1
|
Kim SW, Baik S, Hyun J, Lee J, Lim D, Lee TJ, Jeong GJ, Im GB, Seo I, Kim YH, Pang C, Bhang SH. Facile Size Tunable Skin-Adaptive Patch for Accelerating Wound Healing. Adv Healthc Mater 2024:e2304435. [PMID: 39235562 DOI: 10.1002/adhm.202304435] [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: 12/13/2023] [Revised: 05/12/2024] [Indexed: 09/06/2024]
Abstract
Owing to the moist and curved interfaces of skin wounds, enhancing the adhesiveness while maintaining delivery efficacy of biomolecules has drawn significant attention in advanced wound dressings. Despite tremendous trials to load biomolecules with sound adhesiveness, the complicated fabricating processes and abnormal allergic responses that are attributed to chemical moiety-based adhesives remain as major problems. To this end, in this study a one-step fabrication process is developed to manufacture microstructures with both a therapeutic (cylindrical structure for embossed structure human adipose-derived stem cell sheet, ESS) and an adhesive part (octopi-inspired structure of adhesive, OIA), which ESOIA is called. OIA showed the highest adhesion strength in both dry (1.48 N cm-2) and wet pig skin conditions (0.81 N cm-2), maintaining the adhesive properties after repeated attach-detach trials. ESS from the therapeutic part of ESOIA also showed an enhanced angiogenic effect compared with the ones that are normally cultured in vitro. ESS also showed improved in vivo wound healing outcomes following enhanced cell engraftment compared to the cell injection group by means of intact cell-extracellular matrix interactions.
Collapse
Affiliation(s)
- Sung-Won Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sangyul Baik
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dohyun Lim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Tae-Jin Lee
- Department of Medical Biotechnology, Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon-si, 24341, Republic of Korea
| | - Gun-Jae Jeong
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Inwoo Seo
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Changhyun Pang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
2
|
Kavand A, Noverraz F, Gerber-Lemaire S. Recent Advances in Alginate-Based Hydrogels for Cell Transplantation Applications. Pharmaceutics 2024; 16:469. [PMID: 38675129 PMCID: PMC11053880 DOI: 10.3390/pharmaceutics16040469] [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: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
With its exceptional biocompatibility, alginate emerged as a highly promising biomaterial for a large range of applications in regenerative medicine. Whether in the form of microparticles, injectable hydrogels, rigid scaffolds, or bioinks, alginate provides a versatile platform for encapsulating cells and fostering an optimal environment to enhance cell viability. This review aims to highlight recent studies utilizing alginate in diverse formulations for cell transplantation, offering insights into its efficacy in treating various diseases and injuries within the field of regenerative medicine.
Collapse
Affiliation(s)
| | | | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.K.); (F.N.)
| |
Collapse
|
3
|
Thummarati P, Laiwattanapaisal W, Nitta R, Fukuda M, Hassametto A, Kino-oka M. Recent Advances in Cell Sheet Engineering: From Fabrication to Clinical Translation. Bioengineering (Basel) 2023; 10:211. [PMID: 36829705 PMCID: PMC9952256 DOI: 10.3390/bioengineering10020211] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Cell sheet engineering, a scaffold-free tissue fabrication technique, has proven to be an important breakthrough technology in regenerative medicine. Over the past two decades, the field has developed rapidly in terms of investigating fabrication techniques and multipurpose applications in regenerative medicine and biological research. This review highlights the most important achievements in cell sheet engineering to date. We first discuss cell sheet harvesting systems, which have been introduced in temperature-responsive surfaces and other systems to overcome the limitations of conventional cell harvesting methods. In addition, we describe several techniques of cell sheet transfer for preclinical (in vitro and in vivo) and clinical trials. This review also covers cell sheet cryopreservation, which allows short- and long-term storage of cells. Subsequently, we discuss the cell sheet properties of angiogenic cytokines and vasculogenesis. Finally, we discuss updates to various applications, from biological research to clinical translation. We believe that the present review, which shows and compares fundamental technologies and recent advances in cell engineering, can potentially be helpful for new and experienced researchers to promote the further development of tissue engineering in different applications.
