1
|
Hajimirzaei P, Tabatabaei FSA, Nasibi-Sis H, Razavian RS, Nasirinezhad F. Schwann cell transplantation for remyelination, regeneration, tissue sparing, and functional recovery in spinal cord injury: A systematic review and meta-analysis of animal studies. Exp Neurol 2025; 384:115062. [PMID: 39579959 DOI: 10.1016/j.expneurol.2024.115062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a significant global health challenge that results in profound physical and neurological impairments. Despite progress in medical care, the treatment options for SCI are still restricted and often focus on symptom management rather than promoting neural repair and functional recovery. This study focused on clarifying the impact of Schwann cell (SC) transplantation on the molecular, cellular, and functional basis of recovery in animal models of SCI. MATERIAL AND METHODS Relevant studies were identified by conducting searches across multiple databases, which included PubMed, Web of Science, Scopus, and ProQuest. The data were analyzed via comprehensive meta-analysis software. We assessed the risk of bias via the SYRCLE method. RESULTS The analysis included 59 studies, 48 of which provided quantitative data. The results revealed significant improvements in various outcome variables, including protein zero structures (SMD = 1.66, 95 %CI: 0.96-2.36; p < 0.001; I2 = 49.8 %), peripherally myelinated axons (SMD = 1.81, 95 %CI: 0.99-2.63; p < 0.001; I2 = 39.3 %), biotinylated dextran amine-labeled CST only rostral (SMD = 1.31, 95 % CI: 0.50-2.12, p < 0.01, I2 = 49.7 %), fast blue-labeled reticular formation (SMD = 0.96, 95 %CI: 0.43-1.49, p < 0.001, I2 = 0.0 %), 5-hydroxytryptamine caudally (SMD = 0.83, 95 %CI: 0.36-1.29, p < 0.001, I2 = 17.2 %) and epicenter (SMD = 0.85, 95 %CI: 0.17-1.53, p < 0.05, I2 = 62.7 %), tyrosine hydroxylase caudally (SMD = 1.86, 95 %CI: 1.14-2.59, p < 0.001, I2 = 0.0 %) and epicenter (SMD = 1.82, 95 %CI: 1.18-2.47, p < 0.001, I2 = 0.0 %), cavity volume (SMD = -2.07, 95 %CI: -2.90 - -1.24, p < 0.001, I2 = 67.2 %), and Basso, Beattie, and Bresnahan (SMD = 1.26, 95 %CI: 0.93-1.58; p < 0.001; I2 = 79.4 %). CONCLUSIONS This study demonstrates the promising potential of SC transplantation as a therapeutic approach for SCI, clarifying its impact on various biological processes critical for recovery.
Collapse
Affiliation(s)
- Pooya Hajimirzaei
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Nasibi-Sis
- Department of Medical Library and Information Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Farinaz Nasirinezhad
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Physiology Research Center, Iran University of Medical sciences, Tehran, Iran; Center of Experimental and Comparative Study, Iran University of Medical sciences, Tehran, Iran.
| |
Collapse
|
2
|
Guest JD, Santamaria AJ, Solano JP, de Rivero Vaccari JP, Dietrich WD, Pearse DD, Khan A, Levi AD. Challenges in advancing Schwann cell transplantation for spinal cord injury repair. Cytotherapy 2025; 27:36-50. [PMID: 39387736 DOI: 10.1016/j.jcyt.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AIMS In this article we aimed to provide an expert synthesis of the current status of Schwann cell (SC)therapeutics and potential steps to increase their clinical utility. METHODS We provide an expert synthesis based on preclinical, clinical and manufacturing experience. RESULTS Schwann cells (SCs) are essential for peripheral nerve regeneration and are of interest in supporting axonal repair after spinal cord injury (SCI). SCs can be isolated and cultivated in tissue culture from adult nerve biopsies or generated from precursors and neural progenitors using specific differentiation protocols leading to expanded quantities. In culture, they undergo dedifferentiation to a state similar to "repair" SCs. The known repertoire of SC functions is increasing beyond axon maintenance, myelination, and axonal regeneration to include immunologic regulation and the release of potentially therapeutic extracellular vesicles. Recently, autologous human SC cultures purified under cGMP conditions have been tested in both nerve repair and subacute and chronic SCI clinical trials. Although the effects of SCs to support nerve regeneration are indisputable, their efficacy for clinical SCI has been limited according to the outcomes examined. CONCLUSIONS This review discusses the current limitations of transplanted SCs within the damaged spinal cord environment. Limitations include limited post-transplant cell survival, the inability of SCs to migrate within astrocytic parenchyma, and restricted axonal regeneration out of SC-rich graft regions. We describe steps to amplify the survival and integration of transplanted SCs and to expand the repertoire of uses of SCs, including SC-derived extracellular vesicles. The relative merits of transplanting autologous versus allogeneic SCs and the role that endogenous SCs play in spinal cord repair are described. Finally, we briefly describe the issues requiring solutions to scale up SC manufacturing for commercial use.
Collapse
Affiliation(s)
- James D Guest
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Andrea J Santamaria
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P Solano
- Pediatric Critical Care, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P de Rivero Vaccari
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - William D Dietrich
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Aisha Khan
- The Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D Levi
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
3
|
Chiang B, Heng K, Jang K, Dalal R, Liao YJ, Myung D, Goldberg JL. Development of a novel SupraChoroidal-to-Optic-NervE (SCONE) drug delivery system. Drug Deliv 2024; 31:2379369. [PMID: 39010743 PMCID: PMC467098 DOI: 10.1080/10717544.2024.2379369] [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: 10/18/2023] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
PURPOSE Targeted drug delivery to the optic nerve head may be useful in the preclinical study and later clinical management of optic neuropathies, however, there are no FDA-approved drug delivery systems to achieve this. The purpose of this work was to develop an optic nerve head drug delivery technique. METHODS Different strategies to approach the optic nerve head were investigated, including standard intravitreal and retroorbital injections. A novel SupraChoroidal-to-Optic-NervE (SCONE) delivery was optimized by creating a sclerotomy and introducing a catheter into the suprachoroidal space. Under direct visualization, the catheter was guided to the optic nerve head. India ink was injected. The suprachoroidal approach was performed in New Zealand White rabbit eyes in vivo (25 animals total). Parameters, including microneedle size and design, catheter design, and catheter tip angle, were optimized ex vivo and in vivo. RESULTS Out of the candidate optic nerve head approaches, intravitreal, retroorbital, and suprachoroidal approaches were able to localize India ink to within 2 mm of the optic nerve. The suprachoroidal approach was further investigated, and after optimization, was able to deposit India ink directly within the optic nerve head in up to 80% of attempts. In eyes with successful SCONE delivery, latency and amplitude of visual evoked potentials was not different than the naïve untreated eye. CONCLUSIONS SCONE delivery can be used for targeted drug delivery to the optic nerve head of rabbits without measurable toxicity measured anatomically or functionally. Successful development of this system may yield novel opportunities to study optic nerve head-specific drug delivery in animal models, and paradigm-shifting management strategies for treating optic neuropathies. TRANSLATIONAL RELEVANCE Here we demonstrate data on a new method for targeted delivery to the optic nerve head, addressing a significant unmet need in therapeutics for optic neuropathies.
Collapse
Affiliation(s)
- Bryce Chiang
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, CA, USA
| | - Kathleen Heng
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, CA, USA
- Department of Comparative Medicine, Stanford University, Stanford, CA, USA
| | - Kyeongwoo Jang
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, CA, USA
| | - Roopa Dalal
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, CA, USA
| | - Yaping Joyce Liao
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, CA, USA
- Department of Neurology, Stanford University, Palo Alto, CA, USA
| | - David Myung
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, CA, USA
- Department of Chemical Engineering, Stanford University, Palo Alto, CA, USA
| | - Jeffrey L Goldberg
- Department of Ophthalmology, Spencer Center for Vision Research, Byers Eye Institute, Palo Alto, CA, USA
| |
Collapse
|
4
|
Paterson TE, Owen R, Sherborne C, Bahmaee H, Harding AL, Green NH, Reilly GC, Claeyssens F. Highly porous polycaprolactone microspheres for skeletal repair promote a mature bone cell phenotype in vitro. J Mater Chem B 2024; 12:11746-11758. [PMID: 39415638 DOI: 10.1039/d4tb01532k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Improving our ability to treat skeletal defects is a critical medical challenge that necessitates the development of new biomaterials. One promising approach involves the use of degradable polymer microparticles with an interconnected internal porosity. Here, we employed a double emulsion to generate such round microparticles (also known as microspheres) from a polycaprolactone-based polymerised high internal phase emulsion (polyHIPE). These microspheres effectively supported the growth of mesenchymal progenitors over a 30-day period, and when maintained in osteogenic media, cells deposited a bone-like extracellular matrix, as determined by histological staining for calcium and collagen. Interestingly, cells with an osteocyte-like morphology were observed within the core of the microspheres indicating the role of a physical environment comparable to native bone for this phenotype to occur. At later timepoints, these cultures had significantly increased mRNA expression of the osteocyte-specific markers dentin matrix phosphoprotein-1 (Dmp-1) and sclerostin, with sclerostin also observed at the protein level. Cells pre-cultured on porous microspheres exhibited enhanced survival rates compared to those pre-cultured on non-porous counterparts when injected. Cells precultured on both porous and non-porous microspheres promoted angiogenesis in a chorioallantoic membrane (CAM) assay. In summary, the polycaprolactone polyHIPE microspheres developed in this study exhibit significant promise as an alternative to traditional synthetic bone graft substitutes, offering a conducive environment for cell growth and differentiation, with the potential for better clinical outcomes in bone repair and regeneration.
Collapse
Affiliation(s)
- Thomas E Paterson
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield, UK.
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Robert Owen
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| | - Colin Sherborne
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield, UK.
| | - Hossein Bahmaee
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield, UK.
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Amy L Harding
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Nicola H Green
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield, UK.
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Gwendolen C Reilly
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield, UK.
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Frederik Claeyssens
- Department of Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield, UK.
| |
Collapse
|
5
|
Bedar M, Pulos NA, Shin AY. Dynamic Seeding versus Microinjection of Adipose-Derived Mesenchymal Stem Cells to Acellular Nerve Allograft Reconstructions. Plast Reconstr Surg 2024; 154:114e-125e. [PMID: 37537724 PMCID: PMC10838349 DOI: 10.1097/prs.0000000000010970] [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] [Indexed: 08/05/2023]
Abstract
BACKGROUND Functional recovery after acellular nerve allograft (ANA) reconstruction remains inferior to that after autologous nerve grafting, but improved outcomes have been demonstrated with the addition of adipose-derived mesenchymal stem cells (MSCs). Controversy exists regarding the optimal cell-delivery method to enhance ANA reconstructions. The authors investigated the functional recovery of ANAs after dynamic seeding versus microinjection of MSCs. METHODS Forty Lewis rats underwent reconstruction of a 10-mm sciatic nerve defect. Animals were divided into 4 groups: reversed autograft, ANA alone, dynamically seeded ANA, or ANA injected with MSCs. During the survival period, ultrasound measurements of the tibialis anterior muscle cross-sectional area were performed. At 12 weeks, functional recovery was evaluated using measurements of ankle contracture, compound muscle action potential, maximum isometric tetanic force, muscle mass, histomorphometry, and immunofluorescence. RESULTS The dynamic seeding and microinjection groups demonstrated higher cross-sectional tibialis anterior muscle area recovery than autografts and ANAs alone at week 8 and weeks 4 and 8, respectively. The ankle contracture and compound muscle action potential amplitude recovery were superior in autografts and both seeding methods compared with ANAs alone. The microinjection group demonstrated significantly higher isometric tetanic force, muscle mass, and number of axons compared with ANAs alone. Both seeding methods showed higher CD34 densities compared with ANAs alone. No significant differences between dynamic seeding and microinjection were observed in functional or histologic outcomes. CONCLUSIONS The addition of MSCs to ANAs demonstrated earlier motor regeneration compared with autografts and ANAs alone. Both seeding methods improved functional outcomes in the rat sciatic nerve defect model.
