Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 11(14)2022 07 08.
Article in English | MEDLINE | ID: mdl-35883596

ABSTRACT

(1) Background: the use of Mesenchymal Stromal Cells (MSC) in emerging therapies for spinal cord injury (SCI) hold the potential to improve functional recovery. However, the development of cell-based medicines is challenging and preclinical studies addressing quality, safety and efficacy must be conducted prior to clinical testing; (2) Methods: herein we present (i) the characterization of the quality attributes of MSC from the Wharton's jelly (WJ) of the umbilical cord, (ii) safety of intrathecal infusion in a 3-month subchronic toxicity assessment study, and (iii) efficacy in a rat SCI model by controlled impaction (100 kdynes) after single (day 7 post-injury) and repeated dose of 1 × 106 MSC,WJ (days 7 and 14 post-injury) with 70-day monitoring by electrophysiological testing, motor function assessment and histology evaluation; (3) Results: no toxicity associated to MSC,WJ infusion was observed. Regarding efficacy, recovery of locomotion was promoted at early time points. Persistence of MSC,WJ was detected early after administration (day 2 post-injection) but not at days 14 and 63 post-injection. (4) Conclusions: the safety profile and signs of efficacy substantiate the suitability of the presented data for inclusion in the Investigational Medicinal Product Dossier for further consideration by the competent Regulatory Authority to proceed with clinical trials.


Subject(s)
Mesenchymal Stem Cells , Spinal Cord Injuries , Wharton Jelly , Animals , Cells, Cultured , Humans , Rats , Spinal Cord Injuries/therapy , Umbilical Cord
2.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073117

ABSTRACT

We currently lack effective treatments for the devastating loss of neural function associated with spinal cord injury (SCI). In this study, we evaluated a combination therapy comprising human neural stem cells derived from induced pluripotent stem cells (iPSC-NSC), human mesenchymal stem cells (MSC), and a pH-responsive polyacetal-curcumin nanoconjugate (PA-C) that allows the sustained release of curcumin. In vitro analysis demonstrated that PA-C treatment protected iPSC-NSC from oxidative damage in vitro, while MSC co-culture prevented lipopolysaccharide-induced activation of nuclear factor-κB (NF-κB) in iPSC-NSC. Then, we evaluated the combination of PA-C delivery into the intrathecal space in a rat model of contusive SCI with stem cell transplantation. While we failed to observe significant improvements in locomotor function (BBB scale) in treated animals, histological analysis revealed that PA-C-treated or PA-C and iPSC-NSC + MSC-treated animals displayed significantly smaller scars, while PA-C and iPSC-NSC + MSC treatment induced the preservation of ß-III Tubulin-positive axons. iPSC-NSC + MSC transplantation fostered the preservation of motoneurons and myelinated tracts, while PA-C treatment polarized microglia into an anti-inflammatory phenotype. Overall, the combination of stem cell transplantation and PA-C treatment confers higher neuroprotective effects compared to individual treatments.


Subject(s)
Curcumin/pharmacology , Mesenchymal Stem Cell Transplantation , Nanoconjugates/therapeutic use , Neuroprotective Agents/pharmacology , Recovery of Function , Spinal Cord Injuries/therapy , Acetals/therapeutic use , Animals , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Neural Stem Cells , Polymers/therapeutic use , Rats , Rats, Sprague-Dawley
3.
Front Mol Neurosci ; 14: 582497, 2021.
Article in English | MEDLINE | ID: mdl-34093123

ABSTRACT

Research on microglia has established the differentiation between the so-called M1 and M2 phenotypes. However, new frameworks have been proposed attempting to discern between meaningful microglia profiles. We have set up an in vitro microglial activation model by adding an injured spinal cord (SCI) lysate to microglial cultures, obtained from postnatal rats, in order to mimic the environment of the spinal cord after injury. We found that under the presence of the SCI lysate microglial cells changed their phenotype, developing less ramified but longer processes, and proliferated. The SCI lysate also led to upregulation of pro-inflammatory cytokines, such as IL-1ß, IL-6, and TNF-α, downregulation of the anti-inflammatory cytokines IL-10 and IL-4, and a biphasic profile of iNOS. In addition, a latex beads phagocytosis assay revealed the SCI lysate stimulated the phagocytic capacity of microglia. Flow cytometry analysis indicated that microglial cells showed a pro-inflammatory profile in the presence of SCI lysate. Finally, characterization of the microglial activation in the spinal cord on day 7 after contusion injury, we showed that these cells have a pro-inflammatory phenotype. Overall, these results indicate that the use of SCI lysates could be a useful tool to skew microglia towards a closer phenotype to that observed after the spinal cord contusion injury than the use of LPS or IFNγ.

