Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Neural Regen Res ; 18(2): 299-305, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35900407

ABSTRACT

Currently, there is no cure for traumatic spinal cord injury but one therapeutic approach showing promise is gene therapy. In this systematic review and meta-analysis, we aim to assess the efficacy of gene therapies in pre-clinical models of spinal cord injury and the risk of bias. In this meta-analysis, registered at PROSPERO (Registration ID: CRD42020185008), we identified relevant controlled in vivo studies published in English by searching the PubMed, Web of Science, and Embase databases. No restrictions of the year of publication were applied and the last literature search was conducted on August 3, 2020. We then conducted a random-effects meta-analysis using the restricted maximum likelihood estimator. A total of 71 studies met our inclusion criteria and were included in the systematic review. Our results showed that overall, gene therapies were associated with improvements in locomotor score (standardized mean difference [SMD]: 2.07, 95% confidence interval [CI]: 1.68-2.47, Tau2 = 2.13, I2 = 83.6%) and axonal regrowth (SMD: 2.78, 95% CI: 1.92-3.65, Tau2 = 4.13, I2 = 85.5%). There was significant asymmetry in the funnel plots of both outcome measures indicating the presence of publication bias. We used a modified CAMARADES (Collaborative Approach to Meta-Analysis and Review of Animal Data in Experimental Studies) checklist to assess the risk of bias, finding that the median score was 4 (IQR: 3-5). In particular, reports of allocation concealment and sample size calculations were lacking. In conclusion, gene therapies are showing promise as therapies for spinal cord injury repair, but there is no consensus on which gene or genes should be targeted.

2.
Neural Regen Res ; 17(4): 773-774, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34472465
3.
Glia ; 69(12): 2963-2980, 2021 12.
Article in English | MEDLINE | ID: mdl-34472629

ABSTRACT

Microglial proliferation and activation and macrophage accumulation are implicated in neuropathic pain development. In this study, we aim to suppress microgliosis and macrophage accumulation by over-expressing a non-functional soluble colony stimulating factor-1 receptor (sCSF1R) using an adeno-associated virus 9 vector (AAV9). AAV9/sCSF1R and the control vector AAV9/GFP were intrathecally administered into the lumbar spine of adult C57BL/6 mice. Two weeks later, these mice underwent partial sciatic nerve ligation to induce neuropathic pain. GFP and sCSF1R were highly expressed in lumbar dorsal root ganglia (DRG) and spinal cord of AAV9-injected mice. A significant increase in microglia densities in the dorsal and ventral horns of lumbar spinal cords and macrophage densities in DRG and sciatic nerves were observed in the mice with either ligation alone or pre-treated with AAV9/GFP. In nerve-ligated mice pre-treated with AAV9/sCSF1R the microglia densities in the dorsal and ventral horns and macrophage densities in DRG and sciatic nerves were significantly lower compared to nerve-ligated mice pre-treated with AAV9/GFP. Behavioral tests showed that nerve-ligated mice pre-treated with AAV9/sCSF1R had a significantly higher paw withdrawal threshold, indicating the alleviation of neuropathic pain. The results implicate that viral vector-mediated expression of sCSF1R may represent a novel strategy in the alleviation of neuropathic pain.


Subject(s)
Macrophage Colony-Stimulating Factor , Neuralgia , Animals , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neuralgia/metabolism , Neuralgia/therapy , Receptor, Macrophage Colony-Stimulating Factor , Spinal Cord/metabolism
4.
Neural Regen Res ; 16(3): 460-469, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32985466

