Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
2.
Exp Neurol ; 360: 114275, 2023 02.
Article in English | MEDLINE | ID: mdl-36379273

ABSTRACT

We present an updated, clinically relevant model of moderately severe bilateral cervical level 6 contusive spinal cord injury (SCI) in the rat. This model is more clinically relevant than previous models due it its severity, yet animals readily survive the lesion. The C6 bilateral lesion is administered to Fischer 344 rats using the Infinite Horizons impactor adjusted to a 200 kdyne force with a 3.5 mm impactor head. The lesion results in loss of 60 ± 10% of the spinal cord area, including virtually the entire dorsal half of the spinal cord and complete interruption of the main corticospinal tract. Skilled forelimb performance declines by 60 ± 10% compared to the pre-operative baseline and deficits are sustained over time. This model is a substantial step closer to mimicking the most common level (cervical) and more severe form of SCI in humans and should provide a superior tool for assessing the likelihood that experimental interventions may promote motor recovery after SCI in humans.


Subject(s)
Cervical Cord , Spinal Cord Injuries , Humans , Rats , Animals , Spinal Cord/pathology , Pyramidal Tracts/pathology , Forelimb , Upper Extremity , Recovery of Function , Disease Models, Animal
3.
Exp Neurol ; 359: 114259, 2023 01.
Article in English | MEDLINE | ID: mdl-36309123

ABSTRACT

Neural stem cells (NSCs) implanted into sites of spinal cord injury (SCI) extend very large numbers of new axons over very long distances caudal to the lesion site, and support partial functional recovery. Newly extending graft axons distribute throughout host gray and white matter caudal to the injury. We hypothesized that provision of trophic gradients caudal to the injury would provide neurotrophic guidance to newly extending graft-derived axons to specific intermediate and ventral host gray matter regions, thereby potentially further improving neural relay formation. Immunodeficient rats underwent C5 lateral hemisection lesions, following by implants of human NSC grafts two weeks later. After an additional two weeks, animals received injections of AAV2-BDNF expressing vectors three spinal segments (9 mm) caudal to the lesion in host ventral and intermediate gray matter. After 2 months additional survival, we found a striking, 5.5-fold increase in the density of human axons innervating host ventral gray matter (P < 0.05) and 2.7-fold increase in intermediate gray matter (P < 0.01). Moreover, stem cell-derived axons formed a substantially greater number of putative synaptic connections with host motor neurons (P < 0.01). Thus, trophic guidance is an effective means of enhancing and guiding neural stem cell axon growth after SCI and will be used in future experiments to determine whether neural relay formation and functional outcomes can be improved.


Subject(s)
Neural Stem Cells , Spinal Cord Injuries , Rats , Humans , Animals , Brain-Derived Neurotrophic Factor , Axons/pathology , Neural Stem Cells/transplantation , Motor Neurons/pathology , Interneurons/pathology , Spinal Cord/pathology , Nerve Regeneration/physiology
5.
JCI Insight ; 7(16)2022 08 22.
Article in English | MEDLINE | ID: mdl-35993363

ABSTRACT

We reported previously that neural progenitor cell (NPC) grafts form neural relays across sites of subacute spinal cord injury (SCI) and support functional recovery. Here, we examine whether NPC grafts after chronic delays also support recovery and whether intensive rehabilitation further enhances recovery. One month after severe bilateral cervical contusion, rats received daily intensive rehabilitation, NPC grafts, or both rehabilitation and grafts. Notably, only the combination of rehabilitation and grafting significantly improved functional recovery. Moreover, improved functional outcomes were associated with a rehabilitation-induced increase in host corticospinal axon regeneration into grafts. These findings identify a critical and synergistic role of rehabilitation and neural stem cell therapy in driving neural plasticity to support functional recovery after chronic and severe SCI.


