1
|
Gu X, Zhang S, Ma W. Prussian blue nanotechnology in the treatment of spinal cord injury: application and challenges. Front Bioeng Biotechnol 2024; 12:1474711. [PMID: 39323764 PMCID: PMC11422158 DOI: 10.3389/fbioe.2024.1474711] [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: 08/05/2024] [Accepted: 08/28/2024] [Indexed: 09/27/2024] Open
Abstract
Spinal cord injury (SCI) is a serious neurological condition that currently lacks effective treatments, placing a heavy burden on both patients and society. Prussian blue nanoparticles exhibit great potential for treating spinal cord injuries due to their excellent physicochemical properties and biocompatibility. These nanoparticles have strong anti-inflammatory and antioxidant capabilities, effectively scavenge free radicals, and reduce oxidative stress damage to cells. Prussian blue nanotechnology shows broad application potential in drug delivery, bioimaging, cancer therapy, anti-inflammatory and oxidative stress treatment, and biosensors. This article reviewed the potential applications of Prussian blue nanotechnology in treating spinal cord injuries, explored the challenges and solutions associated with its application, and discussed the future prospects of this technology in SCI treatment.
Collapse
Affiliation(s)
- XiaoPeng Gu
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Orthopedics, NingBo NO.6 Hospital, Ningbo, Zhejiang, China
- Department of Orthopedics, Zhoushan Guhechuan Hospital, Zhoushan, Zhejiang, China
- Zhoushan Institute of Orthopedics and Traumatology, Zhoushan, Zhejiang, China
| | - SongOu Zhang
- Department of Clinical Medicine, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - WeiHu Ma
- Department of Orthopedics, NingBo NO.6 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
2
|
Sharma S, Kalyani N, Dutta T, Velázquez-González JS, Llamas-Garro I, Ung B, Bas J, Dubey R, Mishra SK. Optical Devices for the Diagnosis and Management of Spinal Cord Injuries: A Review. BIOSENSORS 2024; 14:296. [PMID: 38920599 PMCID: PMC11201428 DOI: 10.3390/bios14060296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/21/2024] [Accepted: 06/02/2024] [Indexed: 06/27/2024]
Abstract
Throughout the central nervous system, the spinal cord plays a very important role, namely, transmitting sensory and motor information inwardly so that it can be processed by the brain. There are many different ways this structure can be damaged, such as through traumatic injury or surgery, such as scoliosis correction, for instance. Consequently, damage may be caused to the nervous system as a result of this. There is no doubt that optical devices such as microscopes and cameras can have a significant impact on research, diagnosis, and treatment planning for patients with spinal cord injuries (SCIs). Additionally, these technologies contribute a great deal to our understanding of these injuries, and they are also essential in enhancing the quality of life of individuals with spinal cord injuries. Through increasingly powerful, accurate, and minimally invasive technologies that have been developed over the last decade or so, several new optical devices have been introduced that are capable of improving the accuracy of SCI diagnosis and treatment and promoting a better quality of life after surgery. We aim in this paper to present a timely overview of the various research fields that have been conducted on optical devices that can be used to diagnose spinal cord injuries as well as to manage the associated health complications that affected individuals may experience.
Collapse
Affiliation(s)
- Sonika Sharma
- Department of Physics, Graphic Era Hill University, Dehradun 248002, Uttarakhand, India;
| | - Neeti Kalyani
- Department of Biotechnology and Biomedicine, Denmark Technical University, 2800 Kongens Lyngby, Denmark;
| | - Taposhree Dutta
- Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, Howarh 711103, West Bengal, India;
| | - Jesús Salvador Velázquez-González
- Navigation and Positioning, Center Technologic de Telecomunicacions de Catalunya (CTTC), Avinguda Carl Friedrich Gauss, 11, 08860 Castelldefels, Spain; (J.S.V.-G.); (I.L.-G.)
| | - Ignacio Llamas-Garro
- Navigation and Positioning, Center Technologic de Telecomunicacions de Catalunya (CTTC), Avinguda Carl Friedrich Gauss, 11, 08860 Castelldefels, Spain; (J.S.V.-G.); (I.L.-G.)
| | - Bora Ung
- Electrical Engineering Department, Ecole de Technologie Superieure, Montreal, QC H3C 1K3, Canada;
| | - Joan Bas
- Space and Resilient Communications and Systems (SRCOM), Center Technologic de Telecomunicacions de Catalunya (CTTC), Avinguda Carl Friedrich Gauss, 11, 08860 Castelldefels, Spain;
| | - Rakesh Dubey
- Institute of Physics, University of Szczecin, 70-453 Szczecin, Poland;
| | - Satyendra K. Mishra
- Space and Resilient Communications and Systems (SRCOM), Center Technologic de Telecomunicacions de Catalunya (CTTC), Avinguda Carl Friedrich Gauss, 11, 08860 Castelldefels, Spain;
| |
Collapse
|
3
|
Nguyen VP, Zhe J, Hu J, Ahmed U, Paulus YM. Molecular and cellular imaging of the eye. BIOMEDICAL OPTICS EXPRESS 2024; 15:360-386. [PMID: 38223186 PMCID: PMC10783915 DOI: 10.1364/boe.502350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/25/2023] [Accepted: 12/02/2023] [Indexed: 01/16/2024]
Abstract
The application of molecular and cellular imaging in ophthalmology has numerous benefits. It can enable the early detection and diagnosis of ocular diseases, facilitating timely intervention and improved patient outcomes. Molecular imaging techniques can help identify disease biomarkers, monitor disease progression, and evaluate treatment responses. Furthermore, these techniques allow researchers to gain insights into the pathogenesis of ocular diseases and develop novel therapeutic strategies. Molecular and cellular imaging can also allow basic research to elucidate the normal physiological processes occurring within the eye, such as cell signaling, tissue remodeling, and immune responses. By providing detailed visualization at the molecular and cellular level, these imaging techniques contribute to a comprehensive understanding of ocular biology. Current clinically available imaging often relies on confocal microscopy, multi-photon microscopy, PET (positron emission tomography) or SPECT (single-photon emission computed tomography) techniques, optical coherence tomography (OCT), and fluorescence imaging. Preclinical research focuses on the identification of novel molecular targets for various diseases. The aim is to discover specific biomarkers or molecular pathways associated with diseases, allowing for targeted imaging and precise disease characterization. In parallel, efforts are being made to develop sophisticated and multifunctional contrast agents that can selectively bind to these identified molecular targets. These contrast agents can enhance the imaging signal and improve the sensitivity and specificity of molecular imaging by carrying various imaging labels, including radionuclides for PET or SPECT, fluorescent dyes for optical imaging, or nanoparticles for multimodal imaging. Furthermore, advancements in technology and instrumentation are being pursued to enable multimodality molecular imaging. Integrating different imaging modalities, such as PET/MRI (magnetic resonance imaging) or PET/CT (computed tomography), allows for the complementary strengths of each modality to be combined, providing comprehensive molecular and anatomical information in a single examination. Recently, photoacoustic microscopy (PAM) has been explored as a novel imaging technology for visualization of different retinal diseases. PAM is a non-invasive, non-ionizing radiation, and hybrid imaging modality that combines the optical excitation of contrast agents with ultrasound detection. It offers a unique approach to imaging by providing both anatomical and functional information. Its ability to utilize molecularly targeted contrast agents holds great promise for molecular imaging applications in ophthalmology. In this review, we will summarize the application of multimodality molecular imaging for tracking chorioretinal angiogenesis along with the migration of stem cells after subretinal transplantation in vivo.
Collapse
Affiliation(s)
- Van Phuc Nguyen
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Josh Zhe
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Justin Hu
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Umayr Ahmed
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Yannis M. Paulus
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
4
|
Jhunjhunwala A, Kim J, Kubelick KP, Ethier CR, Emelianov SY. In Vivo Photoacoustic Monitoring of Stem Cell Location and Apoptosis with Caspase-3-Responsive Nanosensors. ACS NANO 2023; 17:17931-17945. [PMID: 37703202 PMCID: PMC10540261 DOI: 10.1021/acsnano.3c04161] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Stem cell therapy has immense potential in a variety of regenerative medicine applications. However, clinical stem cell therapy is severely limited by challenges in assessing the location and functional status of implanted cells in vivo. Thus, there is a great need for longitudinal, noninvasive stem cell monitoring. Here we introduce a multidisciplinary approach combining nanosensor-augmented stem cell labeling with ultrasound guided photoacoustic (US/PA) imaging for the spatial tracking and functional assessment of transplanted stem cell fate. Specifically, our nanosensor incorporates a peptide sequence that is selectively cleaved by caspase-3, the primary effector enzyme in mammalian cell apoptosis; this cleavage event causes labeled cells to show enhanced optical absorption in the first near-infrared (NIR) window. Optimization of labeling protocols and spectral characterization of the nanosensor in vitro showed a 2.4-fold increase in PA signal from labeled cells during apoptosis while simultaneously permitting cell localization. We then successfully tracked the location and apoptotic status of mesenchymal stem cells in a mouse hindlimb ischemia model for 2 weeks in vivo, demonstrating a 4.8-fold increase in PA signal and spectral slope changes in the first NIR window under proapoptotic (ischemic) conditions. We conclude that our nanosensor allows longitudinal, noninvasive, and nonionizing monitoring of stem cell location and apoptosis, which is a significant improvement over current end-point monitoring methods such as biopsies and histological staining of excised tissue.
Collapse
Affiliation(s)
- Anamik Jhunjhunwala
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
| | - Jinhwan Kim
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- School
of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Kelsey P. Kubelick
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- School
of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - C. Ross Ethier
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
| | - Stanislav Y. Emelianov
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- School
of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
5
|
Huang WH, Ding SL, Zhao XY, Li K, Guo HT, Zhang MZ, Gu Q. Collagen for neural tissue engineering: Materials, strategies, and challenges. Mater Today Bio 2023; 20:100639. [PMID: 37197743 PMCID: PMC10183670 DOI: 10.1016/j.mtbio.2023.100639] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/19/2023] Open
Abstract
Neural tissue engineering (NTE) has made remarkable strides in recent years and holds great promise for treating several devastating neurological disorders. Selecting optimal scaffolding material is crucial for NET design strategies that enable neural and non-neural cell differentiation and axonal growth. Collagen is extensively employed in NTE applications due to the inherent resistance of the nervous system against regeneration, functionalized with neurotrophic factors, antagonists of neural growth inhibitors, and other neural growth-promoting agents. Recent advancements in integrating collagen with manufacturing strategies, such as scaffolding, electrospinning, and 3D bioprinting, provide localized trophic support, guide cell alignment, and protect neural cells from immune activity. This review categorises and analyses collagen-based processing techniques investigated for neural-specific applications, highlighting their strengths and weaknesses in repair, regeneration, and recovery. We also evaluate the potential prospects and challenges of using collagen-based biomaterials in NTE. Overall, this review offers a comprehensive and systematic framework for the rational evaluation and applications of collagen in NTE.
