Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Cells ; 11(19)2022 10 09.
Article in English | MEDLINE | ID: mdl-36231129

ABSTRACT

Facial motoneuron (FMN) survival is mediated by CD4+ T cells in an interleukin-10 (IL-10)-dependent manner after facial nerve axotomy (FNA), but CD4+ T cells themselves are not the source of this neuroprotective IL-10. The aims of this study were to (1) identify the temporal and cell-specific induction of IL-10 expression in the facial motor nucleus and (2) elucidate the neuroprotective capacity of this expression after axotomy. Immunohistochemistry revealed that FMN constitutively produced IL-10, whereas astrocytes were induced to make IL-10 after FNA. Il10 mRNA co-localized with microglia before and after axotomy, but microglial production of IL-10 protein was not detected. To determine whether any single source of IL-10 was critical for FMN survival, Cre/Lox mouse strains were utilized to selectively knock out IL-10 in neurons, astrocytes, and microglia. In agreement with the localization data reflecting concerted IL-10 production by multiple cell types, no single cellular source of IL-10 alone could provide neuroprotection after FNA. These findings suggest that coordinated neuronal and astrocytic IL-10 production is necessary for FMN survival and has roles in neuronal homeostasis, as well as neuroprotective trophism after axotomy.


Subject(s)
Facial Nerve Injuries , Facial Nucleus , Animals , Mice , Axotomy , Facial Nerve Injuries/genetics , Facial Nerve Injuries/metabolism , Facial Nucleus/metabolism , Interleukin-10/metabolism , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/metabolism , Neuroprotection , RNA, Messenger/metabolism
2.
J Neurosci ; 42(12): 2474-2491, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35149515

ABSTRACT

In postmitotic neurons, several tumor suppressor genes (TSGs), including p53, Rb, and PTEN, modulate the axon regeneration success after injury. Particularly, PTEN inhibition is a key driver of successful CNS axon regeneration after optic nerve or spinal cord injury. In contrast, in peripheral neurons, TSG influence in neuronal morphology, physiology, and pathology has not been investigated to the same depth. In this study, we conditionally deleted PTEN from mouse facial motoneurons (Chat-Cre/PtenloxP/loxP ) and analyzed neuronal responses in vivo with or without peripheral facial nerve injury in male and female mice. In uninjured motoneurons, PTEN loss induced somatic, axonal, and nerve hypertrophy, synaptic terminal enlargement and reduction in physiological whisker movement. Despite these morphologic and physiological changes, PTEN deletion positively regulated facial nerve regeneration and recovery of whisker movement after nerve injury. Regenerating PTEN-deficient motoneurons upregulated P-CREB and a signaling pathway involving P-Akt, P-PRAS40, P-mTOR, and P-4EBP1. In aged mice (12 months), PTEN deletion induced hair loss and facial hyperplasia of the epidermis. This suggests a time window in younger mice with PTEN loss stimulating axon growth after injury, however, at the risk of hyperplasia formation at later time points in the old animal. Overall, our data highlight a dual TSG function with PTEN loss impairing physiological neuron function but furthermore underscoring the positive effects of PTEN ablation in axon regeneration also for the PNS.SIGNIFICANCE STATEMENT Tumor suppressor genes (TSGs) restrict cell proliferation and growth. TSG inhibition, including p53 and PTEN, stimulates axon regeneration after CNS injury. In contrast, in PNS axon regeneration, TSGs have not been analyzed in great depth. Herein we show enhanced peripheral axon regeneration after PTEN deletion from facial motoneurons. This invokes a signaling cascade with novel PTEN partners, including CREB and PRAS40. In adult mice, PTEN loss induces hyperplasia of the skin epidermis, suggesting detrimental consequences when reaching adulthood in contrast to a beneficial TSG role for regeneration in young adult mice. Thus, our data highlight the double-edged sword nature of interfering with TSG function.


Subject(s)
Facial Nerve Injuries , Nerve Regeneration , PTEN Phosphohydrolase/metabolism , Animals , Axons/physiology , Facial Nerve Injuries/genetics , Facial Nerve Injuries/pathology , Female , Hyperplasia/pathology , Hypertrophy/pathology , Male , Mice , Motor Neurons/metabolism , Nerve Regeneration/genetics , Tumor Suppressor Protein p53
3.
Mol Cell Neurosci ; 113: 103628, 2021 06.
Article in English | MEDLINE | ID: mdl-33962001

ABSTRACT

Facial nerves are frequently injured during cosmetic or other types of facial surgery. However, information on the genes involved in the damage and recovery of the facial nerves is limited. Here, we aimed to identify the genes affected by facial nerve injury and repair using next-generation sequencing. We established a rat axotomy model and a parallel epineurial neurorrhaphy model, in which gene expression was analyzed from 3 days to 8 weeks after surgery. We discovered that ARRB1, SGK1, and GSK3B genes associated with neuronal cell death were upregulated in the axotomy model. In contrast, MFRP, MDK, and ACE genes involved in neural recovery and regeneration exhibited higher expression in the neurorrhaphy model. In the present study, the analysis of the big data obtained from the next-generation sequencing (RNA-seq) technology reveals that the expression of genes involved in neuronal cell death is induced during nerve damage, and those associated with neural recovery are more abundantly expressed during repair processes. These results are considered to be useful for the establishment of the treatment of related diseases and basic research in various neuroscience fields by utilizing damage and recovery mechanism of facial nerves.


