenziale Integrato Cardio Toraco-Vascolare, Chirurgia e Trapianti d'Organo, Azienda Ospedaliera Universitaria Policlinico Umberto I. Dipartimento Universitario Chirurgia Generale e Specialistica "Paride Stefanini", Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy
| | - Paolo Bruzzone
- Dipartimento Assistenziale Integrato Cardio Toraco-Vascolare, Chirurgia e Trapianti d'Organo, Azienda Ospedaliera Universitaria Policlinico Umberto I. Dipartimento Universitario Chirurgia Generale e Specialistica "Paride Stefanini", Sapienza Università di Roma, Viale del Policlinico 155, 00161 Rome, Italy.
| |
Collapse
|
37
|
Piejko M, Jablonska A, Walczak P, Janowski M. Proteolytic Rafts for Improving Intraparenchymal Migration of Minimally Invasively Administered Hydrogel-Embedded Stem Cells. Int J Mol Sci 2019; 20:E3083. [PMID: 31238564 PMCID: PMC6628268 DOI: 10.3390/ijms20123083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 11/23/2022] Open
Abstract
The physiological spaces (lateral ventricles, intrathecal space) or pathological cavities (stroke lesion, syringomyelia) may serve as an attractive gateway for minimally invasive deployment of stem cells. Embedding stem cells in injectable scaffolds is essential when transplanting into the body cavities as they secure favorable microenvironment and keep cells localized, thereby preventing sedimentation. However, the limited migration of transplanted cells from scaffold to the host tissue is still a major obstacle, which prevents this approach from wider implementation for the rapidly growing field of regenerative medicine. Hyaluronan, a naturally occurring polymer, is frequently used as a basis of injectable scaffolds. We hypothesized that supplementation of hyaluronan with activated proteolytic enzymes could be a viable approach for dissolving the connective tissue barrier on the interface between the scaffold and the host, such as pia mater or scar tissue, thus demarcating lesion cavity. In a proof-of-concept study, we have found that collagenase and trypsin immobilized in hyaluronan-based hydrogel retain 60% and 28% of their proteolytic activity compared to their non-immobilized forms, respectively. We have also shown that immobilized enzymes do not have a negative effect on the viability of stem cells (glial progenitors and mesenchymal stem cells) in vitro. In conclusion, proteolytic rafts composed of hyaluronan-based hydrogels and immobilized enzymes may be an attractive strategy to facilitate migration of stem cells from injectable scaffolds into the parenchyma of surrounding tissue.
Collapse
Affiliation(s)
- Marcin Piejko
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- 3rd Department of General Surgery, Jagiellonian University Medical College, 31202 Krakow, Poland.
| | - Anna Jablonska
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Piotr Walczak
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Miroslaw Janowski
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
38
|
Modo M, Badylak SF. A roadmap for promoting endogenous in situ tissue restoration using inductive bioscaffolds after acute brain injury. Brain Res Bull 2019; 150:136-149. [PMID: 31128250 DOI: 10.1016/j.brainresbull.2019.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 02/08/2023]
Abstract
The regeneration of brain tissue remains one of the greatest unsolved challenges in medicine and by many is considered unfeasible. Indeed, the adult mammalian brain does not regenerate tissue, but there is ongoing endogenous neurogenesis, which is upregulated after injury and contributes to tissue repair. This endogenous repair response is a conditio sine que non for tissue regeneration. However, scarring around the lesion core and cavitation provide unfavorable conditions for tissue regeneration in the brain. Based on the success of using extracellular matrix (ECM)-based bioscaffolds in peripheral soft tissue regeneration, it is plausible that the provision of an inductive ECM-based hydrogel inside the volumetric tissue loss can attract neural cells and create a de novo viable tissue. Following perturbation theory of these successes in peripheral tissues, we here propose 9 perturbation parts (i.e. requirements) that can be solved independently to create an integrated series to build a functional and integrated de novo neural tissue. Necessities for tissue formation, anatomical and functional connectivity are further discussed to provide a new substrate to support the improvement of behavioral impairments after acute brain injury. We also consider potential parallel developments of this tissue engineering effort that can support therapeutic benefits in the absence of de novo tissue formation (e.g. structural support to veterate brain tissue). It is envisaged that eventually top-down inductive "natural" bioscaffolds composed of decellularized tissues (i.e. ECM) will be replaced by bottom-up synthetic designer hydrogels that will provide very defined structural and signaling properties, potentially even opening up opportunities we currently do not envisage using natural materials.