Collapse
Affiliation(s)
- Wen-Hui Huang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Sheng-Long Ding
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, PR China
| | - Xi-Yuan Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Kai Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
| | - Hai-Tao Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Ming-Zhu Zhang
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, PR China
- Corresponding author.
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
- Corresponding author. Institute of Zoology, Chinese Academy of Sciences, No. 5 of Courtyard 1, Beichen West Road, Chaoyang District, Beijing 100101, PR China.
| |
Collapse
|
6
|
Su G, Lei X, Wang Z, Xie W, Wen D, Wu Y. Mesenchymal Stem Cell-derived Exosomes Affect Macrophage Phenotype: A Cell-free Strategy for the Treatment of Skeletal Muscle Disorders. Curr Mol Med 2023; 23:350-357. [PMID: 35546766 DOI: 10.2174/1566524022666220511123625] [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: 02/27/2022] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022]
Abstract
The process of tissue damage, repair, and regeneration in the skeletal muscle system involves complex inflammatory processes. Factors released in the inflammatory microenvironment can affect the phenotypic changes of macrophages, thereby changing the inflammatory process, making macrophages an important target for tissue repair treatment. Mesenchymal stem cells exert anti-inflammatory effects by regulating immune cells. In particular, exosomes secreted by mesenchymal stem cells have become a new cell-free treatment strategy due to their low tumorigenicity and immunogenicity. This article focuses on the mechanism of the effect of exosomes derived from mesenchymal stem cells on the phenotype of macrophages after skeletal muscle system injury and explores the possible mechanism of macrophages as potential therapeutic targets after tissue injury.
Collapse
Affiliation(s)
- Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaoting Lei
- Department of Orthopedics, The First People's Hospital of Tianshui City, Tianshui, China
| | - Zhenyu Wang
- Department of Orthopedics, The First People's Hospital of Tianshui City, Tianshui, China
| | - Weiqiang Xie
- Department of Orthopedics, The First People's Hospital of Tianshui City, Tianshui, China
| | - Donghong Wen
- Department of Orthopedics, The First People's Hospital of Tianshui City, Tianshui, China
| | - Yucheng Wu
- Department of Orthopedics, The First People's Hospital of Tianshui City, Tianshui, China
| |
Collapse
|
7
|
Cadena M, Ning L, King A, Hwang B, Jin L, Serpooshan V, Sloan SA. 3D Bioprinting of Neural Tissues. Adv Healthc Mater 2021; 10:e2001600. [PMID: 33200587 PMCID: PMC8711131 DOI: 10.1002/adhm.202001600] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/19/2020] [Indexed: 02/06/2023]
Abstract
The human nervous system is a remarkably complex physiological network that is inherently challenging to study because of obstacles to acquiring primary samples. Animal models offer powerful alternatives to study nervous system development, diseases, and regenerative processes, however, they are unable to address some species-specific features of the human nervous system. In vitro models of the human nervous system have expanded in prevalence and sophistication, but still require further advances to better recapitulate microenvironmental and cellular features. The field of neural tissue engineering (TE) is rapidly adopting new technologies that enable scientists to precisely control in vitro culture conditions and to better model nervous system formation, function, and repair. 3D bioprinting is one of the major TE technologies that utilizes biocompatible hydrogels to create precisely patterned scaffolds, designed to enhance cellular responses. This review focuses on the applications of 3D bioprinting in the field of neural TE. Important design parameters are considered when bioprinting neural stem cells are discussed. The emergence of various bioprinted in vitro platforms are also reviewed for developmental and disease modeling and drug screening applications within the central and peripheral nervous systems, as well as their use as implants for in vivo regenerative therapies.
Collapse
Affiliation(s)
- Melissa Cadena
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Alexia King
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Boeun Hwang
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Linqi Jin
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Steven A. Sloan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
8
|
Chen W, Zhang Y, Yang S, Sun J, Qiu H, Hu X, Niu X, Xiao Z, Zhao Y, Zhou Y, Dai J, Chu T. NeuroRegen Scaffolds Combined with Autologous Bone Marrow Mononuclear Cells for the Repair of Acute Complete Spinal Cord Injury: A 3-Year Clinical Study. Cell Transplant 2021; 29:963689720950637. [PMID: 32862715 PMCID: PMC7784506 DOI: 10.1177/0963689720950637] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Spinal cord injury (SCI) remains among the most challenging pathologies worldwide and has limited therapeutic possibilities and a very bleak prognosis. Biomaterials and stem cell transplantation are promising treatments for functional recovery in SCI. Seven patients with acute complete SCI diagnosed by a combination of methods were included in the study, and different lengths (2.0–6.0 cm) of necrotic spinal cord tissue were surgically cleaned under intraoperative neurophysiological monitoring. Subsequently, NeuroRegen scaffolds loaded with autologous bone marrow mononuclear cells (BMMCs) were implanted into the cleaned site. All patients participated in 6 months of rehabilitation and at least 3 years of clinical follow-up. No adverse symptoms associated with stem cell or functional scaffold implantation were observed during the 3-year follow-up period. Additionally, partial shallow sensory and autonomic nervous functional improvements were observed in some patients, but no motor function recovery was observed. Magnetic resonance imaging suggested that NeuroRegen scaffold implantation supported injured spinal cord continuity after treatment. These findings indicate that implantation of NeuroRegen scaffolds combined with stem cells may serve as a safe and promising clinical treatment for patients with acute complete SCI. However, determining the therapeutic effects and exact application methods still requires further study.
Collapse
Affiliation(s)
- Wugui Chen
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China.,* Both the authors contributed equally as first author
| | - Ying Zhang
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China.,* Both the authors contributed equally as first author
| | - Sizhen Yang
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China
| | - Jing Sun
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China
| | - Hao Qiu
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China
| | - Xu Hu
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China
| | - Xiaojian Niu
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, 53019Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, 53019Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yue Zhou
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, 53019Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Tongwei Chu
- 105785Xinqiao Hospital, Department of Orthopedics, Army Medical University, Chongqing, China
| |
Collapse
|
9
|
Lee HL, Yeum CE, Lee H, Oh J, Kim JT, Lee WJ, Ha Y, Yang YI, Kim KN. Peripheral Nerve-Derived Stem Cell Spheroids Induce Functional Recovery and Repair after Spinal Cord Injury in Rodents. Int J Mol Sci 2021; 22:ijms22084141. [PMID: 33923671 PMCID: PMC8072978 DOI: 10.3390/ijms22084141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 01/09/2023] Open
Abstract
Stem cell therapy is one of the most promising candidate treatments for spinal cord injury. Research has shown optimistic results for this therapy, but clinical limitations remain, including poor viability, engraftment, and differentiation. Here, we isolated novel peripheral nerve-derived stem cells (PNSCs) from adult peripheral nerves with similar characteristics to neural-crest stem cells. These PNSCs expressed neural-crest specific markers and showed multilineage differentiation potential into Schwann cells, neuroglia, neurons, and mesodermal cells. In addition, PNSCs showed therapeutic potential by releasing the neurotrophic factors, including glial cell-line-derived neurotrophic factor, insulin-like growth factor, nerve growth factor, and neurotrophin-3. PNSC abilities were also enhanced by their development into spheroids which secreted neurotrophic factors several times more than non-spheroid PNSCs and expressed several types of extra cellular matrix. These features suggest that the potential for these PNSC spheroids can overcome their limitations. In an animal spinal cord injury (SCI) model, these PNSC spheroids induced functional recovery and neuronal regeneration. These PNSC spheroids also reduced the neuropathic pain which accompanies SCI after remyelination. These PNSC spheroids may represent a new therapeutic approach for patients suffering from SCI.
Collapse
Affiliation(s)
- Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Chung-Eun Yeum
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - HyeYeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Jinsoo Oh
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Jong-Tae Kim
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - Won-Jin Lee
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Korea
| | - Young-Il Yang
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
- Correspondence: (Y.-I.Y.); (K.-N.K.)
| | - Keung-Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
- Correspondence: (Y.-I.Y.); (K.-N.K.)
| |
Collapse
|
10
|
Cellular therapy for treatment of spinal cord injury in Zebrafish model. Mol Biol Rep 2021; 48:1787-1800. [PMID: 33459959 DOI: 10.1007/s11033-020-06126-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/24/2020] [Indexed: 02/08/2023]
Abstract
Spinal cord injury is a serious problem with a high rate of morbidity and mortality for all persons, especially young people (15-25 years old). Due to the large burden and the costs incurred on the government, finding the best therapeutic approach is necessary. In this respect, treatment strategies based on the disease mechanism can be effective. After the first trauma of spinal cord cascades, cellular events happen one after the other known as secondary trauma. The mechanism of secondary events of spinal cord injury could be helpful for target therapy as trying to stop the secondary trauma. Herein, some medical and surgical therapy has been introduced and cell therapy strategy was considered as a recent method. Actually, cell therapy is defined as the application of different cells including mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells, and some others to replace or reconstruct the damaged tissues and restore their functions. However, as a newly emerged therapeutic method, cell therapy should be used through various subclinical studies in animal models to assess the efficacy of the treatment under controlled conditions. In this review, the role of Zebrafish as a recommended model has been discussed and combinatory approach as the probably most useful treatment has been suggested.
Collapse
|
11
|
Jevans B, James ND, Burnside E, McCann CJ, Thapar N, Bradbury EJ, Burns AJ. Combined treatment with enteric neural stem cells and chondroitinase ABC reduces spinal cord lesion pathology. Stem Cell Res Ther 2021; 12:10. [PMID: 33407795 PMCID: PMC7789480 DOI: 10.1186/s13287-020-02031-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) presents a significant challenge for the field of neurotherapeutics. Stem cells have shown promise in replenishing the cells lost to the injury process, but the release of axon growth-inhibitory molecules such as chondroitin sulfate proteoglycans (CSPGs) by activated cells within the injury site hinders the integration of transplanted cells. We hypothesised that simultaneous application of enteric neural stem cells (ENSCs) isolated from the gastrointestinal tract, with a lentivirus (LV) containing the enzyme chondroitinase ABC (ChABC), would enhance the regenerative potential of ENSCs after transplantation into the injured spinal cord. METHODS ENSCs were harvested from the GI tract of p7 rats, expanded in vitro and characterised. Adult rats bearing a contusion injury were randomly assigned to one of four groups: no treatment, LV-ChABC injection only, ENSC transplantation only or ENSC transplantation+LV-ChABC injection. After 16 weeks, rats were sacrificed and the harvested spinal cords examined for evidence of repair. RESULTS ENSC cultures contained a variety of neuronal subtypes suitable for replenishing cells lost through SCI. Following injury, transplanted ENSC-derived cells survived and ChABC successfully degraded CSPGs. We observed significant reductions in the injured tissue and cavity area, with the greatest improvements seen in the combined treatment group. ENSC-derived cells extended projections across the injury site into both the rostral and caudal host spinal cord, and ENSC transplantation significantly increased the number of cells extending axons across the injury site. Furthermore, the combined treatment resulted in a modest, but significant functional improvement by week 16, and we found no evidence of the spread of transplanted cells to ectopic locations or formation of tumours. CONCLUSIONS Regenerative effects of a combined treatment with ENSCs and ChABC surpassed either treatment alone, highlighting the importance of further research into combinatorial therapies for SCI. Our work provides evidence that stem cells taken from the adult gastrointestinal tract, an easily accessible source for autologous transplantation, could be strongly considered for the repair of central nervous system disorders.