Subject(s)
Neural Stem Cells , Spinal Cord Injuries , Animals , Axons , Nerve Regeneration , Rats , Spinal Cord Injuries/therapy , Stem Cell Transplantation
6.
Neural Regen Res ; 17(8): 1633-1639, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35017408

ABSTRACT

Amyotrophic lateral sclerosis is a motor neuron degenerative disease that is also known as Lou Gehrig's disease in the United States, Charcot's disease in France, and motor neuron disease in the UK. The loss of motor neurons causes muscle wasting, paralysis, and eventually death, which is commonly related to respiratory failure, within 3-5 years after onset of the disease. Although there are a limited number of drugs approved for amyotrophic lateral sclerosis, they have had little success at treating the associated symptoms, and they cannot reverse the course of motor neuron degeneration. Thus, there is still a lack of effective treatment for this debilitating neurodegenerative disorder. Stem cell therapy for amyotrophic lateral sclerosis is a very attractive strategy for both basic and clinical researchers, particularly as transplanted stem cells and stem cell-derived neural progenitor/precursor cells can protect endogenous motor neurons and directly replace the lost or dying motor neurons. Stem cell therapies may also be able to re-establish the motor control of voluntary muscles. Here, we review the recent progress in the use of neural stem cells and neural progenitor cells for the treatment of amyotrophic lateral sclerosis. We focus on MN progenitor cells derived from fetal central nervous system tissue, embryonic stem cells, and induced pluripotent stem cells. In our recent studies, we found that transplanted human induced pluripotent stem cell-derived motor neuron progenitors survive well, differentiate into motor neurons, and extend axons into the host white matter, not only in the rostrocaudal direction, but also along motor axon tracts towards the ventral roots in the immunodeficient rat spinal cord. Furthermore, the significant motor axonal extension after neural progenitor cell transplantation in amyotrophic lateral sclerosis models demonstrates that motor neuron replacement therapy could be a promising therapeutic strategy for amyotrophic lateral sclerosis, particularly as a variety of stem cell derivatives, including induced pluripotent stem cells, are being considered for clinical trials for various diseases.

7.
Cells ; 10(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34943804

ABSTRACT

Spinal cord injury (SCI) leads to irreversible functional impairment caused by neuronal loss and the disruption of neuronal connections across the injury site. While several experimental strategies have been used to minimize tissue damage and to enhance axonal growth and regeneration, the corticospinal projection, which is the most important voluntary motor system in humans, remains largely refractory to regenerative therapeutic interventions. To date, one of the most promising pre-clinical therapeutic strategies has been neural stem cell (NSC) therapy for SCI. Over the last decade we have found that host axons regenerate into spinal NSC grafts placed into sites of SCI. These regenerating axons form synapses with the graft, and the graft in turn extends very large numbers of new axons from the injury site over long distances into the distal spinal cord. Here we discuss the pathophysiology of SCI that makes the spinal cord refractory to spontaneous regeneration, the most recent findings of neural stem cell therapy for SCI, how it has impacted motor systems including the corticospinal tract and the implications for sensory feedback.


Subject(s)
Axons/physiology , Nerve Net/physiology , Nerve Regeneration/physiology , Neural Stem Cells/physiology , Spinal Cord/physiology , Humans , Neural Stem Cells/transplantation , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy
8.
eNeuro ; 8(1)2021.
Article in English | MEDLINE | ID: mdl-33441399