Collapse
Affiliation(s)
- Michel Modo
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA; University of Pittsburgh, Department of Bioengineering, Pittsburgh, PA, USA; University of Pittsburgh, Department of Radiology, Pittsburgh, PA, USA.
| | - Stephen F Badylak
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA; University of Pittsburgh, Department of Bioengineering, Pittsburgh, PA, USA; University of Pittsburgh, Department of Surgery, Pittsburgh, PA, USA
| |
Collapse
|
39
|
Srivastava RK, Jablonska A, Chu C, Gregg L, Bulte JWM, Koehler RC, Walczak P, Janowski M. Biodistribution of Glial Progenitors in a Three Dimensional-Printed Model of the Piglet Cerebral Ventricular System. Stem Cells Dev 2019; 28:515-527. [PMID: 30760110 DOI: 10.1089/scd.2018.0172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
White matter damage persists in hypoxic-ischemic newborns even when treated with hypothermia. We have previously shown that intraventricular delivery of human glial progenitors (GPs) at the neonatal stage is capable of replacing abnormal host glia and rescuing the lifespan of dysmyelinated mice. However, such transplantation in the human brain poses significant challenges as related to high-volume ventricles and long cell migration distances. These challenges can only be studied in large animal model systems. In this study, we developed a three dimensional (3D)-printed model of the ventricular system sized to a newborn pig to investigate the parameters that can maximize a global biodistribution of injected GPs within the ventricular system, while minimizing outflow to the subarachnoid space. Bioluminescent imaging and magnetic resonance imaging were used to image the biodistribution of luciferase-transduced GPs in simple fluid containers and a custom-designed, 3D-printed model of the piglet ventricular system. Seven independent variables were investigated. The results demonstrated that a low volume (0.1 mL) of cell suspension is essential to keep cells within the ventricular system. If higher volumes (1 mL) are needed, a very slow infusion speed (0.01 mL/min) is necessary. Real-time magnetic resonance imaging demonstrated that superparamagnetic iron oxide (SPIO) labeling significantly alters the rheological properties of the GP suspension, such that, even at high speeds and high volumes, the outflow to the subarachnoid space is reduced. Several other factors, including GP species (human vs. mouse), type of catheter tip (end hole vs. side hole), catheter length (0.3 vs. 7.62 m), and cell concentration, had less effect on the overall distribution of GPs. We conclude that the use of a 3D-printed phantom model represents a robust, reproducible, and cost-saving alternative to in vivo large animal studies for determining optimal injection parameters.
