1
|
Kwokdinata C, Ramanujam V, Chen J, de Oliveira PN, Nai MH, Chooi WH, Lim CT, Ng SY, David L, Chew SY. Encapsulation of Human Spinal Cord Progenitor Cells in Hyaluronan-Gelatin Hydrogel for Spinal Cord Injury Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50679-50692. [PMID: 37751213 DOI: 10.1021/acsami.3c07419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Transplanting human induced pluripotent stem cells (iPSCs)-derived spinal cord progenitor cells (SCPCs) is a promising approach to treat spinal cord injuries. However, stem cell therapies face challenges in cell survival, cell localization to the targeted site, and the control of cell differentiation. Here, we encapsulated SCPCs in thiol-modified hyaluronan-gelatin hydrogels and optimized scaffold mechanical properties and cell encapsulation density to promote cell viability and neuronal differentiation in vitro and in vivo. Different compositions of hyaluronan-gelatin hydrogels formulated by varying concentrations of poly(ethylene glycol) diacrylate were mechanically characterized by using atomic force microscopy. In vitro SCPC encapsulation study showed higher cell viability and proliferation with lower substrate Young's modulus (200 Pa vs 580 Pa) and cell density. Moreover, the soft hydrogels facilitated a higher degree of neuronal differentiation with extended filament structures in contrast to clumped cellular morphologies obtained in stiff hydrogels (p < 0.01). When transplanted in vivo, the optimized SCPC-encapsulated hydrogels resulted in higher cell survival and localization at the transplanted region as compared to cell delivery without hydrogel encapsulation at 2 weeks postimplantation within the rat spinal cord (p < 0.01). Notably, immunostaining demonstrated that the hydrogel-encapsulated SCPCs differentiated along the neuronal and oligodendroglial lineages in vivo. The lack of pluripotency and proliferation also supported the safety of the SCPC transplantation approach. Overall, the injectable hyaluronan-gelatin hydrogel shows promise in supporting the survival and neural differentiation of human SCPCs after transplantation into the spinal cord.
Collapse
Affiliation(s)
- Christy Kwokdinata
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | - Vaibavi Ramanujam
- CNRS@CREATE, Create Tower #08-01, 1 Create Way, Singapore 138602, Singapore
| | - Jiahui Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | | | - Mui Hoon Nai
- Department of Biomedical Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Wai Hon Chooi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117576, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Shi Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Laurent David
- CNRS@CREATE, Create Tower #08-01, 1 Create Way, Singapore 138602, Singapore
- Ingénierie des Matériaux Polymères IMP UMR 5223, CNRS, Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, Université de Lyon, Villeurbanne F69622, France
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
2
|
Papa S, Pizzetti F, Perale G, Veglianese P, Rossi F. Regenerative medicine for spinal cord injury: focus on stem cells and biomaterials. Expert Opin Biol Ther 2020; 20:1203-1213. [PMID: 32421405 DOI: 10.1080/14712598.2020.1770725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a dramatic medical pathology consequence of a trauma (primary injury). However, most of the post-traumatic degeneration of the tissue is caused by the so-called secondary injury, which is known to be a multifactorial process. This, indeed, includes a wide spectrum of events: blood-brain barrier dysfunction, local inflammation, neuronal death, demyelination and disconnection of nerve pathways. AREAS COVERED Cell therapy represents a promising cure to target diseases and disorders at the cellular level, by restoring cell population or using cells as carriers of therapeutic cargo. In particular, regenerative medicine with stem cells represents the most appealing category to be used, thanks to their peculiar features. EXPERT OPINION Many preclinical research studies demonstrated that cell treatment can improve animal sensory/motor functions and so demonstrated to be very promising for clinical trials. In particular, recent advances have led to the development of biomaterials aiming to promote in situ cell delivery. This review digs into this topic discussing the possibility of cell treatment to improve medical chances in SCI repair.
Collapse
Affiliation(s)
- Simonetta Papa
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy
| | - Fabio Pizzetti
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy.,Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Milan, Italy
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI) , Lugano, Switzerland.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology , Vienna, Austria
| | - Pietro Veglianese
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Milan, Italy
| |
Collapse
|
3
|
Mahmoodi N, Ai J, Ebrahimi‐Barough S, Hassannejad Z, Hasanzadeh E, Basiri A, Vaccaro AR, Rahimi‐Movaghar V. Microtubule stabilizer epothilone B as a motor neuron differentiation agent for human endometrial stem cells. Cell Biol Int 2020; 44:1168-1183. [DOI: 10.1002/cbin.11315] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/02/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Narges Mahmoodi
- Sina Trauma and Surgery Research Center, Sina HospitalTehran University of Medical Sciences Hasan‐Abad Square, Imam Khomeini Ave. Tehran 11365‐3876 Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Somayeh Ebrahimi‐Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of ExcellenceTehran University of Medical Sciences No. 62, Dr. Gharibs Street, Keshavarz Boulevard Tehran 1419733151 Iran
| | - Elham Hasanzadeh
- Department of Tissue Engineering, School of Advanced Technologies in MedicineMazandaran University of Medical Sciences Next to Tooba Medical Building, Khazar Boulevard Sari 48471‐91971 Iran
| | - Arefeh Basiri
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Alexander R. Vaccaro
- Department of Orthopedic Surgery, Rothman InstituteThomas Jefferson University 1925 Chestnut Street, 5th Floor Philadelphia Pennsylvania 19107 USA
| | - Vafa Rahimi‐Movaghar
- Sina Trauma and Surgery Research Center, Sina HospitalTehran University of Medical Sciences Hasan‐Abad Square, Imam Khomeini Ave. Tehran 11365‐3876 Iran
| |
Collapse
|
4
|
New PW. A Narrative Review of Pediatric Nontraumatic Spinal Cord Dysfunction. Top Spinal Cord Inj Rehabil 2019; 25:112-120. [PMID: 31068743 DOI: 10.1310/sci2502-112] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This article provides a narrative review of seven key issues relevant to pediatric onset of spinal cord damage not due to trauma, or spinal cord dysfunction (SCDys). The first topic discussed is terminology issues. There is no internationally accepted term for spinal cord damage not due to trauma. The implications of this terminology issue and an approach to addressing this are discussed. Second, a brief history of SCDys is presented, focusing on conditions relevant to pediatrics. Third, the classification of SCDys is outlined, based on the International Spinal Cord Injury Data Sets for Non-Traumatic Spinal Cord Injury. Following this, a summary is given of the epidemiology of pediatric SCDys, primarily focused on the incidence, prevalence, and etiology, with a comparison to traumatic spinal cord injury. Next, important clinical rehabilitation principles unique to children with SCDys are highlighted. Then important prevention opportunities for SCDys in children are discussed. Finally, trends, challenges, and opportunities regarding research in SCDys are mentioned.
Collapse
Affiliation(s)
- Peter Wayne New
- Spinal Rehabilitation Service, Department of Rehabilitation, Caulfield Hospital, Alfred Health, Victoria, Australia.,Epworth-Monash Rehabilitation Medicine Unit, Monash University, Melbourne, Victoria, Australia.,Department of Epidemiology and Preventive Medicine, School of Public Health & Preventive Medicine, Monash University, Victoria, Australia
| |
Collapse
|
5
|
Effects of Different Doses of Mesenchymal Stem Cells on Functional Recovery After Compressive Spinal-Cord Injury in Mice. Neuroscience 2019; 400:17-32. [DOI: 10.1016/j.neuroscience.2018.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 02/07/2023]
|
6
|
Aging alters glucose uptake in the naïve and injured rodent spinal cord. Neurosci Lett 2018; 690:23-28. [PMID: 30296507 DOI: 10.1016/j.neulet.2018.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/29/2018] [Accepted: 10/03/2018] [Indexed: 11/20/2022]
Abstract
Aging results in increased activation of inflammatory glial cells and decreased neuronal viability following spinal cord injury (SCI). Metabolism and transport of glucose is also decreased with age, although the influence of age on glucose transporter (GLUT) expression or glucose uptake in SCI is currently unknown. We therefore performed [18F]Fluorodeoxyglucose (FDG) PET imaging of young (3 month) and middle-aged (12 month) rats. Glucose uptake in middle-aged rats was decreased compared to young rats at baseline, followed by increased uptake 14 days post contusion SCI. qRT-PCR and protein analysis revealed an association between 14 day glucose uptake and 14 day post-injury inflammation. Further, gene expression analysis of neuron-specific GLUT3 and non-specific GLUT4 (present on glial cells) revealed an inverse relationship between GLUT3/4 gene expression and glucose uptake patterns. Protein expression revealed increased GLUT3 in 3 month rats only, consistent with age related decreases in glucose uptake, and increased GLUT4 in 12 month rats only, consistent with age related increases in inflammatory activity and glucose uptake. Inconsistencies between gene and protein suggest an influence of age-related impairment of translation and/or protein degradation. Overall, our findings show that age alters glucose uptake and GLUT3/4 expression profiles before and after SCI, which may be dependent on level of inflammatory response, and may suggest a therapeutic avenue in addressing glucose uptake in the aging population.