Subject(s)
Facial Nerve Injuries/genetics , Nerve Regeneration/genetics , Neurons/metabolism , Transcriptome , Animals , Cell Death , Facial Nerve Injuries/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Male , Midkine/genetics , Midkine/metabolism , Neurons/physiology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism
4.
J Cell Physiol ; 235(12): 9609-9622, 2020 12.
Article in English | MEDLINE | ID: mdl-32391604

ABSTRACT

Facial paralysis can result in severe implications for patients. A good prognosis depends on the degree of nerve regeneration. Schwann cells (SCs) play an important role in facial nerve development and regeneration through migration. Forkhead box C1 (Foxc1), a member of the forkhead transcription factor family, is implicated in cell migration. However, the role of Foxc1 in the progression after facial nerve crush remains unknown. Our aim was to evaluate the effect of Foxc1 overexpression on SC migration and recovery of facial nerves after crush injury. The rat facial nerve crush injury model was established through the use of unilateral surgery. The results showed that the expression of Foxc1 was increased in the surgery group compared to that of the control group. SCs were isolated from the sciatic nerves and cultured. Foxc1, delivered by an adeno-associated virus in vivo, or adenovirus in vitro, both induced overexpression of Foxc1, and increased the expression of CXCL12 and ß-catenin. After the transfection of Foxc1, the migration of SC was increased both in vitro and in vivo, was reduced by the inhibition of CXCL12 or ß-catenin. The facial nerve function and the nerve axon remyelination of the rats transfected with Foxc1 were significantly improved after nerve crush injury. Overall, the results demonstrated that overexpression of Foxc1 promoted SC migration by regulating CXCL12 via the Wnt/ß-catenin pathway, thus contributing to improved facial nerve function after crush injury.


Subject(s)
Facial Nerve Injuries/therapy , Facial Nerve/surgery , Forkhead Transcription Factors/genetics , Nerve Regeneration/genetics , Animals , Cell Movement/genetics , Chemokine CXCL12/genetics , Facial Nerve/pathology , Facial Nerve Injuries/genetics , Facial Nerve Injuries/pathology , Forkhead Transcription Factors/pharmacology , Gene Expression Regulation/genetics , Humans , Rats , Schwann Cells/cytology , Schwann Cells/metabolism , Sciatic Nerve/cytology , Sciatic Nerve/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics
5.
J Neuroinflammation ; 17(1): 121, 2020 Apr 17.
Article in English | MEDLINE | ID: mdl-32303238

ABSTRACT

BACKGROUND: After peripheral nerve transection, facial motoneuron (FMN) survival depends on an intact CD4+ T cell population and a central source of interleukin-10 (IL-10). However, it has not been determined previously whether CD4+ T cells participate in the central neuroprotective IL-10 cascade after facial nerve axotomy (FNA). METHODS: Immunohistochemical labeling of CD4+ T cells, pontine vasculature, and central microglia was used to determine whether CD4+ T cells cross the blood-brain barrier and enter the facial motor nucleus (FMNuc) after FNA. The importance of IL-10 signaling in CD4+ T cells was assessed by performing adoptive transfer of IL-10 receptor beta (IL-10RB)-deficient CD4+ T cells into immunodeficient mice prior to injury. Histology and qPCR were utilized to determine the impact of IL-10RB-deficient T cells on FMN survival and central gene expression after FNA. Flow cytometry was used to determine whether IL-10 signaling in T cells was necessary for their differentiation into neuroprotective subsets. RESULTS: CD4+ T cells were capable of crossing the blood-brain barrier and associating with reactive microglial nodules in the axotomized FMNuc. Full induction of central IL-10R gene expression after FNA was dependent on CD4+ T cells, regardless of their own IL-10R signaling capability. Surprisingly, CD4+ T cells lacking IL-10RB were incapable of mediating neuroprotection after axotomy and promoted increased central expression of genes associated with microglial activation, antigen presentation, T cell co-stimulation, and complement deposition. There was reduced differentiation of IL-10RB-deficient CD4+ T cells into regulatory CD4+ T cells in vitro. CONCLUSIONS: These findings support the interdependence of IL-10- and CD4+ T cell-mediated mechanisms of neuroprotection after axotomy. CD4+ T cells may potentiate central responsiveness to IL-10, while IL-10 signaling within CD4+ T cells is necessary for their ability to rescue axotomized motoneuron survival. We propose that loss of IL-10 signaling in CD4+ T cells promotes non-neuroprotective autoimmunity after FNA.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Facial Nerve Injuries/metabolism , Facial Nerve/metabolism , Motor Neurons/metabolism , Receptors, Interleukin-10/biosynthesis , Animals , Axotomy/methods , Cell Survival/physiology , Cells, Cultured , Facial Nerve Injuries/genetics , Female , Gene Expression , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-10/genetics
6.
Int J Biol Macromol ; 124: 460-468, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30391592