Collapse
Affiliation(s)
- Meiwand Bedar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Plastic Surgery, Nijmegen, The Netherlands
| | | | | |
Collapse
|
6
|
Frost OG, Ramkilawan P, Rebbaa A, Stolzing A. A systematic review of lifespan studies in rodents using stem cell transplantations. Ageing Res Rev 2024; 97:102295. [PMID: 38588866 DOI: 10.1016/j.arr.2024.102295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Organismal aging involves the progressive decline in organ function and increased susceptibility to age-associated diseases. Regardless of its origin, cellular aging is consequently reflected at the level of organ and associated systems dysfunction. Aging of stem cell populations within the body and their decreased ability to self-renew, differentiate, and regenerate damaged tissues, is a key contributor to organismal decline. Based on this, supplementing young stem cells may delay tissue aging, improve frailty and extend health and lifespan. This review investigates studies in rodents using stem cell transplantation from either mice or human donors. The aim is to consolidate available information on the efficacy of stem cell therapies in rodent models and provide insights to guide further research efforts. Out of the 21 studies included in this review, the methodology varied significantly including the lifespan measurement. To enable comparison the median lifespan was calculated using WebPlotDigitizer 4.6 if not provided by the literature. A total of 18 out of 21 studies evidenced significant lifespan extension post stem cell transplant, with 7 studies demonstrating benefits in reduced frailty and other aging complications.
Collapse
Affiliation(s)
- Oliver G Frost
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK; SENS Research Foundation, Mountain View, CA 94041, USA
| | | | | | - Alexandra Stolzing
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK.
| |
Collapse
|
7
|
Rolsma JL, Darch W, Higgins NC, Morgan JT. The tardigrade-derived mitochondrial abundant heat soluble protein improves adipose-derived stem cell survival against representative stressors. Sci Rep 2024; 14:11834. [PMID: 38783150 PMCID: PMC11116449 DOI: 10.1038/s41598-024-62693-w] [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: 03/14/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
Human adipose-derived stem cell (ASC) grafts have emerged as a powerful tool in regenerative medicine. However, ASC therapeutic potential is hindered by stressors throughout their use. Here we demonstrate the transgenic expression of the tardigrade-derived mitochondrial abundant heat soluble (MAHS) protein for improved ASC resistance to metabolic, mitochondrial, and injection shear stress. In vitro, MAHS-expressing ASCs demonstrate up to 61% increased cell survival following 72 h of incubation in phosphate buffered saline containing 20% media. Following up to 3.5% DMSO exposure for up to 72 h, a 14-49% increase in MAHS-expressing ASC survival was observed. Further, MAHS expression in ASCs is associated with up to 39% improved cell viability following injection through clinically relevant 27-, 32-, and 34-gauge needles. Our results reveal that MAHS expression in ASCs supports survival in response to a variety of common stressors associated with regenerative therapies, thereby motivating further investigation into MAHS as an agent for stem cell stress resistance. However, differentiation capacity in MAHS-expressing ASCs appears to be skewed in favor of osteogenesis over adipogenesis. Specifically, activity of the early bone formation marker alkaline phosphatase is increased by 74% in MAHS-expressing ASCs following 14 days in osteogenic media. Conversely, positive area of the neutral lipid droplet marker BODIPY is decreased by up to 10% in MAHS-transgenic ASCs following 14 days in adipogenic media. Interestingly, media supplementation with up to 40 mM glucose is sufficient to restore adipogenic differentiation within 14 days, prompting further analysis of mechanisms underlying interference between MAHS and differentiation processes.
Collapse
Affiliation(s)
- Jordan L Rolsma
- Department of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - William Darch
- Department of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - Nicholas C Higgins
- Department of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - Joshua T Morgan
- Department of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA.
| |
Collapse
|
8
|
Heuer M, Stiti M, Eras V, Scholz J, Ahmed N, Berrocal E, Brune JC. High-Speed Fluorescence Imaging Corroborates Biological Data on the Influence of Different Nozzle Types on Cell Spray Viability and Formation. J Funct Biomater 2024; 15:126. [PMID: 38786637 PMCID: PMC11122036 DOI: 10.3390/jfb15050126] [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: 04/04/2024] [Revised: 04/29/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Treating severe dermal disruptions often presents significant challenges. Recent advancements have explored biological cell sprays as a promising treatment, but their success hinges on efficient cell delivery and complete wound coverage. This requires a good spray distribution with a small droplet size, high particle number, and ample surface coverage. The type of nozzle used with the spray device can impact these parameters. To evaluate the influence of different nozzles on spray characteristics, we compared air-assisted and unassisted nozzles. The unassisted nozzle displayed small particle size, high particle number, good overall coverage, high cell viability, preserved cell metabolic activity, and low cytotoxicity. Air-assisted nozzles did not perform well regarding cell viability and metabolic activity. Flow visualization analysis comparing two different unassisted nozzles using high-speed imaging (100 kHz frame rate) revealed a tulip-shaped spray pattern, indicating optimal spray distribution. High-speed imaging showed differences between the unassisted nozzles. One unassisted nozzle displayed a bi-modal distribution of the droplet diameter while the other unassisted nozzle displayed a mono-modal distribution. These findings demonstrate the critical role of nozzle selection in successful cell delivery. A high-quality, certified nozzle manufactured for human application omits the need for an air-assisted nozzle and provides a simple system to use with similar or better performance characteristics than those of an air-assisted system.
Collapse
Affiliation(s)
- Miriam Heuer
- German Institute for Cell and Tissue Replacement (DIZG, gemeinnützige GmbH), Haus 42, Köpenicker Str. 325, 12555 Berlin, Germany
| | - Mehdi Stiti
- Division of Combustion Physics, Department of Physics, Lund University, P.O. Box 118, 22100 Lund, Sweden
- Institut de Mécanique des Fluides de Toulouse (IMFT), CNRS, Université de Toulouse, 31400 Toulouse, France
| | - Volker Eras
- German Institute for Cell and Tissue Replacement (DIZG, gemeinnützige GmbH), Haus 42, Köpenicker Str. 325, 12555 Berlin, Germany
| | - Julia Scholz
- German Institute for Cell and Tissue Replacement (DIZG, gemeinnützige GmbH), Haus 42, Köpenicker Str. 325, 12555 Berlin, Germany
| | - Norus Ahmed
- German Institute for Cell and Tissue Replacement (DIZG, gemeinnützige GmbH), Haus 42, Köpenicker Str. 325, 12555 Berlin, Germany
| | - Edouard Berrocal
- Division of Combustion Physics, Department of Physics, Lund University, P.O. Box 118, 22100 Lund, Sweden
| | - Jan C Brune
- German Institute for Cell and Tissue Replacement (DIZG, gemeinnützige GmbH), Haus 42, Köpenicker Str. 325, 12555 Berlin, Germany
| |
Collapse
|
9
|
Fırlak Demirkan M, Öztürk D, Çifçibaşı ZS, Ertan F, Hardy JG, Nurşeval Oyunlu A, Darıcı H. Controlled Sr(ii) ion release from in situ crosslinking electroactive hydrogels with potential for the treatment of infections. RSC Adv 2024; 14:4324-4334. [PMID: 38304567 PMCID: PMC10828636 DOI: 10.1039/d3ra07061a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/26/2023] [Indexed: 02/03/2024] Open
Abstract
The development of electrochemical stimuli-responsive drug delivery systems is of both academic and industrial interest due to the ease with which it is possible to trigger payload release, providing drug delivery in a controllable manner. Herein, the preparation of in situ forming hydrogels including electroactive polypyrrole nanoparticles (PPy-NPs) where Sr2+ ions are electrochemically loaded for electrically triggered release of Sr2+ ions is reported. The hydrogels were characterized by a variety of techniques including Fourier-transform infrared (FTIR) spectroscopy, scanning electron microscopy (SEM), energy dispersive X-ray spectroscopy (EDX), thermogravimetric analysis (TGA), X-ray diffraction (XRD), cyclic voltammetry (CV), etc. The cytocompatibility towards human mesenchymal stem cells (MSCs) and fibroblasts were also studied. The Sr2+ ion loaded PEC-ALD/CS/PPy-NPs hydrogel showed no significant cytotoxicity towards human mesenchymal stem cells (MSCs) and fibroblasts. Sr2+ ions were electrochemically loaded and released from the electroactive hydrogels, and the application of an electrical stimulus enhanced the release of Sr2+ ions from gels by ca. 2-4 fold relative to the passive release control experiment. The antibacterial activity of Sr2+ ions against E. coli and S. aureus was demonstrated in vitro. Although these prototypical examples of Sr2+ loaded electroactive gels don't release sufficient Sr2+ ions to show antibacterial activity against E. coli and S. aureus, we believe future iterations with optimised physical properties of the gels will be capable of doing so.
Collapse
Affiliation(s)
| | - Dilek Öztürk
- Department of Chemistry, Gebze Technical University Gebze Kocaeli 41400 Turkey
| | | | - Fatma Ertan
- Department of Chemistry, Gebze Technical University Gebze Kocaeli 41400 Turkey
| | | | | | - Hakan Darıcı
- HD Bioink Biotechnology Corp. İstanbul Turkey
- 3D Bioprinting Design & Prototyping R&D Center, Istinye University Istanbul Turkey
- Faculty of Medicine, Dept. of Histology & Embryology, Istinye University Istanbul Turkey
- Stem Cell, and Tissue Engineering R&D Center, Istinye University Istanbul Turkey
| |
Collapse
|
10
|
Maheshwari S, Akram H, Bulstrode H, Kalia SK, Morizane A, Takahashi J, Natalwala A. Dopaminergic Cell Replacement for Parkinson's Disease: Addressing the Intracranial Delivery Hurdle. JOURNAL OF PARKINSON'S DISEASE 2024; 14:415-435. [PMID: 38457149 PMCID: PMC11091588 DOI: 10.3233/jpd-230328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/16/2024] [Indexed: 03/09/2024]
Abstract
Parkinson's disease (PD) is an increasingly prevalent neurological disorder, affecting more than 8.5 million individuals worldwide. α-Synucleinopathy in PD is considered to cause dopaminergic neuronal loss in the substantia nigra, resulting in characteristic motor dysfunction that is the target for current medical and surgical therapies. Standard treatment for PD has remained unchanged for several decades and does not alter disease progression. Furthermore, symptomatic therapies for PD are limited by issues surrounding long-term efficacy and side effects. Cell replacement therapy (CRT) presents an alternative approach that has the potential to restore striatal dopaminergic input and ameliorate debilitating motor symptoms in PD. Despite promising pre-clinical data, CRT has demonstrated mixed success clinically. Recent advances in graft biology have renewed interest in the field, resulting in several worldwide ongoing clinical trials. However, factors surrounding the effective neurosurgical delivery of cell grafts have remained under-studied, despite their significant potential to influence therapeutic outcomes. Here, we focus on the key neurosurgical factors to consider for the clinical translation of CRT. We review the instruments that have been used for cell graft delivery, highlighting current features and limitations, while discussing how future devices could address these challenges. Finally, we review other novel developments that may enhance graft accessibility, delivery, and efficacy. Challenges surrounding neurosurgical delivery may critically contribute to the success of CRT, so it is crucial that we address these issues to ensure that CRT does not falter at the final hurdle.