Collapse
|
7
|
New PW, Guilcher SJT, Jaglal SB, Biering-Sørensen F, Noonan VK, Ho C. Trends, Challenges, and Opportunities Regarding Research in Non-traumatic Spinal Cord Dysfunction. Top Spinal Cord Inj Rehabil 2018; 23:313-323. [PMID: 29339907 DOI: 10.1310/sci2304-313] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Spinal cord dysfunction (SCDys) is caused by heterogeneous health conditions, and the incidence is increasing. Despite the growing interest in rehabilitation research for SCDys, research into SCDys faces many challenges. Objective: The objective of this project was to perform a clinical review of changes in SCDys research over the last 4 decades; identify challenges to conducting research in SCDys; and propose opportunities for improving research in SCDys. Methods: A triangulation approach was used for obtaining evidence: literature search (January 2017) using MEDLINE and Embase databases for publications in English (1974-2016) regarding SCDys; workshop discussions at the International Spinal Cord Society annual meeting, September 16, 2016, Vienna, Austria; and our collective expertise in SCDys clinical rehabilitation research. Results: There has been a substantial increase in publications on SCDys over the 4 decades, from 1,825 in 1974-1983 to 11,887 in the decade 2004-2013, along with an improvement in research methodology. Numerous challenges to research in SCDys rehabilitation were grouped into the following themes: (a) identification of cases; (b) study design and data collection; and (c) funding, preclinical, and international research. Opportunities for addressing these were identified. Conclusions: The increase in scientific publications on SCDys highlights the importance of this heterogeneous group among the research community. The overall lack of good quality epidemiological studies regarding incidence, prevalence, and survival in these patients serves as a benchmark for guiding improvements to inform evidence-based care and policy.
Collapse
Affiliation(s)
- Peter Wayne New
- Spinal Rehabilitation Service, Department of Rehabilitation, Caulfield Hospital, Alfred Health, Caulfield, Victoria, Australia.,Epworth-Monash Rehabilitation Medicine Unit, Southern Medical School, Monash University, Victoria, Australia.,Department of Epidemiology and Preventive Medicine, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Sara J T Guilcher
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Susan B Jaglal
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada.,Department of Physical Therapy, University of Toronto, Toronto. Ontario, Canada.,Toronto Rehabilitation Institute, University Health Network, Ontario, Canada
| | - Fin Biering-Sørensen
- Clinic for Spinal Cord Injuries, Neuroscience Center, Rigshospitalet, University of Copenhagen, Denmark
| | | | - Chester Ho
- Division of Physical Medicine & Rehabilitation, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
8
|
Wood CR, Al Delfi IRT, Innes JF, Myint P, Johnson WEB. Exposing mesenchymal stem cells to chondroitin sulphated proteoglycans reduces their angiogenic and neuro-adhesive paracrine activity. Biochimie 2018; 155:26-36. [PMID: 29680669 DOI: 10.1016/j.biochi.2018.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/13/2018] [Indexed: 01/04/2023]
Abstract
The multifactorial complexity of spinal cord injuries includes the formation of a glial scar, of which chondroitin sulphated proteoglycans (CSPG) are an integral component. Previous studies have shown CSPG to have inhibitory effects on endothelial and neuronal cell growth, highlighting the difficulty of spinal cord regeneration. Mesenchymal stem/stromal cells (MSC) are widely used as a cell therapy, and there is mounting evidence for their angiogenic and neurotrophic paracrine properties. However, in vivo studies have observed poor engraftment and survival of MSC when injected into SCI. Currently, it is not known whether increasing CSPG concentrations seen after SCI may affect MSC; therefore we have investigated the effects of CSPG exposure to MSC in vitro. CSPG-mediated inhibition of MSC adhesion was observed when MSC were cultured on substrates of increasing CSPG concentration, however MSC viability was not affected even up to five days of culture. Culture conditioned medium harvested from these cultures (primed MSC CM) was used as both culture substrata and soluble medium for EA.hy926 endothelial cells and SH-SY5Y neuronal cells. MSC CM was angiogenic, promoting endothelial cell adhesion, proliferation and tubule formation. However, exposing MSC to CSPG reduced the effects of CSPG-primed MSC CM on endothelial cell adhesion and proliferation, but did not reduce MSC-induced endothelial tubule formation. Primed MSC CM also promoted neuronal cell adhesion, which was reduced following exposure to CSPG. There were no marked differences in neurite outgrowth in MSC CM from CSPG primed MSC cultures versus control conditions, although non-primed MSC CM from the same donors was found to significantly enhance neurite outgrowth. Taken together, these studies demonstrate that MSC are resilient to CSPG exposure, but that there is a marked effect of CSPG on their paracrine regenerative activity. The findings increase our understanding of how the wound microenvironment after SCI can mitigate the beneficial effects of MSC transplantation.
Collapse
Affiliation(s)
- Chelsea R Wood
- Biological Sciences, Faculty of Medicine, Dentistry and Life Sciences, University of Chester, Parkgate Road, Chester, CH1 4BJ, United Kingdom.
| | - Ibtesam R T Al Delfi
- Centre for Experimental Medicine, Queen's University, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland, UK.
| | - John F Innes
- Veterinary Tissue Bank Ltd, Brynkinalt Business Centre, Wrexham, LL14 5NS, United Kingdom.
| | - Peter Myint
- Veterinary Tissue Bank Ltd, Brynkinalt Business Centre, Wrexham, LL14 5NS, United Kingdom.
| | - William E B Johnson
- Biological Sciences, Faculty of Medicine, Dentistry and Life Sciences, University of Chester, Parkgate Road, Chester, CH1 4BJ, United Kingdom.
| |
Collapse
|
9
|
von Leden RE, Khayrullina G, Moritz KE, Byrnes KR. Age exacerbates microglial activation, oxidative stress, inflammatory and NOX2 gene expression, and delays functional recovery in a middle-aged rodent model of spinal cord injury. J Neuroinflammation 2017; 14:161. [PMID: 28821269 PMCID: PMC5563003 DOI: 10.1186/s12974-017-0933-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/04/2017] [Indexed: 12/17/2022] Open
Abstract
Background Spinal cord injury (SCI) among people over age 40 has been steadily increasing since the 1980s and is associated with worsened outcome than injuries in young people. Age-related increases in reactive oxygen species (ROS) are suggested to lead to chronic inflammation. The NADPH oxidase 2 (NOX2) enzyme is expressed by microglia and is a primary source of ROS. This study aimed to determine the effect of age on inflammation, oxidative damage, NOX2 gene expression, and functional performance with and without SCI in young adult (3 months) and middle-aged (12 months) male rats. Methods Young adult and middle-aged rats were assessed in two groups—naïve and moderate contusion SCI. Functional recovery was determined by weekly assessment with the Basso, Beattie, and Breshnahan general motor score (analyzed two-way ANOVA) and footprint analysis (analyzed by Chi-square analysis). Tissue was analyzed for markers of oxidative damage (8-OHdG, Oxyblot, and 3-NT), microglial-related inflammation (Iba1), NOX2 component (p47PHOX, p22PHOX, and gp91PHOX), and inflammatory (CD86, CD206, TNFα, and NFκB) gene expression (all analyzed by unpaired Student’s t test). Results In both naïve and injured aged rats, compared to young rats, tissue analysis revealed significant increases in 8-OHdG and Iba1, as well as inflammatory and NOX2 component gene expression. Further, injured aged rats showed greater lesion volume rostral and caudal to the injury epicenter. Finally, injured aged rats showed significantly reduced Basso–Beattie–Bresnahan (BBB) scores and stride length after SCI. Conclusions These results show that middle-aged rats demonstrate increased microglial activation, oxidative stress, and inflammatory gene expression, which may be related to elevated NOX2 expression, and contribute to worsened functional outcome following injury. These findings are essential to elucidating the mechanisms of age-related differences in response to SCI and developing age-appropriate therapeutics.
Collapse
Affiliation(s)
- Ramona E von Leden
- Neuroscience Program, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA. .,Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Room C2099, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Guzal Khayrullina
- Neuroscience Program, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Room C2099, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kasey E Moritz
- Neuroscience Program, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kimberly R Byrnes
- Neuroscience Program, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Room C2099, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| |
Collapse
|
10
|
Shroff G. Human Embryonic Stem Cell Therapy in Chronic Spinal Cord Injury: A Retrospective Study. Clin Transl Sci 2016; 9:168-75. [PMID: 27144379 PMCID: PMC5351327 DOI: 10.1111/cts.12394] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/28/2016] [Indexed: 12/21/2022] Open
Abstract
Human embryonic stem cells (hESCs) have a role in treating neurological disorders. The efficacy and safety of hESC in treating spinal cord injury (SCI) was reported in our previous study. In the present study, we have evaluated the efficacy and safety of hESC therapy in 226 patients with SCI. In the first treatment phase (T1), 0.25 mL hESCs were administered intramuscularly twice daily, 1 mL every 10 days i.v., and 1-5 mL every 7 days. Of 153 patients in the American Spinal Injury Association (ASIA) scale A at the beginning of T1, a significant number of patients (n = 80; 52.3%) moved to lower scales at the end of T1 (p = 0.01). At the end of T2, of 32 patients in ASIA scale A, 12 patients (37.5%) moved to scale B (p = 0.01). Of 19 patients, 3 patients (37.5%) moved to scale B at the end of T3 (p = 0.02). No serious adverse events (AEs) were observed. hESC transplantation is safe and effective.
