Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 346
1.
Cell Transplant ; 31: 9636897221125685, 2022.
Article En | MEDLINE | ID: mdl-36124646

Olfactory ensheathing cell (OEC) transplantation is emerging as a promising treatment option for injuries of the nervous system. OECs can be obtained relatively easily from nasal biopsies, and exhibit several properties such as secretion of trophic factors, and phagocytosis of debris that facilitate neural regeneration and repair. But a major limitation of OEC-based cell therapies is the poor survival of transplanted cells which subsequently limit their therapeutic efficacy. There is an unmet need for approaches that enable the in vitro production of OECs in a state that will optimize their survival and integration after transplantation into the hostile injury site. Here, we present an overview of the strategies to modulate OECs focusing on oxygen levels, stimulating migratory, phagocytic, and secretory properties, and on bioengineering a suitable environment in vitro.


Neuroglia , Olfactory Bulb , Cell Transplantation , Cellular Microenvironment , Neuroglia/transplantation , Oxygen
2.
Sci Rep ; 12(1): 4016, 2022 03 07.
Article En | MEDLINE | ID: mdl-35256767

Amyotrophic lateral sclerosis (ALS) is a progressive motor neuronal disorder characterized by neuronal degeneration and currently no effective cure is available to stop or delay the disease from progression. Transplantation of murine glial-restricted precursors (mGRPs) is an attractive strategy to modulate ALS development and advancements such as the use of immune modulators could potentially extend graft survival and function. Using a well-established ALS transgenic mouse model (SOD1G93A), we tested mGRPs in combination with the immune modulators synthetic PreImplantation Factor (sPIF), Tacrolimus (Tac), and Costimulatory Blockade (CB). We report that transplantation of mGRPs into the cisterna magna did not result in increased mice survival. The addition of immunomodulatory regimes again did not increase mice lifespan but improved motor functions and sPIF was superior compared to other immune modulators. Immune modulators did not affect mGRPs engraftment significantly but reduced pro-inflammatory cytokine production. Finally, sPIF and CB reduced the number of microglial cells and prevented neuronal number loss. Given the safety profile and a neuroprotective potential of sPIF, we envision its clinical application in near future.


Amyotrophic Lateral Sclerosis , Motor Disorders , Neuroglia , Peptides , Stem Cell Transplantation , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Inflammation , Mice , Mice, Transgenic , Motor Disorders/drug therapy , Motor Disorders/therapy , Neuroglia/cytology , Neuroglia/transplantation , Peptides/pharmacology , Stem Cells/cytology
3.
J Neuroinflammation ; 18(1): 104, 2021 Apr 30.
Article En | MEDLINE | ID: mdl-33931070

BACKGROUND: Cell transplantation-based treatments for neurological disease are promising, yet graft rejection remains a major barrier to successful regenerative therapies. Our group and others have shown that long-lasting tolerance of transplanted stem cells can be achieved in the brain with systemic application of monoclonal antibodies blocking co-stimulation signaling. However, it is unknown if subsequent injury and the blood-brain barrier breach could expose the transplanted cells to systemic immune system spurring fulminant rejection and fatal encephalitis. Therefore, we investigated whether delayed traumatic brain injury (TBI) could trigger graft rejection. METHODS: Glial-restricted precursor cells (GRPs) were intracerebroventricularly transplanted in immunocompetent neonatal mice and co-stimulation blockade (CoB) was applied 0, 2, 4, and 6 days post-grafting. Bioluminescence imaging (BLI) was performed to monitor the grafted cell survival. Mice were subjected to TBI 12 weeks post-transplantation. MRI and open-field test were performed to assess the brain damage and behavioral change, respectively. The animals were decapitated at week 16 post-transplantation, and the brains were harvested. The survival and distribution of grafted cells were verified from brain sections. Hematoxylin and eosin staining (HE) was performed to observe TBI-induced brain legion, and neuroinflammation was evaluated immunohistochemically. RESULTS: BLI showed that grafted GRPs were rejected within 4 weeks after transplantation without CoB, while CoB administration resulted in long-term survival of allografts. BLI signal had a steep rise following TBI and subsequently declined but remained higher than the preinjury level. Open-field test showed TBI-induced anxiety for all animals but neither CoB nor GRP transplantation intensified the symptom. HE and MRI demonstrated a reduction in TBI-induced lesion volume in GRP-transplanted mice compared with non-transplanted mice. Brain sections further validated the survival of grafted GRPs and showed more GRPs surrounding the injured tissue. Furthermore, the brains of post-TBI shiverer mice had increased activation of microglia and astrocytes compared to post-TBI wildtype mice, but infiltration of CD45+ leukocytes remained low. CONCLUSIONS: CoB induces sustained immunological tolerance towards allografted cerebral GRPs which is not disrupted following TBI, and unexpectedly TBI may enhance GRPs engraftment and contribute to post-injury brain tissue repair.