ABSTRACT

Astrocytes play several critical roles in the normal functioning of the mammalian brain, including ion homeostasis, synapse formation, and synaptic plasticity. Following injury and infection or in the setting of neurodegeneration, astrocytes become hypertrophic and reactive, a process termed astrogliosis. Although acute reactive gliosis is beneficial in limiting further tissue damage, chronic gliosis becomes detrimental for neuronal recovery and regeneration. Several extracellular factors have been identified that generate reactive astrocytes; however, very little is known about the cell-autonomous transcriptional mechanisms that regulate the maintenance of astrocytes in the normal non-reactive state. Here, we show that conditional deletion of the stimulus-dependent transcription factor, serum response factor (SRF) in astrocytes (SrfGFAPCKO) results in astrogliosis marked by hypertrophic morphology and increased expression of GFAP, vimentin, and nestin. These reactive astrocytes were not restricted to any specific brain region and were seen in both white and gray matter in the entire brain. This astrogliosis persisted throughout adulthood concomitant with microglial activation. Importantly, the Srf mutant mouse brain did not exhibit any cell death or blood brain barrier (BBB) deficits suggesting that apoptosis and leaky BBB are not the causes for the reactive phenotype. The mutant astrocytes expressed more A2 reactive astrocyte marker genes and the SrfGFAPCKO mice exhibited normal neuronal numbers indicating that SRF-deficient gliosis astrocytes are not neurotoxic. Together, our findings suggest that SRF plays a critical role in astrocytes to maintain them in a non-reactive state.


Subject(s)
Astrocytes , Serum Response Factor , Animals , Astrocytes/metabolism , Brain/metabolism , Central Nervous System , Glial Fibrillary Acidic Protein/metabolism , Gliosis , Mice
9.
Precis Clin Med ; 4(3): 168-175, 2021 Sep.
Article in English | MEDLINE | ID: mdl-35693220

ABSTRACT

Somatic gene therapy remains technically challenging, especially in the central nervous system (CNS). Efficiency of gene delivery, efficacy in recipient cells, and proportion of cells required for overall benefit are the key points needed to be considered in any therapeutic approach. Recent efforts have demonstrated the efficacy of RNA-guided nucleases such as CRISPR/Cas9 in correcting point mutations or removing dominant mutations. Here we used viral delivered Cas9 plasmid and two guide RNAs to remove a recessive insertional mutation, vibrator (vb), in the mouse brain. The vb mice expressed ∼20% of normal levels of phosphatidylinositol transfer protein, α (PITPα) RNA and protein due to an endogenous retrovirus inserted in intron 4, resulting in early-onset tremor, degeneration of brainstem and spinal cord neurons, and juvenile death. The in situ CRISPR/Cas9 viral treatment effectively delayed neurodegeneration, attenuated tremor, and bypassed juvenile death. Our studies demonstrate the potential of CRISPR/Cas9-mediated gene therapy for insertional mutations in the postnatal brain.

10.
Acad Med ; 96(3): 460-469, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33298696

ABSTRACT

PURPOSE: Stakeholders have expressed concerns regarding the impact of visiting trainees and physicians from high-income countries (HICs) providing education and/or short-term clinical care in low- and middle-income countries (LMICs). This systematic review aimed to summarize LMIC host perceptions of visiting trainees and physicians from HICs during short-term experiences in global health (STEGH). METHOD: In September 2018 then again in August 2020, the authors searched 7 databases (PubMed, Embase, Scopus, Web of Science, ERIC, Cochrane Library, Global Index Medicus) for peer-reviewed studies that described LMIC host perceptions of STEGH. They extracted information pertaining to study design, participant demographics, participant perceptions, representation of LMICs and HICs, and HIC visitors' roles and used thematic synthesis to code the text, develop descriptive themes, and generate analytical themes. RESULTS: Of the 4,020 studies identified, 17 met the inclusion criteria. In total, the studies included 448 participants, of which 395 (88%) represented LMICs. The authors identified and organized 42 codes under 8 descriptive themes. They further organized these descriptive themes into 4 analytical themes related to STEGH: (1) sociocultural and contextual differences, (2) institutional and programmatic components, (3) impact on host institutions and individuals, and (4) visitor characteristics and conduct. CONCLUSIONS: STEGH can have both beneficial and detrimental effects on LMIC host institutions and individuals. The authors translated these findings into a set of evidence-based best practices for STEGH that provide specific guidance for LMIC and HIC stakeholders. Moving forward, LMIC and HIC institutions must work together to focus on the quality of their relationships and create conditions in which all stakeholders feel empowered to openly communicate to ensure equity and mutual benefit for all parties.