Collapse
Affiliation(s)
- G Shroff
- Nutech Mediworld, New Delhi, India
| |
Collapse
|
11
|
Peng WS, Qi C, Zhang H, Gao ML, Wang H, Ren F, Li XQ. Distribution of paired immunoglobulin-like receptor B in the nervous system related to regeneration difficulties after unilateral lumbar spinal cord injury. Neural Regen Res 2015; 10:1139-46. [PMID: 26330840 PMCID: PMC4541248 DOI: 10.4103/1673-5374.160111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2015] [Indexed: 12/21/2022] Open
Abstract
Paired immunoglobulin-like receptor B (PirB) is a functional receptor of myelin-associated inhibitors for axonal regeneration and synaptic plasticity in the central nervous system, and thus suppresses nerve regeneration. The regulatory effect of PirB on injured nerves has received a lot of attention. To better understand nerve regeneration inability after spinal cord injury, this study aimed to investigate the distribution of PirB (via immunofluorescence) in the central nervous system and peripheral nervous system 10 days after injury. Immunoreactivity for PirB increased in the dorsal root ganglia, sciatic nerves, and spinal cord segments. In the dorsal root ganglia and sciatic nerves, PirB was mainly distributed along neuronal and axonal membranes. PirB was found to exhibit a diffuse, intricate distribution in the dorsal and ventral regions. Immunoreactivity for PirB was enhanced in some cortical neurons located in the bilateral precentral gyri. Overall, the findings suggest a pattern of PirB immunoreactivity in the nervous system after unilateral spinal transection injury, and also indicate that PirB may suppress repair after injury.
Collapse
Affiliation(s)
- Wan-Shu Peng
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Chao Qi
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hong Zhang
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Mei-Ling Gao
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hong Wang
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Fei Ren
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xia-Qing Li
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
12
|
Praet J, Santermans E, Daans J, Le Blon D, Hoornaert C, Goossens H, Hens N, Van Der Linden A, Berneman Z, Ponsaerts P. Early Inflammatory Responses following Cell Grafting in the CNS Trigger Activation of the Subventricular Zone: A Proposed Model of Sequential Cellular Events. Cell Transplant 2015; 24:1481-92. [DOI: 10.3727/096368914x682800] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
While multiple rodent preclinical studies, and to a lesser extent human clinical trials, claim the feasibility, safety, and potential clinical benefit of cell grafting in the central nervous system (CNS), currently only little convincing knowledge exists regarding the actual fate of the grafted cells and their effect on the surrounding environment (or vice versa). Our preceding studies already indicated that only a minor fraction of the initially grafted cell population survives the grafting process, while the surviving cell population becomes invaded by highly activated microglia/macrophages and surrounded by reactive astrogliosis. In the current study, we further elaborate on early cellular and inflammatory events following syngeneic grafting of eGFP mouse embryonic fibroblasts (mEFs) in the CNS of immunocompetent mice. Based on obtained quantitative histological data, we here propose a detailed mathematically derived working model that sequentially comprises hypoxia-induced apoptosis of grafted mEFs, neutrophil invasion, neoangiogenesis, microglia/macrophage recruitment, astrogliosis, and eventually survival of a limited number of grafted mEFs. Simultaneously, we observed that the cellular events following mEF grafting activates the subventricular zone neural stem and progenitor cell compartment. This proposed model therefore further contributes to our understanding of cell graft-induced cellular responses and will eventually allow for successful manipulation of this intervention.
Collapse
Affiliation(s)
- Jelle Praet
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- BioImaging Laboratory, University of Antwerp, Wilrijk, Belgium
| | - Eva Santermans
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium
| | - Jasmijn Daans
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Debbie Le Blon
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Chloé Hoornaert
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium
- Centre for Health Economic Research and Modeling Infectious Diseases (Chermid), University of Antwerp, Wilrijk, Belgium
| | | | - Zwi Berneman
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Peter Ponsaerts
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
13
|
Costa R, Bergwerf I, Santermans E, De Vocht N, Praet J, Daans J, Le Blon D, Hoornaert C, Reekmans K, Hens N, Goossens H, Berneman Z, Parolini O, Alviano F, Ponsaerts P. Distinct In Vitro Properties of Embryonic and Extraembryonic Fibroblast-Like Cells are Reflected in their in Vivo Behavior following Grafting in the Adult Mouse Brain. Cell Transplant 2015; 24:223-33. [DOI: 10.3727/096368913x676196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Although intracerebral transplantation of various fibroblast(-like) cell populations has been shown feasible, little is known about the actual in vivo remodeling of these cellular grafts and their environment. In this study, we aimed to compare the in vitro and in vivo behavior of two phenotypically similar—but developmentally distinct—fibroblast-like cell populations, namely, mouse embryonic fibroblasts (mEFs) and mouse fetal membrane-derived stromal cells (mFMSCs). While both mEFs and mFMSCs are readily able to reduce TNF-α secretion by LPS/IFN-γ-activated BV-2 microglia, mFMSCs and mEFs display strikingly opposite behavior with regard to VEGF production under normal and inflammatory conditions. Whereas mFMSCs downregulate VEGF production upon coculture with LPS/IFN-γ-activated BV-2 microglia, mEFs upregulate VEGF production in the presence of LPS/IFN-γ-activated BV-2 microglia. Subsequently, in vivo grafting of mFMSCs and mEFs revealed no difference in microglial and astroglial responses toward the cellular grafts. However, mFMSC grafts displayed a lower degree of neoangiogenesis compared to mEF grafts, thereby potentially explaining the lower cell number able to survive in mFMSC grafts. In summary, our results suggest that physiological differences between fibroblast-like cell populations might lie at the basis of variations in histopathological and/or clinical outcome following cell grafting in mouse brain.
Collapse
Affiliation(s)
- Roberta Costa
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Irene Bergwerf
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Eva Santermans
- Center for Statistics, I-Biostat, Hasselt University, Hasselt, Belgium
| | - Nathalie De Vocht
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Jelle Praet
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Jasmijn Daans
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Debbie Le Blon
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Chloé Hoornaert
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Kristien Reekmans
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Niel Hens
- Center for Statistics, I-Biostat, Hasselt University, Hasselt, Belgium
- Centre for Health Economic Research and Modeling Infectious Diseases (Chermid), University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Ornella Parolini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| | - Francesco Alviano
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Peter Ponsaerts
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
14
|
Evaluation of Motor Neuron-Like Cell Differentiation of hEnSCs on Biodegradable PLGA Nanofiber Scaffolds. Mol Neurobiol 2014; 52:1704-1713. [DOI: 10.1007/s12035-014-8931-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
|
15
|
Abstract
Spinal cord injury is a complex pathology often resulting in functional impairment and paralysis. Gene therapy has emerged as a possible solution to the problems of limited neural tissue regeneration through the administration of factors promoting axonal growth, while also offering long-term local delivery of therapeutic molecules at the injury site. Of note, gene therapy is our response to the requirements of neural and glial cells following spinal cord injury, providing, in a time-dependent manner, growth substances for axonal regeneration and eliminating axonal growth inhibitors. Herein, we explore different gene therapy strategies, including targeting gene expression to modulate the presence of neurotrophic growth or survival factors and increase neural tissue plasticity. Special attention is given to describing advances in viral and non-viral gene delivery systems, as well as the available routes of gene delivery. Finally, we discuss the future of combinatorial gene therapies and give consideration to the implementation of gene therapy in humans.
Collapse
|
16
|
Distinct spatial distribution of microglia and macrophages following mesenchymal stem cell implantation in mouse brain. Immunol Cell Biol 2014; 92:650-8. [DOI: 10.1038/icb.2014.49] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/09/2014] [Accepted: 05/23/2014] [Indexed: 12/18/2022]
|
17
|
Epidermal stem cells in orthopaedic regenerative medicine. Int J Mol Sci 2013; 14:11626-42. [PMID: 23727934 PMCID: PMC3709750 DOI: 10.3390/ijms140611626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 05/15/2013] [Accepted: 05/20/2013] [Indexed: 01/01/2023] Open
Abstract
In the last decade, great advances have been made in epidermal stem cell studies at the cellular and molecular level. These studies reported various subpopulations and differentiations existing in the epidermal stem cell. Although controversies and unknown issues remain, epidermal stem cells possess an immune-privileged property in transplantation together with easy accessibility, which is favorable for future clinical application. In this review, we will summarize the biological characteristics of epidermal stem cells, and their potential in orthopedic regenerative medicine. Epidermal stem cells play a critical role via cell replacement, and demonstrate significant translational potential in the treatment of orthopedic injuries and diseases, including treatment for wound healing, peripheral nerve and spinal cord injury, and even muscle and bone remodeling.