Collapse
Affiliation(s)
- Parichut Thummarati
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
- Biosensors and Bioanalytical Technology for Cells and Innovative Testing Device Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wanida Laiwattanapaisal
- Biosensors and Bioanalytical Technology for Cells and Innovative Testing Device Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Rikiya Nitta
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Megumi Fukuda
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Artchaya Hassametto
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
4
|
Honda N, Watanabe Y, Tokuoka Y, Hanajima R. Roles of microglia/macrophage and antibody in cell sheet transplantation in the central nervous system. Stem Cell Res Ther 2022; 13:470. [PMID: 36089602 PMCID: PMC9465875 DOI: 10.1186/s13287-022-03168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Background We previously established a human mesenchymal stem cell (MSC) line that was modified to express trophic factors. Transplanting a cell sheet produced from this line in an amyotrophic lateral sclerosis mouse model showed a beneficial trend for mouse life spans. However, the sheet survived for less than 14 days, and numerous microglia and macrophages were observed within and adjacent to the sheet. Here, we examined the roles of microglia and macrophages as well as acquired antibodies in cell sheet transplantation. Methods We observed the effects of several MSC lines on macrophages in vitro, that is, phenotype polarization (M1 or M2) and migration. We then investigated how phenotypic polarization affected MSC survival using antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). We also confirmed the role of complement on cytotoxicity. Lastly, we selectively eliminated microglia and macrophages in vivo to determine whether these cells were cytoprotective to the donor sheet. Results In vitro co-culture with MSCs induced M2 polarization in macrophages and facilitated their migration toward MSCs in vitro. There was no difference between M1 and M2 phenotypes on ADCC and ADCP. Cytotoxicity was observed even in the absence of complement. Eliminating microglia/macrophage populations in vivo resulted in increased survival of donor cells after transplantation. Conclusions Acquired antibodies played a role in ADCC and ADCP. MSCs induced M2 polarization in macrophages and facilitated their migration toward MSCs in vitro. Despite these favorable characteristics of microglia and macrophages, deletion of these cells was advantageous for the survival of donor cells in vivo. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03168-5.
Collapse
|
5
|
Wang Y, Zhao Z, Liu S, Luo W, Wang G, Zhu Z, Ma Q, Liu Y, Wang L, Lu S, Zhang Y, Qian J, Zhang Y. Application of vancomycin-impregnated calcium sulfate hemihydrate/nanohydroxyapatite/carboxymethyl chitosan injectable hydrogels combined with BMSC sheets for the treatment of infected bone defects in a rabbit model. BMC Musculoskelet Disord 2022; 23:557. [PMID: 35681160 PMCID: PMC9185966 DOI: 10.1186/s12891-022-05499-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The choice of bone substitutes for the treatment of infected bone defects (IBDs) has attracted the attention of surgeons for years. However, single-stage bioabsorbable materials that are used as carriers for antibiotic release, as well as scaffolds for BMSC sheets, need further exploration. Our study was designed to investigate the effect of vancomycin-loaded calcium sulfate hemihydrate/nanohydroxyapatite/carboxymethyl chitosan (CSH/n-HA/CMCS) hydrogels combined with BMSC sheets as bone substitutes for the treatment of IBDs. METHODS BMSCs were harvested and cultured into cell sheets. After the successful establishment of an animal model with chronic osteomyelitis, 48 New Zealand white rabbits were randomly divided into 4 groups. Animals in Group A were treated with thorough debridement as a control. Group B was treated with BMSC sheets. CSH/n-HA/CMCS hydrogels were implanted in the treatment of Group C, and Group D was treated with CSH/n-HA/CMCS+BMSC sheets. Gross observation and micro-CT 3D reconstruction were performed to assess the osteogenic and infection elimination abilities of the treatment materials. Histological staining (haematoxylin and eosin and Van Gieson) was used to observe inflammatory cell infiltration and the formation of collagen fibres at 4, 8, and 12 weeks after implantation. RESULTS The bone defects of the control group were not repaired at 12 weeks, as chronic osteomyelitis was still observed. HE staining showed a large amount of inflammatory cell infiltration around the tissue, and VG staining showed no new collagen fibres formation. In the BMSC sheet group, although new bone formation was observed by gross observation and micro-CT scanning, infection was not effectively controlled due to unfilled cavities. Some neutrophils and only a small amount of collagen fibres could be observed. Both the hydrogel and hydrogel/BMSCs groups achieved satisfactory repair effects and infection control. Micro-CT 3D reconstruction at 4 weeks showed that the hydrogel/BMSC sheet group had higher reconstruction efficiency and better bone modelling with normal morphology. HE staining showed little aggregation of inflammatory cells, and VG staining showed a large number of new collagen fibres. CONCLUSIONS Our preliminary results suggested that compared to a single material, the novel antibiotic-impregnated hydrogels acted as superior scaffolds for BMSC sheets and excellent antibiotic vectors against infection, which provided a basis for applying tissue engineering technology to the treatment of chronic osteomyelitis.