Subject(s)
Developing Countries/economics , Evidence-Based Practice/standards , Global Health/education , Perception/physiology , Cross-Cultural Comparison , Evidence-Based Practice/trends , Female , Global Health/statistics & numerical data , Humans , International Cooperation , International Educational Exchange/trends , Male , Peer Review , Publication Bias , Quality Improvement , Stakeholder Participation/psychology , Thematic Apperception Test/standards
11.
Adv Exp Med Biol ; 1266: 1-8, 2020.
Article in English | MEDLINE | ID: mdl-33105491

ABSTRACT

Neurodegenerative diseases (NDs) are a group of neurological diseases caused by the progressive degeneration of neurons and glial cells in the brain and spinal cords. Usually there is a selective loss of specific neuronal cells in a restricted brain area from any neurodegenerative diseases, such as dopamine (DA) neuron death in Parkinson disease (PD) and motor neuron loss in amyotrophic lateral sclerosis (ALS), or a widespread degeneration affecting many types of neurons in Alzheimer's disease (AD). As there is no effective treatment to stop the progression of these neurodegenerative diseases, stem cell-based therapies have provided great potentials for these disorders. Currently transplantation of different stem cells or their derivatives has improved neural function in animal models of neurodegenerative diseases by replacing the lost neural cells, releasing cytokines, modulation of inflammation, and mediating remyelination. With the advance in somatic cell reprogramming to generate induced pluripotent stem cells (iPS cells) and directly induced neural stem cells or neurons, pluripotent stem cell can be induced to differentiate to any kind of neural cells and overcome the immune rejection of the allogeneic transplantation. Recent studies have proved the effectiveness of transplanted stem cells in animal studies and some clinical trials on patients with NDs. However, some significant hurdles need to be resolved before these preclinical results can be translated to clinic. In particular, we need to better understand the molecular mechanisms of stem cell transplantation and develop new approaches to increase the directed neural differentiation, migration, survival, and functional connections of transplanted stem cells in the pathological environment of the patient's central nerve system.


Subject(s)
Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Pluripotent Stem Cells , Stem Cell Transplantation , Amyotrophic Lateral Sclerosis/therapy , Animals , Humans , Neural Stem Cells , Neurodegenerative Diseases/therapy , Parkinson Disease/therapy
12.
Adv Exp Med Biol ; 1266: 71-97, 2020.
Article in English | MEDLINE | ID: mdl-33105496

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a motor neuronal degeneration disease, in which the death of motor neurons causes lost control of voluntary muscles. The consequence is weakness of muscles with a wide range of disabilities and eventually death. Most patients died within 5 years after diagnosis, and there is no cure for this devastating neurodegenerative disease up to date. Stem cells, including non-neural stem cells and neural stem cells (NSCs) or neural progenitor cells (NPCs), are very attractive cell sources for potential neuroprotection and motor neuron replacement therapy which bases on the idea that transplant-derived and newly differentiated motor neurons can replace lost motor neurons to re-establish voluntary motor control of muscles in ALS. Our recent studies show that transplanted NSCs or NPCs not only survive well in injured spinal cord, but also function as neuronal relays to receive regenerated host axonal connection and extend their own axons to host for connectivity, including motor axons in ventral root. This reciprocal connection between host neurons and transplanted neurons provides a strong rationale for neuronal replacement therapy for ALS to re-establish voluntary motor control of muscles. In addition, a variety of new stem cell resources and the new methodologies to generate NSCs or motor neuron-specific progenitor cells have been discovered and developed. Together, it provides the basis for motor neuron replacement therapy with NSCs or NPCs in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Neural Stem Cells , Stem Cell Transplantation , Amyotrophic Lateral Sclerosis/therapy , Animals , Disease Models, Animal , Humans , Motor Neurons/pathology
13.
Adv Exp Med Biol ; 1266: 99-115, 2020.
Article in English | MEDLINE | ID: mdl-33105497