Collapse
|
18
|
Park DY, Mayle RE, Smith RL, Corcoran-Schwartz I, Kharazi AI, Cheng I. Combined Transplantation of Human Neuronal and Mesenchymal Stem Cells following Spinal Cord Injury. Global Spine J 2013; 3:1-6. [PMID: 24436845 PMCID: PMC3854610 DOI: 10.1055/s-0033-1337118] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 11/12/2012] [Indexed: 10/27/2022] Open
Abstract
Transplantation of human fetal neural stem cells (hNSCs) previously demonstrated significant functional recovery after spinal cord contusion in rats. Other studies indicated that human mesenchymal stem cells (hMSCs) can home to areas of damage and cross the blood-brain barrier. The purpose of this article is to determine if combined administration of mesenchymal stem cells and neuronal stem cells improves functional outcomes in rats. The study design was a randomized controlled animal trial. Female adult Long-Evans hooded rats underwent laminectomy at T10 level. Moderate spinal cord contusion at T10 level was induced by the MASCIS Impactor. Four groups were identified. The MSC + NSC group received hMSCs intravenously (IV) immediately after spinal cord injury (acute) and returned 1 week later (subacute) for injection of hNSC directly at site of injury. The MSC-only group received hMSC IV acutely and cell media subacutely. The NSC-only group received cell media IV acutely and hNSC subacutely. The control group received cell media IV acutely and subacutely. Subjects were assessed for 6 weeks using Basso, Beattie, Bresnahan Locomotor Rating Score. Twenty-four subjects were utilized, six subjects in each group. Statistically significant functional improvement was seen in the MSC + NSC group and the NSC-only group versus controls (p = 0.027, 0.042, respectively). The MSC-only group did not demonstrate a significant improvement over control (p = 0.145). Comparing the MSC + NSC group and the NSC-only group, there was no significant difference (p = 0.357). Subacute transplantation of hNSCs into contused spinal cord of rats led to significant functional recovery when injected either with or without acute IV administration of hMSCs. Neither hMSCs nor addition of hMSC to hNSC resulted in significant improvement.
Collapse
Affiliation(s)
- D. Y. Park
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - R. E. Mayle
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - R. L. Smith
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - I. Corcoran-Schwartz
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | | | - I. Cheng
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
19
|
Jain KK. Regenerative Therapy for Central Nervous System Trauma. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
20
|
Reekmans K, De Vocht N, Praet J, Le Blon D, Hoornaert C, Daans J, Van der Linden A, Berneman Z, Ponsaerts P. Quantitative evaluation of stem cell grafting in the central nervous system of mice by in vivo bioluminescence imaging and postmortem multicolor histological analysis. Methods Mol Biol 2013; 1052:125-141. [PMID: 23733539 DOI: 10.1007/7651_2013_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Stem cell transplantation in the central nervous system (CNS) is currently under intensive investigation as a novel therapeutic approach for a variety of brain disorders and/or injuries. However, one of the main hurdles at the moment is the lack of standardized procedures to evaluate cell graft survival and behavior following transplantation into CNS tissue, thereby leading to the publication of confusing and/or conflicting research results. In this chapter, we therefore provide validated in vivo bioluminescence and postmortem histological procedures to quantitatively determine: (a) the survival of grafted stem cells, and (b) the microglial and astroglial cell responses following cell grafting.
Collapse
Affiliation(s)
- Kristien Reekmans
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Campus Drie Eiken (CDE-S6.51), Universiteitsplein 1, Antwerp (Wilrijk), 2610, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Reekmans K, De Vocht N, Praet J, Fransen E, Le Blon D, Hoornaert C, Daans J, Goossens H, Van der Linden A, Berneman Z, Ponsaerts P. Spatiotemporal evolution of early innate immune responses triggered by neural stem cell grafting. Stem Cell Res Ther 2012; 3:56. [PMID: 23241452 PMCID: PMC3580486 DOI: 10.1186/scrt147] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/12/2012] [Indexed: 12/12/2022] Open
Abstract
Introduction Transplantation of neural stem cells (NSCs) is increasingly suggested to become part of future therapeutic approaches to improve functional outcome of various central nervous system disorders. However, recently it has become clear that only a small fraction of grafted NSCs display long-term survival in the (injured) adult mouse brain. Given the clinical invasiveness of NSC grafting into brain tissue, profound characterisation and understanding of early post-transplantation events is imperative to claim safety and efficacy of cell-based interventions. Methods Here, we applied in vivo bioluminescence imaging (BLI) and post-mortem quantitative histological analysis to determine the localisation and survival of grafted NSCs at early time points post-transplantation. Results An initial dramatic cell loss (up to 80% of grafted cells) due to apoptosis could be observed within the first 24 hours post-implantation, coinciding with a highly hypoxic NSC graft environment. Subsequently, strong spatiotemporal microglial and astroglial cell responses were initiated, which stabilised by day 5 post-implantation and remained present during the whole observation period. Moreover, the increase in astrocyte density was associated with a high degree of astroglial scarring within and surrounding the graft site. During the two-week follow up in this study, the NSC graft site underwent extensive remodelling with NSC graft survival further declining to around 1% of the initial number of grafted cells. Conclusions The present study quantitatively describes the early post-transplantation events following NSC grafting in the adult mouse brain and warrants that such intervention is directly associated with a high degree of cell loss, subsequently followed by strong glial cell responses.
Collapse
|
22
|
Marinho PAN, Vareschini DT, Gomes IC, Paulsen BDS, Furtado DR, Castilho LDR, Rehen SK. Xeno-free production of human embryonic stem cells in stirred microcarrier systems using a novel animal/human-component-free medium. Tissue Eng Part C Methods 2012; 19:146-55. [PMID: 22834864 DOI: 10.1089/ten.tec.2012.0141] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Currently, stem cell research faces a major bottleneck related to the low efficiency of methods to produce large quantities of human embryonic stem cells (ESC) for use in clinical trials. Most culture media currently employed for human ESC cultivation contain animal compounds, and cells are grown in static flasks. Besides the immediate contamination with nonhuman compounds, cell expansion in flasks tends to be laborious and nonefficient. Here we cultured human ESC in stirred microcarrier (MC) systems using an animal/human-component-free medium, to overcome both issues. The method developed to culture cells on suspended beads combined the use of polymeric MCs in stirred vessels with an optimized culture medium free of supplements of animal and human origin. This approach generated approximately 160 million cells within 6 days, which were shown to remain pluripotent. The process developed herein provides a step forward toward therapy due to the economic advantages in the production of human ESC and to their consequent low immunogenic potential.
Collapse
Affiliation(s)
- Paulo André Nobrega Marinho
- National Laboratory for Embryonic Stem Cell Research, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | |
Collapse
|
23
|
Grumbles RM, Almeida VW, Casella GTB, Wood PM, Hemstapat K, Thomas CK. Motoneuron replacement for reinnervation of skeletal muscle in adult rats. J Neuropathol Exp Neurol 2012; 71:921-30. [PMID: 22964786 PMCID: PMC3760019 DOI: 10.1097/nen.0b013e31826cf69a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Reinnervation is needed to rescue muscle when motoneurons die in disease or injury. Embryonic ventral spinal cord cells transplanted into peripheral nerve reinnervate muscle and reduce atrophy, but low motoneuron survival may limit motor unit formation. We tested whether transplantation of a purified population of embryonic motoneurons into peripheral nerve (mean ± SE, 146,458 ± 4,011 motoneurons) resulted in more motor units and reinnervation than transplantation of a mixed population of ventral spinal cord cells (72,075 ± 12,329 motoneurons). Ten weeks after either kind of transplant, similar numbers of neurons expressed choline acetyl transferase and/or Islet-1. Motoneuron numbers always exceeded the reinnervated motor unit count. Most motor end plate were simple plaques. Reinnervation significantly reduced muscle fiber atrophy. These data show that the number of transplanted motoneurons and motoneuron survival do not limit muscle reinnervation. Incomplete differentiation of motoneurons in nerve and lack of muscle activity may result in immature neuromuscular junctions that limit reinnervation and function.