Collapse
Affiliation(s)
- Yanjun Wang
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Zihou Zhao
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Shiyu Liu
- Institute of Oral Tissue Engineering, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Wen Luo
- Department of Ultrasound, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Guoliang Wang
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Zhenfeng Zhu
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Qiong Ma
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Yunyan Liu
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Linhu Wang
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Shuaikun Lu
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Yong Zhang
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China.
| | - Jixian Qian
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China.
| | - Yunfei Zhang
- Department of Orthopaedics, Second affiliated hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
6
|
Adeyemi SA, Choonara YE. Current advances in cell therapeutics: A biomacromolecules application perspective. Expert Opin Drug Deliv 2022; 19:521-538. [PMID: 35395914 DOI: 10.1080/17425247.2022.2064844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Many chronic diseases have evolved and to circumvent the limitations of using conventional drug therapies, smart cell encapsulating delivery systems have been explored to customize the treatment with alignment to disease longevity. Cell therapeutics has advanced in tandem with improvements in biomaterials that can suitably deliver therapeutic cells to achieve targeted therapy. Among the promising biomacromolecules for cell delivery are those that share bio-relevant architecture with the extracellular matrix and display extraordinary compatibility in the presence of therapeutic cells. Interestingly, many biomacromolecules that fulfil these tenets occur naturally and can form hydrogels. AREAS COVERED This review provides a concise incursion into the paradigm shift to cell therapeutics using biomacromolecules. Advances in the design and use of biomacromolecules to assemble smart therapeutic cell carriers is discussed in light of their pivotal role in enhancing cell encapsulation and delivery. In addition, the principles that govern the application of cell therapeutics in diabetes, neuronal disorders, cancers and cardiovascular disease are outlined. EXPERT OPINION Cell therapeutics promises to revolutionize the treatment of various secretory cell dysfunctions. Current and future advances in designing functional biomacromolecules will be critical to ensure that optimal delivery of therapeutic cells is achieved with desired biosafety and potency.
Collapse
Affiliation(s)
- Samson A Adeyemi
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Science, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown, 2193, South Africa
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Science, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown, 2193, South Africa
| |
Collapse
|
7
|
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: 0.5] [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
|
8
|
Mesenchymal Stem Cell Sheet Promotes Functional Recovery and Palliates Neuropathic Pain in a Subacute Spinal Cord Injury Model. Stem Cells Int 2021; 2021:9964877. [PMID: 34306098 PMCID: PMC8285204 DOI: 10.1155/2021/9964877] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/16/2021] [Accepted: 06/23/2021] [Indexed: 11/25/2022] Open
Abstract
Stem cell therapy has been shown to reverse the sequelae of spinal cord injury (SCI). Although the ideal treatment route remains unknown, providing a large number of stem cells to the injured site using less invasive techniques is critical to achieving maximal recovery. This study was conducted to determine whether administration of bone marrow stem cell (BMSC) sheet made on its own without a scaffold is superior to intramedullary cell transplantation in a rat subacute SCI model. Adult female Sprague-Dawley rats were subjected to SCI by 30 g clip compression at the level of Th6 and Th7 and were administered BMSC cell sheet (7 × 104 cells, subdural), cell suspension (7 × 104 cells, intramedullary), or control seven days after the injury. Motor and sensory assessments, as well as histological evaluation, were performed to determine the efficacy of the different cell transplantation procedures. While both the cell sheet and cell intramedullary injection groups showed significant motor recovery compared to the control group, the cell sheet group showed better results. Furthermore, the cell sheet group displayed a significant sensory recovery compared to the other groups. A histological evaluation revealed that the cell sheet group showed smaller injury lesion volume, less inflammation, and gliosis compared to other groups. Sensory-related fibers of μ-opioid receptors (MOR, interneuron) and hydroxytryptamine transporters (HTT, descending pain inhibitory pathway), located around the dorsal horn of the spinal cord at the caudal side of the SCI, were preserved only in the cell sheet group. Stem cells could also be found inside the peri-injured spinal cord in the cell sheet group. BMSC cell sheets were able to promote functional recovery and palliate neuropathic pain more effectively than intramedullary injections, thus serving as a good treatment option for SCI.
Collapse
|
9
|
Xue F, Bai Y, Jiang Y, Liu J, Jian K. Construction and a preliminary study of paracrine effect of bone marrow-derived endothelial progenitor cell sheet. Cell Tissue Bank 2021; 23:185-197. [PMID: 34052984 PMCID: PMC8854320 DOI: 10.1007/s10561-021-09932-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/21/2021] [Indexed: 12/14/2022]
Abstract
The release of paracrine factors from endothelial progenitor cell (EPC) sheet is a central mechanism of tissue repair. The purpose of this study was to constuct the rat bone marrow derived-endothelial progenitor cell (BM-EPCs) sheet and investigate invest the role of stromal cell-derived factor-1α (SDF-1α)/CXCR4 axis in the biological function of BM-EPCs sheet. BM-EPC cells were identified by the cell-surface markers-CD34/CD133/VE-cadherin/KDR using flow cytometry and dual affinity for acLDL and UEA-1. After 7 days of incubation, the BM-EPC single-cell suspensions were seeded on thermo-sensitive plate to harvest the BM-EPC cell sheets. The expression levels of SDF-1α/CXCR4 axis-associated genes and proteins were examined using RT-qPCR and western blot analysis, and enzyme-linked immunosorbent assay (ELISA) was applied to determine the concentration of vascular endothelial growth factor (VEGF), epidermal growth factor (EGF) and SDF-1α in the cell culture medium. The BM-EPC cell sheets were successfully harvested. Moreover, BM-EPC cell sheets have superior migration and tube formation activity when compared with single cell suspension. When capillary-like tube were formed from EPCs sheets, the releasing of paracrine factors such as VEGF, EGF and SDF-1α were increased. To reveal the mechanism of tube formation of BM-EPCs sheets, our research showed that the activation of PI3K/AKT/eNOS pathway was involved in the process, because the phosphorylation of CXCR, PI3K, AKT and eNOS were increased. BM-EPC cell sheets have superior paracrine and tube formation activity than the BM-EPC single-cell. The strong ability to secrete paracrine factors was be potentially related to the SDF-1α/CXCR4 axis through PI3K/AKT/eNOS pathway.