Collapse
Affiliation(s)
- Saumya Maheshwari
- The Medical School, University of Edinburgh, Edinburgh BioQuarter, UK
| | - Harith Akram
- Unit of Functional Neurosurgery, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Harry Bulstrode
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, Division of Academic Neurosurgery, University of Cambridge, Cambridge, UK
| | - Suneil K. Kalia
- Division of Neurosurgery, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Asuka Morizane
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Regenerative Medicine, Center for Clinical Research and Innovation, Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ammar Natalwala
- Unit of Functional Neurosurgery, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
- Department for Neuromuscular Diseases, Institute of Neurology, University College London, London, UK
| |
Collapse
|
11
|
Mills SJ, Kirby GT, Hofma BR, Smith LE, Statham P, Vaes B, Ting AE, Short R, Cowin AJ. Delivery of multipotent adult progenitor cells via a functionalized plasma polymerized surface accelerates healing of murine diabetic wounds. Front Bioeng Biotechnol 2023; 11:1213021. [PMID: 37675407 PMCID: PMC10477914 DOI: 10.3389/fbioe.2023.1213021] [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: 04/27/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction: Stem cell therapies have been investigated as potential treatment modalities for chronic wounds however there has been limited success to date. Multipotent Adult Progenitor Cells (MAPCs©) have been identified as having potential as an allogenic stem cell product due to their high population doubling number and their characteristic dampening of T-cell proliferation. This helps to prevent autoimmunity and graft/cell rejection. Methods: We have developed a dressing, consisting of medical grade silicone coated with a heptylamine plasma polymer, which supports the growth and transfer of MAPCs to skin. To determine if the dressing can deliver functional stem cells into diabetic wounds, they were loaded with MAPCs and then placed over excisional wounds in both normal and diabetic mice. Results and discussion: Accelerated healing was observed in both the normal and diabetic wounds with wound gape being significantly smaller at day 3 when compared to controls. Wound analysis showed that treatment with the MAPC dressings dampened the inflammatory response with reduced numbers of neutrophils and macrophages observed. Additionally, an increase in pro-angiogenic VEGF and CD31 positive endothelial cells was observed indicating improved new blood vessel formation. The MAPC dressings had no effect on fibrosis with collagen I and III being equally affected in both control and treated wounds. Overall, the functionalized MAPC dressings improve healing responses particularly in diabetic mice with impaired healing responses and therefore, show potential for development as an advanced therapeutic approach for the treatment of chronic diabetic wounds.
Collapse
Affiliation(s)
- S. J. Mills
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - G. T. Kirby
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - B. R. Hofma
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - L. E. Smith
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - P. Statham
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - B. Vaes
- ReGenesys BV, Bio-Incubator Leuven, Leuven, Belgium
| | - A. E. Ting
- Athersys Inc., Cleveland, OH, United States
| | - R. Short
- Material Science Institute, Lancaster University, Lancaster, United Kingdom
| | - A. J. Cowin
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| |
Collapse
|
12
|
Zhu F, Nie G, Liu C. Engineered biomaterials in stem cell-based regenerative medicine. LIFE MEDICINE 2023; 2:lnad027. [PMID: 39872549 PMCID: PMC11749850 DOI: 10.1093/lifemedi/lnad027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/17/2023] [Indexed: 01/30/2025]
Abstract
Stem cell-based regenerative therapies, which harness the self-renewal and differentiation properties of stem cells, have been in the spotlight due to their widespread applications in treating degenerative, aging, and other, generally intractable diseases. Therapeutically effective hematopoietic stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells have been used in numerous basic and translational studies with exciting results. However, pre-/post-transplantation issues of poor cell survival and retention, uncontrolled differentiation, and insufficient numbers of cells engrafted into host tissues are the major challenges in stem cell-based regenerative therapies. Engineered biomaterials have adjustable biochemical and biophysical properties that significantly affect cell behaviors, such as cell engraftment, survival, migration, and differentiation outcomes, thereby enhancing the engraftment of implanted stem cells and guiding tissue regeneration. Therefore, the combination of stem cell biology with bioengineered materials is a promising strategy to improve the therapeutic outcomes of stem cell-based regenerative therapy. In this review, we summarize the advances in the modulation of behaviors of stem cells via engineered biomaterials. We then present different approaches to harnessing bioengineered materials to enhance the transplantation of stem cells. Finally, we will provide future directions in regenerative therapy using stem cells.
Collapse
Affiliation(s)
- Fei Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
13
|
Schluck M, Weiden J, Verdoes M, Figdor CG. Insights in the host response towards biomaterial-based scaffolds for cancer therapy. Front Bioeng Biotechnol 2023; 11:1149943. [PMID: 37342507 PMCID: PMC10277494 DOI: 10.3389/fbioe.2023.1149943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Immunotherapeutic strategies have shown promising results in the treatment of cancer. However, not all patients respond, and treatments can have severe side-effects. Adoptive cell therapy (ACT) has shown remarkable therapeutic efficacy across different leukaemia and lymphoma types. But the treatment of solid tumours remains a challenge due to limited persistence and tumour infiltration. We believe that biomaterial-based scaffolds are promising new tools and may address several of the challenges associated with cancer vaccination and ACT. In particular, biomaterial-based scaffold implants allow for controlled delivery of activating signals and/or functional T cells at specific sites. One of the main challenges for their application forms the host response against these scaffolds, which includes unwanted myeloid cell infiltration and the formation of a fibrotic capsule around the scaffold, thereby limiting cell traffic. In this review we provide an overview of several of the biomaterial-based scaffolds designed for cancer therapy to date. We will discuss the host responses observed and we will highlight design parameters that influence this response and their potential impact on therapeutic outcome.
Collapse
Affiliation(s)
- Marjolein Schluck
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Jorieke Weiden
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Carl G. Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| |
Collapse
|
14
|
Risser GE, Machour M, Hernaez-Estrada B, Li D, Levenberg S, Spiller KL. Effects of Interleukin-4 (IL-4)-releasing microparticles and adoptive transfer of macrophages on immunomodulation and angiogenesis. Biomaterials 2023; 296:122095. [PMID: 36989737 PMCID: PMC10085857 DOI: 10.1016/j.biomaterials.2023.122095] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/16/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
Macrophages are major regulators of angiogenesis in response to injury, but the mechanisms behind their diverse and phenotypically specific functions are still poorly understood. In particular, the effects of interleukin-4 (IL-4) on macrophage behavior have been well studied in vitro, but it remains unclear whether the release of IL-4 from biomaterials can be used to control macrophage phenotype and subsequent effects on angiogenesis in vivo. We used the murine hindlimb ischemia model to investigate the effects of IL-4-releasing poly(lactic-co-glycolic acid) microparticles co-delivered with IL-4-polarized macrophages on host macrophage phenotype and angiogenesis in vivo. We established a minimum dose of IL-4 required to modulate macrophage phenotype in vivo and evaluated effects on macrophage subpopulation diversity using multidimensional flow cytometry and pseudotime analysis. The delivery of IL-4-releasing microparticles did not affect the density or size of blood vessels as measured by immunohistochemical (IHC) analysis, but it did increase perfused tissue volume as measured by 3D microcomputed tomography (microCT). In contrast, the co-delivery of IL-4-releasing microparticles and exogenously IL-4-polarized macrophages increased the size of blood vessels as measured by IHC, but without effects on perfused tissue volume via microCT. These effects occurred in spite of low recovery of adoptively transferred macrophages at 4 days after administration. Spatial analysis of macrophage-blood vessel interactions via IHC showed that macrophages closely interacted with blood vessels, which was slightly influenced by treatment, and that blood vessel size was positively correlated with number of macrophages in close proximity. Altogether, these findings indicate that delivery of IL-4-releasing microparticles and exogenously IL-4-polarized macrophages can be beneficial for angiogenesis, but further mechanistic investigations are required.
Collapse
Affiliation(s)
- Gregory E Risser
- School of Biomedical Engineering, Sciences and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Majd Machour
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Beatriz Hernaez-Estrada
- School of Biomedical Engineering, Sciences and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Dong Li
- Shanghai Key Tissue Engineering Laboratory, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Kara L Spiller
- School of Biomedical Engineering, Sciences and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Ingle RG, Fang WJ. An Overview of the Stability and Delivery Challenges of Commercial Nucleic Acid Therapeutics. Pharmaceutics 2023; 15:pharmaceutics15041158. [PMID: 37111643 PMCID: PMC10143938 DOI: 10.3390/pharmaceutics15041158] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Nucleic acid (NA)-based biopharmaceuticals have emerged as promising therapeutic modalities. NA therapeutics are a diverse class of RNA and DNA and include antisense oligonucleotides, siRNA, miRNA, mRNA, small activating RNA, and gene therapies. Meanwhile, NA therapeutics have posed significant stability and delivery challenges and are expensive. This article discusses the challenges and opportunities for achieving stable formulations of NAs with novel drug delivery systems (DDSs). Here we review the current progress in the stability issues and the significance of novel DDSs associated with NA-based biopharmaceuticals, as well as mRNA vaccines. We also highlight the European Medicines Agency (EMA) and US Food and Drug Administration (FDA)-approved NA-based therapeutics with their formulation profiles. NA therapeutics could impact future markets if the remaining challenges and requirements are addressed. Regardless of the limited information available for NA therapeutics, reviewing and collating the relevant facts and figures generates a precious resource for formulation experts familiar with the NA therapeutics' stability profile, their delivery challenges, and regulatory acceptance.
Collapse
Affiliation(s)
- Rahul G Ingle
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310027, China
- Dr. Rajendra Gode College of Pharmacy, Amravati 444602, India
| | - Wei-Jie Fang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
16
|
Karaz S, Senses E. Liposomes Under Shear: Structure, Dynamics, and Drug Delivery Applications. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Selcan Karaz
- Department of Chemical and Biological Engineering Koç University Istanbul 34450 Turkey
| | - Erkan Senses
- Department of Chemical and Biological Engineering Koç University Istanbul 34450 Turkey
| |
Collapse
|
17
|
Shin HS, Thakore A, Tada Y, Pedroza AJ, Ikeda G, Chen IY, Chan D, Jaatinen KJ, Yajima S, Pfrender EM, Kawamura M, Yang PC, Wu JC, Appel EA, Fischbein MP, Woo YJ, Shudo Y. Angiogenic stem cell delivery platform to augment post-infarction neovasculature and reverse ventricular remodeling. Sci Rep 2022; 12:17605. [PMID: 36266453 PMCID: PMC9584918 DOI: 10.1038/s41598-022-21510-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 09/28/2022] [Indexed: 01/13/2023] Open
Abstract
Many cell-based therapies are challenged by the poor localization of introduced cells and the use of biomaterial scaffolds with questionable biocompatibility or bio-functionality. Endothelial progenitor cells (EPCs), a popular cell type used in cell-based therapies due to their robust angiogenic potential, are limited in their therapeutic capacity to develop into mature vasculature. Here, we demonstrate a joint delivery of human-derived endothelial progenitor cells (EPC) and smooth muscle cells (SMC) as a scaffold-free, bi-level cell sheet platform to improve ventricular remodeling and function in an athymic rat model of myocardial infarction. The transplanted bi-level cell sheet on the ischemic heart provides a biomimetic microenvironment and improved cell-cell communication, enhancing cell engraftment and angiogenesis, thereby improving ventricular remodeling. Notably, the increased density of vessel-like structures and upregulation of biological adhesion and vasculature developmental genes, such as Cxcl12 and Notch3, particularly in the ischemic border zone myocardium, were observed following cell sheet transplantation. We provide compelling evidence that this SMC-EPC bi-level cell sheet construct can be a promising therapy to repair ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Hye Sook Shin
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Akshara Thakore
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Yuko Tada
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Albert J Pedroza
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Gentaro Ikeda
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Ian Y Chen
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Doreen Chan
- Department of Chemistry, Department of Materials Science & Engineering, Stanford University, Stanford University, Stanford, USA
| | - Kevin J Jaatinen
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Shin Yajima
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Eric M Pfrender
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Masashi Kawamura
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Phillip C Yang
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Joseph C Wu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Eric A Appel
- Department of Materials Science & Engineering, Department of Bioengineering, Department of Pediatric (Endocrinology), Stanford University, Stanford, USA
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - YJoseph Woo
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Yasuhiro Shudo
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA.