Collapse
Affiliation(s)
- Rohit K Srivastava
- 1 Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,2 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anna Jablonska
- 1 Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,2 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chengyan Chu
- 1 Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,2 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lydia Gregg
- 3 Visualization Core Laboratory, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jeff W M Bulte
- 1 Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,2 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Raymond C Koehler
- 4 Department of Anesthesiology and Critical Care Medicine, Translational Tissue Engineering Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Piotr Walczak
- 1 Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,2 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,5 Department of Neurology and Neurosurgery, University of Warmia and Mazury, Olsztyn, Poland
| | - Miroslaw Janowski
- 1 Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,2 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
40
|
Mao X, Cheng R, Zhang H, Bae J, Cheng L, Zhang L, Deng L, Cui W, Zhang Y, Santos HA, Sun X. Self-Healing and Injectable Hydrogel for Matching Skin Flap Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801555. [PMID: 30775235 PMCID: PMC6364594 DOI: 10.1002/advs.201801555] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/21/2018] [Indexed: 05/05/2023]
Abstract
The fabrication of highly biocompatible hydrogels with multiple unique healing abilities for the whole healing process, for example, multifunctional hydrogels with injectable, degradation, antibacterial, antihypoxic, and wound healing-promoting properties that match the dynamic healing process of skin flap regeneration, is currently a research challenge. Here, a multifunctional and dynamic coordinative polyethylene glycol (PEG) hydrogel with mangiferin liposomes (MF-Lip@PEG) is developed for clinical applications through Ag-S coordination of four-arm-PEG-SH and Ag+. Compared to MF-PEG, MF-Lip@PEG exhibits self-healing properties, lower swelling percentages, and a longer endurance period. Moreover, the hydrogel exhibits excellent drug dispersibility and release characteristics for slow and persistent drug delivery. In vitro studies show that the hydrogel is biocompatible and nontoxic to cells, and exerts an outstanding neovascularization-promoting effect. The MF-Lip@PEG also exhibits a strong cytoprotective effect against hypoxia-induced apoptosis through regulation of the Bax/Bcl-2/caspase-3 pathway. In a random skin flap animal model, the MF-Lip@PEG is injectable and convenient to deliver into the skin flap, providing excellent anti-inflammation, anti-infection, and proneovascularization effects and significantly reducing the skin flap necrosis rate. In general, the MF-Lip@PEG possesses outstanding multifunctionality for the dynamic healing process of skin flap regeneration.
Collapse
Affiliation(s)
- Xiyuan Mao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of Medicine639 Zhi Zao Ju RoadShanghai200011P. R. China
| | - Ruoyu Cheng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | | | - Jinhong Bae
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of Medicine639 Zhi Zao Ju RoadShanghai200011P. R. China
| | - Liying Cheng
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of Medicine639 Zhi Zao Ju RoadShanghai200011P. R. China
| | - Lu Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of Medicine639 Zhi Zao Ju RoadShanghai200011P. R. China
| | - Lianfu Deng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan UniversityNo. 220 Handan RoadShanghai200433China
| | - Yuguang Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of Medicine639 Zhi Zao Ju RoadShanghai200011P. R. China
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFI‐00014Finland
| | - Xiaoming Sun
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of Medicine639 Zhi Zao Ju RoadShanghai200011P. R. China
| |
Collapse
|
41
|
Walczak P, Janowski M. Chemobrain as a Product of Growing Success in Chemotherapy - Focus on Glia as both a Victim and a Cure. ACTA ACUST UNITED AC 2019; 9:2207-2216. [PMID: 31316584 DOI: 10.4172/neuropsychiatry.1000565] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chemotherapy-induced cognitive impairment or chemobrain is a frequent consequence of cancer treatment with many psychiatric features. Ironically, the increasing efficacy of chemotherapy leaves growing number of patients alive with chemobrain. Therefore, there is an urgent need for strategies capable of returning cancer survivors back to their pre-morbid quality of life. Molecular mechanisms of chemobrain are largely unknown. Over the last decade there was a lot of emphasis in preclinical research on inflammatory consequences of chemotherapy and oxidative stress but so far none of these approaches were translated into clinical scenario. The co-administration of chemotherapy with protective agents was evaluated preclinically but it should be introduced with caution as potential interference was not yet studied and that could blunt therapeutic efficacy. Stem cell-based regenerative medicine approach has so far been exploited very sparsely in the context of chemobrain and the focus was on indirect mechanisms or neuronal replacement in the hippocampus. However, there is evidence for widespread white matter abnormalities in patients with chemobrain. This is quite logical considering life-long proliferation and turnover of glial cells, which makes them vulnerable to chemotherapeutic agents. Feasibility of glia replacement has been established in mice with global dysmyelination where profound therapeutic effect has been observed but only in case of global cell engraftment (across the entire brain). While global glia replacement has been achieved in mice translation to clinical setting might be challenging due to much larger brain size. Therefore, a lot of attention should be directed towards the route of administration to accomplish widespread cell delivery. Techniques facilitating that broad cell distribution including intra-arterial and intrathecal methods should be considered as very compelling options. Summarizing, chemobrain is a rapidly growing medical problem and global glia replacement should be considered as worthwhile therapeutic strategy.