ABSTRACT

Millions of people worldwide are affected by traumatic spinal cord injury, which usually results in permanent sensorimotor disability. Damage to the spinal cord leads to a series of detrimental events including ischaemia, haemorrhage and neuroinflammation, which over time result in further neural tissue loss. Eventually, at chronic stages of traumatic spinal cord injury, the formation of a glial scar, cystic cavitation and the presence of numerous inhibitory molecules act as physical and chemical barriers to axonal regrowth. This is further hindered by a lack of intrinsic regrowth ability of adult neurons in the central nervous system. The intracellular signalling molecule, cyclic adenosine 3',5'-monophosphate (cAMP), is known to play many important roles in the central nervous system, and elevating its levels as shown to improve axonal regeneration outcomes following traumatic spinal cord injury in animal models. However, therapies directly targeting cAMP have not found their way into the clinic, as cAMP is ubiquitously present in all cell types and its manipulation may have additional deleterious effects. A downstream effector of cAMP, exchange protein directly activated by cAMP 2 (Epac2), is mainly expressed in the adult central nervous system, and its activation has been shown to mediate the positive effects of cAMP on axonal guidance and regeneration. Recently, using ex vivo modelling of traumatic spinal cord injury, Epac2 activation was found to profoundly modulate the post-lesion environment, such as decreasing the activation of astrocytes and microglia. Pilot data with Epac2 activation also suggested functional improvement assessed by in vivo models of traumatic spinal cord injury. Therefore, targeting Epac2 in traumatic spinal cord injury could represent a novel strategy in traumatic spinal cord injury repair, and future work is needed to fully establish its therapeutic potential.

5.
J Neurosci ; 39(42): 8330-8346, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31409666

ABSTRACT

Millions of patients suffer from debilitating spinal cord injury (SCI) without effective treatments. Elevating cAMP promotes CNS neuron growth in the presence of growth-inhibiting molecules. cAMP's effects on neuron growth are partly mediated by Epac, comprising Epac1 and Epac2; the latter predominantly expresses in postnatal neural tissue. Here, we hypothesized that Epac2 activation would enhance axonal outgrowth after SCI. Using in vitro assays, we demonstrated, for the first time, that Epac2 activation using a specific soluble agonist (S-220) significantly enhanced neurite outgrowth of postnatal rat cortical neurons and markedly overcame the inhibition by chondroitin sulfate proteoglycans and mature astrocytes on neuron growth. We further investigated the novel potential of Epac2 activation in promoting axonal outgrowth by an ex vivo rat model of SCI mimicking post-SCI environment in vivo and by delivering S-220 via a self-assembling Fmoc-based hydrogel that has suitable properties for SCI repair. We demonstrated that S-220 significantly enhanced axonal outgrowth across the lesion gaps in the organotypic spinal cord slices, compared with controls. Furthermore, we elucidated, for the first time, that Epac2 activation profoundly modulated the lesion environment by reducing astrocyte/microglial activation and transforming astrocytes into elongated morphology that guided outgrowing axons. Finally, we showed that S-220, when delivered by the gel at 3 weeks after contusion SCI in male adult rats, resulted in significantly better locomotor performance for up to 4 weeks after treatment. Our data demonstrate a promising therapeutic potential of S-220 in SCI, via beneficial effects on neurons and glia after injury to facilitate axonal outgrowth.SIGNIFICANCE STATEMENT During development, neuronal cAMP levels decrease significantly compared with the embryonic stage when the nervous system is established. This has important consequences following spinal cord injury, as neurons fail to regrow. Elevating cAMP levels encourages injured CNS neurons to sprout and extend neurites. We have demonstrated that activating its downstream effector, Epac2, enhances neurite outgrowth in vitro, even in the presence of an inhibitory environment. Using a novel biomaterial-based drug delivery system in the form of a hydrogel to achieve local delivery of an Epac2 agonist, we further demonstrated that specific activation of Epac2 enhances axonal outgrowth and minimizes glial activation in an ex vivo model of spinal cord injury, suggesting a new strategy for spinal cord repair.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Guanine Nucleotide Exchange Factors/agonists , Neuronal Outgrowth/drug effects , Neurons/drug effects , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Animals , Astrocytes/metabolism , Neurites/physiology , Neurons/metabolism , Rats , Spinal Cord Regeneration/physiology
6.
Neural Regen Res ; 13(12): 2077-2079, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30323125
7.
Glia ; 66(10): 2108-2125, 2018 10.
Article in English | MEDLINE | ID: mdl-30144320