| |
Collapse
|
18
|
Jury M, Matthiesen I, Rasti Boroojeni F, Ludwig SL, Civitelli L, Winkler TE, Selegård R, Herland A, Aili D. Bioorthogonally Cross-Linked Hyaluronan-Laminin Hydrogels for 3D Neuronal Cell Culture and Biofabrication. Adv Healthc Mater 2022; 11:e2102097. [PMID: 35114074 PMCID: PMC11468931 DOI: 10.1002/adhm.202102097] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/31/2022] [Indexed: 12/13/2022]
Abstract
Laminins (LNs) are key components in the extracellular matrix of neuronal tissues in the developing brain and neural stem cell niches. LN-presenting hydrogels can provide a biologically relevant matrix for the 3D culture of neurons toward development of advanced tissue models and cell-based therapies for the treatment of neurological disorders. Biologically derived hydrogels are rich in fragmented LN and are poorly defined concerning composition, which hampers clinical translation. Engineered hydrogels require elaborate and often cytotoxic chemistries for cross-linking and LN conjugation and provide limited possibilities to tailor the properties of the materials. Here a modular hydrogel system for neural 3D cell cultures, based on hyaluronan and poly(ethylene glycol), that is cross-linked and functionalized with human recombinant LN-521 using bioorthogonal copper-free click chemistry, is shown. Encapsulated human neuroblastoma cells demonstrate high viability and grow into spheroids. Long-term neuroepithelial stem cells (lt-NES) cultured in the hydrogels can undergo spontaneous differentiation to neural fate and demonstrate significantly higher viability than cells cultured without LN. The hydrogels further support the structural integrity of 3D bioprinted structures and maintain high viability of bioprinted and syringe extruded lt-NES, which can facilitate biofabrication and development of cell-based therapies.
Collapse
Affiliation(s)
- Michael Jury
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
| | - Isabelle Matthiesen
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyStockholm100 44Sweden
| | - Fatemeh Rasti Boroojeni
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
| | - Saskia L. Ludwig
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyStockholm100 44Sweden
| | - Livia Civitelli
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
- Nuffield Department of Clinical NeurosciencesJohn Radcliffe HospitalWest WingUniversity of OxfordOxfordOX3 9DUUK
| | - Thomas E. Winkler
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyStockholm100 44Sweden
- Institute of MicrotechnologyCenter of Pharmaceutical EngineeringTechnische Universität BraunschweigBraunschweig38106Germany
| | - Robert Selegård
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
| | - Anna Herland
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyStockholm100 44Sweden
- AIMES, Center for Integrated Medical and Engineering ScienceDepartment of NeuroscienceKarolinska InstituteSolna171 65Sweden
- Division of NanobiotechnologyDepartment of Protein Science, Science for Life LaboratoryKTH Royal Institute of TechnologyStockholm17165Sweden
| | - Daniel Aili
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
| |
Collapse
|
19
|
Dynamic seeding versus microinjection of mesenchymal stem cells for acellular nerve allograft: an in vitro comparison. J Plast Reconstr Aesthet Surg 2022; 75:2821-2830. [PMID: 35570113 PMCID: PMC9391259 DOI: 10.1016/j.bjps.2022.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/17/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-supplemented acellular nerve allografts (ANA) are a potential strategy to improve the treatment of segmental nerve defects. Prior to clinical translation, optimal cell delivery methods must be defined. While two techniques, dynamic seeding and microinjection, have been described, the seeding efficiency, cell viability, and distribution of MSCs in ANAs are yet to be compared. METHODS Sciatic nerve segments of Sprague-Dawley rats were decellularized, and MSCs were harvested from the adipose tissue of Lewis rats. Cell viability was evaluated after injection of MSCs through a 27-gauge needle at different flow rates (10, 5, and 1 µL/min). MSCs were dynamically seeded or longitudinally injected into ANAs. Cell viability, seeding efficiency, and distribution were evaluated using LIVE/DEAD and MTS assays, scanning electron microscopy, and Hoechst staining. RESULTS No statistically significant difference in cell viability after injection at different flow rates was seen. After cell delivery, 84.1 ± 3.7% and 87.8 ± 2.8% of MSCs remained viable in the dynamic seeding and microinjection group, respectively (p = 0.41). The seeding efficiency of microinjection (100.4%±5.6) was significantly higher than dynamic seeding (48.1%±8.6) on day 1 (p = 0.001). Dynamic seeding demonstrated a significantly more uniform cell distribution over the course of the ANA compared to microinjection (p = 0.02). CONCLUSION MSCs remain viable after both dynamic seeding and microinjection in ANAs. Higher seeding efficiency was observed with microinjection, but dynamic seeding resulted in a more uniform distribution. In vivo studies are required to assess the effect on gene expression profiles and functional motor outcomes.
Collapse
|
20
|
Geng R, Knoll J, Harland N, Amend B, Enderle MD, Linzenbold W, Abruzzese T, Kalbe C, Kemter E, Wolf E, Schenk M, Stenzl A, Aicher WK. Replacing Needle Injection by a Novel Waterjet Technology Grants Improved Muscle Cell Delivery in Target Tissues. Cell Transplant 2022; 31:9636897221080943. [PMID: 35466714 PMCID: PMC9036380 DOI: 10.1177/09636897221080943] [Citation(s) in RCA: 4] [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
Current regimen to treat patients suffering from stress urinary incontinence often seems not to yield satisfactory improvement or may come with severe side effects. To overcome these hurdles, preclinical studies and clinical feasibility studies explored the potential of cell therapies successfully and raised high hopes for better outcome. However, other studies were rather disappointing. We therefore developed a novel cell injection technology to deliver viable cells in the urethral sphincter complex by waterjet instead of using injection needles. We hypothesized that the risk of tissue injury and loss of cells could be reduced by a needle-free injection technology. Muscle-derived cells were obtained from young male piglets and characterized. Upon expansion and fluorescent labeling, cells were injected into cadaveric tissue samples by either waterjet or injection needle. In other experiments, labeled cells were injected by waterjet in the urethra of living pigs and incubated for up to 7 days of follow-up. The analyses documented that the cells injected by waterjet in vitro were viable and proliferated well. Upon injection in live animals, cells appeared undamaged, showed defined cellular somata with distinct nuclei, and contained intact chromosomal DNA. Most importantly, by in vivo waterjet injections, a significantly wider cell distribution was observed when compared with needle injections (P < .05, n ≥ 12 samples). The success rates of waterjet cell application in living animals were significantly higher (≥95%, n = 24) when compared with needle injections, and the injection depth of cells in the urethra could be adapted to the need by adjusting waterjet pressures. We conclude that the novel waterjet technology injects viable muscle cells in tissues at distinct and predetermined depth depending on the injection pressure employed. After waterjet injection, loss of cells by full penetration or injury of the tissue targeted was reduced significantly in comparison with our previous studies employing needle injections.
Collapse
Affiliation(s)
- Ruizhi Geng
- Department of Urology, Center for Medical Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Jasmin Knoll
- Department of Urology, Center for Medical Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Niklas Harland
- Department of Urology, University of Tübingen Hospital, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Bastian Amend
- Department of Urology, University of Tübingen Hospital, Eberhard Karl University of Tübingen, Tübingen, Germany
| | | | | | - Tanja Abruzzese
- Department of Urology, Center for Medical Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Claudia Kalbe
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Elisabeth Kemter
- Department of Molecular Animal Breeding and Biotechnology, LMU Munich, Oberschleißheim, Germany.,Center for Innovative Medical Models, LMU Munich, Oberschleißheim, Germany
| | - Eckhard Wolf
- Department of Molecular Animal Breeding and Biotechnology, LMU Munich, Oberschleißheim, Germany.,Center for Innovative Medical Models, LMU Munich, Oberschleißheim, Germany
| | - Martin Schenk
- Department of Surgery, University of Tübingen Hospital, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tübingen Hospital, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Wilhelm K Aicher
- Department of Urology, Center for Medical Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| |
Collapse
|
21
|
Nguyen Thanh T, Laowattanatham N, Ratanavaraporn J, Sereemaspun A, Yodmuang S. Hyaluronic acid crosslinked with alginate hydrogel: A versatile and biocompatible bioink platform for tissue engineering. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
22
|
Yan X, Yang B, Chen Y, Song Y, Ye J, Pan Y, Zhou B, Wang Y, Mao F, Dong Y, Liu D, Yu J. Anti-Friction MSCs Delivery System Improves the Therapy for Severe Osteoarthritis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2104758. [PMID: 34657320 DOI: 10.1002/adma.202104758] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/12/2021] [Indexed: 06/13/2023]
Abstract
Osteoarthritis (OA) is a musculoskeletal disorder disease affecting about 500 million people worldwide and mesenchymal sem cells (MSCs) therapy has been demonstrated as a potential strategy to treat OA. However, the shear forces during direct injection and the harsher shear condition of OA environments would lead to significant cell damage and inhibit the therapeutic efficacy. Herein, DNA supramolecular hydrogel has been applied as delivering material for MSCs to treat severe OA model, which perform extraordinary protection in MSCs against the shear force both in vitro and in vivo. It is demonstrated that the DNA supramolecular hydrogel can promote formation of quality cartilage, reduce osteophyte, and normalize subchondral bone under the high friction condition of OA, whose molecular mechanisms underlying therapeutic effects are also investigated. It can be anticipated that DNA supramolecular hydrogel would be a promising cell delivery system for multiple potential MSCs therapy.
Collapse
Affiliation(s)
- Xin Yan
- Department of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
- Institute of Sports Medicine, Peking University, Beijing, 100191, China
| | - Bo Yang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yourong Chen
- Department of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
- Institute of Sports Medicine, Peking University, Beijing, 100191, China
| | - Yifan Song
- Department of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
- Institute of Sports Medicine, Peking University, Beijing, 100191, China
| | - Jing Ye
- Department of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
- Institute of Sports Medicine, Peking University, Beijing, 100191, China
| | - Yufan Pan
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Bini Zhou
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yuqing Wang
- Tsinghua Laboratory of Brain and Intelligence, Tsinghua University, Beijing, 100084, China
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Yuanchen Dong
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Dongsheng Liu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Jiakuo Yu
- Department of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
- Institute of Sports Medicine, Peking University, Beijing, 100191, China
| |
Collapse
|
23
|
Li J, Ding Z, Zheng X, Lu G, Lu Q, Kaplan DL. Injectable silk nanofiber hydrogels as stem cell carriers to accelerate wound healing. J Mater Chem B 2021; 9:7771-7781. [PMID: 34586152 PMCID: PMC8486307 DOI: 10.1039/d1tb01320c] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stem cells have potential utility in wound therapy, however the benefits are often limited due to cell injury from shear stress during injection and poor retention at the wound site. Here, shear-thinning silk nanofiber hydrogels were used to load bone marrow derived mesenchymal stem cells (BMSCs) and inject into wound sites to optimize cell retention and accelerate wound healing. The BMSCs in the silk nanofiber hydrogels maintained stemness better than the cells cultured on plates, and the expression of wound healing-related genes was significantly higher in the hydrogels with higher silk concentrations (2 wt%). The silk nanofibers physically prevented migration of BMSCs from the deposition site in the wound bed. In addition to faster wound healing, these BMSC-loaded hydrogels mediated angiogenesis and inflammation and improved collagen deposition and hair follicle regeneration in vivo in rats. Considering that these silk nanofiber hydrogels were successfully used here as carriers for stem cells to accelerate wound healing, further study for skin regeneration may be warranted.