Brain Injuries, Traumatic , Graft Rejection/immunology , Immune Tolerance/immunology , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Allografts , Animals , Antibodies, Monoclonal/pharmacology , B7-1 Antigen/antagonists & inhibitors , B7-2 Antigen/antagonists & inhibitors , CD28 Antigens/antagonists & inhibitors , CD40 Antigens/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Neuroglia/transplantation
4.
ACS Appl Mater Interfaces ; 13(20): 23423-23437, 2021 May 26.
Article En | MEDLINE | ID: mdl-33978398

Transplanted glial-restricted progenitor (GRP) cells have potential to focally replace defunct astrocytes and produce remyelinating oligodendrocytes to avert neuronal death and dysfunction. However, most central nervous system cell therapeutic paradigms are hampered by high initial cell death and a host anti-graft immune response. We show here that composite hyaluronic acid-based hydrogels of tunable mechanical strengths can significantly improve transplanted GRP survival and differentiation. Allogeneic GRPs expressing green fluorescent protein and firefly luciferase were scaffolded in optimized hydrogel formulations and transplanted intracerebrally into immunocompetent BALB/c mice followed by serial in vivo bioluminescent imaging and chemical exchange saturation transfer magnetic resonance imaging (CEST MRI). We demonstrate that gelatin-sensitive CEST MRI can be exploited to monitor hydrogel scaffold degradation in vivo for ∼5 weeks post transplantation without necessitating exogenous labeling. Hydrogel scaffolding of GRPs resulted in a 4.5-fold increase in transplanted cell survival at day 32 post transplantation compared to naked cells. Histological analysis showed significant enhancement of cell proliferation as well as Olig2+ and GFAP+ cell differentiation for scaffolded cells compared to naked cells, with reduced host immunoreactivity. Hence, hydrogel scaffolding of transplanted GRPs in conjunction with serial in vivo imaging of cell survival and hydrogel degradation has potential for further advances in glial cell therapy.


Cell Survival/physiology , Hydrogels/chemistry , Neuroglia , Optical Imaging/methods , Stem Cells , Animals , Cell Differentiation/physiology , Cell Tracking , Hyaluronic Acid/chemistry , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred BALB C , Neuroglia/cytology , Neuroglia/physiology , Neuroglia/transplantation , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/physiology
5.
J Biomed Mater Res A ; 109(5): 779-787, 2021 05.
Article En | MEDLINE | ID: mdl-32720459

Spinal cord decellularized (DC) scaffolds can promote axonal regeneration and restore hindlimb motor function of spinal cord defect rats. However, scarring caused by damage to the astrocytes at the margin of injury can hinder axon regeneration. Olfactory ensheathing cells (OECs) integrate and migrate with astrocytes at the site of spinal cord injury, providing a bridge for axons to penetrate the scars and grow into lesion cores. The purpose of this study was to evaluate whether DC scaffolds carrying OECs could better promote axon growth. For these studies, DC scaffolds were cocultured with primary extracted and purified OECs. Immunofluorescence and electron microscopy were used for verification of cells adhere and growth on the scaffold. Scaffolds with OECs were transplanted into rat spinal cord defects to evaluate axon regeneration and functional recovery of hind limbs. Basso, Beattie, and Bresnahan (BBB) scoring was used to assess motor function recovery, and glial fibrillary acidic protein (GFAP) and NF200-stained tissue sections were used to evaluate axonal regeneration and astrological scar distribution. Our results indicated that spinal cord DC scaffolds have good histocompatibility and spatial structure, and can promote the proliferation of adherent OECs. In animal experiments, scaffolds carrying OECs have better axon regeneration promoting protein expression than the SCI model, and improve the proliferation and distribution of astrocytes at the site of injury. These results proved that the spinal cord DC scaffold with OECs can promote axon regeneration at the site of injury, providing a new basis for clinical application.