Collapse
Affiliation(s)
- Fenlong Xue
- Department of Cardiovascular Surgery, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Yunpeng Bai
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, 300051, China
| | - Yiyao Jiang
- Department of Cardiovascular Surgery, Tianjin First Central Hospital, Tianjin, 300192, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, China
| | - Jianshi Liu
- Department of Cardiovascular Surgery, DeltaHealth Hospital Shanghai, Shanghai, 200336, China
| | - Kaitao Jian
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, 300051, China.
- Department of Cardiovascular Surgery, DeltaHealth Hospital Shanghai, Shanghai, 200336, China.
| |
Collapse
|
10
|
Gao X, Cao Z. Gingiva-derived Mesenchymal Stem Cells and Their Potential Applications in Oral and Maxillofacial Diseases. Curr Stem Cell Res Ther 2020; 15:43-53. [PMID: 31702517 DOI: 10.2174/1574888x14666191107100311] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/02/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Stem cells are undifferentiated cells with multilineage differentiation potential. They can be collected from bone marrow, fat, amniotic fluid, and teeth. Stem cell-based therapies have been widely used to treat multiple diseases, such as cardiac disease, and hematological disorders. The cells may also be beneficial for controlling the disease course and promoting tissue regeneration in oral and maxillofacial diseases. Oral-derived gingival mesenchymal stem cells are easy to access and the donor sites heal rapidly without a scar. Such characteristics demonstrate the beneficial role of GMSCs in oral and maxillofacial diseases. OBJECTIVE We summarize the features of GMSCs, including their self-renewal, multipotent differentiation, immunomodulation, and anti-inflammation properties. We also discuss their applications in oral and maxillofacial disease treatment and tissue regeneration. CONCLUSION GMSCs are easily harvestable adult stem cells with outstanding proliferation, differentiation, and immunomodulation characteristics. A growing body of evidence indicates that GMSCs have strong potential use in accelerating wound healing and promoting the regeneration of bone defects, periodontium, oral neoplasms, salivary glands, peri-implantitis, and nerves. Moreover, alginate, polylactic acid and polycaprolactone can be used as biodegradable scaffolds for GMSC encapsulation. Various growth factors can be applied to the corresponding scaffolds to obtain the desired GMSC differentiation and phenotypes. Three-dimensional spheroid culture systems could optimize GMSC properties and improve the performance of the cells in tissue engineering. The immunomodulatory property of GMSCs in controlling oral and maxillofacial inflammation needs further research.
Collapse
Affiliation(s)
- Xudong Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory for Oral Biomedical Engineering of Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Cai Y, Liu W, Lian L, Xu Y, Bai X, Xu S, Zhang J. Stroke treatment: Is exosome therapy superior to stem cell therapy? Biochimie 2020; 179:190-204. [PMID: 33010339 DOI: 10.1016/j.biochi.2020.09.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023]
Abstract
Stroke is one of the most common causes of disability and death, and currently, ideal clinical treatment is lacking. Stem cell transplantation is a widely-used treatment approach for stroke. When compared with other types of stem cells, bone marrow mesenchymal stem cells (BMSCs) have been widely studied because of their many advantages. The paracrine effect is the primary mechanism for stem cells to play their role, and exosomes play an essential role in the paracrine effect. When compared with cell therapy, cell-free exosome therapy can prevent many risks and difficulties, and therefore, represents a promising and novel approach for treatment. In this study, we reviewed the research progress in the application of BMSCs-derived exosomes (BMSCs-exos) and BMSCs in the treatment of stroke. In addition, the advantages and disadvantages of cell therapy and cell-free exosome therapy were described, and the possible factors that hinder the introduction of these two treatments into the clinic were analyzed. Furthermore, we reviewed the current optimization methods of cell therapy and cell-free exosome therapy. Taken together, we hypothesize that cell-free exosome therapy will have excellent research prospects in the future, and therefore, it is worth further exploring. There are still some issues that need to be further addressed. For example, differences between the in vivo microenvironment and in vitro culture conditions will affect the paracrine effect of stem cells. Most importantly, we believe that more preclinical and clinical design studies are required to compare the efficacy of stem cells and exosomes.