Collapse
Affiliation(s)
- Benjamin Jevans
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
- Present Address: German Centre for Neurodegenerative diseases (DZNE), Bonn, Germany
| | - Nicholas D James
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Emily Burnside
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
- Neurogastroenterology and Motility Unit, Department of Gastroenterology, Great Ormond Street Hospital, London, UK
- Present Address: Department of Paediatric Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, Brisbane, Australia
| | - Elizabeth J Bradbury
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
- Present Address: Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals International, Cambridge, USA.
| |
Collapse
|
12
|
Kubelick KP, Emelianov SY. A Trimodal Ultrasound, Photoacoustic and Magnetic Resonance Imaging Approach for Longitudinal Post-operative Monitoring of Stem Cells in the Spinal Cord. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:3468-3474. [PMID: 32988671 PMCID: PMC7709928 DOI: 10.1016/j.ultrasmedbio.2020.08.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/10/2020] [Accepted: 08/25/2020] [Indexed: 06/11/2023]
Abstract
Longitudinal monitoring of stem cells in the spinal cord could unveil critical information needed to understand regenerative processes, thereby expediting therapy development and translation. We introduce a post-operative trimodal imaging approach to monitor stem cells in the spinal cord over time. A key aspect of the approach is to label the stem cells with Prussian blue nanocubes (PBNCs), which simultaneously possess optical and magnetic properties for ultrasound-guided photoacoustic (US/PA) and magnetic resonance imaging (MRI) contrast. PBNC-Labeled stem cells were injected into the spinal cord of immunodeficient rats and tracked with US/PA imaging and MRI up to 14 d post-injection. Good agreement was observed between imaging modalities in vivo. Our results suggest that further development of the US/PA/MR imaging approach may create a powerful tool to aid development of regenerative therapies of the spinal cord, and the non-invasive imaging approach can ultimately be deployed in intra- and post-operative environments.
Collapse
Affiliation(s)
- Kelsey P Kubelick
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, USA; School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
| | - Stanislav Y Emelianov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, USA; School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
| |
Collapse
|
13
|
Ahuja CS, Mothe A, Khazaei M, Badhiwala JH, Gilbert EA, van der Kooy D, Morshead CM, Tator C, Fehlings MG. The leading edge: Emerging neuroprotective and neuroregenerative cell-based therapies for spinal cord injury. Stem Cells Transl Med 2020; 9:1509-1530. [PMID: 32691994 PMCID: PMC7695641 DOI: 10.1002/sctm.19-0135] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/01/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injuries (SCIs) are associated with tremendous physical, social, and financial costs for millions of individuals and families worldwide. Rapid delivery of specialized medical and surgical care has reduced mortality; however, long-term functional recovery remains limited. Cell-based therapies represent an exciting neuroprotective and neuroregenerative strategy for SCI. This article summarizes the most promising preclinical and clinical cell approaches to date including transplantation of mesenchymal stem cells, neural stem cells, oligodendrocyte progenitor cells, Schwann cells, and olfactory ensheathing cells, as well as strategies to activate endogenous multipotent cell pools. Throughout, we emphasize the fundamental biology of cell-based therapies, critical features in the pathophysiology of spinal cord injury, and the strengths and limitations of each approach. We also highlight salient completed and ongoing clinical trials worldwide and the bidirectional translation of their findings. We then provide an overview of key adjunct strategies such as trophic factor support to optimize graft survival and differentiation, engineered biomaterials to provide a support scaffold, electrical fields to stimulate migration, and novel approaches to degrade the glial scar. We also discuss important considerations when initiating a clinical trial for a cell therapy such as the logistics of clinical-grade cell line scale-up, cell storage and transportation, and the delivery of cells into humans. We conclude with an outlook on the future of cell-based treatments for SCI and opportunities for interdisciplinary collaboration in the field.
Collapse
Affiliation(s)
- Christopher S. Ahuja
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Andrea Mothe
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Mohamad Khazaei
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Jetan H. Badhiwala
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
| | - Emily A. Gilbert
- Division of Anatomy, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
| | - Derek van der Kooy
- Department of Molecular GeneticsUniversity of TorontoTorontoOntarioCanada
| | - Cindi M. Morshead
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Division of Anatomy, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Charles Tator
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Michael G. Fehlings
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| |
Collapse
|
14
|
Yang X, Tian DC, He W, Lv W, Fan J, Li H, Jin WN, Meng X. Cellular and molecular imaging for stem cell tracking in neurological diseases. Stroke Vasc Neurol 2020; 6:121-127. [PMID: 33122254 PMCID: PMC8005893 DOI: 10.1136/svn-2020-000408] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/27/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
Stem cells (SCs) are cells with strong proliferation ability, multilineage differentiation potential and self-renewal capacity. SC transplantation represents an important therapeutic advancement for the treatment strategy of neurological diseases, both in the preclinical experimental and clinical settings. Innovative and breakthrough SC labelling and tracking technologies are widely used to monitor the distribution and viability of transplanted cells non-invasively and longitudinally. Here we summarised the research progress of the main tracers, labelling methods and imaging technologies involved in current SC tracking technologies for various neurological diseases. Finally, the applications, challenges and unresolved problems of current SC tracing technologies were discussed.
Collapse
Affiliation(s)
- Xiaoxia Yang
- China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing Tiantan Hospital, Beijing, China
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - De-Cai Tian
- China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing Tiantan Hospital, Beijing, China
| | - Wenyan He
- China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing Tiantan Hospital, Beijing, China
| | - Wei Lv
- China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing Tiantan Hospital, Beijing, China
| | - Junwan Fan
- China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing Tiantan Hospital, Beijing, China
| | - Haowen Li
- China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing Tiantan Hospital, Beijing, China
| | - Wei-Na Jin
- China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing Tiantan Hospital, Beijing, China
| | - Xia Meng
- China National Clinical Research Center for Neurological Diseases, Capital Medical University, Beijing Tiantan Hospital, Beijing, China
| |
Collapse
|
15
|
Kubelick KP, Emelianov SY. In vivo photoacoustic guidance of stem cell injection and delivery for regenerative spinal cord therapies. NEUROPHOTONICS 2020; 7:030501. [PMID: 32743015 PMCID: PMC7388074 DOI: 10.1117/1.nph.7.3.030501] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 07/14/2020] [Indexed: 05/16/2023]
Abstract
Significance: Stem cell therapies are of interest for treating a variety of neurodegenerative diseases and injuries of the spinal cord. However, the lack of techniques for longitudinal monitoring of stem cell therapy progression is inhibiting clinical translation. Aim: The goal of this study is to demonstrate an intraoperative imaging approach to guide stem cell injection to the spinal cord in vivo. Results may ultimately support the development of an imaging tool that spans intra- or postoperative environments to guide therapy throughout treatment. Approach: Stem cells were labeled with Prussian blue nanocubes (PBNCs) to facilitate combined ultrasound and photoacoustic (US/PA) imaging to visualize stem cell injection and delivery to the spinal cord in vivo. US/PA results were confirmed by magnetic resonance imaging (MRI) and histology. Results: Real-time intraoperative US/PA image-guided injection of PBNC-labeled stem cells and three-dimensional volumetric images of injection provided feedback necessary for successful delivery of therapeutics into the spinal cord. Postoperative MRI confirmed delivery of PBNC-labeled stem cells. Conclusions: The nanoparticle-augmented US/PA approach successfully detected injection and delivery of stem cells into the spinal cord, confirmed by MRI. Our work demonstrated in vivo feasibility, which is a critical step toward the development of a US/PA/MRI platform to monitor regenerative spinal cord therapies.
Collapse
Affiliation(s)
- Kelsey P. Kubelick
- Georgia Institute of Technology, Emory University School of Medicine, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Georgia Institute of Technology, School of Electrical and Computer Engineering, Atlanta, Georgia, United States
| | - Stanislav Y. Emelianov
- Georgia Institute of Technology, Emory University School of Medicine, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Georgia Institute of Technology, School of Electrical and Computer Engineering, Atlanta, Georgia, United States
| |
Collapse
|
16
|
Gao Y, Jin SZ. Strategies for treating oesophageal diseases with stem cells. World J Stem Cells 2020; 12:488-499. [PMID: 32742566 PMCID: PMC7360987 DOI: 10.4252/wjsc.v12.i6.488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/02/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
There is a wide range of oesophageal diseases, the most general of which are inflammation, injury and tumours, and treatment methods are constantly being developed and updated. With an increasingly comprehensive understanding of stem cells and their characteristics of multilineage differentiation, self-renewal and homing as well as the combination of stem cells with regenerative medicine, tissue engineering and gene therapy, stem cells are playing an important role in the treatment of a variety of diseases. Mesenchymal stem cells have many advantages and are most commonly applied; however, most of these applications have been in experimental studies, with few related clinical trials for comparison. Therefore, the methods, positive significance and limitations of stem cells in the treatment of oesophageal diseases remain incompletely understood. Thus, the purpose of this paper is to review the current literature and summarize the efficacy of stem cells in the treatment of oesophageal diseases, including oesophageal ulceration, acute radiation-induced oesophageal injury, corrosive oesophageal injury, oesophageal stricture formation after endoscopic submucosal dissection and oesophageal reconstruction, as well as gene therapy for oesophageal cancer.