ABSTRACT

Multiple sclerosis (MS) is the most frequent demyelinating disease of the central nervous system (CNS) associated with inflammatory plaques of white matter demyelination, oligodendrocyte destruction, reactive gliosis and axonal degeneration. In this chapter, we first review the pathological process of axonal degeneration in MS and discuss how these changes cause clinical symptoms of MS. We then discuss the pharmacological treatment to improve the clinical symptoms. Finally, we highlight how the autologous hematopoietic stem cell transplantation (AHSCT) can be effective for aggressive MS patients, who fail to respond to drug therapies, and also propose the future challenges of AHSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Sclerosis , Humans , Multiple Sclerosis/therapy , Oligodendroglia/pathology , Transplantation, Autologous
14.
Adv Exp Med Biol ; 1266: 141-145, 2020.
Article in English | MEDLINE | ID: mdl-33105500

ABSTRACT

Stem cell-based therapy has shown exciting efficacy in pre-clinical studies on different neurodegenerative diseases (NDs). However, no clinically applicable stem-cell-derived neurons are available to the patients with NDs. There exist some obstacles associated with stem cell therapy, which need to be overcome in future clinical studies. In this chapter, more challenges and new strategies will be explored to accelerate the clinical translation of a human embryonic stem cell (hESC)/induced pluripotent stem cell (iPSC)-derived neural cell product to patients with NDs.


Subject(s)
Neurodegenerative Diseases , Stem Cell Transplantation , Cell Differentiation , Embryonic Stem Cells , Humans , Induced Pluripotent Stem Cells , Neurodegenerative Diseases/therapy , Neurons
15.
Nature ; 581(7806): 77-82, 2020 05.
Article in English | MEDLINE | ID: mdl-32376949

ABSTRACT

Grafts of spinal-cord-derived neural progenitor cells (NPCs) enable the robust regeneration of corticospinal axons and restore forelimb function after spinal cord injury1; however, the molecular mechanisms that underlie this regeneration are unknown. Here we perform translational profiling specifically of corticospinal tract (CST) motor neurons in mice, to identify their 'regenerative transcriptome' after spinal cord injury and NPC grafting. Notably, both injury alone and injury combined with NPC grafts elicit virtually identical early transcriptomic responses in host CST neurons. However, in mice with injury alone this regenerative transcriptome is downregulated after two weeks, whereas in NPC-grafted mice this transcriptome is sustained. The regenerative transcriptome represents a reversion to an embryonic transcriptional state of the CST neuron. The huntingtin gene (Htt) is a central hub in the regeneration transcriptome; deletion of Htt significantly attenuates regeneration, which shows that Htt has a key role in neural plasticity after injury.


Subject(s)
Cell Proliferation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Nerve Regeneration/genetics , Neural Stem Cells/cytology , Neurons/metabolism , Neurons/pathology , Transcription, Genetic , Animals , Axons/pathology , Axons/physiology , Disease Models, Animal , Female , Gene Expression Profiling , Huntingtin Protein/genetics , Mice , Neural Stem Cells/transplantation , Neuronal Plasticity , Neurons/cytology , Neurons/transplantation , Protein Biosynthesis , Pyramidal Tracts/cytology , Pyramidal Tracts/metabolism , Pyramidal Tracts/pathology , RNA-Seq , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Transcriptome
16.
Stem Cell Reports ; 13(1): 105-114, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31204300

ABSTRACT

Neural progenitor cells (NPCs) transplanted into sites of spinal cord injury (SCI) extend large numbers of axons into the caudal host spinal cord. We determined the precise locations of neurons in the graft that extend axons into the caudal host spinal cord using AAV9-Cre-initiated retrograde tracing into floxed-TdTomato-expressing NPC grafts. 7,640 ± 630 grafted neurons extended axons to a single caudal host spinal cord site located 2 mm beyond the lesion, 5 weeks post injury. While caudally projecting axons arose from neurons located in all regions of the graft, the majority of caudally projecting graft neurons (53%) were located within the caudal one-third of the graft. Numerous host corticospinal axons formed monosynaptic projections onto caudally projecting graft neurons; however, we find that the majority of host axonal neuronal projections formed by neural progenitor cell interneuronal "relays" across sites of SCI are likely polysynaptic in nature.