Neuroglia/transplantation , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Tissue Scaffolds , Animals , Axons/physiology , Biomarkers , Cells, Cultured , Coculture Techniques , Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/prevention & control , Gliosis/etiology , Materials Testing , Neuroglia/physiology , Olfactory Bulb/cytology , Random Allocation , Rats , Rats, Sprague-Dawley , Spinal Cord , Spinal Cord Injuries/complications
6.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article En | MEDLINE | ID: mdl-33255323

The translation of new therapies for spinal cord injury to clinical trials can be facilitated with large animal models close in morpho-physiological scale to humans. Here, we report functional restoration and morphological reorganization after spinal contusion in pigs, following a combined treatment of locomotor training facilitated with epidural electrical stimulation (EES) and cell-mediated triple gene therapy with umbilical cord blood mononuclear cells overexpressing recombinant vascular endothelial growth factor, glial-derived neurotrophic factor, and neural cell adhesion molecule. Preliminary results obtained on a small sample of pigs 2 months after spinal contusion revealed the difference in post-traumatic spinal cord outcomes in control and treated animals. In treated pigs, motor performance was enabled by EES and the corresponding morpho-functional changes in hind limb skeletal muscles were accompanied by the reorganization of the glial cell, the reaction of stress cell, and synaptic proteins. Our data demonstrate effects of combined EES-facilitated motor training and cell-mediated triple gene therapy after spinal contusion in large animals, informing a background for further animal studies and clinical translation.


Electric Stimulation Therapy , Glial Cell Line-Derived Neurotrophic Factor/genetics , Neural Cell Adhesion Molecules/genetics , Spinal Cord Injuries/therapy , Vascular Endothelial Growth Factor A/genetics , Adenoviridae/genetics , Animals , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Epidural Space , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Humans , Motor Activity/genetics , Motor Activity/physiology , Neural Cell Adhesion Molecules/therapeutic use , Neuroglia/transplantation , Recovery of Function/genetics , Recovery of Function/radiation effects , Spinal Cord/physiopathology , Spinal Cord/radiation effects , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Swine/genetics , Vascular Endothelial Growth Factor A/therapeutic use
7.
Int J Mol Sci ; 21(19)2020 09 30.
Article En | MEDLINE | ID: mdl-33008128

Among the numerous candidates for cell therapy of the central nervous system (CNS), olfactory progenitors (OPs) represent an interesting alternative because they are free of ethical concerns, are easy to collect, and allow autologous transplantation. In the present study, we focused on the optimization of neuron production and maturation. It is known that plated OPs respond to various trophic factors, and we also showed that the use of Nerve Growth Factor (NGF) allowed switching from a 60/40 neuron/glia ratio to an 80/20 one. Nevertheless, in order to focus on the integration of OPs in mature neural circuits, we cocultured OPs in primary cultures obtained from the cortex and hippocampus of newborn mice. When dissociated OPs were plated, they differentiated into both glial and neuronal phenotypes, but we obtained a 1.5-fold higher viability in cortex/OP cocultures than in hippocampus/OP ones. The fate of OPs in cocultures was characterized with different markers such as BrdU, Map-2, and Synapsin, indicating a healthy integration. These results suggest that the integration of transplanted OPs might by affected by trophic factors and the environmental conditions/cell phenotypes of the host tissue. Thus, a model of coculture could provide useful information on key cell events for the use of progenitors in cell therapy.