Collapse
Affiliation(s)
- Yang Gao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Shi-Zhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| |
Collapse
|
17
|
Kubelick KP, Emelianov SY. Prussian blue nanocubes as a multimodal contrast agent for image-guided stem cell therapy of the spinal cord. PHOTOACOUSTICS 2020; 18:100166. [PMID: 32211291 PMCID: PMC7082547 DOI: 10.1016/j.pacs.2020.100166] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/20/2020] [Accepted: 02/03/2020] [Indexed: 05/16/2023]
Abstract
Translation of stem cell therapies to treat injuries and diseases of the spinal cord is hindered by lack of real-time monitoring techniques to guide regenerative therapies intra- and postoperatively. Thus, we developed an ultrasound (US), photoacoustic (PA), and magnetic resonance (MR) imaging approach augmented with Prussian blue nanocubes (PBNCs) to guide stem cell injections intraoperatively and monitor stem cell therapies in the spinal cord postoperatively. Per the clinical procedure, a multi-level laminectomy was performed in rats ex vivo, and PBNC-labeled stem cells were injected directly into the spinal cord while US/PA images were acquired. US/PA/MR images were also acquired post-surgery. Several features of the imaging approach were demonstrated including detection of low stem cell concentrations, real-time needle guidance and feedback on stem cell delivery, and good agreement between US/PA/MR images. These benefits span intra- and postoperative environments to support future development of this imaging tool.
Collapse
Key Words
- AuNS, gold nanosphere
- DIUF, deionized ultra-filtered water
- IACUC, Institutional Animal Care and Use Committee
- LOD, limit of detection
- MRI, magnetic resonance imaging
- MSC, mesenchymal stem cell
- Magnetic resonance imaging
- Multimodal imaging
- Nanoparticles
- OR, operating room
- PA, photoacoustic
- PBNC, Prussian blue nanocube
- PBS, phosphate buffered saline
- Photoacoustic imaging
- SPION, superparamagnetic iron oxide nanoparticle
- Spinal cord
- Stem cells
- TE, echo time
- TEM, transmission electron microscopy
- TR, repetition time
- US, ultrasound
- Ultrasound
Collapse
Affiliation(s)
- Kelsey P. Kubelick
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, 777 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Stanislav Y. Emelianov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, 777 Atlantic Drive, Atlanta, GA, 30332, USA
| |
Collapse
|
18
|
Qian K, Xu TY, Wang X, Ma T, Zhang KX, Yang K, Qian TD, Shi J, Li LX, Wang Z. Effects of neural stem cell transplantation on the motor function of rats with contusion spinal cord injuries: a meta-analysis. Neural Regen Res 2020; 15:748-758. [PMID: 31638100 PMCID: PMC6975148 DOI: 10.4103/1673-5374.266915] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective To judge the efficacies of neural stem cell (NSC) transplantation on functional recovery following contusion spinal cord injuries (SCIs). Data sources Studies in which NSCs were transplanted into a clinically relevant, standardized rat model of contusion SCI were identified by searching the PubMed, Embase and Cochrane databases, and the extracted data were analyzed by Stata 14.0. Data selection Inclusion criteria were that NSCs were used in in vivo animal studies to treat contusion SCIs and that behavioral assessment of locomotor functional recovery was performed using the Basso, Beattie, and Bresnahan lo-comotor rating scale. Exclusion criteria included a follow-up of less than 4 weeks and the lack of control groups. Outcome measures The restoration of motor function was assessed by the Basso, Beattie, and Bresnahan locomotor rating scale. Results We identified 1756 non-duplicated papers by searching the aforementioned electronic databases, and 30 full-text articles met the inclusion criteria. A total of 37 studies reported in the 30 articles were included in the meta-analysis. The meta-analysis results showed that transplanted NSCs could improve the motor function recovery of rats following contusion SCIs, to a moderate extent (pooled standardized mean difference (SMD) = 0.73; 95% confidence interval (CI): 0.47-1.00; P < 0.001). NSCs obtained from different donor species (rat: SMD = 0.74; 95% CI: 0.36-1.13; human: SMD = 0.78; 95% CI: 0.31-1.25), at different donor ages (fetal: SMD = 0.67; 95% CI: 0.43-0.92; adult: SMD = 0.86; 95% CI: 0.50-1.22) and from different origins (brain-derived: SMD = 0.59; 95% CI: 0.27-0.91; spinal cord-derived: SMD = 0.51; 95% CI: 0.22-0.79) had similar efficacies on improved functional recovery; however, adult induced pluripotent stem cell-derived NSCs showed no significant efficacies. Furthermore, the use of higher doses of transplanted NSCs or the administration of immunosuppressive agents did not promote better locomotor function recovery (SMD = 0.45; 95% CI: 0.21-0.70). However, shorter periods between the contusion induction and the NSC transplantation showed slightly higher efficacies (acute: SMD = 1.22; 95% CI: 0.81-1.63; subacute: SMD = 0.75; 95% CI: 0.42-1.09). For chronic injuries, NSC implantation did not significantly improve functional recovery (SMD = 0.25; 95% CI: -0.16 to 0.65). Conclusion NSC transplantation alone appears to be a positive yet limited method for the treatment of contusion SCIs.
Collapse
Affiliation(s)
- Kai Qian
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tuo-Ye Xu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xi Wang
- Department of Intensive Care Unit, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tao Ma
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing; Department of Neurosurgery, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Kai-Xin Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province; Department of Neurosurgery, Huangshan City People's Hospital, Huangshan, Anhui Province, China
| | - Kun Yang
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University; Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Teng-Da Qian
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing; Department of Neurosurgery, Jintan Hospital Affiliated to Jiangsu University, Jintan, Jiangsu Province, China
| | - Jing Shi
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li-Xin Li
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zheng Wang
- Department of Gerontology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
19
|
Borhani-Haghighi M, Navid S, Mohamadi Y. The Therapeutic Potential of Conditioned Medium from Human Breast Milk Stem Cells in Treating Spinal Cord Injury. Asian Spine J 2019; 14:131-138. [PMID: 31711062 PMCID: PMC7113460 DOI: 10.31616/asj.2019.0026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/05/2019] [Indexed: 12/22/2022] Open
Abstract
Study Design Experimental animal study. Purpose This study investigated the therapeutic effects of human breast milk stem cell (BMSC)-conditioned medium (BMSC-CM) in a model of spinal cord injury (SCI) in male Sprague-Dawley rats. Overview of Literature SCI is one of the leading causes of disability in addition to sensory and motor impairment. So far, there have been no successful treatments for SCI. Given the positive outcomes associated with using stem cells and their derivatives as a treatment for various diseases, there is a growing interest in using them as an SCI treatment. Recent research has demonstrated that CM from stem cells has therapeutic advantages. Methods Human BMSCs were isolated and characterized, and CM was subsequently collected. Animals received an intrathecal administration of BMSC-CM after SCI. The activity of caspase-3 was measured to assess apoptosis, and levels of tumor necrosis factor-α and interleukin-1β were measured to assess inflammation. Also, sensory and locomotor performances were assessed after SCI and BMSC-CM administration. Results Administration of CM from BMSC reduced apoptosis and inflammation at the site of injury in a rat model of SCI (p<0.05). Motor, sensory, locomotor, and sensorimotor performances were significantly improved in rats that received BMSC-CM after SCI. Conclusions Intrathecal administration of BMSC-CM improved recovery in a rat model of SCI.
Collapse
Affiliation(s)
- Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shadan Navid
- Department of Anatomy, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Yousef Mohamadi
- Department of Anatomy, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
20
|
CSF transplantation of a specific iPSC-derived neural stem cell subpopulation ameliorates the disease phenotype in a mouse model of spinal muscular atrophy with respiratory distress type 1. Exp Neurol 2019; 321:113041. [DOI: 10.1016/j.expneurol.2019.113041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/21/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022]
|
21
|
Piejko M, Jablonska A, Walczak P, Janowski M. Proteolytic Rafts for Improving Intraparenchymal Migration of Minimally Invasively Administered Hydrogel-Embedded Stem Cells. Int J Mol Sci 2019; 20:E3083. [PMID: 31238564 PMCID: PMC6628268 DOI: 10.3390/ijms20123083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 11/23/2022] Open
Abstract
The physiological spaces (lateral ventricles, intrathecal space) or pathological cavities (stroke lesion, syringomyelia) may serve as an attractive gateway for minimally invasive deployment of stem cells. Embedding stem cells in injectable scaffolds is essential when transplanting into the body cavities as they secure favorable microenvironment and keep cells localized, thereby preventing sedimentation. However, the limited migration of transplanted cells from scaffold to the host tissue is still a major obstacle, which prevents this approach from wider implementation for the rapidly growing field of regenerative medicine. Hyaluronan, a naturally occurring polymer, is frequently used as a basis of injectable scaffolds. We hypothesized that supplementation of hyaluronan with activated proteolytic enzymes could be a viable approach for dissolving the connective tissue barrier on the interface between the scaffold and the host, such as pia mater or scar tissue, thus demarcating lesion cavity. In a proof-of-concept study, we have found that collagenase and trypsin immobilized in hyaluronan-based hydrogel retain 60% and 28% of their proteolytic activity compared to their non-immobilized forms, respectively. We have also shown that immobilized enzymes do not have a negative effect on the viability of stem cells (glial progenitors and mesenchymal stem cells) in vitro. In conclusion, proteolytic rafts composed of hyaluronan-based hydrogels and immobilized enzymes may be an attractive strategy to facilitate migration of stem cells from injectable scaffolds into the parenchyma of surrounding tissue.
Collapse
Affiliation(s)
- Marcin Piejko
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- 3rd Department of General Surgery, Jagiellonian University Medical College, 31202 Krakow, Poland.
| | - Anna Jablonska
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Piotr Walczak
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Miroslaw Janowski
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
22
|
Donnelly EM, Kubelick KP, Dumani DS, Emelianov SY. Photoacoustic Image-Guided Delivery of Plasmonic-Nanoparticle-Labeled Mesenchymal Stem Cells to the Spinal Cord. NANO LETTERS 2018; 18:6625-6632. [PMID: 30160124 DOI: 10.1021/acs.nanolett.8b03305] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Regenerative therapies using stem cells have great potential for treating neurodegenerative diseases and traumatic injuries in the spinal cord. In spite of significant research efforts, many therapies fail at the clinical phase. As stem cell technologies advance toward clinical use, there is a need for a minimally invasive, safe, affordable, and real-time imaging technique that allows for the accurate and safe monitoring of stem cell delivery in the operating room. In this work, we present a combined ultrasound and photoacoustic imaging tool to provide image-guided needle placement and monitoring of nanoparticle-labeled stem cell delivery into the spinal cord. We successfully tagged stem cells using gold nanospheres and provided image-guided delivery of stem cells into the spinal cord in real-time, detecting as few as 1000 cells. Ultrasound and photoacoustic imaging was used to guide needle placement for direct stem cell injection to minimize the risk of needle shear and accidental injury and to improve therapeutic outcomes with accurate, localized stem cell delivery. Following injections of various volumes of cells, three-dimensional ultrasound and photoacoustic images allowed the visualization of stem cell distribution along the spinal cord, showing the potential to monitor the migration of the cells in the future. The feasibility of quantitative imaging was also shown by correlating the total photoacoustic signal over the imaging volume to the volume of cells injected. Overall, the presented method may allow clinicians to utilize imaged-guided delivery for more accurate and safer stem cell delivery to the spinal cord.