Subject(s)
Cell Differentiation , Neural Stem Cells/metabolism , Neuronal Outgrowth , Neurons/metabolism , Spinal Cord Injuries/metabolism , Animals , Biological Transport , Fluorescent Antibody Technique/methods , Gene Expression , Genes, Reporter , Mice , Nerve Regeneration , Neural Stem Cells/cytology , Pyramidal Tracts , Rats , Spinal Cord Injuries/etiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Transduction, Genetic
17.
Cell Rep ; 26(9): 2329-2339.e4, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30811984

ABSTRACT

Neural progenitor cell grafts form new relays across sites of spinal cord injury (SCI). Using a panel of neuronal markers, we demonstrate that spinal neural progenitor grafts to sites of rodent SCI adopt diverse spinal motor and sensory interneuronal fates, representing most neuronal subtypes of the intact spinal cord, and spontaneously segregate into domains of distinct cell clusters. Host corticospinal motor axons regenerating into neural progenitor grafts innervate appropriate pre-motor interneurons, based on trans-synaptic tracing with herpes simplex virus. A human spinal neural progenitor cell graft to a non-human primate also received topographically appropriate corticospinal axon regeneration. Thus, grafted spinal neural progenitor cells give rise to a variety of neuronal progeny that are typical of the normal spinal cord; remarkably, regenerating injured adult corticospinal motor axons spontaneously locate appropriate motor domains in the heterogeneous, developing graft environment, without a need for additional exogenous guidance.


Subject(s)
Interneurons/physiology , Motor Neurons/physiology , Nerve Regeneration , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Spine/innervation , Animals , Axons/physiology , Female , Humans , Macaca mulatta , Male , Neural Stem Cells/physiology , Neurons/physiology , Phenotype , Rats , Rats, Inbred F344 , Spinal Cord Injuries/physiopathology
18.
Nat Med ; 25(2): 263-269, 2019 02.
Article in English | MEDLINE | ID: mdl-30643285

ABSTRACT

Current methods for bioprinting functional tissue lack appropriate biofabrication techniques to build complex 3D microarchitectures essential for guiding cell growth and promoting tissue maturation1. 3D printing of central nervous system (CNS) structures has not been accomplished, possibly owing to the complexity of CNS architecture. Here, we report the use of a microscale continuous projection printing method (µCPP) to create a complex CNS structure for regenerative medicine applications in the spinal cord. µCPP can print 3D biomimetic hydrogel scaffolds tailored to the dimensions of the rodent spinal cord in 1.6 s and is scalable to human spinal cord sizes and lesion geometries. We tested the ability of µCPP 3D-printed scaffolds loaded with neural progenitor cells (NPCs) to support axon regeneration and form new 'neural relays' across sites of complete spinal cord injury in vivo in rodents1,2. We find that injured host axons regenerate into 3D biomimetic scaffolds and synapse onto NPCs implanted into the device and that implanted NPCs in turn extend axons out of the scaffold and into the host spinal cord below the injury to restore synaptic transmission and significantly improve functional outcomes. Thus, 3D biomimetic scaffolds offer a means of enhancing CNS regeneration through precision medicine.