ABSTRACT

Microglia contribute to pathophysiology at all stages of multiple sclerosis. Colony-stimulating factor-1 (CSF1) is crucial for microglial proliferation and activation. In this study we measured the CSF1 levels and studied its cellular expression in the mouse spinal cords with experimental autoimmune encephalomyelitis (EAE) to explore the potential contribution of CSF1 in neuronal death. ELISA data showed that CSF1 levels were significantly higher in the spinal cords with acute and chronic EAE than those of normal and adjuvant-injected mice. Immunohistochemical studies demonstrated that CSF1 was expressed in astrocytes and neurons in normal mouse spinal cord. In acute EAE, CSF1 expression was significantly increased, especially in astrocytes in peripheral white matter and large motoneurons. High density of activated microglia was observed in the gray matter where motoneurons expressed high-level CSF1 in acute EAE. Significant large motoneuron loss was seen in chronic EAE and the remaining motoneurons with high-level CSF1 were enwrapped by microglia. Viral vector mediated over-expression of CSF1 in spinal neurons induced profound proliferation and activation of microglia at the injection site and microglia enwrapped CSF1-transduced neurons and their neurites. Significant loss of large CSF1-transduced neurons was seen at 2 and 3 weeks post-viral injection. Demyelination in the CSF1-transduced areas was also significant. These results implicate that CSF1 upregulation in CNS may play an important role in the proliferation and activation of microglia in EAE, contributing to neuroinflammation and neurodegeneration. © 2018 Wiley Periodicals, Inc.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Microglia/metabolism , Neurons/metabolism , Spinal Cord/metabolism , Acute Disease , Animals , Cell Proliferation/physiology , Chronic Disease , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Gray Matter/metabolism , Gray Matter/pathology , HEK293 Cells , Humans , Interleukins/metabolism , Male , Mice , Microglia/pathology , Neurons/pathology , Spinal Cord/pathology , White Matter/metabolism , White Matter/pathology
8.
J Neurosci ; 38(6): 1351-1365, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29279307

ABSTRACT

Injury to the peripheral axons of sensory neurons strongly enhances the regeneration of their central axons in the spinal cord. It remains unclear on what molecules that initiate such conditioning effect. Because ATP is released extracellularly by nerve and other tissue injury, we hypothesize that injection of ATP into a peripheral nerve might mimic the stimulatory effect of nerve injury on the regenerative state of the primary sensory neurons. We found that a single injection of 6 µl of 150 µm ATP into female rat sciatic nerve quadrupled the number of axons growing into a lesion epicenter in spinal cord after a concomitant dorsal column transection. A second boost ATP injection 1 week after the first one markedly reinforced the stimulatory effect of a single injection. Single ATP injection increased expression of phospho-STAT3 and GAP43, two markers of regenerative activity, in sensory neurons. Double ATP injections sustained the activation of phospho-STAT3 and GAP43, which may account for the marked axonal growth across the lesion epicenter. Similar studies performed on P2X7 or P2Y2 receptor knock-out mice indicate P2Y2 receptors are involved in the activation of STAT3 after ATP injection or conditioning lesion, whereas P2X7 receptors are not. Injection of ATP at 150 µm caused little Wallerian degeneration and behavioral tests showed no significant long-term adverse effects on sciatic nerve functions. The results in this study reveal possible mechanisms underlying the stimulation of regenerative programs and suggest a practical strategy for stimulating axonal regeneration following spinal cord injury.SIGNIFICANCE STATEMENT Injury of peripheral axons of sensory neurons has been known to strongly enhance the regeneration of their central axons in the spinal cord. In this study, we found that injection of ATP into a peripheral nerve can mimic the effect of peripheral nerve injury and significantly increase the number of sensory axons growing across lesion epicenter in the spinal cord. ATP injection increased expression of several markers for regenerative activity in sensory neurons, including phospho-STAT3 and GAP43. ATP injection did not cause significant long-term adverse effects on the functions of the injected nerve. These results may lead to clinically applicable strategies for enhancing neuronal responses that support regeneration of injured axons.


Subject(s)
Adenosine Triphosphate/pharmacology , Axons/drug effects , Nerve Regeneration/drug effects , Neurons/drug effects , Sensory Receptor Cells/drug effects , Spinal Cord/drug effects , Adenosine Triphosphate/administration & dosage , Animals , Behavior, Animal , Female , GAP-43 Protein/biosynthesis , GAP-43 Protein/genetics , Injections , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/pathology , Rats , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2Y2/genetics , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Sciatic Nerve , Spinal Cord Injuries/pathology , Wallerian Degeneration/genetics , Wallerian Degeneration/physiopathology
9.
Neurobiol Dis ; 58: 270-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23811498