Collapse
Affiliation(s)
- Eleanor M Donnelly
- School of Electrical and Computer Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Kelsey P Kubelick
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Diego S Dumani
- School of Electrical and Computer Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Stanislav Y Emelianov
- School of Electrical and Computer Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| |
Collapse
|
23
|
Bhat IA, T B S, Somal A, Pandey S, Bharti MK, Panda BSK, B I, Verma M, J A, Sonwane A, Kumar GS, Amarpal, Chandra V, Sharma GT. An allogenic therapeutic strategy for canine spinal cord injury using mesenchymal stem cells. J Cell Physiol 2018; 234:2705-2718. [PMID: 30132873 DOI: 10.1002/jcp.27086] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022]
Abstract
This study was conducted to characterize canine bone marrow-derived mesenchymal stem cells (BMSCs); in vivo tracking in mice, and therapeutic evaluation in canine clinical paraplegia cases. Canine BMSCs were isolated, cultured, and characterized in vitro as per International Society for Cellular Therapy criteria, and successfully differentiated to chondrogenic, osteogenic, and adipogenic lineages. To demonstrate the homing property, the pGL4.51 vector that contained luciferase reporter gene was used to transfect BMSCs. Successfully transfected cells were injected around the skin wound in mice and in vivo imaging was done at 6, 12 and 24 hr post MSCs delivery. In vivo imaging revealed that transfected BMSCs migrated and concentrated predominantly toward the center of the wound. BMSCs were further evaluated for allogenic therapeutic potential in 44 clinical cases of spinal cord injuries (SCI) and compared with conventional therapy (control). Therapeutic potential as evaluated by different body reflexes and recovery score depicted significantly better results in stem cell-treated group compared to control group. In conclusion, allogenic canine BMSCs can serve as potent therapeutic candidate in cell-based therapies, especially for diseases like SCI, where the conventional medication is not so promising.
Collapse
Affiliation(s)
- Irfan A Bhat
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Sivanarayanan T B
- Division of Veterinary Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Anjali Somal
- Department of Veterinary Physiology and Biochemistry, CSK HPKV Palampur (H.P.)
| | - Sriti Pandey
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Mukesh K Bharti
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Bibhudatta S K Panda
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Indu B
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Megha Verma
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Anand J
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Arvind Sonwane
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - G Sai Kumar
- Division of Veterinary Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Amarpal
- Division of Veterinary Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Vikash Chandra
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| | - G Taru Sharma
- Division of Physiology and Climatology, ICAR-Indian Veter inary Research Institute, Izatnagar, Uttar Pradesh, India
| |
Collapse
|
24
|
Liu XY, Zhou CB, Fang C. Nanomaterial-involved neural stem cell research: Disease treatment, cell labeling, and growth regulation. Biomed Pharmacother 2018; 107:583-597. [PMID: 30114642 DOI: 10.1016/j.biopha.2018.08.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) have been widely investigated for their potential in the treatment of various diseases and transplantation therapy. However, NSC growth regulation, labeling, and its application to disease diagnosis and treatment are outstanding challenges. Recently, nanomaterials have shown promise for various applications including genetic modification, imaging, and controlled drug release. Here we summarize the recent progress in the use of nanomaterials in combination with NSCs for disease treatment and diagnosis, cell labeling, and NSC growth regulation. The toxicity of nanomaterials to NSCs is also discussed.
Collapse
Affiliation(s)
- Xiang-Yu Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Cheng-Bin Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240 China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
25
|
Foster AA, Marquardt LM, Heilshorn SC. The Diverse Roles of Hydrogel Mechanics in Injectable Stem Cell Transplantation. Curr Opin Chem Eng 2017; 15:15-23. [PMID: 29085771 PMCID: PMC5659597 DOI: 10.1016/j.coche.2016.11.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Stem cell delivery by local injection has tremendous potential as a regenerative therapy but has seen limited clinical success. Several mechanical challenges hinder therapeutic efficacy throughout all stages of cell transplantation, including mechanical forces during injection and loss of mechanical support post-injection. Recent studies have begun exploring the use of biomaterials, in particular hydrogels, to enhance stem cell transplantation by addressing the often-conflicting mechanical requirements associated with each stage of the transplantation process. This review explores recent biomaterial approaches to improve the therapeutic efficacy of stem cells delivered through local injection, with a focus on strategies that specifically address the mechanical challenges that result in cell death and/or limit therapeutic function throughout the stages of transplantation.
Collapse
Affiliation(s)
- Abbygail A Foster
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305
| | - Laura M Marquardt
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305
| |
Collapse
|
26
|
|
27
|
|
28
|
Stem Cells and Labeling for Spinal Cord Injury. Int J Mol Sci 2016; 18:ijms18010006. [PMID: 28035961 PMCID: PMC5297641 DOI: 10.3390/ijms18010006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that usually results in sudden and long-lasting locomotor and sensory neuron degeneration below the lesion site. During the last two decades, the search for new therapies has been revolutionized with the improved knowledge of stem cell (SC) biology. SCs therapy offers several attractive strategies for spinal cord repair. The transplantation of SCs promotes remyelination, neurite outgrowth and axonal elongation, and activates resident or transplanted progenitor cells across the lesion cavity. However, optimized growth and differentiation protocols along with reliable safety assays should be established prior to the clinical application of SCs. Additionally, the ideal method of SCs labeling for efficient cell tracking after SCI remains a challenging issue that requires further investigation. This review summarizes the current findings on the SCs-based therapeutic strategies, and compares different SCs labeling approaches for SCI.
Collapse
|
29
|
Lee JH, Lee T, Choi JW. Nano-Biosensor for Monitoring the Neural Differentiation of Stem Cells. NANOMATERIALS 2016; 6:nano6120224. [PMID: 28335352 PMCID: PMC5302715 DOI: 10.3390/nano6120224] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/07/2016] [Accepted: 11/17/2016] [Indexed: 01/06/2023]
Abstract
In tissue engineering and regenerative medicine, monitoring the status of stem cell differentiation is crucial to verify therapeutic efficacy and optimize treatment procedures. However, traditional methods, such as cell staining and sorting, are labor-intensive and may damage the cells. Therefore, the development of noninvasive methods to monitor the differentiation status in situ is highly desirable and can be of great benefit to stem cell-based therapies. Toward this end, nanotechnology has been applied to develop highly-sensitive biosensors to noninvasively monitor the neural differentiation of stem cells. Herein, this article reviews the development of noninvasive nano-biosensor systems to monitor the neural differentiation of stem cells, mainly focusing on optical (plasmonic) and eletrochemical methods. The findings in this review suggest that novel nano-biosensors capable of monitoring stem cell differentiation are a promising type of technology that can accelerate the development of stem cell therapies, including regenerative medicine.
Collapse
Affiliation(s)
- Jin-Ho Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, Seoul 121-742, Korea.
- Institute of Integrated Biotechnology, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, Seoul 121-742, Korea.
| | - Taek Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, Seoul 121-742, Korea.
- Institute of Integrated Biotechnology, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, Seoul 121-742, Korea.
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, Seoul 121-742, Korea.
- Institute of Integrated Biotechnology, Sogang University, 35 Baekbeom-ro (Sinsu-dong), Mapo-gu, Seoul 121-742, Korea.
| |
Collapse
|
30
|
Cheng Z, Bosco DB, Sun L, Chen X, Xu Y, Tai W, Didier R, Li J, Fan J, He X, Ren Y. Neural Stem Cell-Conditioned Medium Suppresses Inflammation and Promotes Spinal Cord Injury Recovery. Cell Transplant 2016; 26:469-482. [PMID: 27737726 DOI: 10.3727/096368916x693473] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) causes functional impairment as a result of the initial injury followed by secondary injury mechanism. SCI provokes an inflammatory response that causes secondary tissue damage and neurodegeneration. While the use of neural stem cell (NSC) engraftment to mitigate secondary injury has been of interest to many researchers, it still faces several limitations. As such, we investigated if NSC-conditioned medium (NSC-M) possesses therapeutic potential for the treatment of SCI. It has been proposed that many of the beneficial effects attributed to stem cell therapies are due to secreted factors. Utilizing primary cell culture and murine models of SCI, we determined that systemic treatment with NSC-M was able to significantly improve motor function and lesion healing. In addition, NSC-M demonstrated significant anti-inflammatory potential in vitro and in vivo, reducing inflammatory cytokine expression in both activated macrophages and injured spinal cord tissues. NSC-M was also able to reduce the expression of inducible nitric oxide synthase (iNOS) within the spleen of injured animals, indicating an ability to reduce systemic inflammation. Thus, we believe that NSC-M offers a possible alternative to direct stem cell engraftment for the treatment of SCI.
Collapse
|
31
|
Stem cells in canine spinal cord injury--promise for regenerative therapy in a large animal model of human disease. Stem Cell Rev Rep 2015; 11:180-93. [PMID: 25173879 DOI: 10.1007/s12015-014-9553-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The use of cell transplantation for spinal cord injury is a rapidly evolving field in regenerative medicine. Numerous animal models are currently being used. However, translation to human patients is still a challenging step. Dogs are of increasing importance as a translational model for human disease since there is a greater awareness of the need to increase the quality of preclinical data. The use of dogs ultimately brings benefit to both human and veterinary medicine. In this review we analyze experimental and clinical studies using cell transplantation for canine spinal cord injury. Overall, in experimental studies, transplantation groups showed improvement over control groups. Improvements were measured at the functional, electrophysiological, histological, RNA and protein levels. Most clinical studies support beneficial effects of cell transplantation despite the fact that methodological limitations preclude definitive conclusions. However, the mechanisms of action and underlying the behavior of transplanted cells in the injured spinal cord remain unclear. Overall, we conclude here that stem cell interventions are a promising avenue for the treatment of spinal cord injury. Canines are a promising model that may help bridge the gap between translational research and human clinical trials.