Brain/metabolism , Neurons/metabolism , Olfactory Cortex/metabolism , Stem Cell Transplantation , Stem Cells/cytology , Animals , Brain/cytology , Brain/growth & development , Cell Differentiation/genetics , Cell Lineage/genetics , Central Nervous System/metabolism , Coculture Techniques , Humans , Mice , Nerve Growth Factor/genetics , Neuroglia/cytology , Neuroglia/metabolism , Neuroglia/transplantation , Neurons/transplantation , Olfactory Cortex/cytology , Olfactory Cortex/transplantation , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oligodendroglia/transplantation , Stem Cells/metabolism
8.
Cell Mol Life Sci ; 77(24): 5171-5188, 2020 Dec.
Article En | MEDLINE | ID: mdl-32617639

Glial cells have been identified more than 100 years ago, and are known to play a key role in the central nervous system (CNS) function. A recent piece of evidence is emerging showing that in addition to the capacity of CNS modulation and homeostasis, glial cells are also being looked like as a promising cell source not only to study CNS pathologies initiation and progression but also to the establishment and development of new therapeutic strategies. Thus, in the present review, we will discuss the current evidence regarding glial cells' contribution to neurodegenerative diseases as Parkinson's disease, providing cellular, molecular, functional, and behavioral data supporting its active role in disease initiation, progression, and treatment. As so, considering their functional relevance, glial cells may be important to the understanding of the underlying mechanisms regarding neuronal-glial networks in neurodegeneration/regeneration processes, which may open new research opportunities for their future use as a target or treatment in human clinical trials.


Cell- and Tissue-Based Therapy , Neuroglia/transplantation , Neurons/transplantation , Parkinson Disease/therapy , Central Nervous System/pathology , Humans , Nerve Degeneration/pathology , Nerve Degeneration/therapy , Neurons/pathology , Parkinson Disease/pathology
9.
Exp Neurol ; 329: 113316, 2020 07.
Article En | MEDLINE | ID: mdl-32304749

The development and translation of cell therapies have been hindered by an inability to predict and evaluate their efficacy after transplantation. Using an experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis (MS), we studied attenuation of the diffuse injury characteristic of EAE and MS by transplanted glial-restricted precursor cells (GRPs). We assessed the potential of on-resonance variable delay multiple pulse (onVDMP) chemical exchange saturation transfer (CEST) MRI to visualize this attenuation. Allogeneic GRPs transplanted in the motor cortex or lateral ventricles attenuated paralysis in EAE mice and attenuated differences compared to naïve mice in onVDMP CEST signal 5 days after transplantation near the transplantation site. Histological analysis revealed that transplanted GRPs co-localized with attenuated astrogliosis. Hence, diffuse injury-sensitive onVDMP CEST MRI may complement conventional MRI to locate and monitor tissue regions responsive to GRP therapy.


Cell Transplantation/methods , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Encephalomyelitis, Autoimmune, Experimental/therapy , Magnetic Resonance Imaging/methods , Neuroglia/transplantation , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Luminescent Measurements/methods , Mice , Mice, Transgenic , Neuroglia/metabolism
10.
Methods Mol Biol ; 2140: 159-170, 2020.
Article En | MEDLINE | ID: mdl-32207111

Bioprinting cells with an electrically conductive bioink provides an opportunity to produce three-dimensional (3D) cell-laden constructs with the option of electrically stimulating cells in situ during and after tissue development. We and others have demonstrated the use of electrical stimulation (ES) to influence cell behavior and function for a more biomimetic approach to tissue engineering. Here, we detail a previously published method for 3D printing an electrically conductive bioink with human neural stem cells (hNSCs) that are subsequently differentiated. The differentiated tissue constructs comprise functional neurons and supporting neuroglia and are amenable to ES for the purposeful modulation of neural activity. Importantly, the method could be adapted to fabricate and stimulate neural and nonneural tissues from other cell types, with the potential to be applied for both research- and clinical-product development.