ABSTRACT

Nogo receptor 1 (NgR1) mediates the inhibitory effects of several myelin-associated inhibitors (MAIs) on axonal regeneration in the central nervous system. A truncated soluble NgR1 (sNgR) has been reported to act as a decoy receptor to block the actions of MAIs. In this study, we fused the sNgR to nerve growth factor (NGF) and used NGF as a carrier to deliver sNgR to the intercellular space to neutralize MAIs. NGF in NGF-sNgR remained biologically active and induced sprouting of calcitonin gene related peptide containing axons when expressed in the spinal cord using a lentiviral vector (LV). Secreted NGF-sNgR promoted neurite outgrowth of dissociated dorsal root ganglion neurons on myelin protein substrate. In a rat dorsal column transection model, regenerating sensory axons were found to grow into the lesion cavity in animals injected with LV/NGF-sNgR, while in animals injected with LV/GFP or LV/NGF-GFP few sensory axons entered the lesion cavity. The results indicate that NGF-sNgR fusion protein can reduce the inhibition of MAIs and facilitate sensory axon regeneration. The fusion constructs may be modified to target other molecules to promote axonal regeneration and the concept may also be adapted to develop gene therapy strategies to treat other disorders.


Subject(s)
Axons/drug effects , Lentivirus/physiology , Myelin Proteins/administration & dosage , Nerve Growth Factor/metabolism , Nerve Regeneration/drug effects , Spinal Cord Injuries/therapy , Animals , Axons/physiology , Calcitonin Gene-Related Peptide/metabolism , Cell Differentiation/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Lentivirus/genetics , Male , Myelin Basic Protein/metabolism , Myelin Proteins/biosynthesis , Nerve Growth Factor/biosynthesis , Nerve Regeneration/physiology , Neurites/drug effects , Nogo Proteins , PC12 Cells , Rats , Rats, Wistar , Recombinant Fusion Proteins/administration & dosage , Serotonin/metabolism , Spinal Cord Injuries/complications
10.
Curr Gene Ther ; 11(2): 101-15, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21291358

ABSTRACT

Recent understanding in pathophysiological mechanisms of spinal cord and spinal root injuries has facilitated the development of new strategies to promote neural repair. Gene therapy approaches have been viewed as the ideal means to achieve long-term local delivery of therapeutic molecules in the central nervous system (CNS). Ex vivo gene delivery offers the additional advantage of providing cellular support for regenerating axons. In this review, we summarize the studies on viral vector-mediated gene delivery to spinal cord in animal models, both in vivo and ex vivo. Most of the studies reported so far are aimed at delivery of various growth factors, such as neurotrophins and neuropoietic cytokines. Other molecules tested include those that interfere with intracellular processes to prevent cell death, or increase intrinsic regenerating state of injured neurons, or modify the CNS environment to make it permissive for axon growth. Several different combinatorial strategies involving gene delivery are also discussed as it has been recognized that successful neural repair may require the synergistic actions of multiple therapeutic managements.


Subject(s)
Axons/metabolism , Genetic Therapy/methods , Nerve Regeneration/genetics , Neurons/metabolism , Spinal Cord Injuries/therapy , Spinal Nerve Roots/injuries , Animals , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Nerve Growth Factors/genetics , Recovery of Function , Spinal Nerve Roots/physiopathology
11.
Glia ; 59(3): 424-34, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21264949

ABSTRACT

The poor survival and migration of transplanted Schwann cells (SCs) are major drawbacks for their clinical application in cell therapy for neurotrauma. To overcome such drawbacks we genetically modified SCs to over-express polysialic acid (PSA) by lentiviral delivery of polysialyltransferase (PST) to study whether over-expression of PSA could enhance their survival, migration, and integration when transplanted into the spinal cord. It was found that more PSA-expressing SCs (PST/SCs) survived than GFP-expressing SCs (GFP/SCs) after transplantation, although cell loss was still quite significant. PSA expression did not enhance the motility of transplanted SCs in uninjured spinal cord. However, in a spinal cord crush injury model PST/SCs transplanted caudal to the lesion showed that increased number of PST/SCs migrated to the injury site compared with that of GFP/SCs. Induced expression of PSA in spinal cord can further facilitate the infiltration of PST/SCs into the lesion site. PST/SCs were also shown to intermingle well with host spinal cells while GFP/SCs formed boundaries with host tissue. This was confirmed by an in vitro confrontation assay showing that more PST/SCs crossed over to astrocyte territory than GFP/SCs. Furthermore, PST/SCs induced much less expression of glial fibrillary acidic protein and chondroitin sulfate proteoglycan in the surrounding tissues than GFP/SCs, indicating that expression of PSA on SCs do not cause significant stress response of astrocytes. These results demonstrate that expression of PSA on SCs significantly changes their biological properties and makes them more feasible for neural repair after neurotrauma.