Collapse
Affiliation(s)
- Jiadai Li
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China.
- Nanjng University of Chinese Medicine, Nanjng 210000, P. R. China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China.
| | - Xin Zheng
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou 318000, P. R. China
| | - Guozhong Lu
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Wuxi 214041, P. R. China.
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| |
Collapse
|
24
|
Correa S, Grosskopf AK, Lopez Hernandez H, Chan D, Yu AC, Stapleton LM, Appel EA. Translational Applications of Hydrogels. Chem Rev 2021; 121:11385-11457. [PMID: 33938724 PMCID: PMC8461619 DOI: 10.1021/acs.chemrev.0c01177] [Citation(s) in RCA: 413] [Impact Index Per Article: 103.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Advances in hydrogel technology have unlocked unique and valuable capabilities that are being applied to a diverse set of translational applications. Hydrogels perform functions relevant to a range of biomedical purposes-they can deliver drugs or cells, regenerate hard and soft tissues, adhere to wet tissues, prevent bleeding, provide contrast during imaging, protect tissues or organs during radiotherapy, and improve the biocompatibility of medical implants. These capabilities make hydrogels useful for many distinct and pressing diseases and medical conditions and even for less conventional areas such as environmental engineering. In this review, we cover the major capabilities of hydrogels, with a focus on the novel benefits of injectable hydrogels, and how they relate to translational applications in medicine and the environment. We pay close attention to how the development of contemporary hydrogels requires extensive interdisciplinary collaboration to accomplish highly specific and complex biological tasks that range from cancer immunotherapy to tissue engineering to vaccination. We complement our discussion of preclinical and clinical development of hydrogels with mechanical design considerations needed for scaling injectable hydrogel technologies for clinical application. We anticipate that readers will gain a more complete picture of the expansive possibilities for hydrogels to make practical and impactful differences across numerous fields and biomedical applications.
Collapse
Affiliation(s)
- Santiago Correa
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Abigail K. Grosskopf
- Chemical
Engineering, Stanford University, Stanford, California 94305, United States
| | - Hector Lopez Hernandez
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Doreen Chan
- Chemistry, Stanford University, Stanford, California 94305, United States
| | - Anthony C. Yu
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | | | - Eric A. Appel
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
- Bioengineering, Stanford University, Stanford, California 94305, United States
- Pediatric
Endocrinology, Stanford University School
of Medicine, Stanford, California 94305, United States
- ChEM-H Institute, Stanford
University, Stanford, California 94305, United States
- Woods
Institute for the Environment, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
25
|
Woods WA, Chowdhury F, Tzerakis N, Adams CF, Chari DM. Developing a New Strategy for Delivery of Neural Transplant Populations Using Precursor Cell Sprays and Specialized Cell Media. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- William A. Woods
- Neural Tissue Engineering Group School of Medicine Keele University ST5 5BG UK
| | - Farhana Chowdhury
- Neural Tissue Engineering Group School of Medicine Keele University ST5 5BG UK
| | - Nikolaos Tzerakis
- Department of Neurosurgery University Hospital of North Midlands ST4 6QG UK
| | | | - Divya M. Chari
- Neural Tissue Engineering Group School of Medicine Keele University ST5 5BG UK
| |
Collapse
|
26
|
Novel Techniques to Improve Precise Cell Injection. Int J Mol Sci 2021; 22:ijms22126367. [PMID: 34198683 PMCID: PMC8232276 DOI: 10.3390/ijms22126367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022] Open
Abstract
We noted recently that the injection of cells with a needle through a cystoscope in the urethral sphincter muscle of pigs failed to deposit them nearby or at the intended target position in about 50% of all animals investigated (n > 100). Increasing the chance for precise cell injection by shotgun approaches employing several circumferential injections into the sphincter muscle bears the risk of tissue injury. In this study, we developed and tested a novel needle-free technique to precisely inject cells in the urethral sphincter tissue, or other tissues, using a water-jet system. This system was designed to fit in the working channels of endoscopes and cystoscopes, allowing a wide range of minimally invasive applications. We analyze key features, including the physical parameters of the injector design, pressure ranges applicable for tissue penetration and cell injections and biochemical parameters, such as different compositions of injection media. Our results present settings that enable the high viability of cells post-injection. Lastly, the method is suitable to inject cells in the superficial tissue layer and in deeper layers, required when the submucosa or the sphincter muscle of the urethra is targeted.
Collapse
|
27
|
Stem cell sprays for neurological injuries: a perspective. Emerg Top Life Sci 2021; 5:519-522. [PMID: 34096585 DOI: 10.1042/etls20210113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/11/2021] [Accepted: 05/19/2021] [Indexed: 11/17/2022]
Abstract
Injuries to the brain and spinal cord have major clinical consequences with high costs for healthcare systems. Neural cell transplantation therapies have significant translational potential to promote regeneration post-injury with clinical trials commencing for various pathologies. However, there are challenges associated with current clinical approaches used for systemic or direct delivery of transplant cells to neural tissue in regenerative applications. These include risks associated with surgical microinjection into neural tissue (e.g. haemorrhage, cell clumping) and high cell loss due to systemic clearance or with cell passage through fine gauge needles into densely packed neural tissue. This article presents lines of evidence supporting the concept that cell spray delivery technology can offer significant translational benefits for neural transplantation therapy, versus current cell delivery methods. Potential benefits include rapid/homogenous cell delivery, release over large surface areas, minimal invasiveness, compatibility with neurosurgical procedures in acute injury, no predictable clinical complications and the capacity to combine cell therapies with drug/biomolecule delivery. Accordingly, we consider that the development of cell spray delivery technology represents a key goal to develop advanced cell therapies for regenerative neurology.
Collapse
|
28
|
Danalache M, Knoll J, Linzenbold W, Enderle M, Abruzzese T, Stenzl A, Aicher WK. Injection of Porcine Adipose Tissue-Derived Stromal Cells by a Novel Waterjet Technology. Int J Mol Sci 2021; 22:ijms22083958. [PMID: 33921246 PMCID: PMC8070533 DOI: 10.3390/ijms22083958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022] Open
Abstract
Previously, we developed a novel, needle-free waterjet (WJ) technology capable of injecting viable cells by visual guided cystoscopy in the urethral sphincter. In the present study, we aimed to investigate the effect of WJ technology on cell viability, surface markers, differentiation and attachment capabilities, and biomechanical features. Porcine adipose tissue-derived stromal cells (pADSCs) were isolated, expanded, and injected by WJ technology. Cell attachment assays were employed to investigate cell-matrix interactions. Cell surface molecules were analyzed by flow cytometry. Cells injected by Williams Needle (WN), normal cannula, or not injected cells served as controls. Biomechanical properties were assessed by atomic force microscopy (AFM). pADSCs injected by the WJ were viable (85.9%), proliferated well, and maintained their in vitro adipogenic and osteogenic differentiation capacities. The attachment of pADSCs was not affected by WJ injection and no major changes were noted for cell surface markers. AFM measurements yielded a significant reduction of cellular stiffness after WJ injections (p < 0.001). WJ cell delivery satisfies several key considerations required in a clinical context, including the fast, simple, and reproducible delivery of viable cells. However, the optimization of the WJ device may be necessary to further reduce the effects on the biomechanical properties of cells.
Collapse
Affiliation(s)
- Marina Danalache
- Department of Orthopaedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany;
| | - Jasmin Knoll
- Department of Urology, University Hospital Tübingen, Waldhörnlestrasse 22, 72072 Tübingen, Germany; (J.K.); (T.A.); (A.S.)
| | - Walter Linzenbold
- ERBE Elektromedizin GmbH Tübingen, 72072 Tübingen, Germany; (W.L.); (M.E.)
| | - Markus Enderle
- ERBE Elektromedizin GmbH Tübingen, 72072 Tübingen, Germany; (W.L.); (M.E.)
| | - Tanja Abruzzese
- Department of Urology, University Hospital Tübingen, Waldhörnlestrasse 22, 72072 Tübingen, Germany; (J.K.); (T.A.); (A.S.)
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, Waldhörnlestrasse 22, 72072 Tübingen, Germany; (J.K.); (T.A.); (A.S.)
| | - Wilhelm K. Aicher
- Department of Urology, University Hospital Tübingen, Waldhörnlestrasse 22, 72072 Tübingen, Germany; (J.K.); (T.A.); (A.S.)
- Correspondence: ; Tel.: +49-7071-298-7021; Fax: +49-7071-292-5072
| |
Collapse
|
29
|
Eigel D, Werner C, Newland B. Cryogel biomaterials for neuroscience applications. Neurochem Int 2021; 147:105012. [PMID: 33731275 DOI: 10.1016/j.neuint.2021.105012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022]
Abstract
Biomaterials in the form of 3D polymeric scaffolds have been used to create structurally and functionally biomimetic constructs of nervous system tissue. Such constructs can be used to model defects and disease or can be used to supplement neuronal tissue regeneration and repair. One such group of biomaterial scaffolds are hydrogels, which have been widely investigated for cell/tissue culture and as cell or molecule delivery systems in the field of neurosciences. However, a subset of hydrogels called cryogels, have shown to possess several distinct structural advantages over conventional hydrogel networks. Their macroporous structure, created via the time and resource efficient fabrication process (cryogelation) not only allows mass fluid transport throughout the structure, but also creates a high surface area to volume ratio for cell growth or drug loading. In addition, the macroporous structure of cryogels is ideal for applications in the central nervous system as they are very soft and spongey, yet also robust, which makes them a user-friendly and reproducible tool to address neuroscience challenges. In this review, we aim to provide the neuroscience community, who may not be familiar with the fundamental concepts of cryogels, an accessible summary of the basic information that pertain to their use in the brain and nervous tissue. We hope that this review shall initiate creative ways that cryogels could be further adapted and employed to tackle unsolved neuroscience challenges.
Collapse
Affiliation(s)
- Dimitri Eigel
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany; Technische Universität Dresden, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ben Newland
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Str. 6, 01069, Dresden, Germany; School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF10 3NB, Cardiff, Wales, UK.
| |
Collapse
|
30
|
Rosell-Valle C, Antúnez C, Campos F, Gallot N, García-Arranz M, García-Olmo D, Gutierrez R, Hernán R, Herrera C, Jiménez R, Leyva-Fernández L, Maldonado-Sanchez R, Muñoz-Fernández R, Nogueras S, Ortiz L, Piudo I, Ranchal I, Rodríguez-Acosta A, Segovia C, Fernández-Muñoz B. Evaluation of the effectiveness of a new cryopreservation system based on a two-compartment vial for the cryopreservation of cell therapy products. Cytotherapy 2021; 23:740-753. [PMID: 33714705 DOI: 10.1016/j.jcyt.2020.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND AIMS Successful cell cryopreservation and banking remain a major challenge for the manufacture of cell therapy products, particularly in relation to providing a hermetic, sterile cryovial that ensures optimal viability and stability post-thaw while minimizing exposure to toxic cryoprotective agents, typically dimethyl sulfoxide (Me2SO). METHODS In the present study, the authors evaluated the effectiveness and functionality of Limbo technology (Cellulis S.L., Santoña, Spain). This system provides a hermetic vial with two compartments (one for adding cells with the cryoprotective agent solution and the other for the diluent solution) and an automated defrosting device. Limbo technology (Cellulis S.L.) allows reduction of the final amount of Me2SO, sidestepping washing and dilution steps and favoring standardization. The study was performed in several Good Manufacturing Practice laboratories manufacturing diverse cell therapy products (human mesenchymal stromal cells, hematopoietic progenitor cells, leukapheresis products, fibroblasts and induced pluripotent stem cells). Laboratories compared Limbo technology (Cellulis S.L.) with their standard cryopreservation procedure, analyzing cell recovery, viability, phenotype and functionality. RESULTS Limbo technology (Cellulis S.L.) maintained the viability and functionality of most of the cell products and preserved sterility while reducing the final concentration of Me2SO. CONCLUSIONS Results showed that use of Limbo technology (Cellulis S.L.) offers an overall safe alternative for cell banking and direct infusion of cryopreserved cell products into patients.