Biocompatible Materials , Bioprinting , Neural Stem Cells , Printing, Three-Dimensional , Tissue Engineering/methods , Calcium/analysis , Cells, Cultured , Electric Conductivity , Electric Stimulation , Fluorescent Dyes , Humans , Immunophenotyping , Microscopy, Confocal/methods , Neural Stem Cells/transplantation , Neurogenesis , Neuroglia/transplantation , Neurons/transplantation , Single-Cell Analysis
11.
J Neurotrauma ; 37(5): 817-829, 2020 03 01.
Article En | MEDLINE | ID: mdl-32056492

Cell transplantation constitutes an important avenue for development of new treatments for spinal cord injury (SCI). These therapies are aimed at supporting neural repair and/or replacing lost cells at the injury site. To date, various cell types have been trialed, with most studies focusing on different types of stem cells or glial cells. Here, we review commonly used cell transplantation approaches for spinal cord injury (SCI) repair, with focus on transplantation of olfactory ensheathing cells (OECs), the glial cells of the primary olfactory nervous system. OECs are promising candidates for promotion of neural repair given that they support continuous regeneration of the olfactory nerve that occurs throughout life. Further, OECs can be accessed from the nasal mucosa (olfactory neuroepithelium) at the roof of the nasal cavity and can be autologously transplanted. OEC transplantation has been trialed in many animal models of SCI, as well as in human clinical trials. While several studies have been promising, outcomes are variable and the method needs improvement to enhance aspects such as cell survival, integration, and migration. As a case study, we include the approaches used by our team (the Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia) to address the current problems with OEC transplantation and discuss how the therapeutic potential of OEC transplantation can be improved. Our approach includes discovery research to improve our knowledge of OEC biology, identifying natural and synthetic compounds to stimulate the neural repair properties of OECs, and designing three-dimensional cell constructs to create stable and transplantable cell structures.


Cell Transplantation/methods , Neuroglia/transplantation , Spinal Cord Injuries/therapy , Spinal Cord Regeneration/physiology , Animals , Humans , Nerve Regeneration/physiology , Olfactory Bulb/transplantation
12.
Cell Transplant ; 28(1_suppl): 132S-159S, 2019 Dec.
Article En | MEDLINE | ID: mdl-31726863

Olfactory ensheathing cells (OECs), the glial cells of the primary olfactory nervous system, support the natural regeneration of the olfactory nerve that occurs throughout life. OECs thus exhibit unique properties supporting neuronal survival and growth. Transplantation of OECs is emerging as a promising treatment for spinal cord injury; however, outcomes in both animals and humans are variable and the method needs improvement and standardization. A major reason for the discrepancy in functional outcomes is the variability in survival and integration of the transplanted cells, key factors for successful spinal cord regeneration. Here, we review the outcomes of OEC transplantation in rodent models over the last 10 years, with a focus on survival and integration of the transplanted cells. We identify the key factors influencing OEC survival: injury type, source of transplanted cells, co-transplantation with other cell types, number and concentration of cells, method of delivery, and time of transplantation after the injury. We found that two key issues are hampering optimization and standardization of OEC transplantation: lack of (1) reliable methods for identifying transplanted cells, and (2) three-dimensional systems for OEC delivery. To develop OEC transplantation as a successful and standardized therapy for spinal cord injury, we must address these issues and increase our understanding of the complex parameters influencing OEC survival.


Neuroglia/transplantation , Olfactory Bulb/cytology , Olfactory Nerve/cytology , Spinal Cord Injuries/therapy , Animals , Cell Survival , Cell Transplantation/methods , Cell Transplantation/standards , Cells, Cultured , Disease Models, Animal , Humans , Nerve Regeneration , Neuroglia/cytology , Olfactory Nerve/pathology , Spinal Cord Regeneration , Time Factors
14.
Mol Imaging Biol ; 21(6): 1071-1078, 2019 12.
Article En | MEDLINE | ID: mdl-30850968