Subject(s)
Cell Movement/physiology , Cell Transplantation/methods , Graft Survival/physiology , Schwann Cells/transplantation , Sialic Acids/biosynthesis , Sialic Acids/genetics , Spinal Cord Injuries/surgery , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Female , Mice , Rats , Rats, Wistar , Schwann Cells/cytology , Schwann Cells/metabolism , Sialic Acids/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
12.
Neurosurgery ; 65(4 Suppl): A74-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19927082

ABSTRACT

OBJECTIVE: Peripheral nerve injury causes retrograde changes in the damaged neurons, which are beneficial to axonal regeneration. Better understanding of the mechanisms of induction and mediation of these conditioning responses would help to design strategies to invoke stronger regenerative responses in neurons in situations when these responses are inadequate. METHODS: Relevant literature is reviewed. RESULTS: Experimental preparations that measure the influence of peripheral axotomy on regeneration in the central axons of primary sensory neurons are useful to examine mechanisms of conditioning neurons. Despite 4 decades of speculation, the nature of the damage signals from injured nerves that initiate axonal signals to the nerve cell body remains elusive. Members of the family of neuropoietic cytokines are clearly implicated, but what induces them is unknown. Multiple changes in gene regulation in axotomized neurons have been described, and dozens of growth-associated genes have been identified: neurotrophic factors, transcription factors, molecules participating in axonal transport, and molecules active in the growth cone. The mechanisms of interaction of a few regeneration-associated molecules with the signaling cascades that lead to actin and tubulin remodeling at the growth cone are understood in some detail. In animals, viral gene therapy to deliver regeneration-associated genes to neurons or other local measures to induce these genes can improve regeneration. A few pharmacological agents, administered systemically, have small beneficial effects on axonal regeneration. CONCLUSION: Advances in laboratory research have provided knowledge of cell body responses to axotomy with clinical relevance.


Subject(s)
Axotomy/adverse effects , Neurons/metabolism , Peripheral Nerve Injuries , Peripheral Nerves/metabolism , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/surgery , Animals , Genetic Therapy/methods , Genetic Therapy/trends , Growth Cones/metabolism , Growth Cones/ultrastructure , Humans , Microtubules/genetics , Microtubules/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Nerve Regeneration/genetics , Neurons/cytology , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/physiopathology , Recovery of Function/genetics
13.
Mol Ther ; 17(12): 2020-30, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19623163

ABSTRACT

Many axonal growth inhibitors that contribute to the usual failure of axon regeneration in the central nervous system (CNS) exert their effects via the RhoA-Rho kinase (ROCK) signal pathway. In this study, we investigated whether lentiviral vector (LV)-mediated neuron-specific expression of a dominant negative mutant of ROCK (DNROCK) could promote axon outgrowth in vitro and in vivo. Dissociated adult rat dorsal root ganglion (DRG) neurons were seeded on solubilized myelin proteins and transduced with either LV/DNROCK or LV/green fluorescent protein (GFP). DNROCK-expressing neurons were shown to have a greater chance of generating neurites and a longer mean length of neurite than GFP-expressing neurons. In the in vivo studies, lentiviruses were injected into the adult rat red nucleus followed by unilateral rubrospinal tract (RST) transection at the fourth cervical level. Rats in the DNROCK group showed better functional recovery in the affected hindlimbs and forelimbs than those in the GFP group. Examination of the spinal cord sections revealed more rubrospinal axonal profiles growing to the spinal cord caudal to the lesion in the DNROCK group than in the GFP group. These results indicate that blocking the RhoA-ROCK signal pathway by expressing DNROCK can enhance regenerative axonal sprouting and lead to partial recovery of limb function.