Collapse
Affiliation(s)
- Cristina Rosell-Valle
- Unidad de Producción y Reprogramación Celular, Red Andaluza Para el Diseño y Traslación de Terapias Avanzadas, Sevilla, Spain.
| | - Cristina Antúnez
- Unidad de Expansión y Terapia Celular. Centro de Transfusión, Tejidos y Células, Málaga, Spain
| | - Fernando Campos
- Unidad de Producción y Reprogramación Celular, Red Andaluza Para el Diseño y Traslación de Terapias Avanzadas, Sevilla, Spain
| | | | | | | | - Rosario Gutierrez
- Unidad de Terapia Celular, Hospital Universitario Reina Sofía, Córdoba, Spain
| | | | - Concha Herrera
- Unidad de Terapia Celular, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain
| | - Rosario Jiménez
- Unidad de Terapia Celular, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Laura Leyva-Fernández
- Unidad de Producción Celular, Hospital Regional Universitario de Málaga, Málaga, Spain
| | | | | | - Sonia Nogueras
- Unidad de Terapia Celular, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Lourdes Ortiz
- Unidad de Terapia Celular, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Inmaculada Piudo
- Unidad de Producción y Reprogramación Celular, Red Andaluza Para el Diseño y Traslación de Terapias Avanzadas, Sevilla, Spain
| | - Isidora Ranchal
- Unidad de Producción y Reprogramación Celular, Red Andaluza Para el Diseño y Traslación de Terapias Avanzadas, Sevilla, Spain
| | | | - Cristina Segovia
- Unidad de Expansión y Terapia Celular. Centro de Transfusión, Tejidos y Células, Málaga, Spain
| | - Beatriz Fernández-Muñoz
- Unidad de Producción y Reprogramación Celular, Red Andaluza Para el Diseño y Traslación de Terapias Avanzadas, Sevilla, Spain.
| |
Collapse
|
31
|
Dahn ML, Marcato P. In Vivo Genome-Wide Pooled RNAi Screens in Cancer Cells to Identify Determinants of Chemotherapy/Drug Response. Methods Mol Biol 2021; 2381:189-200. [PMID: 34590277 DOI: 10.1007/978-1-0716-1740-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Large-scale RNAi screens (i.e., genome-wide arrays and pools) can reveal the essential biological functions of previously uncharacterized genes. Due to the nature of the selection process involved in screens, RNAi screens are also very useful for identifying genes involved in drug responses. The information gained from these screens could be used to predict a cancer patient's response to a specific drug (i.e., precision medicine) or identify anti-cancer drug resistance genes, which could be targeted to improve treatment outcomes. In this capacity, screens have been most often performed in vitro. However, there is limitation to performing these screens in vitro: genes which are required in only an in vivo setting (e.g., rely on the tumor microenvironment for function) will not be identified. As such, it can be desirable to perform RNAi screens in vivo. Here we outline the additional technical details that should be considered for performing genome-wide RNAi drug screens of cancer cells under in vivo conditions (i.e., tumor xenografts).
Collapse
Affiliation(s)
- Margaret L Dahn
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS, Canada. .,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
32
|
Lee JSJ, Kim SJ, Choi JS, Eom MR, Shin H, Kwon SK. Adipose-derived mesenchymal stem cell spheroid sheet accelerates regeneration of ulcerated oral mucosa by enhancing inherent therapeutic properties. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2020.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
Syromiatnikova V, Idrisova K, Masgutova G, Gomzikova M, Kabwe E, Bek J, Andreeva D, Masgutov R, Mullakhmetova A, James V, Rizvanov A. Analyzing the Effectiveness of Adipose Tissue Stem Cell and Microvesicle Therapy in Premature Skin Aging Caused by Chronic Exposure to Ultraviolet Radiation. BIONANOSCIENCE 2020. [DOI: 10.1007/s12668-020-00793-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
34
|
Flegeau K, Toquet C, Rethore G, d'Arros C, Messager L, Halgand B, Dupont D, Autrusseau F, Lesoeur J, Veziers J, Bordat P, Bresin A, Guicheux J, Delplace V, Gautier H, Weiss P. In Situ Forming, Silanized Hyaluronic Acid Hydrogels with Fine Control Over Mechanical Properties and In Vivo Degradation for Tissue Engineering Applications. Adv Healthc Mater 2020; 9:e2000981. [PMID: 32864869 DOI: 10.1002/adhm.202000981] [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: 06/09/2020] [Revised: 08/09/2020] [Indexed: 12/19/2022]
Abstract
In situ forming hydrogels that can be injected into tissues in a minimally-invasive fashion are appealing as delivery vehicles for tissue engineering applications. Ideally, these hydrogels should have mechanical properties matching those of the host tissue, and a rate of degradation adapted for neo-tissue formation. Here, the development of in situ forming hyaluronic acid hydrogels based on the pH-triggered condensation of silicon alkoxide precursors into siloxanes is reported. Upon solubilization and pH adjustment, the low-viscosity precursor solutions are easily injectable through fine-gauge needles prior to in situ gelation. Tunable mechanical properties (stiffness from 1 to 40 kPa) and associated tunable degradability (from 4 days to more than 3 weeks in vivo) are obtained by varying the degree of silanization (from 4.3% to 57.7%) and molecular weight (120 and 267 kDa) of the hyaluronic acid component. Following cell encapsulation, high cell viability (> 80%) is obtained for at least 7 days. Finally, the in vivo biocompatibility of silanized hyaluronic acid gels is verified in a subcutaneous mouse model and a relationship between the inflammatory response and the crosslink density is observed. Silanized hyaluronic acid hydrogels constitute a tunable hydrogel platform for material-assisted cell therapies and tissue engineering applications.
Collapse
Affiliation(s)
- Killian Flegeau
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Claire Toquet
- Department of Pathology University Hospital of Nantes Nantes F‐44042 France
| | - Gildas Rethore
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Cyril d'Arros
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Léa Messager
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Boris Halgand
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Davy Dupont
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Florent Autrusseau
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Julie Lesoeur
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- SC3M SFR Santé F. Bonamy FED 4203 UMS Inserm 016 CNRS 3556 Nantes F‐44042 France
| | - Joëlle Veziers
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
- SC3M SFR Santé F. Bonamy FED 4203 UMS Inserm 016 CNRS 3556 Nantes F‐44042 France
| | | | | | - Jérôme Guicheux
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Vianney Delplace
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Hélène Gautier
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- Université de Nantes Faculté de Pharmacie Laboratoire de Pharmacie Galénique Nantes F‐44042 France
| | - Pierre Weiss
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| |
Collapse
|
35
|
Shen Y. Stem cell therapies for retinal diseases: from bench to bedside. J Mol Med (Berl) 2020; 98:1347-1368. [PMID: 32794020 DOI: 10.1007/s00109-020-01960-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/02/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022]
Abstract
As the human retina has no regenerative ability, stem cell interventions represent potential therapies for various blinding retinal diseases. This type of therapy has been extensively studied in the human eyes through decades of preclinical studies. The safety profiles shown in clinical trials thus far have indicated that these strategies should be further explored. There are still challenges with regard to cell source, cell delivery, immuno-related adverse events and long-term maintenance of the therapeutic effects. Retinal stem cell therapy is likely to be most successful with a combination of multiple technologies, such as gene therapy. The purpose of this review is to present a synthetical and systematic coverage of stem cell therapies that target retinal diseases from bench to bedside, intending to appeal to both junior specialists and the broader community of clinical investigators alike. This review will only focus on therapies that have already been studied in clinical trials. This review summarizes key concepts, highlights the main studies in human patients and discusses the current challenges and potential methods to reduce safety concerns while enhancing the therapeutic effects.
Collapse
Affiliation(s)
- Yuening Shen
- Institute of Ophthalmology, University College London , 11-43 Bath St, London, EC1V 9EL, UK. .,Department of Medical Retina, Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London, EC1V 2PD, UK.
| |
Collapse
|
36
|
Zheng X, Ding Z, Cheng W, Lu Q, Kong X, Zhou X, Lu G, Kaplan DL. Microskin-Inspired Injectable MSC-Laden Hydrogels for Scarless Wound Healing with Hair Follicles. Adv Healthc Mater 2020; 9:e2000041. [PMID: 32338466 PMCID: PMC7473495 DOI: 10.1002/adhm.202000041] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/26/2020] [Indexed: 12/20/2022]
Abstract
Scarless skin regeneration with functional tissue remains a challenge for full-thickness wounds. Here, mesenchymal stem cell (MSC)-laden hydrogels are developed for scarless wound healing with hair follicles. Microgels composed of aligned silk nanofibers are used to load MSCs to modulate the paracrine. MSC-laden microgels are dispersed into injectable silk nanofiber hydrogels, forming composites biomaterials containing the cells. The injectable hydrogels protect and stabilize the MSCs in the wounds. The synergistic action of silk-based composite hydrogels and MSCs stimulated angiogenesis and M1-M2 phenotype switching of macrophages, provides a suitable niche for functional recovery of wounds. Compared to skin defects treated with MSC-free hydrogels, the defects treated with the MSC-laden composite hydrogels heal faster and form scarless tissues with hair follicles. Wound healing can be further improved by adjusting the ratio of silk nanofibers and particles and the loaded MSCs, suggesting tunability of the system. To the best of current knowledge, this is the first time scarless skin regeneration with hair follicles based on silk material systems is reported. The improved wound healing capacity of the systems suggests future in vivo studies to compare to other biomaterial systems related to clinical goals in skin regeneration in the absence of scarring.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, P. R. China
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou, 318000, P. R. China
| | - Zhaozhao Ding
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, 214041, P. R. China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi, 214041, P. R. China
| | - Weinan Cheng
- Department of Orthopedics, The First Affiliated Hospital of Xiamen University, Xiamen, 361000, P. R. China
| | - Qiang Lu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, P. R. China
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, 214041, P. R. China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi, 214041, P. R. China
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, 215123, P. R. China
| | - Xiangdong Kong
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, P. R. China
| | - Guozhong Lu
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, 214041, P. R. China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Wuxi, 214041, P. R. China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
37
|
Zhang S, Huang D, Lin H, Xiao Y, Zhang X. Cellulose Nanocrystal Reinforced Collagen-Based Nanocomposite Hydrogel with Self-Healing and Stress-Relaxation Properties for Cell Delivery. Biomacromolecules 2020; 21:2400-2408. [DOI: 10.1021/acs.biomac.0c00345] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Shuang Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Danyang Huang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Hai Lin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yun Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
38
|
Muralidhara BK, Wong M. Critical considerations in the formulation development of parenteral biologic drugs. Drug Discov Today 2020; 25:574-581. [DOI: 10.1016/j.drudis.2019.12.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/02/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022]
|
39
|
Jäger L, Linzenbold W, Fech A, Enderle M, Abruzzese T, Stenzl A, Aicher WK. A novel waterjet technology for transurethral cystoscopic injection of viable cells in the urethral sphincter complex. Neurourol Urodyn 2019; 39:594-602. [PMID: 31873953 DOI: 10.1002/nau.24261] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/14/2019] [Indexed: 12/22/2022]
Abstract
AIMS In a recent preclinical study, we noticed that injection of cells in the urethral sphincter by needle through a cystoscope under visual control frequently yielded in misplacement or loss of cells. We, therefore, investigated if a needle-free waterjet device delivers viable cells under defined settings, including injection volume and pressure, fluid velocity and transportation media, precisely through the urothelium and connective tissue close to the sphincter muscle without full penetration of the sphincter apparatus. METHODS Mesenchymal stromal cells (MSCs) were prepared for needle-free waterjet injections. Upon injections into liquids cell viability and yield were investigated by trypan blue dye exclusion. Upon injection into cadaveric urethral tissue samples, cells were isolated from the urethrae and expanded to prove that this novel method delivered viable cells into the tissue. MSC injections by William's needle served as controls. RESULTS Waterjet injections of MSCs into isotonic cell culture medium resulted in equal or better yields of viable cells when compared with needle injections. Upon injection in urethral tissue samples, the waterjet technology facilitated fast and precise injections of viable cells through urothelial, mucosal and submucosal layers to reach the sphincter muscle. By controlling the injection pressure, loss of cells due to insufficient thrust or unintended full penetration was avoided. CONCLUSIONS Needle-free waterjet injections deliver cells in the urethra faster and more precisely when compared with needle injections without compromising their viability. This is the first proof-of-concept study providing evidence that a waterjet transports viable cells precisely into the targeted tissue.