Collapse
|
32
|
Developing Extracellular Matrix Technology to Treat Retinal or Optic Nerve Injury(1,2,3). eNeuro 2015; 2:eN-REV-0077-15. [PMID: 26478910 PMCID: PMC4603254 DOI: 10.1523/eneuro.0077-15.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 12/30/2022] Open
Abstract
Adult mammalian CNS neurons often degenerate after injury, leading to lost neurologic functions. In the visual system, retinal or optic nerve injury often leads to retinal ganglion cell axon degeneration and irreversible vision loss. CNS axon degeneration is increasingly linked to the innate immune response to injury, which leads to tissue-destructive inflammation and scarring. Extracellular matrix (ECM) technology can reduce inflammation, while increasing functional tissue remodeling, over scarring, in various tissues and organs, including the peripheral nervous system. However, applying ECM technology to CNS injuries has been limited and virtually unstudied in the visual system. Here we discuss advances in deriving fetal CNS-specific ECMs, like fetal porcine brain, retina, and optic nerve, and fetal non-CNS-specific ECMs, like fetal urinary bladder, and the potential for using tissue-specific ECMs to treat retinal or optic nerve injuries in two platforms. The first platform is an ECM hydrogel that can be administered as a retrobulbar, periocular, or even intraocular injection. The second platform is an ECM hydrogel and polymer "biohybrid" sheet that can be readily shaped and wrapped around a nerve. Both platforms can be tuned mechanically and biochemically to deliver factors like neurotrophins, immunotherapeutics, or stem cells. Since clinical CNS therapies often use general anti-inflammatory agents, which can reduce tissue-destructive inflammation but also suppress tissue-reparative immune system functions, tissue-specific, ECM-based devices may fill an important need by providing naturally derived, biocompatible, and highly translatable platforms that can modulate the innate immune response to promote a positive functional outcome.
Collapse
|
33
|
McMahill BG, Spriet M, Sisó S, Manzer MD, Mitchell G, McGee J, Garcia TC, Borjesson DL, Sieber-Blum M, Nolta JA, Sturges BK. Feasibility Study of Canine Epidermal Neural Crest Stem Cell Transplantation in the Spinal Cords of Dogs. Stem Cells Transl Med 2015; 4:1173-86. [PMID: 26273065 DOI: 10.5966/sctm.2015-0018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/17/2015] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED This pilot feasibility study aimed to determine the outcome of canine epidermal neural crest stem cell (cEPI-NCSC) grafts in the normal spinal cords of healthy bred-for-research dogs. This included developing novel protocols for (a) the ex vivo expansion of cEPI-NCSCs, (b) the delivery of cEPI-NCSCs into the spinal cord, and (c) the labeling of the cells and subsequent tracing of the graft in the live animal by magnetic resonance imaging. A total of four million cEPI-NCSCs were injected into the spinal cord divided in two locations. Differences in locomotion at baseline and post-treatment were evaluated by gait analysis and compared with neurological outcome and behavioral exams. Histopathological analyses of the spinal cords and cEPI-NCSC grafts were performed at 3 weeks post-transplantation. Neurological and gait parameters were minimally affected by the stem cell injection. cEPI-NCSCs survived in the canine spinal cord for the entire period of investigation and did not migrate or proliferate. Subsets of cEPI-NCSCs expressed the neural crest stem cell marker Sox10. There was no detectable expression of markers for glial cells or neurons. The tissue reaction to the cell graft was predominantly vascular in addition to a degree of reactive astrogliosis and microglial activation. In the present study, we demonstrated that cEPI-NCSC grafts survive in the spinal cords of healthy dogs without major adverse effects. They persist locally in the normal spinal cord, may promote angiogenesis and tissue remodeling, and elicit a tissue response that may be beneficial in patients with spinal cord injury. SIGNIFICANCE It has been established that mouse and human epidermal neural crest stem cells are somatic multipotent stem cells with proved innovative potential in a mouse model of spinal cord injury (SCI) offering promise of a valid treatment for SCI. Traumatic SCI is a common neurological problem in dogs with marked similarities, clinically and pathologically, to the syndrome in people. For this reason, dogs provide a readily accessible, clinically realistic, spontaneous model for evaluation of epidermal neural crest stem cells therapeutic intervention. The results of this study are expected to give the baseline data for a future clinical trial in dogs with traumatic SCI.
Collapse
Affiliation(s)
- Barbara G McMahill
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Mathieu Spriet
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Sílvia Sisó
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Michael D Manzer
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Gaela Mitchell
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jeannine McGee
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Tanya C Garcia
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Dori L Borjesson
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Maya Sieber-Blum
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jan A Nolta
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Beverly K Sturges
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
34
|
Vanoli F, Rinchetti P, Porro F, Parente V, Corti S. Clinical and molecular features and therapeutic perspectives of spinal muscular atrophy with respiratory distress type 1. J Cell Mol Med 2015; 19:2058-66. [PMID: 26095024 PMCID: PMC4568910 DOI: 10.1111/jcmm.12606] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 04/02/2015] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy with respiratory distress (SMARD1) is an autosomal recessive neuromuscular disease caused by mutations in the IGHMBP2 gene, encoding the immunoglobulin μ-binding protein 2, leading to motor neuron degeneration. It is a rare and fatal disease with an early onset in infancy in the majority of the cases. The main clinical features are muscular atrophy and diaphragmatic palsy, which requires prompt and permanent supportive ventilation. The human disease is recapitulated in the neuromuscular degeneration (nmd) mouse. No effective treatment is available yet, but novel therapeutical approaches tested on the nmd mouse, such as the use of neurotrophic factors and stem cell therapy, have shown positive effects. Gene therapy demonstrated effectiveness in SMA, being now at the stage of clinical trial in patients and therefore representing a possible treatment for SMARD1 as well. The significant advancement in understanding of both SMARD1 clinical spectrum and molecular mechanisms makes ground for a rapid translation of pre-clinical therapeutic strategies in humans.
Collapse
Affiliation(s)
- Fiammetta Vanoli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Rinchetti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Porro
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Parente
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
35
|
Abstract
Human T-lymphotropic virus 1 (HTLV-1)-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a progressive disease of the CNS that causes weakness or paralysis of the legs, lower back pain and urinary symptoms. HAM/TSP was first described in Jamaica in the nineteenth century, but the aetiology of the condition, infection with the retrovirus HTLV-1, was only identified in the 1980s. HAM/TSP causes chronic disability and, accordingly, imposes a substantial health burden in areas where HTLV-1 infection is endemic. Since the discovery of the cause of HAM/TSP, considerable advances have been made in the understanding of the virology, immunology, cell biology and pathology of HTLV-1 infection and its associated diseases. However, progress has been limited by the lack of accurate animal models of the disease. Moreover, the treatment of HAM/TSP remains highly unsatisfactory: antiretroviral drugs have little impact on the infection and, although potential disease-modifying therapies are widely used, their value is unproved. At present, clinical management is focused on symptomatic treatment and counselling. Here, we summarize current knowledge on the epidemiology, pathogenesis and treatment of HAM/TSP and identify areas in which further research is needed. For an illustrated summary of this Primer, visit: http://go.nature.com/tjZCFM.
Collapse
|
36
|
Burns TC, Verfaillie CM. From mice to mind: Strategies and progress in translating neuroregeneration. Eur J Pharmacol 2015; 759:90-100. [PMID: 25814255 DOI: 10.1016/j.ejphar.2015.03.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/18/2015] [Accepted: 03/12/2015] [Indexed: 12/20/2022]
Abstract
Decisions about what experimental therapies are advanced to clinical trials are based almost exclusively on findings in preclinical animal studies. Over the past 30 years, animal models have forecast the success of hundreds of neuroprotective pharmacological therapies for stroke, Alzheimer׳s disease, spinal cord injury, traumatic brain injury and amyotrophic lateral sclerosis. Yet almost without exception, all have failed. Rapid advances in stem cell technologies have raised new hopes that these neurological diseases may one day be treatable. Still, how can neuroregenerative therapies be translated into clinical realities if available animal models are such poor surrogates of human disease? To address this question we discuss human and rodent neurogenesis, evaluate mechanisms of action for cellular therapies and describe progress in translating neuroregeneration to date. We conclude that not only are appropriate animal models critical to the development of safe and effective therapies, but that the multiple mechanisms of stem cell-mediated therapies may be particularly well suited to the mechanistically diverse nature of central nervous system diseases in mice and man.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA.
| | | |
Collapse
|
37
|
Nezakati T, Cousins BG, Seifalian AM. Toxicology of chemically modified graphene-based materials for medical application. Arch Toxicol 2014; 88:1987-2012. [PMID: 25234085 PMCID: PMC4201927 DOI: 10.1007/s00204-014-1361-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 09/03/2014] [Indexed: 12/12/2022]
Abstract
This review article aims to provide an overview of chemically modified graphene, and graphene oxide (GO), and their impact on toxicology when present in biological systems. Graphene is one of the most promising nanomaterials due to unique physicochemical properties including enhanced optical, thermal, and electrically conductive behavior in addition to mechanical strength and high surface-to-volume ratio. Graphene-based nanomaterials have received much attention over the last 5 years in the biomedical field ranging from their use as polymeric conduits for nerve regeneration, carriers for targeted drug delivery and in the treatment of cancer via photo-thermal therapy. Both in vitro and in vivo biological studies of graphene-based nanomaterials help understand their relative toxicity and biocompatibility when used for biomedical applications. Several studies investigating important material properties such as surface charge, concentration, shape, size, structural defects, and chemical functional groups relate to their safety profile and influence cyto- and geno-toxicology. In this review, we highlight the most recent studies of graphene-based nanomaterials and outline their unique properties, which determine their interactions under a range of environmental conditions. The advent of graphene technology has led to many promising new opportunities for future applications in the field of electronics, biotechnology, and nanomedicine to aid in the diagnosis and treatment of a variety of debilitating diseases.
Collapse
Affiliation(s)
- Toktam Nezakati
- UCL Centre for Nanotechnology and Regeneration Medicine, Division of Surgery and Interventional Science, University College London, London, UK
| | - Brian G. Cousins
- UCL Centre for Nanotechnology and Regeneration Medicine, Division of Surgery and Interventional Science, University College London, London, UK
| | - Alexander M. Seifalian
- UCL Centre for Nanotechnology and Regeneration Medicine, Division of Surgery and Interventional Science, University College London, London, UK
- Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
38
|
Simone C, Nizzardo M, Rizzo F, Ruggieri M, Riboldi G, Salani S, Bucchia M, Bresolin N, Comi GP, Corti S. iPSC-Derived neural stem cells act via kinase inhibition to exert neuroprotective effects in spinal muscular atrophy with respiratory distress type 1. Stem Cell Reports 2014; 3:297-311. [PMID: 25254343 PMCID: PMC4176534 DOI: 10.1016/j.stemcr.2014.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 12/01/2022] Open
Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a motor neuron disease caused by mutations in the IGHMBP2 gene, without a cure. Here, we demonstrate that neural stem cells (NSCs) from human-induced pluripotent stem cells (iPSCs) have therapeutic potential in the context of SMARD1. We show that upon transplantation NSCs can appropriately engraft and differentiate in the spinal cord of SMARD1 animals, ameliorating their phenotype, by protecting their endogenous motor neurons. To evaluate the effect of NSCs in the context of human disease, we generated human SMARD1-iPSCs motor neurons that had a significantly reduced survival and axon length. Notably, the coculture with NSCs ameliorate these disease features, an effect attributable to the production of neurotrophic factors and their dual inhibition of GSK-3 and HGK kinases. Our data support the role of iPSC as SMARD1 disease model and their translational potential for therapies in motor neuron disorders.