Subject(s)
Axons/physiology , Lentivirus/genetics , Neurons/cytology , Spinal Cord Injuries/therapy , rho-Associated Kinases/physiology , Animals , Behavior, Animal , Cells, Cultured , Female , Genes, Dominant , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , NIH 3T3 Cells , Nerve Regeneration , Neurons/physiology , Rats , Rats, Wistar , Recovery of Function , Spinal Cord Injuries/pathology
14.
Restor Neurol Neurosci ; 26(2-3): 81-96, 2008.
Article in English | MEDLINE | ID: mdl-18820404

ABSTRACT

Cell adhesion molecules (CAMs) play important roles in cell-cell and cell-extracellular matrix interactions in both mature and developing nervous system. During development, they are involved in cell migration, axon guidance, target recognition, and synapse formation; while in the mature nervous system, they maintain synaptic connections, cell-cell contacts, and neuron-glial interactions. Injuries to the nervous systems break the stable state of the tissues and the repair of damaged tissues and regeneration of axons require the participation of CAMs both as adhesion molecules and as signal transduction molecules. One group of the well-studied CAMs in the nervous system is the immunoglobulin superfamily including L1 and neural cell adhesion molecule (NCAM). This review will be focussed on the involvement of L1, NCAM, and polysialylated NCAM in neural repair and axon regeneration after nerve injury and their potential applications in the treatment of CNS injury.


Subject(s)
Central Nervous System/metabolism , Growth Cones/metabolism , Nerve Regeneration/physiology , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecules/metabolism , Animals , Central Nervous System/injuries , Genetic Therapy/methods , Humans , Immunoglobulins/classification , Immunoglobulins/metabolism , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecules/genetics , Neuronal Plasticity/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy
15.
Mol Cell Neurosci ; 37(2): 367-75, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18055217

ABSTRACT

Transduction and activation of an inducible form of STAT3 (signal transducer and activator of transcription) sufficed to increase VIP (vasoactive intestinal protein) mRNA concentrations in neuroblastoma cells. Overexpression of SOCS3 (suppressor of cytokine signaling) inhibited and mutant SOCS3 (with an inactivating point mutation in amino acid 25) enhanced the induction of VIP mRNA by CNTF (ciliary neurotrophic factor). Because mutant SOCS3 did not augment the increase in STAT transcriptional activity following CNTF stimulation, the enhancement by mutant SOCS3 of the actions of CNTF cannot be attributed to changes in STAT3 signaling. Mutant SOCS3 increased AP-1 (activator protein) transcriptional activity and JNK (c-Jun N-terminal kinase) activity and SOCS3 bound to the scaffolding protein, JNK-interacting protein-1: these observations provide a plausible explanation for the enhancement by mutant SOCS3 of the actions of CNTF. We conclude that endogenous SOCS3 inhibits AP-1 activity through blocking of JNK phosphorylation.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/pharmacology , Down-Regulation/genetics , Feedback, Physiological/genetics , Gene Expression Regulation/genetics , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Neuroblastoma , Phosphorylation , RNA, Messenger/metabolism , Rats , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Transcription Factor AP-1/genetics , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
16.
Mol Ther ; 15(10): 1796-804, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17551503

ABSTRACT

In adult mammals, sensory axons that regenerate in the dorsal root are unable to grow across the dorsal root entry zone (DREZ) into the spinal cord. In this study we examined whether, by inducing expression of polysialic acid (PSA) (a large carbohydrate attached to molecules on the cell surface), in the spinal cord by lentiviral vector (LV) delivery of polysialyltransferase (PST), DREZ could be rendered permeable to regenerating sensory axons. High-level PSA expression was observed in astrocytes and many other cell types after LV/PST injection into the spinal cord. In animals receiving LV/PST injection in combination with a conditioning lesion, many axons penetrated the DREZ following L4-5 dorsal rhizotomy. Some axons reached lamina IV-V and extended rostrally and caudally in the degenerating dorsal column. In LV/green fluorescent protein (GFP)-injected animals, most of the regenerating axons were halted before DREZ, even with a conditioning lesion. More Schwann cells migrated into the LV/PST-transduced spinal cord, many of them accompanying the regenerating axons. A Schwann cell-astrocyte-dorsal root ganglion (DRG) neuron co-culture experiment confirmed that induced PSA expression on astrocytes facilitates the crossing of DRG axons from Schwann cells to astrocytes. These data suggest that over-expression of PSA can create a favorable condition for regenerating axons, and that this approach could form part of a combinational therapeutic strategy for promoting the repair of central nervous system (CNS) injuries.