PURPOSE: We studied the feasibility of labeling hydrogel scaffolds with a fluorine nanoemulsion for 19F- magnetic resonance imaging (MRI) to enable non-invasive visualization of their precise placement and potential degradation. PROCEDURE: Hyaluronan-based hydrogels (activated hyaluronan, HA) with increasing concentrations of fluorine nanoemulsion (V-sense) were prepared to measure the gelation time and oscillatory stress at 1 h and 7 days after the beginning of gelation. All biomechanical measurements were conducted with an ARES 2 rheometer. Diffusion of fluorine from the hydrogel: Three hydrogels in various Vs to HA volumetric ratios (1:50, 1:10, and 1:5) were prepared in duplicate. Hydrogels were incubated at 37 °C. To induce diffusion, three hydrogels were agitated at 1000 rpm. 1H and 19F MRI scans were acquired at 1, 3, 7 days and 2 months after gel preparation on a Bruker Ascend 750 scanner. To quantify fluorine content, scans were analyzed using Voxel Tracker 2.0. Assessment of cell viability in vitro and in vivo: Luciferase-positive mouse glial-restricted progenitors (GRPs) were embedded in 0:1, 1:50, 1:10, and 1:5 Vs:HA mixtures (final cell concentration  =1 × 107/ml). For the in vitro assay, mixtures were placed in 96-wells plate in triplicate and bioluminescence was measured after 1, 3, 7, 14, 21, and 28 days. For in vivo experiments, Vs/HA mixtures containing GRPs were injected subcutaneously in SCID mice and BLI was acquired at 1, 3, 7, and 14 days post-injection. RESULTS: Mixing of V-sense at increasing ratios of 1:50, 1:10, and 1:5 v/v of fluorine/activated hyaluronan (HA) hydrogel gradually elongated the gelation time from 194 s for non-fluorinated controls to 304 s for 1:5 V-sense:HA hydrogels, while their elastic properties slightly decreased. There was no release of V-sense from hydrogels maintained in stationary conditions over 2 months. The addition of V-sense positively affected in vitro survival of scaffolded GRPs in a dose-dependent manner. CONCLUSIONS: These results show that hydrogel fluorination does not impair its beneficial properties for scaffolded cells, which may be used to visualize scaffolded GRP transplants with 19F MRI.


Emulsions/chemistry , Fluorine/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Nanoparticles/chemistry , Neuroglia/transplantation , Staining and Labeling , Animals , Cell Survival , Elastic Modulus , Fluorocarbons/chemistry , Injections, Spinal , Mice, SCID , Mice, Transgenic , Nanoparticles/ultrastructure , Polyethylene Glycols/chemistry , Rheology
15.
Sci Rep ; 8(1): 16490, 2018 11 07.
Article En | MEDLINE | ID: mdl-30405160

Disseminated diseases of the central nervous system such as amyotrophic lateral sclerosis (ALS) require that therapeutic agents are delivered and distributed broadly. Intrathecal route is attractive in that respect, but to date there was no methodology available allowing for optimization of this technique to assure safety and efficacy in a clinically relevant setting. Here, we report on interventional, MRI-guided approach for delivery of hydrogel-embedded glial progenitor cells facilitating cell placement over extended surface of the spinal cord in pigs and in naturally occurring ALS-like disease in dogs. Glial progenitors used as therapeutic agent were embedded in injectable hyaluronic acid-based hydrogel to support their survival and prevent sedimentation or removal. Intrathecal space was reached through lumbar puncture and the catheter was advanced under X-ray guidance to the cervical part of the spine. Animals were then transferred to MRI suite for MRI-guided injection. Interventional and follow-up MRI as well as histopathology demonstrated successful and predictable placement of embedded cells and safety of the procedure.