Collapse
Affiliation(s)
- Chiara Simone
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Monica Nizzardo
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Federica Rizzo
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Margherita Ruggieri
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Giulietta Riboldi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Sabrina Salani
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Monica Bucchia
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Nereo Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Giacomo P Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
39
|
Dolezalova D, Hruska-Plochan M, Bjarkam CR, Sørensen JCH, Cunningham M, Weingarten D, Ciacci JD, Juhas S, Juhasova J, Motlik J, Hefferan MP, Hazel T, Johe K, Carromeu C, Muotri A, Bui J, Strnadel J, Marsala M. Pig models of neurodegenerative disorders: Utilization in cell replacement-based preclinical safety and efficacy studies. J Comp Neurol 2014; 522:2784-801. [DOI: 10.1002/cne.23575] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Dasa Dolezalova
- Department of Anesthesiology; University of California; San Diego La Jolla CA USA
| | | | - Carsten R. Bjarkam
- Department of Neurosurgery; Aalborg University Hospital; Aalborg Denmark
- Department of Biomedicine; Institute of Anatomy, University of Aarhus; Aarhus Denmark
| | | | - Miles Cunningham
- MRC 312, McLean Hospital, Harvard Medical School; Belmont MA 02478 USA
| | - David Weingarten
- UCSD Division of Neurosurgery; University of California; San Diego CA USA
| | - Joseph D. Ciacci
- UCSD Division of Neurosurgery; University of California; San Diego CA USA
| | - Stefan Juhas
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences; 277 21 Libechov Czech Republic
| | - Jana Juhasova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences; 277 21 Libechov Czech Republic
| | - Jan Motlik
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences; 277 21 Libechov Czech Republic
| | | | | | | | - Cassiano Carromeu
- Department of Cellular and Molecular Medicine; University of California; San Diego CA USA
| | - Alysson Muotri
- Department of Cellular and Molecular Medicine; University of California; San Diego CA USA
| | - Jack Bui
- Department of Pathology; University of California; San Diego CA USA
| | - Jan Strnadel
- Department of Pathology; University of California; San Diego CA USA
| | - Martin Marsala
- Department of Anesthesiology; University of California; San Diego La Jolla CA USA
- Institute of Neurobiology, Slovak Academy of Sciences; Kosice Slovakia
| |
Collapse
|
40
|
Gericota B, Anderson JS, Mitchell G, Borjesson DL, Sturges BK, Nolta JA, Sieber-Blum M. Canine epidermal neural crest stem cells: characterization and potential as therapy candidate for a large animal model of spinal cord injury. Stem Cells Transl Med 2014; 3:334-45. [PMID: 24443004 PMCID: PMC3952930 DOI: 10.5966/sctm.2013-0129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/23/2013] [Indexed: 02/07/2023] Open
Abstract
The discovery of multipotent neural crest-derived stem cells, named epidermal neural crest stem cells (EPI-NCSC), that persist postnatally in an easy-to-access location-the bulge of hair follicles-opens a spectrum of novel opportunities for patient-specific therapies. We present a detailed characterization of canine EPI-NCSC (cEPI-NCSC) from multiple dog breeds and protocols for their isolation and ex vivo expansion. Furthermore, we provide novel tools for research in canines, which currently are still scarce. In analogy to human and mouse EPI-NCSC, the neural crest origin of cEPI-NCSC is shown by their expression of the neural crest stem cell molecular signature and other neural crest-characteristic genes. Similar to human EPI-NCSC, cEPI-NCSC also expressed pluripotency genes. We demonstrated that cEPI-NCSC can generate all major neural crest derivatives. In vitro clonal analyses established multipotency and self-renewal ability of cEPI-NCSC, establishing cEPI-NCSC as multipotent somatic stem cells. A critical analysis of the literature on canine spinal cord injury (SCI) showed the need for novel treatments and suggested that cEPI-NCSC represent viable candidates for cell-based therapies in dog SCI, particularly for chondrodystrophic dogs. This notion is supported by the close ontological relationship between neural crest stem cells and spinal cord stem cells. Thus, cEPI-NCSC promise to offer not only a potential treatment for canines but also an attractive and realistic large animal model for human SCI. Taken together, we provide the groundwork for the development of a novel cell-based therapy for a condition with extremely poor prognosis and no available effective treatment.
Collapse
|
41
|
Oliveri RS, Bello S, Biering-Sørensen F. Mesenchymal stem cells improve locomotor recovery in traumatic spinal cord injury: systematic review with meta-analyses of rat models. Neurobiol Dis 2013; 62:338-53. [PMID: 24148857 DOI: 10.1016/j.nbd.2013.10.014] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 09/13/2013] [Accepted: 10/10/2013] [Indexed: 12/13/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating event with huge personal and societal costs. A limited number of treatments exist to ameliorate the progressive secondary damage that rapidly follows the primary mechanical impact. Mesenchymal stem or stromal cells (MSCs) have anti-inflammatory and neuroprotective effects and may thus reduce secondary damage after administration. We performed a systematic review with quantitative syntheses to assess the evidence of MSCs versus controls for locomotor recovery in rat models of traumatic SCI, and identified 83 eligible controlled studies comprising a total of 1,568 rats. Between-study heterogeneity was large. Fifty-three studies (64%) were reported as randomised, but only four reported adequate methodologies for randomisation. Forty-eight studies (58%) reported the use of a blinded outcome assessment. A random-effects meta-analysis yielded a difference in behavioural Basso-Beattie-Bresnahan (BBB) locomotor score means of 3.9 (95% confidence interval [CI] 3.2 to 4.7; P<0.001) in favour of MSCs. Trial sequential analysis confirmed the findings of the meta-analyses with the upper monitoring boundary for benefit being crossed by the cumulative Z-curve before reaching the diversity-adjusted required information size. Only time from intervention to last follow-up remained statistically significant after adjustment using multivariate random-effects meta-regression modelling. Lack of other demonstrable explanatory variables could be due to insufficient meta-analytic study power. MSCs would seem to demonstrate a substantial beneficial effect on locomotor recovery in a widely-used animal model of traumatic SCI. However, the animal results should be interpreted with caution concerning the internal and external validity of the studies in relation to the design of future clinical trials.
Collapse
Affiliation(s)
- Roberto S Oliveri
- Cell Therapy Facility, The Blood Bank, Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
| | - Segun Bello
- The Nordic Cochrane Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Fin Biering-Sørensen
- Department of Spinal Cord Injuries, Copenhagen University Hospital Rigshospitalet and Glostrup Hospital, Copenhagen, Denmark
| |
Collapse
|
42
|
3D graphene oxide-encapsulated gold nanoparticles to detect neural stem cell differentiation. Biomaterials 2013; 34:8660-70. [PMID: 23937915 DOI: 10.1016/j.biomaterials.2013.07.101] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 07/28/2013] [Indexed: 11/21/2022]
Abstract
Monitoring of stem cell differentiation and pluripotency is an important step for the practical use of stem cells in the field of regenerative medicine. Hence, a new non-destructive detection tool capable of in situ monitoring of stem cell differentiation is highly needed. In this study, we report a 3D graphene oxide-encapsulated gold nanoparticle that is very effective for the detection of the differentiation potential of neural stem cells (NSCs) based on surface-enhanced Raman spectroscopy (SERS). A new material, 3D GO-encapsulated gold nanoparticle, is developed to induce the double enhancement effect of graphene oxide and gold nanoparticle on SERS signals which is only effective for undifferentiated NSCs. The Raman peaks achieved from undifferentiated NSCs on the graphene oxide (GO)-encapsulated gold nanoparticles were 3.5 times higher than peaks obtained from normal metal structures and were clearly distinguishable from those of differentiated cells. The number of CC bonds and the Raman intensity at 1656 cm(-1) was found to show a positive correlation, which matches the differentiation state of the NSCs. Moreover, the substrate composed of 3D GO-encapsulated gold nanoparticles was also effective at distinguishing the differentiation state of single NSC by using electrochemical and electrical techniques. Hence, the proposed technique can be used as a powerful non-destructive in situ monitoring tool for the identification of the differentiation potential of various kinds of stem cells (mesenchymal, hematopoietic, and neural stem cells).
Collapse
|
43
|
Cocks G, Romanyuk N, Amemori T, Jendelova P, Forostyak O, Jeffries AR, Perfect L, Thuret S, Dayanithi G, Sykova E, Price J. Conditionally immortalized stem cell lines from human spinal cord retain regional identity and generate functional V2a interneurons and motorneurons. Stem Cell Res Ther 2013; 4:69. [PMID: 23759128 PMCID: PMC3706922 DOI: 10.1186/scrt220] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 06/03/2013] [Indexed: 01/24/2023] Open
Abstract
Introduction The use of immortalized neural stem cells either as models of neural development in vitro or as cellular therapies in central nervous system (CNS) disorders has been controversial. This controversy has centered on the capacity of immortalized cells to retain characteristic features of the progenitor cells resident in the tissue of origin from which they were derived, and the potential for tumorogenicity as a result of immortalization. Here, we report the generation of conditionally immortalized neural stem cell lines from human fetal spinal cord tissue, which addresses these issues. Methods Clonal neural stem cell lines were derived from 10-week-old human fetal spinal cord and conditionally immortalized with an inducible form of cMyc. The derived lines were karyotyped, transcriptionally profiled by microarray, and assessed against a panel of spinal cord progenitor markers with immunocytochemistry. In addition, the lines were differentiated and assessed for the presence of neuronal fate markers and functional calcium channels. Finally, a clonal line expressing eGFP was grafted into lesioned rat spinal cord and assessed for survival, differentiation characteristics, and tumorogenicity. Results We demonstrate that these clonal lines (a) retain a clear transcriptional signature of ventral spinal cord progenitors and a normal karyotype after extensive propagation in vitro, (b) differentiate into relevant ventral neuronal subtypes with functional T-, L-, N-, and P/Q-type Ca2+ channels and spontaneous calcium oscillations, and (c) stably engraft into lesioned rat spinal cord without tumorogenicity. Conclusions We propose that these cells represent a useful tool both for the in vitro study of differentiation into ventral spinal cord neuronal subtypes, and for examining the potential of conditionally immortalized neural stem cells to facilitate functional recovery after spinal cord injury or disease.