4.
Cytotherapy ; 23(2): 146-156, 2021 02.
Article in English | MEDLINE | ID: mdl-32981857

ABSTRACT

BACKGROUND AIMS: Spinal cord injury (SCI) represents a devastating condition leading to severe disability related to motor, sensory and autonomic dysfunction. Stem cell transplantation is considered a potential emerging therapy to stimulate neuroplastic and neuroregenerative processes after SCI. In this clinical trial, the authors investigated the safety and clinical recovery effects of intrathecal infusion of expanded Wharton jelly mesenchymal stromal cells (WJ-MSCs) in chronic complete SCI patients. METHODS: The authors designed a randomized, double-blind, crossover, placebo-controlled, phase 1/2a clinical trial (NCT03003364). Participants were 10 patients (7 males, 3 females, age range, 25-47 years) with chronic complete SCI (American Spinal Injury Association A) at dorsal level (T3-11). Patients were randomly assigned to receive a single dose of intrathecal ex vivo-expanded WJ-MSCs (10 × 106 cells) from human umbilical cord or placebo and were then switched to the other arm at 6 months. Clinical evaluation (American Spinal Injury Association impairment scale motor and sensory score, spasticity, neuropathic pain, electrical perception and pain thresholds), lower limb motor evoked potentials (MEPs) and sensory evoked potentials (SEPs), Spinal Cord Independence Measure and World Health Organization Quality of Life Brief Version were assessed at baseline, 1 month, 3 months and 6 months after each intervention. Urodynamic studies and urinary-specific quality of life (Qualiveen questionnaire) as well as anorectal manometry, functional assessment of bowel dysfunction (Rome III diagnostic questionnaire) and severity of fecal incontinence (Wexner score) were conducted at baseline and at 6 months after each intervention. RESULTS: Intrathecal transplantation of WJ-MSCs was considered safe, with no significant side effects. Following MSC infusion, the authors found significant improvement in pinprick sensation in the dermatomes below the level of injury compared with placebo. Other clinically relevant effects, such as an increase in bladder maximum capacity and compliance and a decrease in bladder neurogenic hyperactivity and external sphincter dyssynergy, were observed only at the individual level. No changes in motor function, spasticity, MEPs, SEPs, bowel function, quality of life or independence measures were observed. CONCLUSIONS: Intrathecal transplantation of human umbilical cord-derived WJ-MSCs is a safe intervention. A single intrathecal infusion of WJ-MSCs in patients with chronic complete SCI induced sensory improvement in the segments adjacent to the injury site.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries , Wharton Jelly , Adult , Female , Humans , Male , Middle Aged , Quality of Life , Spinal Cord Injuries/therapy
5.
J Neurosurg Spine ; 28(1): 109-118, 2018 01.
Article in English | MEDLINE | ID: mdl-29125428

ABSTRACT

OBJECTIVE Artificial nerve guides are being developed to substitute for autograft repair after peripheral nerve injuries. However, the use of conduits is limited by the length of the gap that needs to be bridged, with the success of regeneration highly compromised in long gaps. Addition of aligned proregenerative cells and extracellular matrix (ECM) components inside the conduit can be a good strategy to achieve artificial grafts that recreate the natural environment offered by a nerve graft. The purpose of this study was to functionalize chitosan devices with different cell types to support regeneration in limiting gaps in the rat peripheral nerve. METHODS The authors used chitosan devices combined with proteins of the ECM and cells in a rat model of sciatic nerve injury. Combinations of fibronectin and laminin with mesenchymal stem cells (MSCs) or Schwann cells (SCs) were aligned within tethered collagen-based gels, which were placed inside chitosan tubes that were then used to repair a critical-size gap of 15 mm in the rat sciatic nerve. Electrophysiology and algesimetry tests were performed to analyze functional recovery during the 4 months after injury and repair. Histological analysis was performed at the midlevel and distal level of the tubes to assess the number of regenerated myelinated fibers. RESULTS Functional analysis demonstrated that SC-aligned scaffolds resulted in 100% regeneration success in a 15-mm nerve defect in this rat model. In contrast, animals that underwent repair with MSC-aligned constructs had only 90% regeneration success, and those implanted with acellular bridges had only 75% regeneration success. CONCLUSIONS These results indicate that the combination of chitosan conduits with ECM-enriched cellular gels represents a good alternative to the use of autografts for repairing long nerve gaps.