Subject(s)
Axons , Genetic Vectors , Lentivirus/genetics , Regeneration , Sialic Acids/metabolism , Spinal Cord/physiology , Animals , Astrocytes/metabolism , Female , HeLa Cells , Humans , Immunohistochemistry , Mice , Rats , Rats, Wistar , Spinal Cord/metabolism , Spinal Cord/pathology
17.
Mol Cell Neurosci ; 35(1): 109-19, 2007 May.
Article in English | MEDLINE | ID: mdl-17363265

ABSTRACT

After spinal cord injury axonal regeneration is prevented by glial scar formation. In this study we examined whether induced expression of polysialic acid (PSA) in the lesion site would render the glial scar permissive to axonal regeneration after dorsal column transection. PSA was induced by lentiviral vector-mediated expression of polysialyltransferase (LV/PST). PSA expression increased astrocyte infiltration and permitted the penetration of regenerating axons across the caudal border of the lesion and into the lesion cavity. In LV/PST-injected animals with a peripheral nerve-conditioning lesion, 20 times more axons grew into the lesion cavity than those LV/GFP-injected plus conditioning lesion, and some axons grew across the cavity and extended to the rostral cord, while in LV/GFP group most ascending axons terminated at the caudal border of the lesion. Our result suggests that induced expression of PSA can provide a favorable environment for axonal regeneration.


Subject(s)
Genetic Therapy/methods , Nerve Regeneration/physiology , Sialic Acids/metabolism , Spinal Cord Injuries/therapy , Spinal Cord/physiology , Animals , Astrocytes/pathology , Axons/physiology , DNA, Complementary/genetics , Genetic Vectors , Gliosis/metabolism , Gliosis/pathology , Green Fluorescent Proteins/genetics , Lentivirus/genetics , Male , Neurons, Afferent/physiology , Neurons, Afferent/ultrastructure , Rats , Rats, Wistar , Sialyltransferases/genetics , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
18.
Eur J Neurosci ; 25(2): 351-61, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17284175

ABSTRACT

Purkinje axons in adult mammals are generally unable to regenerate after axotomy. Our recent work has shown that over-expression of growth related genes, GAP-43 and L1, in Purkinje cells increased their axonal outgrowth into a predegenerated peripheral nerve graft, but not into a fresh graft [Zhang et al., (2005) Proc. Natl Acad. Sci. USA, 102, 14883-14888]. In the current study we investigated whether engineered expression of growth permissive molecule polysialic acid (PSA) in the glial scar or on transplanted Schwann cells could overcome the inhibitory environment and promote Purkinje axonal regeneration. A stab wound was introduced in the cerebellum of the L1/GAP-43 transgenic mice and a lentiviral vector (LV) carrying the polysialyltransferase (PST) cDNA (LV/PST) was injected into the lesion site to transduce the cells in the glial scar. Regenerating Purkinje axons were examined by calbindin immunostaining. There was increased Purkinje axonal sprouting in the area expressing high-level PSA. However, Purkinje axons were unable to grow into the lesion cavity. In the second set of experiments when LV/PST transduced Schwann cells were transplanted into the lesion site, the number of Purkinje axons growing into the transplant was nine times more than that growing into Schwann cell transplant expressing GFP two months post operation. Our result suggests that transplanted Schwann cells engineered to express PSA support axonal regeneration better than naïve Schwann cells.


Subject(s)
Axons/drug effects , Purkinje Cells/pathology , Regeneration/drug effects , Sialic Acids/metabolism , Sialic Acids/therapeutic use , Transduction, Genetic/methods , Animals , Animals, Newborn , Axons/physiology , Cells, Cultured , Cerebellar Diseases/pathology , Cerebellar Diseases/surgery , Fluorescent Antibody Technique , GAP-43 Protein/genetics , Genetic Vectors/physiology , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/genetics , Purkinje Cells/physiology , Regeneration/physiology , Schwann Cells/physiology , Schwann Cells/transplantation , Sciatic Nerve/cytology , Sialyltransferases/metabolism
19.
Exp Neurol ; 204(1): 66-76, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17112514