Collapse
Affiliation(s)
- Luise Jäger
- ERBE Elektromedizin GmbH, Tuebingen, Germany
| | | | | | | | - Tanja Abruzzese
- Department of Urology, University of Tuebingen Hospital, University of Tuebingen, Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tuebingen Hospital, University of Tuebingen, Tuebingen, Germany
| | - Wilhelm K Aicher
- Department of Urology, University of Tuebingen Hospital, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
40
|
Barakat AH, Elwell VA, Lam KS. Stem cell therapy in discogenic back pain. JOURNAL OF SPINE SURGERY (HONG KONG) 2019; 5:561-583. [PMID: 32043007 PMCID: PMC6989932 DOI: 10.21037/jss.2019.09.22] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2023]
Abstract
Chronic low back pain has both substantial social and economic impacts on patients and healthcare budgets. Adding to the magnitude of the problem is the difficulty in identifying the exact causes of disc degeneration with modern day diagnostic and imaging techniques. With that said, current non-operative and surgical treatment modalities for discogenic low back pain fails to meet the expectations in many patients and hence the challenge. The objective for newly emerging stem cell regenerative therapy is to treat degenerative disc disease (DDD) by restoring the disc's cellularity and modulating the inflammatory response. Appropriate patient selection is crucial for the success of stem cell therapy. Regenerative modalities for discogenic pain currently focus on the use of either primary cells harvested from the intervertebral discs or stem cells from other sources whether autogenic or allogenic. The microenvironment in which stem cells are being cultured has been recognized to play a crucial role in directing or maintaining the production of the desired phenotypes and may enhance their regenerative potential. This has led to a more specific focus on innovating more effective culturing techniques, delivery vehicles and scaffolds for stem cell application. Although stem cell therapy might offer an attractive alternative treatment option, more clinical studies are still needed to establish on the safety and feasibility of such therapy. In this literature review, we aim to present the most recent in vivo and in vitro studies related to the use of stem cell therapy in the treatment of discogenic low back pain.
Collapse
Affiliation(s)
- Ahmed H. Barakat
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | - Vivian A. Elwell
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | | |
Collapse
|
41
|
Fath-Bayati L, Vasei M, Sharif-Paghaleh E. Optical fluorescence imaging with shortwave infrared light emitter nanomaterials for in vivo cell tracking in regenerative medicine. J Cell Mol Med 2019; 23:7905-7918. [PMID: 31559692 PMCID: PMC6850965 DOI: 10.1111/jcmm.14670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/13/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
In vivo tracking and monitoring of adoptive cell transfer has a distinct importance in cell‐based therapy. There are many imaging modalities for in vivo monitoring of biodistribution, viability and effectiveness of transferred cells. Some of these procedures are not applicable in the human body because of low sensitivity and high possibility of tissue damages. Shortwave infrared region (SWIR) imaging is a relatively new technique by which deep biological tissues can be potentially visualized with high resolution at cellular level. Indeed, scanning of the electromagnetic spectrum (beyond 1000 nm) of SWIR has a great potential to increase sensitivity and resolution of in vivo imaging for various human tissues. In this review, molecular imaging modalities used for monitoring of biodistribution and fate of administered cells with focusing on the application of non‐invasive optical imaging at shortwave infrared region are discussed in detail.
Collapse
Affiliation(s)
- Leyla Fath-Bayati
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Department of Tissue Engineering, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Vasei
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Cell-based Therapies Research Institute, Digestive Disease Research Institute (DDRI), Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Ehsan Sharif-Paghaleh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Imaging Chemistry and Biology, Faculty of Life Sciences and Medicine, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
42
|
Freitas GP, Lopes HB, Souza ATP, Oliveira PGFP, Almeida ALG, Souza LEB, Coelho PG, Beloti MM, Rosa AL. Cell Therapy: Effect of Locally Injected Mesenchymal Stromal Cells Derived from Bone Marrow or Adipose Tissue on Bone Regeneration of Rat Calvarial Defects. Sci Rep 2019; 9:13476. [PMID: 31530883 PMCID: PMC6748998 DOI: 10.1038/s41598-019-50067-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/05/2019] [Indexed: 02/07/2023] Open
Abstract
Treatment of large bone defects is a challenging clinical situation that may be benefited from cell therapies based on regenerative medicine. This study was conducted to evaluate the effect of local injection of bone marrow-derived mesenchymal stromal cells (BM-MSCs) or adipose tissue-derived MSCs (AT-MSCs) on the regeneration of rat calvarial defects. BM-MSCs and AT-MSCs were characterized based on their expression of specific surface markers; cell viability was evaluated after injection with a 21-G needle. Defects measuring 5 mm that were created in rat calvaria were injected with BM-MSCs, AT-MSCs, or vehicle-phosphate-buffered saline (Control) 2 weeks post-defect creation. Cells were tracked by bioluminescence, and 4 weeks post-injection, the newly formed bone was evaluated by µCT, histology, nanoindentation, and gene expression of bone markers. BM-MSCs and AT-MSCs exhibited the characteristics of MSCs and maintained their viability after passing through the 21-G needle. Injection of both BM-MSCs and AT-MSCs resulted in increased bone formation compared to that in Control and with similar mechanical properties as those of native bone. The expression of genes associated with bone formation was higher in the newly formed bone induced by BM-MSCs, whereas the expression of genes involved in bone resorption was higher in the AT-MSC group. Cell therapy based on local injection of BM-MSCs or AT-MSCs is effective in delivering cells that induced a significant improvement in bone healing. Despite differences observed in molecular cues between BM-MSCs and AT-MSCs, both cells had the ability to induce bone tissue formation at comparable amounts and properties. These results may drive new cell therapy approaches toward complete bone regeneration.
Collapse
Affiliation(s)
- Gileade P Freitas
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Helena B Lopes
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Alann T P Souza
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Paula G F P Oliveira
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Adriana L G Almeida
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Lucas E B Souza
- Hemotherapy Center of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Paulo G Coelho
- Department of Biomaterials, New York University College of Dentistry, New York, NY, USA.,Hanjorg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, NY, USA
| | - Marcio M Beloti
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Adalberto L Rosa
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
43
|
Byun H, Bin Lee Y, Kim EM, Shin H. Fabrication of size-controllable human mesenchymal stromal cell spheroids from micro-scaled cell sheets. Biofabrication 2019; 11:035025. [PMID: 31096204 DOI: 10.1088/1758-5090/ab21f6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, stromal cell spheroids have been actively studied for use in tissue regeneration. In this study, we report a method for the fabrication of size-controllable stromal cell spheroids in different sizes from micro-scaled cell sheets (μCS) using thermosensitive hydrogels and investigated their effects on stromal cell function. Mesenchymal stromal cells isolated from different tissues such as human turbinate tissue, bone marrow, and adipose tissue were adhered selectively to each micro-pattern (squares with widths of 100 and 400 μm) on the surface of the hydrogel and formed μCS. The diameters of the spheroids were modulated by the size of the patterns (45 ± 5 and 129 ± 4 μm in diameter for the 100 and 400 μm micro-patterns, respectively) and the seeding density (129 ± 4, 149 ± 6, and 163 ± 6 μm for 5.0, 10.0, and 15.0 × 104 cells cm-2, respectively, on 400 μm micro-pattern). In addition, the spheroids were successfully fabricated regardless of stromal cell origin, and the diameter of the spheroids was also affected by cell spreading area on a cell culture dish. Stemness markers were highly expressed in the spheroids regardless of the spheroid size. Furthermore, an increase in E-cadherin and decrease in N-cadherin gene expression showed the stable formation of spheroids of different sizes. Gene expression levels of hypoxia inducible factors and secretion of vascular endothelial growth factor were increased (13.2 ± 1.4, 325 ± 83.4 and 534.3 ± 121.5 pg ng-1 DNA in a monolayer, and 100 and 400 μm micro-patterned spheroids, respectively) proportional to the diameters of the spheroids. The size of spheroids were maintained even after injection, cryopreservation and 7 d of suspension culture with high viability (∼90%). In conclusion, this novel technique to fabricate spheroids with controlled size could be widely applied in various applications that require a controlled size in regenerative medicine.
Collapse
Affiliation(s)
- Hayeon Byun
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea. BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | | | | | | |
Collapse
|
44
|
Masgutov R, Masgutova G, Mullakhmetova A, Zhuravleva M, Shulman A, Rogozhin A, Syromiatnikova V, Andreeva D, Zeinalova A, Idrisova K, Allegrucci C, Kiyasov A, Rizvanov A. Adipose-Derived Mesenchymal Stem Cells Applied in Fibrin Glue Stimulate Peripheral Nerve Regeneration. Front Med (Lausanne) 2019; 6:68. [PMID: 31024916 PMCID: PMC6465797 DOI: 10.3389/fmed.2019.00068] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/19/2019] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) hold a great promise for cell therapy. To date, they represent one of the best choices for the treatment of post-traumatic injuries of the peripheral nervous system. Although autologous can be easily transplanted in the injured area, clinical advances in this filed have been impaired by lack of preservation of graft cells into the injury area after transplantation. Indeed, cell viability is not retained after injection into the blood stream, and cells injected directly into the area of injury either are washed off or inhibit regeneration through scar formation and neuroma development. This study proposes a new way of MSCs delivery to the area of traumatic injury by using fibrin glue, which not only fixes cells at the site of application but also provides extracellular matrix support. Using a sciatic nerve injury model, MSC derived from adipose tissue embedded in fibrin glue were able to enter the nerve and migrate mainly retrogradely after transplantation. They also demonstrated a neuroprotective effect on DRG L5 sensory neurons and stimulated axon growth and myelination. Post-traumatic changes of the sensory neuron phenotype were also improved. Importantly, MSCs stimulated nerve angiogenesis and motor function recovery. Therefore, our data suggest that MSC therapy using fibrin glue is a safe and efficient method of cell transplantation in cases of sciatic nerve injury, and that this method of delivery of regeneration stimulants could be beneficial for the successful treatment of other central and peripheral nervous system conditions.