Collapse
Affiliation(s)
- Yichen Cai
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Wanying Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lu Lian
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yingzhi Xu
- Beijing University of Chinese Medicine (BUCM), Beijing, China
| | - Xiaodan Bai
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China.
| | - Junping Zhang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China.
| |
Collapse
|
12
|
Ryu B, Sekine H, Homma J, Kobayashi T, Kobayashi E, Kawamata T, Shimizu T. Allogeneic adipose-derived mesenchymal stem cell sheet that produces neurological improvement with angiogenesis and neurogenesis in a rat stroke model. J Neurosurg 2020; 132:442-455. [PMID: 30797215 DOI: 10.3171/2018.11.jns182331] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/02/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Stem cell therapy is a promising strategy for the treatment of severe cerebral ischemia. However, targeting sufficient grafted cells to the affected area remains challenging. Choosing an adequate transplantation method for the CNS appears crucial for this therapy to become a clinical reality. The authors used a scaffold-free cell sheet as a translational intervention. This method involves the use of cell sheet layers and allows the transplantation of a large number of cells, locally and noninvasively. The authors evaluated the effectiveness of allogeneic adipose tissue-derived mesenchymal stem cell sheets in a rat model of stroke. METHODS The animals, subjected to middle cerebral artery occlusion, were randomly divided in two groups: one in which a cell sheet was transplanted and the other in which a vehicle was used (n = 10/group). Over a period of 14 days after transplantation, the animals' behavior was evaluated, after which brain tissue samples were removed and fixed, and the extent of angiogenesis and infarct areas was evaluated histologically. RESULTS Compared to the vehicle group, in the cell sheet group functional angiogenesis and neurogenesis were significantly increased, which resulted in behavioral improvement. Transplanted cells were identified within newly formed perivascular walls as pericytes, a proportion of which were functional. Newly formed blood vessels were found within the cell sheet that had anastomosed to the cerebral blood vessels in the host. CONCLUSIONS The transplantation approach described here is expected to provide not only a paracrine effect but also a direct cell effect resulting in cell replacement that protects the damaged neurovascular unit. The behavioral improvement seen with this transplantation approach provides the basis for further research on cell sheet-based regenerative treatment as a translational treatment for patients with stroke.
Collapse
Affiliation(s)
- Bikei Ryu
- 1Institute of Advanced Biomedical Engineering and Science and
- 2Department of Neurosurgery, Tokyo Women's Medical University; and
| | - Hidekazu Sekine
- 1Institute of Advanced Biomedical Engineering and Science and
| | - Jun Homma
- 1Institute of Advanced Biomedical Engineering and Science and
| | | | - Eiji Kobayashi
- 3Department of Organ Fabrication, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | | | - Tatsuya Shimizu
- 1Institute of Advanced Biomedical Engineering and Science and
| |
Collapse
|
13
|
Sun H, Lu J, Li B, Chen S, Xiao X, Wang J, Wang J, Wang X. Partial regeneration of uterine horns in rats through adipose-derived stem cell sheets. Biol Reprod 2019; 99:1057-1069. [PMID: 29931041 DOI: 10.1093/biolre/ioy121] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 06/19/2018] [Indexed: 12/15/2022] Open
Abstract
Severe uterine damage and infection lead to intrauterine adhesions, which result in hypomenorrhea, amenorrhea and infertility. Cell sheet engineering has shown great promise in clinical applications. Adipose-derived stem cells (ADSCs) are emerging as an alternative source of stem cells for cell-based therapies. In the present study, we investigated the feasibility of applying ADSCs as seed cells to form scaffold-free cell sheet. Data showed that ADSC sheets expressed higher levels of FGF, Col I, TGFβ, and VEGF than ADSCs in suspension, while increased expression of this gene set was associated with stemness, including Nanog, Oct4, and Sox2. We then investigated the therapeutic effects of 3D ADSCs sheet on regeneration in a rat model. We found that ADSCs were mainly detected in the basal layer of the regenerating endometrium in the cell sheet group at 21 days after transplantation. Additionally, some ADSCs differentiated into stromal-like cells. Moreover, ADSC sheets transplanted into partially excised uteri promoted regeneration of the endometrium cells, muscle cells and stimulated angiogenesis, and also resulted in better pregnancy outcomes. Therefore, ADSC sheet therapy shows considerable promise as a new treatment for severe uterine damage.