Magnetic Resonance Imaging , Neuroglia/cytology , Neuroglia/transplantation , Stem Cell Transplantation , Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Survival , Hydrogels , Injections, Spinal , Magnetic Resonance Imaging/methods , Spinal Cord/diagnostic imaging , Spinal Cord/metabolism , Spinal Cord/pathology , Surgery, Computer-Assisted , Swine
16.
Cell Transplant ; 27(9): 1320-1327, 2018 09.
Article En | MEDLINE | ID: mdl-30095003

Trauma causes spinal cord injury, and the devastating consequences of the injury are due to the failure of the damaged central nervous system (CNS) axons to regenerate. Previous studies have shown that olfactory ensheathing cells (OECs) are a unique type of glial cell and they can promote regeneration of CNS axons to aid recovery after spinal cord injury. Transplantation of OECs, in particular from the olfactory bulb (OB), is considered one of the most promising therapeutic strategies for the repair of CNS injuries, including spinal cord injury. Transplantation of OECs can be autologous or allogenic. Here we focused on the less invasive and more error-proof allograft approach which needs a collection of donor OB tissue for OEC production. In this study, we investigated the effects on the yield and proportions of OECs and olfactory nerve fibroblasts (ONFs) from storing OB tissue in various media for periods of 24 and 48 hours. The OEC yield contributes to the viability of a successful cell transplant. We concluded that storing OB tissue for a period longer than 24 hours negatively impacted the total cell number and subsequently the OEC population. This study provides useful information for future clinical applications.


Neuroglia/cytology , Olfactory Bulb/cytology , Tissue Preservation/methods , Animals , Cell Count , Cell Culture Techniques , Cell Survival , Cells, Cultured , Male , Neuroglia/transplantation , Rats, Sprague-Dawley , Spinal Cord Injuries/therapy
17.
Exp Neurol ; 309: 119-133, 2018 11.
Article En | MEDLINE | ID: mdl-30056160

Olfactory ensheathing cells (OECs) are unique glia that support axon outgrowth in the olfactory system, and when used as cellular therapy after spinal cord injury, improve recovery and axon regeneration. Here we assessed the effects of combining OEC transplantation with another promising therapy, epidural electrical stimulation during a rehabilitative motor task. Sprague-Dawley rats received a mid-thoracic transection and transplantation of OECs or fibroblasts (FBs) followed by lumbar stimulation while climbing an inclined grid. We injected pseudorabies virus (PRV) into hindlimb muscles 7 months post-injury to assess connectivity across the transection. Analyses showed that the number of serotonergic (5-HT) axons that crossed the rostral scar border and the area of neurofilament-positive axons in the injury site were both greater in OEC- than FB-treated rats. We detected PRV-labeled cells rostral to the transection and remarkable evidence of 5-HT and PRV axons crossing the injury site in 1 OEC- and 1 FB-treated rat. The axons that crossed suggested either axon regeneration (OEC) or small areas of probable tissue sparing (FB). Most PRV-labeled thoracic neurons were detected in laminae VII or X, and ~25% expressed Chx10, a marker for V2a interneurons. These findings suggest potential regeneration or sparing of circuits that connect thoracic interneurons to lumbar somatic motor neurons. Despite evidence of axonal connectivity, no behavioral changes were detected in this small-scale study. Together these data suggest that when supplemented with epidural stimulation and climbing, OEC transplantation can increase axonal growth across the injury site and may promote recovery of propriospinal circuitry.


Axons/physiology , Cell Transplantation/methods , Electric Stimulation Therapy/methods , Neuroglia/physiology , Olfactory Bulb/cytology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy , Animals , Disease Models, Animal , Epidural Space/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Neuroglia/transplantation , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Transduction, Genetic
18.
Cell Transplant ; 27(6): 879-889, 2018 Jun.
Article En | MEDLINE | ID: mdl-29882418

Olfactory ensheathing cells (OECs) are glia reported to sustain the continuous axon extension and successful topographic targeting of the olfactory receptor neurons responsible for the sense of smell (olfaction). Due to this distinctive property, OECs have been trialed in human cell transplant therapies to assist in the repair of central nervous system injuries, particularly those of the spinal cord. Though many studies have reported neurological improvement, the therapy remains inconsistent and requires further improvement. Much of this variability stems from differing olfactory cell populations prior to transplantation into the injury site. While some studies have used purified cells, others have used unpurified transplants. Although both preparations have merits and faults, the latter increases the variability between transplants received by recipients. Without a robust purification procedure in OEC transplantation therapies, the full potential of OECs for spinal cord injury may not be realised.