Subject(s)
Fibronectins , Laminin , Mesenchymal Stem Cells/physiology , Peripheral Nerve Injuries/therapy , Schwann Cells/physiology , Sciatic Nerve/injuries , Animals , Chitosan , Disease Models, Animal , Extracellular Matrix , Female , Nerve Regeneration , Peripheral Nerve Injuries/pathology , Rats , Rats, Wistar , Tissue Scaffolds
6.
Cell Transplant ; 25(10): 1833-1852, 2016 10.
Article in English | MEDLINE | ID: mdl-27075820

ABSTRACT

Spinal cord injury (SCI) causes loss of neural functions below the level of the lesion due to interruption of spinal pathways and secondary neurodegenerative processes. The transplant of neural stem cells (NSCs) is a promising approach for the repair of SCI. Reprogramming of adult somatic cells into induced pluripotent stem cells (iPSCs) is expected to provide an autologous source of iPSC-derived NSCs, avoiding the immune response as well as ethical issues. However, there is still limited information on the behavior and differentiation pattern of transplanted iPSC-derived NSCs within the damaged spinal cord. We transplanted iPSC-derived NSCs, obtained from adult human somatic cells, into rats at 0 or 7 days after SCI, and evaluated motor-evoked potentials and locomotion of the animals. We histologically analyzed engraftment, proliferation, and differentiation of the iPSC-derived NSCs and the spared tissue in the spinal cords at 7, 21, and 63 days posttransplant. Both transplanted groups showed a late decline in functional recovery compared to vehicle-injected groups. Histological analysis showed proliferation of transplanted cells within the tissue and that cells formed a mass. At the final time point, most grafted cells differentiated to neural and astroglial lineages, but not into oligodendrocytes, while some grafted cells remained undifferentiated and proliferative. The proinflammatory tissue microenviroment of the injured spinal cord induced proliferation of the grafted cells and, therefore, there are possible risks associated with iPSC-derived NSC transplantation. New approaches are needed to promote and guide cell differentiation, as well as reduce their tumorigenicity once the cells are transplanted at the lesion site.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Animals , Cell Lineage , Cells, Cultured , Cellular Microenvironment , Evoked Potentials , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Locomotion , Nestin/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord/metabolism , Spinal Cord/pathology , Transplantation, Heterologous
7.
J Neurosci ; 35(28): 10224-35, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26180199

ABSTRACT

Lysophosphatidic acid (LPA) is an extracellular lipid mediator involved in many physiological functions that signals through six known G-protein-coupled receptors (LPA1-LPA6). A wide range of LPA effects have been identified in the CNS, including neural progenitor cell physiology, astrocyte and microglia activation, neuronal cell death, axonal retraction, and development of neuropathic pain. However, little is known about the involvement of LPA in CNS pathologies. Herein, we demonstrate for the first time that LPA signaling via LPA1 contributes to secondary damage after spinal cord injury. LPA levels increase in the contused spinal cord parenchyma during the first 14 d. To model this potential contribution of LPA in the spinal cord, we injected LPA into the normal spinal cord, revealing that LPA induces microglia/macrophage activation and demyelination. Use of a selective LPA1 antagonist or mice lacking LPA1 linked receptor-mediated signaling to demyelination, which was in part mediated by microglia. Finally, we demonstrate that selective blockade of LPA1 after spinal cord injury results in reduced demyelination and improvement in locomotor recovery. Overall, these results support LPA-LPA1 signaling as a novel pathway that contributes to secondary damage after spinal cord contusion in mice and suggest that LPA1 antagonism might be useful for the treatment of acute spinal cord injury. SIGNIFICANCE STATEMENT: This study reveals that LPA signaling via LPA receptor type 1 activation causes demyelination and functional deficits after spinal cord injury.


Subject(s)
Demyelinating Diseases/etiology , Receptors, Lysophosphatidic Acid/metabolism , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology , Spinal Cord/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Demyelinating Diseases/drug therapy , Demyelinating Diseases/pathology , Disease Models, Animal , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/genetics , Female , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/metabolism , Lysophospholipids/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/ultrastructure , Motor Activity/drug effects , Motor Activity/genetics , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Receptors, Lysophosphatidic Acid/deficiency , Spinal Cord/drug effects , Spinal Cord Injuries/etiology , Time Factors
8.
J Neurotrauma ; 32(6): 367-80, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25203134