Subject(s)
Biomimetics , Nerve Regeneration , Printing, Three-Dimensional , Spinal Cord Injuries/therapy , Tissue Scaffolds/chemistry , Animals , Green Fluorescent Proteins/metabolism , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Neural Stem Cells/ultrastructure , Rats
19.
Exp Neurol ; 314: 46-57, 2019 04.
Article in English | MEDLINE | ID: mdl-30653967

ABSTRACT

Neural stem cells (NSCs) can differentiate into both neurons and glia after transplantation into spinal cord injury (SCI) sites. The neuronal component of stem cell grafts has the potential to form functional synaptic relays across the lesion site. The glial component may reform a blood-spinal cord barrier, support neuronal function, and contribute to remyelination. We performed a long-term, 1.5-year time course study focused on astrocyte migration, differentiation, integration, and safety following human NSC transplantation into C5 hemisection sites in immunodeficient rats. NSCs that adopted a neuronal fate did not migrate from the lesion site. In contrast, transplanted cells that adopted astrocyte fates exhibited long distance migration from the lesion site through host white matter in rostrocaudal directions. These cells migrated slowly at a mean rate of 2-3 mm/month, divided as they migrated, and gradually differentiated into astrocytes. After 1.5 years, human astrocytes migrated nine spinal cord segments, caudally to the mid-thoracic level, and rostrally into the brainstem. The migrated human astrocytes joined the endogenous population of astrocytes in the host spinal cord, extended perivascular endfeet towards host pericytes and endothelium, formed interspecies and intraspecies perivascular astrocytic networks connected by gap junctions, and expressed glutamate transporter proteins in perisynaptic processes, suggesting structural and functional integration. No adverse consequences of this extended glial migration were detected. Adjacent to the lesion site, chronic host astrocytic upregulation was significantly attenuated by NSC grafts. Thus, human astrocytes can migrate long distances from sites of SCI and safely integrate into the host central nervous system. SIGNIFICANCE STATEMENT: Neural stem cell (NSC) grafts are a candidate therapy for spinal cord injury (SCI). Here we report an 18-month study of astrocyte survival and migration from sites of SCI in immunodeficient rats. NSC grafts significantly attenuate host astrocyte reactivity at the lesion/host interface. Intra-graft astrocytes integrate into the host blood-spinal cord barrier (BSCB) and widely express glutamate transporter proteins characteristic of neurotransmitter regulation. Notably, astrocytic components of NSC grafts exhibit gradual yet extensive migration after implantation into the mid-cervical injury site; neurons do not migrate at all. This extensive astrocyte migration is not detectably associated with adverse outcomes anatomically or behaviorally.


Subject(s)
Astrocytes , Cell Movement/physiology , Neural Stem Cells , Spinal Cord Injuries/therapy , Animals , Brain Stem/cytology , Cell Survival , Female , Gap Junctions , Heterografts , Humans , Immunocompromised Host , Nerve Net/cytology , Neural Stem Cells/metabolism , Neurogenesis , Rats , Rats, Nude , Spinal Cord/cytology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/psychology , Stem Cell Transplantation , Vesicular Glutamate Transport Proteins/metabolism
20.
Stem Cell Reports ; 11(4): 861-868, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30197116

ABSTRACT

Axonal regeneration after spinal cord injury (SCI) can be enhanced by activation of the intrinsic neuronal growth state and, separately, by placement of growth-enabling neural progenitor cell (NPC) grafts into lesion sites. Indeed, NPC grafts support regeneration of all host axonal projections innervating the normal spinal cord. However, some host axons regenerate only short distances into grafts. We examined whether activation of the growth state of the host injured neuron would elicit greater regeneration into NPC grafts. Rats received NPC grafts into SCI lesions in combination with peripheral "conditioning" lesions. Six weeks later, conditioned host sensory axons exhibited a significant, 9.6-fold increase in regeneration into the lesion/graft site compared with unconditioned axons. Regeneration was further enhanced 1.6-fold by enriching NPC grafts with phenotypically appropriate sensory neuronal targets. Thus, activation of the intrinsic host neuronal growth state and manipulation of the graft environment enhance axonal regeneration after SCI.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Neural Stem Cells/transplantation , Animals , Female , Humans , Macaca mulatta , Male , Phenotype , Rats, Inbred F344 , Sensory Receptor Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...