Collapse
|
44
|
Amemori T, Romanyuk N, Jendelova P, Herynek V, Turnovcova K, Prochazka P, Kapcalova M, Cocks G, Price J, Sykova E. Human conditionally immortalized neural stem cells improve locomotor function after spinal cord injury in the rat. Stem Cell Res Ther 2013; 4:68. [PMID: 23759119 PMCID: PMC3706805 DOI: 10.1186/scrt219] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 06/04/2013] [Indexed: 12/28/2022] Open
Abstract
Introduction A growing number of studies have highlighted the potential of stem cell and more-differentiated neural cell transplantation as intriguing therapeutic approaches for neural repair after spinal cord injury (SCI). Methods A conditionally immortalized neural stem cell line derived from human fetal spinal cord tissue (SPC-01) was used to treat a balloon-induced SCI. SPC-01 cells were implanted into the lesion 1 week after SCI. To determine the feasibility of tracking transplanted stem cells, a portion of the SPC-01 cells was labeled with poly-L-lysine-coated superparamagnetic iron-oxide nanoparticles, and the animals grafted with labeled cells underwent magnetic resonance imaging. Functional recovery was evaluated by using the BBB and plantar tests, and lesion morphology, endogenous axonal sprouting and graft survival, and differentiation were analyzed. Quantitative polymerase chain reaction (qPCR) was used to evaluate the effect of transplanted SPC-01 cells on endogenous regenerative processes. Results Transplanted animals displayed significant motor and sensory improvement 2 months after SCI, when the cells robustly survived in the lesion and partially filled the lesion cavity. qPCR revealed the increased expression of rat and human neurotrophin and motor neuron genes. The grafted cells were immunohistologically positive for glial fibrillary acidic protein (GFAP); however, we found 25% of the cells to be positive for Nkx6.1, an early motor neuron marker. Spared white matter and the robust sprouting of growth-associated protein 43 (GAP43)+ axons were found in the host tissue. Four months after SCI, the grafted cells matured into Islet2+ and choline acetyltransferase (ChAT)+ neurons, and the graft was grown through with endogenous neurons. Grafted cells labeled with poly-L-lysine-coated superparamagnetic nanoparticles before transplantation were detected in the lesion on T2-weighted images as hypointense spots that correlated with histologic staining for iron and the human mitochondrial marker MTCO2. Conclusions The transplantation of SPC-01 cells produced significant early functional improvement after SCI, suggesting an early neurotrophic action associated with long-term restoration of the host tissue, making the cells a promising candidate for future cell therapy in patients with SCI.
Collapse
|
45
|
van Gorp S, Leerink M, Kakinohana O, Platoshyn O, Santucci C, Galik J, Joosten EA, Hruska-Plochan M, Goldberg D, Marsala S, Johe K, Ciacci JD, Marsala M. Amelioration of motor/sensory dysfunction and spasticity in a rat model of acute lumbar spinal cord injury by human neural stem cell transplantation. Stem Cell Res Ther 2013; 4:57. [PMID: 23710605 PMCID: PMC3706882 DOI: 10.1186/scrt209] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/25/2013] [Indexed: 12/15/2022] Open
Abstract
Introduction Intraspinal grafting of human neural stem cells represents a promising approach to promote recovery of function after spinal trauma. Such a treatment may serve to: I) provide trophic support to improve survival of host neurons; II) improve the structural integrity of the spinal parenchyma by reducing syringomyelia and scarring in trauma-injured regions; and III) provide neuronal populations to potentially form relays with host axons, segmental interneurons, and/or α-motoneurons. Here we characterized the effect of intraspinal grafting of clinical grade human fetal spinal cord-derived neural stem cells (HSSC) on the recovery of neurological function in a rat model of acute lumbar (L3) compression injury. Methods Three-month-old female Sprague–Dawley rats received L3 spinal compression injury. Three days post-injury, animals were randomized and received intraspinal injections of either HSSC, media-only, or no injections. All animals were immunosuppressed with tacrolimus, mycophenolate mofetil, and methylprednisolone acetate from the day of cell grafting and survived for eight weeks. Motor and sensory dysfunction were periodically assessed using open field locomotion scoring, thermal/tactile pain/escape thresholds and myogenic motor evoked potentials. The presence of spasticity was measured by gastrocnemius muscle resistance and electromyography response during computer-controlled ankle rotation. At the end-point, gait (CatWalk), ladder climbing, and single frame analyses were also assessed. Syrinx size, spinal cord dimensions, and extent of scarring were measured by magnetic resonance imaging. Differentiation and integration of grafted cells in the host tissue were validated with immunofluorescence staining using human-specific antibodies. Results Intraspinal grafting of HSSC led to a progressive and significant improvement in lower extremity paw placement, amelioration of spasticity, and normalization in thermal and tactile pain/escape thresholds at eight weeks post-grafting. No significant differences were detected in other CatWalk parameters, motor evoked potentials, open field locomotor (Basso, Beattie, and Bresnahan locomotion score (BBB)) score or ladder climbing test. Magnetic resonance imaging volume reconstruction and immunofluorescence analysis of grafted cell survival showed near complete injury-cavity-filling by grafted cells and development of putative GABA-ergic synapses between grafted and host neurons. Conclusions Peri-acute intraspinal grafting of HSSC can represent an effective therapy which ameliorates motor and sensory deficits after traumatic spinal cord injury.
Collapse
|
46
|
Fan J, Tan Y, Jie L, Wu X, Yu R, Zhang M. Biological activity and magnetic resonance imaging of superparamagnetic iron oxide nanoparticles-labeled adipose-derived stem cells. Stem Cell Res Ther 2013; 4:44. [PMID: 23618360 PMCID: PMC3706947 DOI: 10.1186/scrt191] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 04/12/2013] [Indexed: 12/11/2022] Open
Abstract
Introduction No comparative study of adipose-derived stem cells (ADSCs) and bone marrow mesenchymal stem cells (BMSCs) by using superparamagnetic iron oxide nanoparticles (SPIOs)-labeling and magnetic resonance imaging (MRI) has been performed. Methods We studied the biological activity and MRI of ADSCs by labeling them with SPIOs and comparing them with BMSCs. After incubating the cells in culture medium with different levels of SPIOs (control group: 0 μg/ml; Groups 1 to 3: 25, 50, and 100 μg/ml) for 24 hours, we compared ADSCs with BMSCs in terms of intracellular iron content, labeling efficiency, and cell viability. Stem cells in the culture medium containing 50 μg/ml SPIOs were induced into osteoblasts and fat cells. Adipogenic and osteogenic differentiation potentials were compared. R2* values of MRI in vitro were compared. Results The results showed that labeling efficiency was highest in Group 2. Intracellular iron content and R2* values increased with increasing concentrations of SPIOs, whereas cell viability decreased with increasing concentrations of SPIOs, and adipogenic and osteogenic differentiation potentials decreased. However, we found no significant difference between the two kinds of cells for any of these indexes. Conclusions ADSCs can be labeled and traced as easily as BMSCs in vitro. Given their abundance and higher proliferative capacity, as was previously shown, ADSCs may be better suited to stem cell therapy than are BMSCs.
Collapse
|
47
|
Simulated microgravity facilitates cell migration and neuroprotection after bone marrow stromal cell transplantation in spinal cord injury. Stem Cell Res Ther 2013; 4:35. [PMID: 23548163 PMCID: PMC3706926 DOI: 10.1186/scrt184] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/08/2013] [Indexed: 01/01/2023] Open
Abstract
Introduction Recently, cell-based therapy has gained significant attention for the treatment of central nervous system diseases. Although bone marrow stromal cells (BMSCs) are considered to have good engraftment potential, challenges due to in vitro culturing, such as a decline in their functional potency, have been reported. Here, we investigated the efficacy of rat BMSCs (rBMSCs) cultured under simulated microgravity conditions, for transplantation into a rat model of spinal cord injury (SCI). Methods rBMSCs were cultured under two different conditions: standard gravity (1G) and simulated microgravity attained by using the 3D-clinostat. After 7 days of culture, the rBMSCs were analyzed morphologically, with RT-PCR and immunostaining, and were used for grafting. Adult rats were used for constructing SCI models by using a weight-dropping method and were grouped into three experimental groups for comparison. rBMSCs cultured under 1 g and simulated microgravity were transplanted intravenously immediately after SCI. We evaluated the hindlimb functional improvement for 3 weeks. Tissue repair after SCI was examined by calculating the cavity area ratio and immunohistochemistry. Results rBMSCs cultured under simulated microgravity expressed Oct-4 and CXCR4, in contrast to those cultured under 1 g conditions. Therefore, rBMSCs cultured under simulated microgravity were considered to be in an undifferentiated state and thus to possess high migration ability. After transplantation, grafted rBMSCs cultured under microgravity exhibited greater survival at the periphery of the lesion, and the motor functions of the rats that received these grafts improved significantly compared with the rats that received rBMSCs cultured in 1 g. In addition, rBMSCs cultured under microgravity were thought to have greater trophic effects on reestablishment and survival of host spinal neural tissues because cavity formations were reduced, and apoptosis-inhibiting factor expression was high at the periphery of the SCI lesion. Conclusions Here we show that transplantation of rBMSCs cultured under simulated microgravity facilitates functional recovery from SCI rather than those cultured under 1 g conditions.
Collapse
|
48
|
Varma AK, Das A, Wallace G, Barry J, Vertegel AA, Ray SK, Banik NL. Spinal cord injury: a review of current therapy, future treatments, and basic science frontiers. Neurochem Res 2013; 38:895-905. [PMID: 23462880 DOI: 10.1007/s11064-013-0991-6] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/17/2013] [Accepted: 01/29/2013] [Indexed: 12/12/2022]
Abstract
The incidence of acute and chronic spinal cord injury (SCI) in the United States is more than 10,000 per year, resulting in 720 cases per million persons enduring permanent disability each year. The economic impact of SCI is estimated to be more than 4 billion dollars annually. Preclinical studies, case reports, and small clinical trials suggest that early treatment may improve neurological recovery. To date, no proven therapeutic modality exists that has demonstrated a positive effect on neurological outcome. Emerging data from recent preclinical and clinical studies offer hope for this devastating condition. This review gives an overview of current basic research and clinical studies for the treatment of SCI.
Collapse
Affiliation(s)
- Abhay K Varma
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Cramer T, Chelli B, Murgia M, Barbalinardo M, Bystrenova E, de Leeuw DM, Biscarini F. Organic ultra-thin film transistors with a liquid gate for extracellular stimulation and recording of electric activity of stem cell-derived neuronal networks. Phys Chem Chem Phys 2013; 15:3897-905. [DOI: 10.1039/c3cp44251a] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
50
|
Abstract
The remarkable speed with which the field of stem cell biology has evolved is unprecedented and has already changed the way we do science. In this series of articles we have invited leading experts to present their efforts in moving from the bench to the bedside, with the hope that we can learn from the experiences of the pioneers.
Collapse
|