ABSTRACT

Cell therapy for spinal cord injury (SCI) is a promising strategy for clinical application. Mesenchymal stem cells (MSC) have demonstrated beneficial effects following transplantation in animal models of SCI. However, despite the immunoprivilege properties of the MSC, their survival in the injured spinal cord is reduced due to the detrimental milieu in the damaged tissue and immune rejection of the cells. The limited survival of the engrafted cells may determine the therapy success. Therefore, we compared two strategies to increase the presence of the cells in the injured spinal cord in rats: increasing the amount of MSC transplants and using immunosuppressive treatment with FK506 after transplantation. Functional outcomes for locomotion and electrophysiological responses were assessed. The grafted cells survival and the amount of cavity and spared tissue were studied. The findings indicate that immunosuppression improved grafted cells survival. A cell-dose effect was found regarding locomotion recovery and tissue protection independent of immunosuppression. Nevertheless, immunosuppression enhanced the electrophysiological outcomes and allowed filling of the cavity formed after injury by new regenerative tissue and axons. These results indicate that MSC transplantation combined with immunosuppression prolongs the survival of engrafted cells and improves functional and morphological outcomes after SCI.


Subject(s)
Graft Survival/immunology , Immunosuppressive Agents/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Spinal Cord Injuries/immunology , Spinal Cord Injuries/therapy , Animals , Cells, Cultured , Female , Graft Survival/drug effects , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Male , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Tacrolimus/pharmacology , Treatment Outcome
9.
Eur J Neurosci ; 39(10): 1704-17, 2014 May.
Article in English | MEDLINE | ID: mdl-24635194

ABSTRACT

Cell therapy for spinal cord injury (SCI) is a promising strategy for clinical application. Both bone marrow mesenchymal stromal cells (MSCs; also known as bone marrow-derived 'mesenchymal stem cells') and olfactory ensheathing cells (OECs) have demonstrated beneficial effects following transplantation in animal models of SCI. However, due to the large number of affecting parameters that determine the therapy success and the lack of methodological consensus, the comparison of different works is difficult. Therefore, we compared the effects of MSC and OEC transplants at early or delayed time after a spinal cord contusion injury in the rat. Functional outcomes for locomotion, sensory perception and electrophysiological responses were assessed. Moreover, the grafted cells survival and the amount of cavity and spared tissue were studied. The findings indicate that grafted cells survived until 7 days post-injection, but markedly disappeared in the following 2 weeks. Despite the low survival of the cells, MSC and OEC grafts provided tissue protection after early and delayed transplantation. Nevertheless, only acute MSC grafts improved locomotion recovery in treadmill condition and electrophysiological outcomes with respect to the other injured groups. These results, together with previous works, indicate that the MSC seem a better option than OEC for treatment of contusion injuries.


Subject(s)
Ependymoglial Cells/transplantation , Mesenchymal Stem Cell Transplantation , Spinal Cord Injuries/therapy , Spinal Cord/pathology , Spinal Cord/physiopathology , Animals , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Ependymoglial Cells/physiology , Evoked Potentials, Motor/physiology , Female , Male , Motor Activity/physiology , Muscle, Skeletal/physiopathology , Neural Conduction/physiology , Olfactory Bulb/cytology , Rats, Sprague-Dawley , Recovery of Function/physiology , Reflex/physiology , Sensorimotor Cortex/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Thermosensing/physiology , Touch/physiology
10.
PLoS One ; 8(10): e76141, 2013.
Article in English | MEDLINE | ID: mdl-24146830

ABSTRACT

Transplantation of bone marrow derived mesenchymal stromal cells (MSC) or olfactory ensheathing cells (OEC) have demonstrated beneficial effects after spinal cord injury (SCI), providing tissue protection and improving the functional recovery. However, the changes induced by these cells after their transplantation into the injured spinal cord remain largely unknown. We analyzed the changes in the spinal cord transcriptome after a contusion injury and MSC or OEC transplantation. The cells were injected immediately or 7 days after the injury. The mRNA of the spinal cord injured segment was extracted and analyzed by microarray at 2 and 7 days after cell grafting. The gene profiles were analyzed by clustering and functional enrichment analysis based on the Gene Ontology database. We found that both MSC and OEC transplanted acutely after injury induce an early up-regulation of genes related to tissue protection and regeneration. In contrast, cells transplanted at 7 days after injury down-regulate genes related to tissue regeneration. The most important change after MSC or OEC transplant was a marked increase in expression of genes associated with foreign body response and adaptive immune response. These data suggest a regulatory effect of MSC and OEC transplantation after SCI regarding tissue repair processes, but a fast rejection response to the grafted cells. Our results provide an initial step to determine the mechanisms of action and to optimize cell therapy for SCI.