Collapse
Affiliation(s)
- Robert M Grumbles
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136-2104, USA
| | | | | | | | | | | |
Collapse
|
24
|
Wang J, Ma C, Rong W, Jing H, Hu X, Liu X, Jiang L, Wei F, Liu Z. Bog bilberry anthocyanin extract improves motor functional recovery by multifaceted effects in spinal cord injury. Neurochem Res 2012; 37:2814-25. [PMID: 23001399 DOI: 10.1007/s11064-012-0883-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/09/2012] [Accepted: 08/13/2012] [Indexed: 11/29/2022]
Abstract
The aim of this study was to determine the therapeutic efficiency of bog bilberry anthocyanin extract (BBAE) treatment starting 1 d after spinal cord injury (SCI) in rats and to investigate the underlying mechanism. The BBAE contained cyanidin-3-glucoside, malvidin-3-galactoside and malvidin-3-glucoside. SCI models were induced using the weight-drop method in Sprague-Dawley rats and additionally with sham group (laminectomy only). The animals were divided into four groups: vehicle-treated group; 10 mg/kg BBAE-treated group; 20 mg/kg BBAE-treated group; sham group. BBAE-treated or vehicle-treated group was administered orally at one day after SCI and then daily for 8 weeks. Locomotor functional recovery was assessed during the 8 weeks post operation period by performing a Basso, Beattie, and Bresnahan (BBB) locomotor score test. At the end of study, the animals were killed, and 1.5 cm segments of spinal cord encompassing the injury site were removed for immunohistochemistry, histopathological and western blotting analysis. Immunohistochemistry for GFAP, aggrecan, neurocan and NeuN was used to assess the degree of astrocytic glial scar formation and neuron survival. Immunohistochemistry and western blotting analysis for TNF-α, IL-6, IL-1β was used to evaluate the anti-inflammation effect of BBAE. To evaluate its inhibition effect on the astrocytes, we performed the MTT assay and immunohistochemistry for Ki67 in vitro. Results show that the BBAE-treated animals showed significantly better locomotor functional recovery, neuron death and smaller glial scar formation after spinal cord injury in vivo. In addition, BBAE administration could inhibit astrocyte proliferation in vivo and vitro. Therefore, BBAE may be useful as a promising therapeutic agent for SCI.
Collapse
Affiliation(s)
- Jun Wang
- Department of Orthopedics, Peking University Third Hospital, 49 North Garden Rd, Haidian, Beijing 100191, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Reekmans K, Praet J, De Vocht N, Daans J, Van der Linden A, Berneman Z, Ponsaerts P. Stem cell therapy for multiple sclerosis: preclinical evidence beyond all doubt? Regen Med 2012; 7:245-59. [PMID: 22397612 DOI: 10.2217/rme.12.5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stem cell transplantation holds great promise for restoration of neural function in various neurodegenerative disorders, including multiple sclerosis (MS). However, many questions remain regarding the true efficacy and precise mode of action of stem cell-based therapeutic approaches. Therefore, in this article, we will first discuss the ideal route and/or timing of stem cell-based therapies for experimental autoimmune encephalomyelitis (EAE), the most used preclinical animal model for MS. Next, we will provide an overview of the proposed mechanisms that contribute to the beneficial effects of stem cell transplantation observed during the treatment of rodent EAE. Reviews of current and past literature clearly demonstrate conceptual changes in the development of stem cell-based approaches for EAE/MS, leading to the identification of several major challenges to be tackled before (stem) cell therapy for rodent EAE can be safely and successfully translated to human therapy for MS.
Collapse
Affiliation(s)
- Kristien Reekmans
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | | | | | | | | | | | | |
Collapse
|
26
|
[Acute traumatic spinal cord injuries: Epidemiology and prospects]. Neurochirurgie 2012; 58:293-9. [PMID: 22959585 DOI: 10.1016/j.neuchi.2012.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 06/26/2012] [Accepted: 06/27/2012] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Specify the epidemiological data on the acute spinal cord injuries and define a group of patients that could benefit from cellular transplantation therapy designed with the aim of repair and regeneration of damaged spinal cord tissues. MATERIAL AND METHODS Five years monocentric (Gui-de-Chauliac Hospital, Montpellier, France) retrospective analysis of patients suffering from spinal cord injury (SCI). Spinal cord injured-patients, defined as sensory-motor complete, underwent a clinical evaluation following American Spinal Injury Association (ASIA) and functional type 2 Spinal Cord Independence Measure (SCIM2) scorings as well as radiological evaluation through spinal cord magnetic resonance imaging (MRI). RESULTS One hundred and fifty-seven medical records were reviewed and we selected and re-examined 20 patients with complete thoracic spinal cord lesion. Clinical and radiological evaluations of these patients demonstrated, in 75 % of the cases, an absence of clinical progression after a mean of 49months. Radiological abnormalities were constantly present in the initial (at the admission to hospital) and control (re-evaluation) MRI and no reliable predictive criteria of prognosis had been found. DISCUSSION/CONCLUSION We compare our results to the literature and discuss advantages and limits of cellular transplantation strategies for these patients.
Collapse
|
27
|
Roet KCD, Eggers R, Verhaagen J. Noninvasive Bioluminescence Imaging of Olfactory Ensheathing Glia and Schwann Cells following Transplantation into the Lesioned Rat Spinal Cord. Cell Transplant 2012; 21:1853-65. [DOI: 10.3727/096368911x627471] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In this study, we assess the feasibility of bioluminescence imaging to monitor the survival of Schwann cells (SCs) and olfactory ensheathing glia cells (OECs) after implantation in the lesioned spinal cord of adult rats. To this end, purified SCs and OECs were genetically modified with lentiviral vectors encoding luciferase-2 and GFP and implanted in the lesioned dorsal column. The bioluminescent signal was monitored for over 3 months, and at 7 and 98 days postsurgery, the signal was compared to standard histological analysis of GFP expression in the spinal cords. The temporal profile of the bioluminescent signal showed three distinct phases for both cell types. (I) A relatively stable signal in the first week. (II) A progressive decline in signal strength in the second and third week. (III) After the third week, the average bioluminescent signal stabilized for both cell types. Interestingly, in the first week, the peak of the bioluminescent signal after luciferin injection was delayed when compared to later time points. Similar to in vitro, our data indicated a linear relationship between the in vivo bioluminescent signal and the GFP signal of the SCs and OECs in the spinal cords when the results of both the 7 and 98 day time points are combined. This is the first report of the use of in vivo bioluminescence to monitor cell survival in the lesioned rat spinal cord. Bioluminescence could be a potentially powerful, non-invasive strategy to examine the efficacy of treatments that aim to improve the survival of proregenerative cells transplanted in the injured rat spinal cord.
Collapse
Affiliation(s)
- Kasper C. D. Roet
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Ruben Eggers
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| |
Collapse
|
28
|
De Vocht N, Lin D, Praet J, Hoornaert C, Reekmans K, Le Blon D, Daans J, Pauwels P, Goossens H, Hens N, Berneman Z, Van der Linden A, Ponsaerts P. Quantitative and phenotypic analysis of mesenchymal stromal cell graft survival and recognition by microglia and astrocytes in mouse brain. Immunobiology 2012; 218:696-705. [PMID: 22944251 DOI: 10.1016/j.imbio.2012.08.266] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 08/02/2012] [Accepted: 08/05/2012] [Indexed: 01/18/2023]
Abstract
Although cell transplantation is increasingly suggested to be beneficial for the treatment of various neurodegenerative diseases, the therapeutic application of such intervention is currently hindered by the limited knowledge regarding central nervous system (CNS) transplantation immunology. In this study, we aimed to investigate the early post transplantation innate immune events following grafting of autologous mesenchymal stromal cells (MSC) in the CNS of immune competent mice. First, the survival of grafted Luciferase/eGFP-expressing MSC (MSC-Luc/eGFP) was demonstrated to be stable from on day 3 post implantation using in vivo bioluminescence imaging (BLI), which was further confirmed by quantitative histological analysis of MSC-Luc/eGFP graft survival. Additional histological analyses at week 1 and week 2 post grafting revealed the appearance of (i) graft-surrounding/-invading Iba1+ microglia and (ii) graft-surrounding GFAP+ astrocytes, as compared to day 0 post grafting. While the density of graft-surrounding astrocytes and microglia did not change between week 1 and week 2 post grafting, the density of graft-invading microglia significantly decreased between week 1 and week 2 post implantation. However, despite the observed decrease in microglial density within the graft site, additional phenotypic analysis of graft-invading microglia, based on CD11b- and MHCII-expression, revealed >50% of graft-invading microglia at week 2 post implantation to display an activated status. Although microglial expression of CD11b and MHCII is already suggestive for a pro-inflammatory M1-oriented phenotype, the latter was further confirmed by: (i) the expression of NOS2 by microglia within the graft site, and (ii) the absence of arginase 1-expression, an enzyme known to suppress NO activity in M2-oriented microglia, on graft-surrounding and -invading microglia. In summary, we here provide a detailed phenotypic analysis of post transplantation innate immune events in the CNS of mice, and warrant that such intervention is associated with an M1-oriented microglia response and severe astrogliosis.