ABSTRACT

A conditioning lesion to peripheral axons of primary sensory neurons accelerates regeneration of their central axons in vivo or neurite outgrowth if the neurons are grown in vitro. Previous evidence has implicated neuropoietic cytokines and also cyclic AMP in regenerative conditioning. In experiments reported here, delivery through a lentivirus vector of ciliary neurotrophic factor to the appropriate dorsal root ganglion in rats was sufficient to mimic the conditioning effect of peripheral nerve injury on the regeneration of dorsal spinal nerve root axons. Regeneration in this experimental preparation was also stimulated by intraganglionic injection of dibutyryl cyclic AMP but the effects of ciliary neurotrophic factor and dibutyryl cyclic AMP were not additive. Dibutyryl cyclic AMP injection into the dorsal root ganglion induced mRNAs for two other neuropoietic cytokines, interleukin-6 and leukemia inhibitory factor and increased the accumulation of phosphorylated STAT3 in neuronal nuclei. The in vitro conditioning action of dibutyryl cyclic AMP was partially blocked by a pharmacological inhibitor of Janus kinase 2, a neuropoietic cytokine signaling molecule. We suggest that the beneficial actions of increased cyclic AMP activity on axonal regeneration of primary sensory neurons are mediated, at least in part, through the induction of neuropoietic cytokine synthesis within the dorsal root ganglion.


Subject(s)
Conditioning, Psychological/physiology , Cyclic AMP/metabolism , Cytokines/metabolism , Nerve Regeneration/physiology , Neurons, Afferent/physiology , Animals , Axons/physiology , Bucladesine/pharmacology , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/pharmacology , Conditioning, Psychological/drug effects , Enzyme Inhibitors/pharmacology , Ganglia, Spinal , Gene Expression Regulation , Gene Transfer Techniques , Genetic Vectors , Interleukin-6/genetics , Janus Kinase 2/antagonists & inhibitors , Lentivirus/genetics , Leukemia Inhibitory Factor/genetics , Nerve Crush , Nerve Regeneration/genetics , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction , Spinal Nerve Roots , Tyrphostins/pharmacology
20.
J Neurosci ; 26(37): 9512-9, 2006 Sep 13.
Article in English | MEDLINE | ID: mdl-16971535

ABSTRACT

The actions of the neuropoietic cytokines are mediated by the gp130 receptor, which activates several signaling molecules including the transcription factor STAT3 (signal transducer and activator of transcription), which, in turn, is subject to feedback inhibition by SOCS3 (suppressor of cytokine signaling). Activation of the gp130 receptor has been implicated in axonal growth particularly during regeneration, but the specific contribution of STAT3 is the subject of conflicting reports. Measurements of SOCS3 mRNA in rat dorsal root ganglia showed a significant induction in this inhibitory molecule after peripheral nerve injury. The functions of STAT3 and SOCS3 in adult rat primary sensory neurons were investigated in vitro through transduction of lentiviruses yielding a conditionally activated STAT3, native SOCS3, or a mutant SOCS3 with dominant-negative actions. The SOCS3 construct was effective in inhibiting tyrosine phosphorylation of STAT3 in a neuroblastoma cell line and in blocking nuclear accumulation of endogenous STAT3 or of the conditionally activated STAT3 chimera in primary sensory neurons. In such neurons, transduction and activation of STAT3 enhanced neurite growth, transduction with SOCS3 reduced neurite outgrowth, and transduction with mutant SOCS3 enhanced neurite growth, at least under basal conditions. In conclusion, STAT3 signaling is beneficial to axonal growth through activating transcription of unidentified genes, and SOCS3 is detrimental to axonal growth through inhibition of STAT3 and/or other transcription factors.


Subject(s)
Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Neurites/metabolism , Neurons, Afferent/metabolism , STAT3 Transcription Factor/physiology , Suppressor of Cytokine Signaling Proteins/physiology , Active Transport, Cell Nucleus/physiology , Animals , Axotomy , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Feedback, Physiological/physiology , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/physiology , Genetic Vectors/genetics , Humans , Mice , Nerve Regeneration/genetics , Neurites/ultrastructure , Neurons, Afferent/cytology , RNA, Messenger/metabolism , Rats , STAT3 Transcription Factor/genetics , Sciatic Neuropathy/genetics , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...