Neuroglia/transplantation , Olfactory Bulb/cytology , Spinal Cord Injuries/therapy , Animals , Cell Separation/methods , Cell Transplantation/methods , Humans , Nerve Regeneration , Neuroglia/cytology , Olfactory Bulb/transplantation , Spinal Cord/physiopathology , Spinal Cord Injuries/physiopathology
19.
Cell Transplant ; 27(6): 867-878, 2018 Jun.
Article En | MEDLINE | ID: mdl-29852748

Autologous olfactory ensheathing cell (OEC) transplantation is a promising therapy for spinal cord injury; however, the efficacy varies between trials in both animals and humans. The main reason for this variability is that the purity and phenotype of the transplanted cells differs between studies. OECs are susceptible to modulation with neurotrophic factors, and thus, neurotrophins can be used to manipulate the transplanted cells into an optimal, consistent phenotype. OEC transplantation can be divided into 3 phases: (1) cell preparation, (2) cell administration, and (3) continuous support to the transplanted cells in situ. The ideal behaviour of OECs differs between these 3 phases; in the cell preparation phase, rapid cell expansion is desirable to decrease the time between damage and transplantation. In the cell administration phase, OEC survival and integration at the injury site, in particular migration into the glial scar, are the most critical factors, along with OEC-mediated phagocytosis of cellular debris. Finally, continuous support needs to be provided to the transplantation site to promote survival of both transplanted cells and endogenous cells within injury site and to promote long-term integration of the transplanted cells and angiogenesis. In this review, we define the 3 phases of OEC transplantation into the injured spinal cord and the optimal cell behaviors required for each phase. Optimising functional outcomes of OEC transplantation can be achieved by modulation of cell behaviours with neurotrophins. We identify the key growth factors that exhibit the strongest potential for optimizing the OEC phenotype required for each phase.


Nerve Growth Factors/therapeutic use , Neuroglia/transplantation , Olfactory Bulb/cytology , Spinal Cord Injuries/therapy , Animals , Cell Proliferation , Humans , Neuroglia/cytology , Spinal Cord Injuries/physiopathology , Spinal Cord Regeneration , Transplantation, Autologous
20.
Monoclon Antib Immunodiagn Immunother ; 37(2): 110-115, 2018 Apr.
Article En | MEDLINE | ID: mdl-29608408

Programmed cell death ligand-1 (PD-L1) is a type I transmembrane glycoprotein expressed on antigen-presenting cells. It is also expressed in several tumor cells such as melanoma and lung cancer cells. A strong correlation has been reported between human PD-L1 (hPD-L1) expression in tumor cells and negative prognosis in cancer patients. Here, a novel anti-hPD-L1 monoclonal antibody (mAb) L1Mab-13 (IgG1, kappa) was produced using a cell-based immunization and screening (CBIS) method. We investigated hPD-L1 expression in lung cancer using flow cytometry, Western blot, and immunohistochemical analyses. L1Mab-13 specifically reacted hPD-L1 of hPD-L1-overexpressed Chinese hamster ovary (CHO)-K1 cells and endogenous hPD-L1 of KMST-6 (human fibroblast) in flow cytometry and Western blot. Furthermore, L1Mab-13 reacted with lung cancer cell lines (EBC-1, Lu65, and Lu99) in flow cytometry and stained lung cancer tissues in a membrane-staining pattern in immunohistochemical analysis. These results indicate that a novel anti-hPD-L1 mAb, L1Mab-13, is very useful for detecting hPD-L1 of lung cancers in flow cytometry, Western blot, and immunohistochemical analyses.


Antibodies, Monoclonal/chemistry , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Immunoglobulin G/chemistry , Lung Neoplasms/diagnosis , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , B7-H1 Antigen/immunology , Biomarkers, Tumor/immunology , CHO Cells , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cricetulus , Flow Cytometry , Gene Expression , HEK293 Cells , Humans , Immunization/methods , Immunoglobulin G/biosynthesis , Immunoglobulin G/isolation & purification , Immunohistochemistry , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Neuroglia/immunology , Neuroglia/pathology , Neuroglia/transplantation , Prognosis
...