Subject(s)
Gene Expression , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Nerve Regeneration/genetics , Neuroglia/physiology , Neuroglia/transplantation , Spinal Cord Injuries , Animals , Female , Femur/cytology , Femur/physiology , Gene Expression Profiling , Male , Mesenchymal Stem Cells/cytology , Neuroglia/cytology , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries/genetics , Spinal Cord Injuries/therapy , Tibia/cytology , Tibia/physiology
11.
Eur J Neurosci ; 38(12): 3786-98, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24112298

ABSTRACT

Microglial cell plays a crucial role in the development and establishment of chronic neuropathic pain after spinal cord injuries. As neuropathic pain is refractory to many treatments and some drugs only present partial efficacy, it is essential to study new targets and mechanisms to ameliorate pain signs. For this reason we have used glibenclamide (GB), a blocker of KATP channels that are over expressed in microglia under activation conditions. GB has already been used to trigger the early scavenger activity of microglia, so we administer it to promote a better removal of dead cells and myelin debris and support the microglia neuroprotective phenotype. Our results indicate that a single dose of GB (1 µg) injected after spinal cord injury is sufficient to promote long-lasting functional improvements in locomotion and coordination. Nevertheless, the Randall-Selitto test measurements indicate that these improvements are accompanied by enhanced mechanical hyperalgesia. In vitro results indicate that GB may influence microglial phagocytosis and therefore this action may be at the basis of the results obtained in vivo.


Subject(s)
Glyburide/pharmacology , Hyperalgesia/drug therapy , Locomotion , Microglia/drug effects , Neuroprotective Agents/pharmacology , Phagocytosis , Spinal Cord Injuries/drug therapy , Animals , Cells, Cultured , Female , Glyburide/therapeutic use , Hyperalgesia/physiopathology , Microglia/physiology , Neuroprotective Agents/therapeutic use , Phenotype , Rats , Rats, Sprague-Dawley , Sensory Thresholds , Spinal Cord Injuries/physiopathology , Touch
12.
Curr Stem Cell Res Ther ; 6(3): 273-87, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21476980

ABSTRACT

Spinal cord injury (SCI) is a traumatic disorder resulting in a functional deficit that usually leads to severe and permanent paralysis. After the initial insult to the spinal cord, additional structure and function are lost through an active and complex secondary process. Since there is not effective treatment for SCI, several strategies including cellular, pharmacological and rehabilitation therapies have been approached in animal models. Some of them have been proved in clinical trials. In this review we focus on the current state of cell therapies, particularly on cells from adult origin, assayed in preclinical research. Cell types used in SCI therapy include Schwann cells, olfactory ensheathing cells and adult stem cells, such as neural stem cells, umbilical cord blood derived cells, mesenchymal stem cells or induced pluripotent stem cells. There are not yet conclusive evidences on which types of glial or adult stem cells are most effective in SCI treatment. Their ability to incorporate into the damaged spinal cord, to differentiate into neural lineages, to exert neuroprotective effects, to promote regeneration of damaged axons, and to improve functional deficits are still discussed, before translation towards clinical use, as a single therapy or in combination with other strategies.


Subject(s)
Adult Stem Cells/transplantation , Spinal Cord Injuries/therapy , Animals , Cicatrix , Cord Blood Stem Cell Transplantation , Humans , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Inflammation , Mesenchymal Stem Cell Transplantation , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Schwann Cells/transplantation , Spinal Cord/blood supply , Spinal Cord/pathology , Spinal Cord Injuries/physiopathology
13.
J Neurosci Methods ; 198(1): 53-61, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21402104

ABSTRACT

In this work we set up an in vitro model, based on organotypic cultures of spinal cord slices and dorsal root ganglia explants from P7 rats, embedded in a collagen matrix and cultured under the same conditions. As specific reinnervation of end-organs is still an unresolved issue in peripheral nerve research, we characterized a model that allows us to compare under the same conditions motor and sensory neuron regeneration. RT97 labeling was used to visualize the regenerating neurites that extended in the collagen gel from both motor neurons in the spinal cord slices and sensory neurons in the DRG explants after a few days in vitro. By adding different neurotrophic factors in the collagen matrix, we evaluated the reliability of DRG and spinal cord preparations. Moreover, we also set up a co-culture with dissociated Schwann cells to further mimic the permissive environment of the peripheral nerve. Thus, these in vitro models can be useful tools to investigate mechanisms for the selective regeneration of sensory and motor neurons, which can be translated into in vivo models.


Subject(s)
Collagen/metabolism , Motor Neurons/physiology , Neurites/physiology , Sensory Receptor Cells/physiology , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques/methods , Extracellular Matrix/physiology , Ganglia, Spinal/cytology , Humans , In Situ Nick-End Labeling/methods , Motor Neurons/cytology , Motor Neurons/drug effects , Nerve Growth Factors/pharmacology , Neurites/drug effects , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Schwann Cells/physiology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Spinal Cord/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...