Collapse
Affiliation(s)
- Huijun Sun
- Department of Obstetrics and Gynecology, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Rd., Xian 710038, China
| | - Jie Lu
- Department of Obstetrics and Gynecology, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Rd., Xian 710038, China
| | - Bo Li
- Department of Obstetrics and Gynecology, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Rd., Xian 710038, China
| | - Shuqiang Chen
- Department of Obstetrics and Gynecology, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Rd., Xian 710038, China
| | - Xifeng Xiao
- Department of Obstetrics and Gynecology, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Rd., Xian 710038, China
| | - Jun Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Rd., Xian 710038, China
| | - Jingjing Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Rd., Xian 710038, China
| | - Xiaohong Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Rd., Xian 710038, China
| |
Collapse
|
14
|
Dynamics of host and graft after cell sheet transplantation: Basic study for the application of amyotrophic lateral sclerosis. Brain Res 2019; 1724:146444. [PMID: 31518575 DOI: 10.1016/j.brainres.2019.146444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022]
Abstract
Stem cells offer great hope for the therapy of neurological disorders. Using a human artificial chromosome (HAC), we generated modified mesenchymal stem cells (MSCs), termed HAC-MSC that express 3 growth factors and 2 marker proteins including luciferase, and previously demonstrated that intrathecal administration of HAC-MSCs extended the lifespan in a mouse model of amyotrophic lateral sclerosis (ALS). However, donor cells disappeared rapidly after transplantation. To overcome this poor survival, we transplanted the HAC-MSCs as a sheet structure which retained the extracellular matrix. We investigated, here, whether cell sheet showed a longer survival than intrathecal administration. Also, the therapeutic effects on ALS model mice were examined. In vivo imaging showed that luciferase signals increased immediately after transplantation up to 7 days, and these signals were sustained for up to 14 days. In contrast, following intrathecal administration, signals were drastically decreased by day 3. Moreover, cell sheet transplantation successfully prolonged the survival of donor HAC-MSCs. Cell sheet transplantation increased the level of p-Akt at the graft area. Pathologically, none of the donor cells differentiated into neurons, astrocytes or microglial cells. When the cell sheet was transplanted into ALS model mice, there was an encouraging trend in the delayed onset of symptoms and increased lifespan. If each group was subdivided into rapid and slow progressors based on cut-off values for respective median survival, the survival of rapid progressors differed significantly between groups (treated vs. sham-operated = 145.4 ± 1.4 vs. 139.2 ± 1.2). The effect of HAC-MSC sheet transplantation still has a temporally narrow therapeutic window. Further improvement could be achieved by optimization of the transplantation conditions, e.g. co-transplantation of HAC-MSCs with endothelial progenitor cells.
Collapse
|
15
|
Hu Y, Xu Y. Relationship between interleukin‐6 and brain ischemia. IBRAIN 2019. [DOI: 10.1002/j.2769-2795.2019.tb00039.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Yue Hu
- Department of AnesthesiologyThe First People's Hospital of Shuangliu DistrictChengduSichuanChina
| | - Yang Xu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
16
|
Suzuki N, Arimitsu N, Shimizu J, Takai K, Hirotsu C, Ueda Y, Wakisaka S, Fujiwara N, Suzuki T. Neuronal Cell Sheets of Cortical Motor Neuron Phenotype Derived from Human iPSCs. Cell Transplant 2018; 26:1355-1364. [PMID: 28901192 PMCID: PMC5680971 DOI: 10.1177/0963689717720280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Transplantation of stem cells that differentiate into more mature neural cells brings about functional improvement in preclinical studies of stroke. Previous transplant approaches in the diseased brain utilized injection of the cells in a cell suspension. In addition, neural stem cells were preferentially used for grafting. However, these cells had no specific relationship to the damaged tissue of stroke and brain injury patients. The injection of cells in a suspension destroyed the cell–cell interactions that are suggested to be important for promoting functional integrity of cortical motor neurons. In order to obtain suitable cell types for grafting in patients with stroke and brain damage, a protocol was modified for differentiating human induced pluripotent stem cells from cells phenotypically related to cortical motor neurons. Moreover, cell sheet technology was applied to neural cell transplantation, as maintaining the cell–cell communications is regarded important for the repair of host brain architecture. Accordingly, neuronal cell sheets that were positive Forebrain Embryonic Zinc Finger (Fez) family zinc finger 2 (FEZF2), COUP-TF-interacting protein 2, insulin-like growth factor–binding protein 4 (IGFBP4), cysteine-rich motor neuron 1 protein precursor (CRIM1), and forkhead box p2 (FOXP2) were developed. These markers are associated with cortical motoneurons that are appropriate for the transplant location in the lesions. The sheets allowed preservation of cell–cell interactions shown by synapsin1 staining after transplantation to damaged mouse brains. The sheet transplantation brought about partial structural restoration and the improvement of motor functions in hemiplegic mice. Collectively, the novel neuronal cell sheets were transplanted into damaged motor cortices; the cell sheets maintained cell–cell interactions and improved the motor functions in the hemiplegic model mice. The motoneuron cell sheets are possibly applicable for stroke patients and patients with brain damage by using patient-specific induced pluripotent stem cells.