Collapse
Affiliation(s)
- Ruslan Masgutov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Department of Orthopaedics, Republic Clinical Hospital, Kazan, Russia
| | - Galina Masgutova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Adelya Mullakhmetova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Margarita Zhuravleva
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Anna Shulman
- Scientific Department, Republic Clinical Hospital, Kazan, Russia
| | - Alexander Rogozhin
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Department of Neurology, Kazan State Medical Academy, Branch of Russian Medical Academy of Postgraduate Education, Kazan, Russia
| | - Valeriya Syromiatnikova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Dina Andreeva
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Alina Zeinalova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kamilla Idrisova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Cinzia Allegrucci
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Andrey Kiyasov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert Rizvanov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
45
|
Memic A, Colombani T, Eggermont LJ, Rezaeeyazdi M, Steingold J, Rogers ZJ, Navare KJ, Mohammed HS, Bencherif SA. Latest Advances in Cryogel Technology for Biomedical Applications. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201800114] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Adnan Memic
- Center of NanotechnologyKing Abdulaziz University Jeddah 21589 Saudi Arabia
- Center for Biomedical EngineeringDepartment of MedicineBrigham and Women's HospitalHarvard Medical School Cambridge MA 02139 USA
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
| | - Thibault Colombani
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
| | - Loek J. Eggermont
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
- Department of Tumor ImmunologyOncode Institute, Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen 6500 The Netherlands
| | | | - Joseph Steingold
- Department of Pharmaceutical SciencesNortheastern University Boston MA 02115 USA
| | - Zach J. Rogers
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
| | | | | | - Sidi A. Bencherif
- Department of Chemical EngineeringNortheastern University Boston MA 02115 USA
- Department of BioengineeringNortheastern University Boston MA 02115 USA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard University Cambridge MA 02138 USA
- Sorbonne UniversityUTC CNRS UMR 7338Biomechanics and Bioengineering (BMBI)University of Technology of Compiègne Compiègne 60159 France
| |
Collapse
|
46
|
Ashammakhi N, Ahadian S, Darabi MA, El Tahchi M, Lee J, Suthiwanich K, Sheikhi A, Dokmeci MR, Oklu R, Khademhosseini A. Minimally Invasive and Regenerative Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1804041. [PMID: 30565732 PMCID: PMC6709364 DOI: 10.1002/adma.201804041] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/20/2018] [Indexed: 05/03/2023]
Abstract
Advances in biomaterial synthesis and fabrication, stem cell biology, bioimaging, microsurgery procedures, and microscale technologies have made minimally invasive therapeutics a viable tool in regenerative medicine. Therapeutics, herein defined as cells, biomaterials, biomolecules, and their combinations, can be delivered in a minimally invasive way to regenerate different tissues in the body, such as bone, cartilage, pancreas, cardiac, skeletal muscle, liver, skin, and neural tissues. Sophisticated methods of tracking, sensing, and stimulation of therapeutics in vivo using nano-biomaterials and soft bioelectronic devices provide great opportunities to further develop minimally invasive and regenerative therapeutics (MIRET). In general, minimally invasive delivery methods offer high yield with low risk of complications and reduced costs compared to conventional delivery methods. Here, minimally invasive approaches for delivering regenerative therapeutics into the body are reviewed. The use of MIRET to treat different tissues and organs is described. Although some clinical trials have been performed using MIRET, it is hoped that such therapeutics find wider applications to treat patients. Finally, some future perspective and challenges for this emerging field are highlighted.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mario El Tahchi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- LBMI, Department of Physics, Lebanese University - Faculty of Sciences 2, PO Box 90656, Jdeidet, Lebanon
| | - Junmin Lee
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Kasinan Suthiwanich
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Amir Sheikhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mehmet R. Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Rahmi Oklu
- Division of Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Radiological Sciences, University of California - Los Angeles, Los Angeles, California, USA
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, California, USA
- Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
47
|
Adams CF, Delaney AM, Carwardine DR, Tickle J, Granger N, Chari DM. Nanoparticle-Based Imaging of Clinical Transplant Populations Encapsulated in Protective Polymer Matrices. Macromol Biosci 2018; 19:e1800389. [PMID: 30511815 DOI: 10.1002/mabi.201800389] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/18/2018] [Indexed: 01/26/2023]
Abstract
A recent clinical trial proves that autologous olfactory mucosal cell (OMC) transplantation improves locomotion in dogs with naturally occurring spinal injuries comparable to human lesions. However, not all dogs respond to the treatment, likely due to the transplantation procedures involving injections of cell suspensions that are associated with cell death, uneven cell distribution, and cell washout. Encapsulating cells in protective hydrogel matrices offers a tissue engineering solution to safely achieve 3D growth of viable transplant cells for implantation into injury sites, to improve regenerative outcomes. It is shown for the first time that canine OMCs (cOMCs) can be propagated with high viability in 3D collagen matrices. Further, a method to incorporate cOMCs pre-labeled with clinical-grade iron oxide nanoparticles into the constructs is described. Intraconstruct labeled cells are visualized using magnetic resonance imaging, offering substantial promise for in vivo tracking of cOMCs delivered in protective matrices.
Collapse
Affiliation(s)
- Christopher F Adams
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Alexander M Delaney
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | | | - Jacqueline Tickle
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Nicolas Granger
- The Royal Veterinary College, Hawkshead Lane, Hatfield, Hertfordshire, AL9 7TA, UK
| | - Divya M Chari
- Cellular and Neural Engineering Group, Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| |
Collapse
|
48
|
Kim SJ, Park J, Byun H, Park YW, Major LG, Lee DY, Choi YS, Shin H. Hydrogels with an embossed surface: An all-in-one platform for mass production and culture of human adipose-derived stem cell spheroids. Biomaterials 2018; 188:198-212. [PMID: 30368228 DOI: 10.1016/j.biomaterials.2018.10.025] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 10/19/2018] [Indexed: 01/01/2023]
Abstract
Stem cell spheroids have been studied extensively in organoid culture and therapeutic transplantation. Herein, hydrogels with an embossed surface (HES) were developed as an all-in-one platform that can enable the rapid formation and culture of a large quantity of size-controllable stem cell spheroids. The embossed structure on the hydrogel was adjustable according to the grit designation of the sandpaper. Human adipose-derived stem cells (hADSCs) were rapidly assembled into spheroids on the hydrogel, with their size distribution precisely controlled from 95 ± 6 μm to 181 ± 15 μm depending on surface roughness. The hADSC spheroids prepared from the HES demonstrated expression of stemness markers and differentiation capacity. In addition, HES-based spheroids showed significantly greater VEGF secretion than spheroids grown on a commercially available low-attachment culture plate. Exploiting those advantages, the HES-based spheroids were used for 3D bioprinting, and the spheroids within the 3D-printed construct showed improved retention and VEGF secretion compared to the same 3D structure containing single cell suspension. Collectively, HES would offer a useful platform for mass fabrication and culture of stem cell spheroids with controlled sizes for a variety of biomedical applications.
Collapse
Affiliation(s)
- Se-Jeong Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Jaesung Park
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Hayeon Byun
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Young-Woo Park
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Luke G Major
- School of Human Science, University of Western Australia, Perth, WA 6009, Australia
| | - Dong Yun Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; Institute of Nano Science & Technology (INST), Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Yu Suk Choi
- School of Human Science, University of Western Australia, Perth, WA 6009, Australia
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; Institute of Nano Science & Technology (INST), Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea.
| |
Collapse
|
49
|
Leoni G, Lyness A, Ginty P, Schutte R, Pillai G, Sharma G, Kemp P, Mount N, Sharpe M. Preclinical development of an automated injection device for intradermal delivery of a cell-based therapy. Drug Deliv Transl Res 2018; 7:695-708. [PMID: 28812281 PMCID: PMC5574955 DOI: 10.1007/s13346-017-0418-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Current methods for intradermal delivery of therapeutic products in clinical use include manual injection via the Mantoux technique and the use of injection devices, primarily developed for the delivery of vaccines and small molecules. A novel automated injection device is presented specifically designed for accurate delivery of multiple doses of product through a number of adjustable injection parameters, including injection depth, dose volume and needle insertion speed. The device was originally conceived for the delivery of a cell-based therapy to patients with skin wounds caused by epidermolysis bullosa. A series of preclinical studies was conducted (i) to evaluate the performance of the pre-production model (PreCTCDV01) and optimise the final design, (ii) to confirm that a cell therapy product can be effectively delivered through the injection system and (iii) to test whether the device can be safely and effectively operated by potential end-users. Results from these studies confirmed that the device is able to consistently deliver repeated doses of a liquid to the intradermal layer in an ex vivo skin model. In addition, the device can support delivery of a cell therapy product through a customised microbore tubing without compromising cell viability. Finally, the device was shown to be safe and easy to use as evidenced by usability testing. The clinical device has since been granted European market access and plans for clinical use are currently underway. The device is expected to find use in the emerging area of cell therapies and a broad spectrum of traditional parenteral drug delivery applications.
Collapse
Affiliation(s)
- Giulia Leoni
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guys Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Alex Lyness
- Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
| | - Patrick Ginty
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guys Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Rindi Schutte
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guys Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Gopalan Pillai
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guys Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Gayatri Sharma
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guys Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Paul Kemp
- Intercytex Ltd, 5 Vale Road, Stockport, SK6 3LE, UK
| | - Natalie Mount
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guys Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Michaela Sharpe
- Cell and Gene Therapy Catapult, 12th Floor Tower Wing, Guys Hospital, Great Maze Pond, London, SE1 9RT, UK.
| |
Collapse
|
50
|
Wahlberg B, Ghuman H, Liu JR, Modo M. Ex vivo biomechanical characterization of syringe-needle ejections for intracerebral cell delivery. Sci Rep 2018; 8:9194. [PMID: 29907825 PMCID: PMC6004017 DOI: 10.1038/s41598-018-27568-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/05/2018] [Indexed: 02/06/2023] Open
Abstract
Intracerebral implantation of cell suspensions is finding its clinical translation with encouraging results in patients with stroke. However, the survival of cells in the brain remains poor. Although the biological potential of neural stem cells (NSCs) is widely documented, the biomechanical effects of delivering cells through a syringe-needle remain poorly understood. We here detailed the biomechanical forces (pressure, shear stress) that cells are exposed to during ejection through different sized needles (20G, 26G, 32G) and syringes (10, 50, 250 µL) at relevant flow rates (1, 5, 10 µL/min). A comparison of 3 vehicles, Phosphate Buffered Saline (PBS), Hypothermosol (HTS), and Pluronic, indicated that less viscous vehicles are favorable for suspension with a high cell volume fraction to minimize sedimentation. Higher suspension viscosity was associated with greater shear stress. Higher flow rates with viscous vehicle, such as HTS reduced viability by ~10% and also produced more apoptotic cells (28%). At 5 µL/min ejection using a 26G needle increased neuronal differentiation for PBS and HTS suspensions. These results reveal the biological impact of biomechanical forces in the cell delivery process. Appropriate engineering strategies can be considered to mitigate these effects to ensure the efficacious translation of this promising therapy.
Collapse
Affiliation(s)
- Brendon Wahlberg
- Departments of Radiology, University of Pittsburgh, Pittsburgh, USA
| | - Harmanvir Ghuman
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Jessie R Liu
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, USA
| | - Michel Modo
- Departments of Radiology, University of Pittsburgh, Pittsburgh, USA. .,Departments of Bioengineering, University of Pittsburgh, Pittsburgh, USA. .,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA. .,Centre for Neural Basis of Cognition, Pittsburgh, PA15203, USA.
| |
Collapse
|