Collapse
Affiliation(s)
- Piotr Walczak
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology and Neurosurgery, University of Warmia and Mazury, Olsztyn, Poland
| | - Miroslaw Janowski
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
42
|
MRI-guided intrathecal transplantation of hydrogel-embedded glial progenitors in large animals. Sci Rep 2018; 8:16490. [PMID: 30405160 PMCID: PMC6220305 DOI: 10.1038/s41598-018-34723-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022] Open
Abstract
Disseminated diseases of the central nervous system such as amyotrophic lateral sclerosis (ALS) require that therapeutic agents are delivered and distributed broadly. Intrathecal route is attractive in that respect, but to date there was no methodology available allowing for optimization of this technique to assure safety and efficacy in a clinically relevant setting. Here, we report on interventional, MRI-guided approach for delivery of hydrogel-embedded glial progenitor cells facilitating cell placement over extended surface of the spinal cord in pigs and in naturally occurring ALS-like disease in dogs. Glial progenitors used as therapeutic agent were embedded in injectable hyaluronic acid-based hydrogel to support their survival and prevent sedimentation or removal. Intrathecal space was reached through lumbar puncture and the catheter was advanced under X-ray guidance to the cervical part of the spine. Animals were then transferred to MRI suite for MRI-guided injection. Interventional and follow-up MRI as well as histopathology demonstrated successful and predictable placement of embedded cells and safety of the procedure.
Collapse
|
43
|
Dalamagkas K, Tsintou M, Seifalian A, Seifalian AM. Translational Regenerative Therapies for Chronic Spinal Cord Injury. Int J Mol Sci 2018; 19:E1776. [PMID: 29914060 PMCID: PMC6032191 DOI: 10.3390/ijms19061776] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury is a chronic and debilitating neurological condition that is currently being managed symptomatically with no real therapeutic strategies available. Even though there is no consensus on the best time to start interventions, the chronic phase is definitely the most stable target in order to determine whether a therapy can effectively restore neurological function. The advancements of nanoscience and stem cell technology, combined with the powerful, novel neuroimaging modalities that have arisen can now accelerate the path of promising novel therapeutic strategies from bench to bedside. Several types of stem cells have reached up to clinical trials phase II, including adult neural stem cells, human spinal cord stem cells, olfactory ensheathing cells, autologous Schwann cells, umbilical cord blood-derived mononuclear cells, adult mesenchymal cells, and autologous bone-marrow-derived stem cells. There also have been combinations of different molecular therapies; these have been either alone or combined with supportive scaffolds with nanostructures to facilitate favorable cell⁻material interactions. The results already show promise but it will take some coordinated actions in order to develop a proper step-by-step approach to solve impactful problems with neural repair.
Collapse
Affiliation(s)
- Kyriakos Dalamagkas
- The Institute for Rehabilitation and Research, Memorial Hermann Texas Medical Centre, Houston, TX 77030, USA.
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery and Interventional Science, University College of London (UCL), London NW3 2QG, UK.
| | - Magdalini Tsintou
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery and Interventional Science, University College of London (UCL), London NW3 2QG, UK.
- Center for Neural Systems Investigations, Massachusetts General Hospital/HST Athinoula A., Martinos Centre for Biomedical Imaging, Harvard Medical School, Boston, MA 02129, USA.
| | - Amelia Seifalian
- Faculty of Medical Sciences, UCL Medical School, London WC1E 6BT, UK.
| | - Alexander M Seifalian
- NanoRegMed Ltd. (Nanotechnology & Regenerative Medicine Commercialization Centre), The London BioScience Innovation Centre, London NW1 0NH, UK.
| |
Collapse
|