Collapse
Affiliation(s)
- Noboru Suzuki
- 1 Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan.,2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Nagisa Arimitsu
- 1 Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan.,2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Jun Shimizu
- 1 Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan.,2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Kenji Takai
- 1 Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan.,2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Chieko Hirotsu
- 2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Yuji Ueda
- 2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Sueshige Wakisaka
- 2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Naruyoshi Fujiwara
- 1 Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan.,2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Tomoko Suzuki
- 2 Department of Regenerative Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| |
Collapse
|
17
|
Sekiya T, Holley MC. 'Surface Transplantation' for Nerve Injury and Repair: The Quest for Minimally Invasive Cell Delivery. Trends Neurosci 2018; 41:429-441. [PMID: 29625774 DOI: 10.1016/j.tins.2018.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 02/22/2018] [Accepted: 03/07/2018] [Indexed: 12/15/2022]
Abstract
Cell transplantation is an ambitious, but arguably realistic, therapy for repair of the nervous system. Cell delivery is a major challenge for clinical translation, especially given the apparently inhibitory astrogliotic environment in degenerated tissue. However, astrogliotic tissue also contains endogenous structural and biochemical cues that can be harnessed for functional repair. Minimizing damage to these cues during cell delivery could enhance cell integration. This theory is supported by studies with an auditory astrocyte scar model, in which cells delivered onto the surface of the damaged nerve were more successfully integrated in the host than those injected into the tissue. We consider the application of this less invasive approach for nerve injury and its potential application to some neurodegenerative disorders.
Collapse
Affiliation(s)
- Tetsuji Sekiya
- Department of Otolaryngology, Head and Neck Surgery, Kyoto University Graduate School of Medicine, Sakyou-ku, Kyoto, 606-8507, Japan; Hikone Chuo Hospital, Department of Neurological Surgery, Hikone Chuo Hospital, 421 Nishiima-cho, Hikone, 522-0054, Japan.
| | - Matthew C Holley
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, United Kingdom
| |
Collapse
|
18
|
Human Recombinant Peptide Sponge Enables Novel, Less Invasive Cell Therapy for Ischemic Stroke. Stem Cells Int 2018; 2018:4829534. [PMID: 29765415 PMCID: PMC5911312 DOI: 10.1155/2018/4829534] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/17/2017] [Accepted: 03/13/2018] [Indexed: 12/19/2022] Open
Abstract
Bone marrow stromal cell (BMSC) transplantation has the therapeutic potential for ischemic stroke. However, it is unclear which delivery routes would yield both safety and maximal therapeutic benefits. We assessed whether a novel recombinant peptide (RCP) sponge, that resembles human collagen, could act as a less invasive and beneficial scaffold in cell therapy for ischemic stroke. BMSCs from green fluorescent protein-transgenic rats were cultured and Sprague–Dawley rats were subjected to permanent middle cerebral artery occlusion (MCAo). A BMSC-RCP sponge construct was transplanted onto the ipsilateral intact neocortex 7 days after MCAo. A BMSC suspension or vehicle was transplanted into the ipsilateral striatum. Rat motor function was serially evaluated and histological analysis was performed 5 weeks after transplantation. The results showed that BMSCs could proliferate well in the RCP sponge and the BMSC-RCP sponge significantly promoted functional recovery, compared with the vehicle group. Histological analysis revealed that the RCP sponge provoked few inflammatory reactions in the host brain. Moreover, some BMSCs migrated to the peri-infarct area and differentiated into neurons in the BMSC-RCP sponge group. These findings suggest that the RCP sponge may be a promising candidate for animal protein-free scaffolds in cell therapy for ischemic stroke in humans.
Collapse
|
19
|
González-Nieto D, Fernández-García L, Pérez-Rigueiro J, Guinea GV, Panetsos F. Hydrogels-Assisted Cell Engraftment for Repairing the Stroke-Damaged Brain: Chimera or Reality. Polymers (Basel) 2018; 10:polym10020184. [PMID: 30966220 PMCID: PMC6415003 DOI: 10.3390/polym10020184] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 01/07/2023] Open
Abstract
The use of advanced biomaterials as a structural and functional support for stem cells-based therapeutic implants has boosted the development of tissue engineering applications in multiple clinical fields. In relation to neurological disorders, we are still far from the clinical reality of restoring normal brain function in neurodegenerative diseases and cerebrovascular disorders. Hydrogel polymers show unique mechanical stiffness properties in the range of living soft tissues such as nervous tissue. Furthermore, the use of these polymers drastically enhances the engraftment of stem cells as well as their capacity to produce and deliver neuroprotective and neuroregenerative factors in the host tissue. Along this article, we review past and current trends in experimental and translational research to understand the opportunities, benefits, and types of tentative hydrogel-based applications for the treatment of cerebral disorders. Although the use of hydrogels for brain disorders has been restricted to the experimental area, the current level of knowledge anticipates an intense development of this field to reach clinics in forthcoming years.