Collapse
Affiliation(s)
- Nathalie De Vocht
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Reekmans K, Praet J, Daans J, Reumers V, Pauwels P, Van der Linden A, Berneman ZN, Ponsaerts P. Current challenges for the advancement of neural stem cell biology and transplantation research. Stem Cell Rev Rep 2012; 8:262-78. [PMID: 21537994 DOI: 10.1007/s12015-011-9266-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transplantation of neural stem cells (NSC) is hoped to become a promising primary or secondary therapy for the treatment of various neurodegenerative disorders of the central nervous system (CNS), as demonstrated by multiple pre-clinical animal studies in which functional recovery has already been demonstrated. However, for NSC therapy to be successful, the first challenge will be to define a transplantable cell population. In the first part of this review, we will briefly discuss the main features of ex vivo culture and characterisation of NSC. Next, NSC grafting itself may not only result in the regeneration of lost tissue, but more importantly has the potential to improve functional outcome through many bystander mechanisms. In the second part of this review, we will briefly discuss several pre-clinical studies that contributed to a better understanding of the therapeutic potential of NSC grafts in vivo. However, while many pre-clinical animal studies mainly report on the clinical benefit of NSC grafting, little is known about the actual in vivo fate of grafted NSC. Therefore, the third part of this review will focus on non-invasive imaging techniques for monitoring cellular grafts in the brain under in vivo conditions. Finally, as NSC transplantation research has evolved during the past decade, it has become clear that the host micro-environment itself, either in healthy or injured condition, is an important player in defining success of NSC grafting. The final part of this review will focus on the host environmental influence on survival, migration and differentiation of grafted NSC.
Collapse
Affiliation(s)
- Kristien Reekmans
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Praet J, Reekmans K, Lin D, De Vocht N, Bergwerf I, Tambuyzer B, Daans J, Hens N, Goossens H, Pauwels P, Berneman Z, Van der Linden A, Ponsaerts P. Cell type-associated differences in migration, survival, and immunogenicity following grafting in CNS tissue. Cell Transplant 2012; 21:1867-81. [PMID: 22472278 DOI: 10.3727/096368912x636920] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell transplantation has been suggested to display several neuroprotective and/or neuroregenerative effects in animal models of central nervous system (CNS) trauma. However, while most studies report on clinical observations, currently little is known regarding the actual fate of the cell populations grafted and whether or how the brain's innate immune system, mainly directed by activated microglia and astrocytes, interacts with autologous cellular implants. In this study, we grafted well-characterized neural stem cell, mouse embryonic fibroblast, dendritic cell, bone marrow mononuclear cell, and splenocyte populations, all isolated or cultured from C57BL/6-eGFP transgenic mice, below the capsula externa (CE) of healthy C57BL/6 mice and below the inflamed/demyelinated CE of cuprizone-treated C57BL/6 mice. Two weeks postgrafting, an extensive quantitative multicolor histological analysis was performed in order (i) to quantify cell graft localization, migration, survival, and toxicity and (ii) to characterize endogenous CNS immune responses against the different cell grafts. Obtained results indicate dependence on the cell type grafted: (i) a different degree of cell graft migration, survival, and toxicity and (ii) a different organization of the endogenous immune response. Based on these observations, we warrant that further research should be undertaken to understand-and eventually control-cell graft-induced tissue damage and activation of the brain's innate immune system. The latter will be inevitable before cell grafting in the CNS can be performed safely and successfully in clinical settings.
Collapse
Affiliation(s)
- Jelle Praet
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
De Vocht N, Bergwerf I, Vanhoutte G, Daans J, De Visscher G, Chatterjee S, Pauwels P, Berneman Z, Ponsaerts P, Van der Linden A. Labeling of Luciferase/eGFP-expressing bone marrow-derived stromal cells with fluorescent micron-sized iron oxide particles improves quantitative and qualitative multimodal imaging of cellular grafts in vivo. Mol Imaging Biol 2012; 13:1133-45. [PMID: 21246293 DOI: 10.1007/s11307-011-0469-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE Development of multimodal imaging strategies is currently of utmost importance for the validation of preclinical stem cell therapy studies. PROCEDURES We performed a combined labeling strategy for bone marrow-derived stromal cells (BMSC) based on genetic modification with the reporter genes Luciferase and eGFP (BMSC-Luc/eGFP) and physical labeling with blue fluorescent micron-sized iron oxide particles (MPIO) in order to unambiguously identify BMSC localization, survival, and differentiation following engraftment in the central nervous system of mice by in vivo bioluminescence (BLI) and magnetic resonance imaging and postmortem histological analysis. RESULTS Using this combination, a significant increase of in vivo BLI signal was observed for MPIO-labeled BMSC-Luc/eGFP. Moreover, MPIO labeling of BMSC-Luc/eGFP allows for the improved identification of implanted cells within host tissue during histological observation. CONCLUSIONS This study describes an optimized labeling strategy for multimodal stem cell imaging resulting in improved quantitative and qualitative detection of cellular grafts.
Collapse
|
32
|
Chen CS, Soni S, Le C, Biasca M, Farr E, Chen EYT, Chin WC. Human stem cell neuronal differentiation on silk-carbon nanotube composite. NANOSCALE RESEARCH LETTERS 2012; 7:126. [PMID: 22333433 PMCID: PMC3292945 DOI: 10.1186/1556-276x-7-126] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 02/14/2012] [Indexed: 05/20/2023]
Abstract
Human embryonic stem cells [hESCs] are able to differentiate into specific lineages corresponding to regulated spatial and temporal signals. This unique attribute holds great promise for regenerative medicine and cell-based therapy for many human diseases such as spinal cord injury [SCI] and multiple sclerosis [MS]. Carbon nanotubes [CNTs] have been successfully used to promote neuronal differentiation, and silk has been widely applied in tissue engineering. This study aims to build silk-CNT composite scaffolds for improved neuron differentiation efficiency from hESCs.Two neuronal markers (β-III tubulin and nestin) were utilized to determine the hESC neuronal lineage differentiation. In addition, axonal lengths were measured to evaluate the progress of neuronal development. The results demonstrated that cells on silk-CNT scaffolds have a higher β-III tubulin and nestin expression, suggesting augmented neuronal differentiation. In addition, longer axons with higher density were found to associate with silk-CNT scaffolds.Our silk-CNT-based composite scaffolds can promote neuronal differentiation of hESCs. The silk-CNT composite scaffolds developed here can serve as efficient supporting matrices for stem cell-derived neuronal transplants, offering a promising opportunity for nerve repair treatments for SCI and MS patients.
Collapse
Affiliation(s)
- Chi-Shuo Chen
- Bioengineering Program, School of Engineering, University of California, Merced, CA, USA
| | - Sushant Soni
- Bioengineering Program, School of Engineering, University of California, Merced, CA, USA
| | - Catherine Le
- School of Nature Sciences, University of California, Merced, CA, USA
| | - Matthew Biasca
- School of Nature Sciences, University of California, Merced, CA, USA
| | - Erik Farr
- School of Nature Sciences, University of California, Merced, CA, USA
| | - Eric Y-T Chen
- Bioengineering Program, School of Engineering, University of California, Merced, CA, USA
| | - Wei-Chun Chin
- Bioengineering Program, School of Engineering, University of California, Merced, CA, USA
| |
Collapse
|
33
|
Rubin AD, Hogikyan ND, Oh A, Feldman EL. Potential for promoting recurrent laryngeal nerve regeneration by remote delivery of viral gene therapy. Laryngoscope 2012; 122:349-55. [PMID: 22241608 DOI: 10.1002/lary.22436] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/06/2011] [Accepted: 10/13/2011] [Indexed: 12/22/2022]
Abstract
OBJECTIVES/HYPOTHESIS The aims of this study were to demonstrate the ability to enhance nerve regeneration by remote delivery of a viral vector to the crushed recurrent laryngeal nerve (RLN), to demonstrate the usefulness of a crushed RLN model to test the efficacy of viral gene therapy, and to discuss future potential applications of this approach. STUDY DESIGN Animal study. METHODS Adult Sprague-Dawley rats were assigned to two groups. In the experimental group, an adeno-associated viral (AAV) vector carrying a zinc-finger transcription factor, which stimulates endogenous insulinlike growth factor I production (AAV2-TO-6876vp16), was injected into the crushed RLN. In the control group, an AAV vector carrying the gene for green fluorescent protein was injected into the crushed RLN. Unilateral RLN paralysis was confirmed endoscopically. At 1 week, laryngeal endoscopies were repeated and recorded. Larynges were cryosectioned in 15-μm sections and processed for acetylcholine histochemistry (motor endplates) followed by neurofilament immunoperoxidase (nerve fibers). Percentage nerve-endplate contact (PEC) was determined and compared. Vocal fold motion was evaluated by blinded reviewers using a visual analogue scale (VAS). RESULTS The difference between PEC on the crushed and uncrushed sides was statistically less in the experimental group (0.54 ± 0.18 vs. 0.30 ± 0.26, P = .0006). The VAS score at 1 week was significantly better in the experimental group (P = .002). CONCLUSIONS AAV2-TO-6876vp16 demonstrated a neurotrophic effect when injected into the crushed RLN. The RLN offers a conduit for viral gene therapy to the brainstem that could be useful for the treatment of RLN injury or bulbar motor neuron disease.