Collapse
Affiliation(s)
- Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
| | - Laura Fernández-García
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid 28040 Madrid, Spain.
| | - Gustavo V Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid 28040 Madrid, Spain.
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Instituto de Investigación Sanitaria, Hospital Clínico San Carlos Madrid, IdISSC, 28040 Madrid, Spain.
| |
Collapse
|
20
|
Laminin-521 Promotes Rat Bone Marrow Mesenchymal Stem Cell Sheet Formation on Light-Induced Cell Sheet Technology. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9474573. [PMID: 28164129 PMCID: PMC5253502 DOI: 10.1155/2017/9474573] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 12/30/2022]
Abstract
Rat bone marrow mesenchymal stem cell sheets (rBMSC sheets) are attractive for cell-based tissue engineering. However, methods of culturing rBMSC sheets are critically limited. In order to obtain intact rBMSC sheets, a light-induced cell sheet method was used in this study. TiO2 nanodot films were coated with (TL) or without (TN) laminin-521. We investigated the effects of laminin-521 on rBMSCs during cell sheet culturing. The fabricated rBMSC sheets were subsequently assessed to study cell sheet viability, reattachment ability, cell sheet thickness, collagen type I deposition, and multilineage potential. The results showed that laminin-521 could promote the formation of rBMSC sheets with good viability under hyperconfluent conditions. Cell sheet thickness increased from an initial 26.7 ± 1.5 μm (day 5) up to 47.7 ± 3.0 μm (day 10). Moreover, rBMSC sheets maintained their potential of osteogenic, adipogenic, and chondrogenic differentiation. This study provides a new strategy to obtain rBMSC sheets using light-induced cell sheet technology.
Collapse
|
21
|
Feasibility and Efficiency of Human Bone Marrow Stromal Cell Culture with Allogeneic Platelet Lysate-Supplementation for Cell Therapy against Stroke. Stem Cells Int 2016; 2016:6104780. [PMID: 27840648 PMCID: PMC5093274 DOI: 10.1155/2016/6104780] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/16/2016] [Accepted: 09/29/2016] [Indexed: 12/17/2022] Open
Abstract
Currently, there is increasing interest in human bone marrow stromal cells (hBMSCs) as regeneration therapy against cerebral stroke. The aim of the present study was to evaluate the feasibility and validity of hBMSC cultures with allogeneic platelet lysates (PLs). Platelet concentrates (PC) were harvested from healthy volunteers and made into single donor-derived PL (sPL). The PL mixtures (mPL) were made from three different sPL. Some growth factors and platelet cell surface antigens were detected by enzyme-linked immunosorbent assay (ELISA). The hBMSCs cultured with 10% PL were analyzed for their proliferative potential, surface markers, and karyotypes. The cells were incubated with superparamagnetic iron oxide (SPIO) agents and injected into a pig brain. MRI and histological analysis were performed. Consequently, nine lots of sPL and three mPL were prepared. ELISA analysis showed that PL contained adequate growth factors and a particle of platelet surface antigens. Cell proliferation capacity of PLs was equivalent to or higher than that of fetal calf serum (FCS). No contradiction in cell surface markers and no chromosomal aberrations were found. The MRI detected the distribution of SPIO-labeled hBMSCs in the pig brain. In summary, the hBMSCs cultured with allogeneic PL are suitable for cell therapy against stroke.
Collapse
|
22
|
Biomaterial Applications in Cell-Based Therapy in Experimental Stroke. Stem Cells Int 2016; 2016:6810562. [PMID: 27274738 PMCID: PMC4870368 DOI: 10.1155/2016/6810562] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/11/2016] [Accepted: 04/04/2016] [Indexed: 01/08/2023] Open
Abstract
Stroke is an important health issue corresponding to the second cause of mortality and first cause of severe disability with no effective treatments after the first hours of onset. Regenerative approaches such as cell therapy provide an increase in endogenous brain structural plasticity but they are not enough to promote a complete recovery. Tissue engineering has recently aroused a major interesting development of biomaterials for use into the central nervous system. Many biomaterials have been engineered based on natural compounds, synthetic compounds, or a mix of both with the aim of providing polymers with specific properties. The mechanical properties of biomaterials can be exquisitely regulated forming polymers with different stiffness, modifiable physical state that polymerizes in situ, or small particles encapsulating cells or growth factors. The choice of biomaterial compounds should be adapted for the different applications, structure target, and delay of administration. Biocompatibilities with embedded cells and with the host tissue and biodegradation rate must be considerate. In this paper, we review the different applications of biomaterials combined with cell therapy in ischemic stroke and we explore specific features such as choice of biomaterial compounds and physical and mechanical properties concerning the recent studies in experimental stroke.
Collapse
|