Collapse
Affiliation(s)
- Adam D Rubin
- Lakeshore Professional Voice Center, St. Clair Shores, Michigan, University of Michigan, Ann Arbor, Michigan 48081, USA.
| | | | | | | |
Collapse
|
34
|
Wang J, Ye R, Wei Y, Wang H, Xu X, Zhang F, Qu J, Zuo B, Zhang H. The effects of electrospun TSF nanofiber diameter and alignment on neuronal differentiation of human embryonic stem cells. J Biomed Mater Res A 2011; 100:632-45. [PMID: 22213384 DOI: 10.1002/jbm.a.33291] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Revised: 09/11/2011] [Accepted: 10/03/2011] [Indexed: 12/17/2022]
Abstract
Although transplantation of human embryonic stem cells (hESCs)-derived neural precursors (NPs) has been demonstrated with some success for nervous repair in small animal model, control of the survival, and directional differentiation of these cells is still challenging. Meanwhile, the notion that using suitable scaffolding materials to control the growth and differentiation of grafted hESC-derived NPs raises the hope for better clinical nervous repair. In this study, we cultured hESC-derived NPs on Tussah silk fibroin (TSF)-scaffold of different diameter (i.e., 400 and 800 nm) and orientation (i.e., random and aligned) to analyze the effect of fiber diameter and alignment on the cell viability, neuronal differentiation, and neurite outgrowth of hESC-derived NPs. The results show that TSF-scaffold supports the survival, migration, and differentiation of hESC-derived NPs. Aligned TSF-scaffold significantly promotes the neuronal differentiation and neurite outgrowth of hESC-derived neurons compared with random TSF-scaffold. Moreover, on aligned 400 nm fibers cell viability, neuronal differentiation and neurite outgrowth are greater than that on aligned 800 nm fibers. Together, these results demonstrate that aligned 400 nm TSF-scaffold is more suitable for the development of hESC-derived NPs, which shed light on optimization of the therapeutic potential of hESCs to be employed for neural regeneration.
Collapse
Affiliation(s)
- Junxia Wang
- Department of Cell Biology, Medical College of Soochow University, Jiangsu Key Laboratory of Stem Cell Research, Ren Ai Road 199, Suzhou Industrial Park, Suzhou 215123, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Saberi H, Firouzi M, Habibi Z, Moshayedi P, Aghayan HR, Arjmand B, Hosseini K, Razavi HE, Yekaninejad MS. Safety of intramedullary Schwann cell transplantation for postrehabilitation spinal cord injuries: 2-year follow-up of 33 cases. J Neurosurg Spine 2011; 15:515-25. [PMID: 21800956 DOI: 10.3171/2011.6.spine10917] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Many experimental studies on spinal cord injuries (SCIs) support behavioral improvement after Schwann cell treatment. This study was conducted to evaluate safety issues 2 years after intramedullary Schwann cell transplantation in 33 consecutively selected patients with SCI. METHODS Of 356 patients with SCIs who had completed at least 6 months of a conventional rehabilitation program and who were screened for the study criteria, 33 were enrolled. After giving their informed consent, they volunteered for participation. They underwent sural nerve harvesting and intramedullary injection of a processed Schwann cell solution. Outcome assessments included a general health questionnaire, neurological examination, and functional recordings in terms of American Spinal Injury Association (ASIA) and Functional Independence Measure scoring, which were documented by independent observers. There were 24 patients with thoracic and 9 with cervical injuries. Sixteen patients were categorized in ASIA Grade A, and the 17 remaining participants had ASIA Grade B. RESULTS There were no cases of deep infection, and the follow-up MR imaging studies obtained at 2 years did not reveal any deformity related to the procedure. There was no case of permanent neurological worsening or any infectious or viral complications. No new increment in syrinx size or abnormal tissue and/or tumor formation were observed on contrast-enhanced MR imaging studies performed 2 years after the treatment. CONCLUSIONS Preliminary results, especially in terms of safety, seem to be promising, paving the way for future cell therapy trials.
Collapse
Affiliation(s)
- Hooshang Saberi
- Department of Neurosurgery, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bhanot Y, Rao S, Ghosh D, Balaraju S, Radhika CR, Satish Kumar KV. Autologous mesenchymal stem cells in chronic spinal cord injury. Br J Neurosurg 2011; 25:516-22. [PMID: 21749185 DOI: 10.3109/02688697.2010.550658] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Spinal cord injury (SCI) occurs in the most productive part of life. Treatment options for treatment of chronic SCI are few and have limited impact on clinical outcome. Central nervous system (CNS) has limited intrinsic regeneration capability. The study included patients with chronic complete SCI. Previously harvested autologous mesenchymal stem cells were administered at the site of injury after a laminectomy. Follow-up was done by a neutral examiner not involved in the surgery every 3 months. One patient had improvement in motor power. Two patients had a patchy improvement in pin prick sensation below the level of injury. Three different, progressively increasing doses did not result in improvement in the clinical outcome. Though the administration of allogenic human mesenchymal stem cells is safe in patients with SCI, it may not be efficacious; especially in patients with chronic SCI.
Collapse
Affiliation(s)
- Yanish Bhanot
- Department of Neurosurgery and Pediatric Neurosurgery, St Philomena's Hospital, Bangalore, India
| | | | | | | | | | | |
Collapse
|
37
|
Regenerative Therapy for Central Nervous System Trauma. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
38
|
Reekmans KP, Praet J, De Vocht N, Tambuyzer BR, Bergwerf I, Daans J, Baekelandt V, Vanhoutte G, Goossens H, Jorens PG, Ysebaert DK, Chatterjee S, Pauwels P, Van Marck E, Berneman ZN, Van der Linden A, Ponsaerts P. Clinical potential of intravenous neural stem cell delivery for treatment of neuroinflammatory disease in mice? Cell Transplant 2010; 20:851-69. [PMID: 21092405 DOI: 10.3727/096368910x543411] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
While neural stem cells (NSCs) are widely expected to become a therapeutic agent for treatment of severe injuries to the central nervous system (CNS), currently there are only few detailed preclinical studies linking cell fate with experimental outcome. In this study, we aimed to validate whether IV administration of allogeneic NSC can improve experimental autoimmune encephalomyelitis (EAE), a well-established animal model for human multiple sclerosis (MS). For this, we cultured adherently growing luciferase-expressing NSCs (NSC-Luc), which displayed a uniform morphology and expression profile of membrane and intracellular markers, and which displayed an in vitro differentiation potential into neurons and astrocytes. Following labeling with green fluorescent micron-sized iron oxide particles (f-MPIO-labeled NSC-Luc) or lentiviral transduction with the enhanced green fluorescent protein (eGFP) reporter gene (NSC-Luc/eGFP), cell implantation experiments demonstrated the intrinsic survival capacity of adherently cultured NSC in the CNS of syngeneic mice, as analyzed by real-time bioluminescence imaging (BLI), magnetic resonance imaging (MRI), and histological analysis. Next, EAE was induced in C57BL/6 mice followed by IV administration of NSC-Luc/eGFP at day 7 postinduction with or without daily immunosuppressive therapy (cyclosporine A, CsA). During a follow-up period of 20 days, the observed clinical benefit could be attributed solely to CsA treatment. In addition, histological analysis demonstrated the absence of NSC-Luc/eGFP at sites of neuroinflammation. In order to investigate the absence of therapeutic potential, BLI biodistribution analysis of IV-administered NSC-Luc/eGFP revealed cell retention in lung capillaries as soon as 1-min postinjection, resulting in massive inflammation and apoptosis in lung tissue. In summary, we conclude that IV administration of NSCs currently has limited or no therapeutic potential for neuroinflammatory disease in mice, and presumably also for human MS. However, given the fact that grafted NSCs have an intrinsic survival capacity in the CNS, their therapeutic exploitation should be further investigated, and-in contrast to several other reports-will most likely be highly complex.
Collapse
|
39
|
Epidermal neural crest stem cells and their use in mouse models of spinal cord injury. Brain Res Bull 2010; 83:189-93. [DOI: 10.1016/j.brainresbull.2010.07.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Revised: 07/05/2010] [Accepted: 07/06/2010] [Indexed: 12/25/2022]
|
40
|
Epidermal neural crest stem cell (EPI-NCSC)--mediated recovery of sensory function in a mouse model of spinal cord injury. Stem Cell Rev Rep 2010; 6:186-98. [PMID: 20414748 PMCID: PMC2887506 DOI: 10.1007/s12015-010-9152-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Here we show that epidermal neural crest stem cell (EPI-NCSC) transplants in the contused spinal cord caused a 24% improvement in sensory connectivity and a substantial recovery of touch perception. Furthermore we present a novel method for the ex vivo expansion of EPI-NCSC into millions of stem cells that takes advantage of the migratory ability of neural crest stem cells and is based on a new culture medium and the use of microcarriers. Functional improvement was shown by two independent methods, spinal somatosensory evoked potentials (SpSEP) and the Semmes-Weinstein touch test. Subsets of transplanted cells differentiated into myelinating oligodendrocytes. Unilateral injections of EPI-NCSC into the lesion of midline contused mouse spinal cords elicited bilateral improvements. Intraspinal EPI-NCSC did not migrate laterally in the spinal cord or invade the spinal roots and dorsal root ganglia, thus implicating diffusible factors. EPI-NCSC expressed neurotrophic factors, angiogenic factors, and metalloproteases. The strength of EPI-NCSC thus is that they can exert a combination of pertinent functions in the contused spinal cord, including cell replacement, neuroprotection, angiogenesis and modulation of scar formation. EPI-NCSC are uniquely qualified for cell-based therapy in spinal cord injury, as neural crest cells and neural tube stem cells share a higher order stem cell and are thus ontologically closely related.
Collapse
|
41
|
Predifferentiated embryonic stem cells promote functional recovery after spinal cord compressive injury. Brain Res 2010; 1349:115-28. [PMID: 20599835 DOI: 10.1016/j.brainres.2010.06.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 06/09/2010] [Accepted: 06/10/2010] [Indexed: 01/01/2023]
Abstract
We tested the effects of mouse embryonic stem cells (mES) grafts in mice spinal cord injury (SCI). Young adult female C57/Bl6 mice were subjected to laminectomy at T9 and 1-minute compression of the spinal cord with a vascular clip. Four groups were analyzed: laminectomy (Sham), injured (SCI), vehicle (DMEM), and mES-treated (EST). mES pre-differentiated with retinoic acid were injected (8 x 10(5) cells/2 microl) into the lesion epicenter, 10 min after SCI. Basso mouse scale (BMS) and Global mobility test (GMT) were assessed weekly up to 8 weeks, when morphological analyses were performed. GMT analysis showed that EST animals moved faster (10.73+/-0.9076, +/-SEM) than SCI (5.581+/-0.2905) and DMEM (5.705+/-0.2848), but slower than Sham animals (15.80+/-0.3887, p<0.001). By BMS, EST animals reached the final phase of locomotor recovery (3.872+/-0.7112, p<0.01), while animals of the SCI and DMEM groups improved to an intermediate phase (2.037+/-0.3994 and 2.111+/-0.3889, respectively). White matter area and number of myelinated nerve fibers were greater in EST (46.80+/-1.24 and 279.4+/-16.33, respectively) than the SCI group (39.97+/-0.925 and 81.39+/-8.078, p<0.05, respectively). EST group also presented better G-ratio values when compared with SCI group (p<0.001). Immunohistochemical revealed the differentiation of transplanted cells into astrocytes, oligodendrocytes, and Schwann cells, indicating an integration of transplanted cells with host tissue. Ultrastructural analysis showed, in the EST group, better tissue preservation and more remyelination by oligodendrocytes and Schwann cells than the other groups. Our results indicate that acute transplantation of predifferentiated mES into the injured spinal cord increased the spared white matter and number of nerve fibers, improving locomotor function.
Collapse
|
42
|
Pannek J, Göcking K, Bersch U. Long-term effects of repeated intradetrusor botulinum neurotoxin A injections on detrusor function in patients with neurogenic bladder dysfunction. BJU Int 2009; 104:1246-50. [DOI: 10.1111/j.1464-410x.2009.08600.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Chhabra HS, Lima C, Sachdeva S, Mittal A, Nigam V, Chaturvedi D, Arora M, Aggarwal A, Kapur R, Khan TAH. Autologous mucosal transplant in chronic spinal cord injury: an Indian Pilot Study. Spinal Cord 2009; 47:887-95. [DOI: 10.1038/sc.2009.54] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
44
|
Abstract
Cell therapy plays an important role in multidisciplinary management of the two major forms of central nervous system (CNS) injury, traumatic brain injury and spinal cord injury, which are caused by external physical trauma. Cell therapy for CNS disorders involves the use of cells of neural or non-neural origin to replace, repair, or enhance the function of the damaged nervous system and is usually achieved by transplantation of the cells, which are isolated and may be modified, e.g., by genetic engineering, when it may be referred to as gene therapy. Because the adult brain cells have a limited capacity to migrate to and regenerate at sites of injury, the use of embryonic stem cells that can be differentiated into various cell types as well as the use of neural stem cells has been explored. Preclinical studies and clinical trials are reviewed. Advantages as well as limitations are discussed. Cell therapy is promising for the treatment of CNS injury because it targets multiple mechanisms in a sustained manner. It can provide repair and regeneration of damaged tissues as well as prolonged release of neuroprotective and other therapeutic substances.
Collapse
|
45
|
Allogeneic stromal cell implantation in brain tissue leads to robust microglial activation. Immunol Cell Biol 2009; 87:267-73. [PMID: 19290016 DOI: 10.1038/icb.2009.12] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although adult and embryonic stem cell-based therapy for central nervous system (CNS) injury is being developed worldwide, less attention is given to the immunological aspects of allogeneic cell implantation in the CNS. The latter is of major importance because, from a practical point of view, future stem cell-based therapy for CNS injury will likely be performed using well-characterised allogeneic stem cell populations. In this study, we aimed to further describe the immunological mechanism leading to rejection of allogeneic bone marrow-derived stromal cells (BM-SC) after implantation in murine CNS. For this, we first investigated the impact of autologous and allogeneic BM-SC on microglia activation in vitro. Although the results indicate that both autologous and allogeneic BM-SC do not activate microglia themselves in vitro, they also do not inhibit activation of microglia after exogenous stimuli in vitro. Next, we investigated the impact of allogeneic BM-SC on microglia activation in vivo. In contrast to the in vitro observations, microglia become highly activated in vivo after implantation of allogeneic BM-SC in the CNS of immune-competent mice. Moreover, our results suggest that microglia, rather than T-cells, are the major contributors to allograft rejection in the CNS.
Collapse
|
46
|
Bergwerf I, De Vocht N, Tambuyzer B, Verschueren J, Reekmans K, Daans J, Ibrahimi A, Van Tendeloo V, Chatterjee S, Goossens H, Jorens PG, Baekelandt V, Ysebaert D, Van Marck E, Berneman ZN, Linden AVD, Ponsaerts P. Reporter gene-expressing bone marrow-derived stromal cells are immune-tolerated following implantation in the central nervous system of syngeneic immunocompetent mice. BMC Biotechnol 2009; 9:1. [PMID: 19128466 PMCID: PMC2630974 DOI: 10.1186/1472-6750-9-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Accepted: 01/07/2009] [Indexed: 01/09/2023] Open
Abstract
Background Cell transplantation is likely to become an important therapeutic tool for the treatment of various traumatic and ischemic injuries to the central nervous system (CNS). However, in many pre-clinical cell therapy studies, reporter gene-assisted imaging of cellular implants in the CNS and potential reporter gene and/or cell-based immunogenicity, still remain challenging research topics. Results In this study, we performed cell implantation experiments in the CNS of immunocompetent mice using autologous (syngeneic) luciferase-expressing bone marrow-derived stromal cells (BMSC-Luc) cultured from ROSA26-L-S-L-Luciferase transgenic mice, and BMSC-Luc genetically modified using a lentivirus encoding the enhanced green fluorescence protein (eGFP) and the puromycin resistance gene (Pac) (BMSC-Luc/eGFP/Pac). Both reporter gene-modified BMSC populations displayed high engraftment capacity in the CNS of immunocompetent mice, despite potential immunogenicity of introduced reporter proteins, as demonstrated by real-time bioluminescence imaging (BLI) and histological analysis at different time-points post-implantation. In contrast, both BMSC-Luc and BMSC-Luc/eGFP/Pac did not survive upon intramuscular cell implantation, as demonstrated by real-time BLI at different time-points post-implantation. In addition, ELISPOT analysis demonstrated the induction of IFN-γ-producing CD8+ T-cells upon intramuscular cell implantation, but not upon intracerebral cell implantation, indicating that BMSC-Luc and BMSC-Luc/eGFP/Pac are immune-tolerated in the CNS. However, in our experimental transplantation model, results also indicated that reporter gene-specific immune-reactive T-cell responses were not the main contributors to the immunological rejection of BMSC-Luc or BMSC-Luc/eGFP/Pac upon intramuscular cell implantation. Conclusion We here demonstrate that reporter gene-modified BMSC derived from ROSA26-L-S-L-Luciferase transgenic mice are immune-tolerated upon implantation in the CNS of syngeneic immunocompetent mice, providing a research model for studying survival and localisation of autologous BMSC implants in the CNS by real-time BLI and/or histological analysis in the absence of immunosuppressive therapy.
Collapse
Affiliation(s)
- Irene Bergwerf
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|