ABSTRACT

Facial nerve injury is a clinically common disease accompanied by demyelination of damaged nerves. The remyelination of damaged nerves and the unsatisfactory function recovery are problems that have been plaguing people for a long time. The role that CXCL12 plays after facial nerve injury remains unknown. Our experiments found that the expression of CXCL12 was up-regulated in the early stage of facial nerve injury and decreased after two weeks. Further research found that CXCL12 had no effect on Schwann cells proliferation, apoptosis and cell cycle, while significantly promoted Schwann cells migration. Treatment with CXCL12 decreased the phosphorylation of PI3K, AKT and mTOR, but increased autophagy marker LC3II/I. The CXCL12-induced Schwann cells migration was significantly attenuated by inhibition of autophagy and activation of PI3K pathway through pretreatment with 3-MA and IGF-1 respectively, and this effect was enhanced by PI3K pathway inhibitor LY294002. Animal experiment also confirmed that CXCL12 could improve facial nerve function and myelin regeneration. The findings of this study indicate that CXCL12 can promote the migration of Schwann cells and potentially become a key molecule in the repair of facial nerve injury.


Subject(s)
Autophagy/drug effects , Chemokine CXCL12/pharmacology , Facial Nerve Injuries/drug therapy , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromones/pharmacology , Cranial Nerves/drug effects , Cranial Nerves/metabolism , Cranial Nerves/pathology , Disease Models, Animal , Facial Nerve/drug effects , Facial Nerve/metabolism , Facial Nerve/pathology , Facial Nerve Injuries/genetics , Facial Nerve Injuries/metabolism , Facial Nerve Injuries/pathology , Gene Expression Regulation , Humans , Insulin-Like Growth Factor I/pharmacology , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
7.
Neuromolecular Med ; 19(2-3): 357-374, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28689354

ABSTRACT

Traumatic injuries to human peripheral nerves are frequently associated with damage to nerve surrounding tissues including muscles and blood vessels. Currently, most rodent models of peripheral nerve injuries (e.g., facial or sciatic nerve) employ surgical nerve transection with scissors or scalpels. However, such an isolated surgical nerve injury only mildly damages neighboring tissues and weakly activates an immune response. In order to provide a rodent nerve injury model accounting for such nerve-associated tissue damage and immune cell activation, we developed a drop tower-based facial nerve trauma model in mice. We compare nerve regeneration in this novel peripheral nerve trauma model with the established surgical nerve injury along several parameters. These include gene expression, histological and functional facial motoneuron (FMN) regeneration, facial nerve degeneration, immune cell activation and muscle damage. Regeneration-associated genes (RAGs; e.g., Atf3) were strongly induced in FMNs subjected to traumatic and surgical injury. Regeneration of FMNs and functional recovery of whisker movement were faster in traumatic versus complete surgical injury, thus cutting down experimentation time. Wallerian degeneration of distal nerve stumps was readily observed in this novel trauma injury model. Importantly, drop tower-inflicted facial nerve injury resulted in muscle damage, activation of muscle satellite cell markers (PAX7) and pronounced infiltration of immune cells to the injury site only in this model but not upon surgical nerve transection. Thus, we provide a novel rodent PNS trauma model that can be easily adopted to other PNS nerves such as the sciatic nerve. Since this nerve trauma model replicates multiple tissue damage frequently encountered in clinical routine, it will be well suited to identify molecular and cellular mechanisms of PNS nerve repair in wild-type and genetically modified rodents.


Subject(s)
Facial Nerve Injuries/physiopathology , Facial Nerve/physiology , Models, Animal , Nerve Regeneration , Animals , Facial Nerve/surgery , Facial Nerve Injuries/genetics , Facial Nerve Injuries/immunology , Facial Nerve Injuries/pathology , Female , Gene Expression Regulation , Male , Masseter Muscle/pathology , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Vibrissae/physiology , Wallerian Degeneration , Wounds, Nonpenetrating/physiopathology
8.
J Biol Regul Homeost Agents ; 31(1): 147-152, 2017.
Article in English | MEDLINE | ID: mdl-28337884

ABSTRACT

Orofacial pain is associated with peripheral and central sensitization of trigeminal nociceptive neurons. Nerve injury results in release of chemical mediators that contribute to persistent pain conditions. The activation of the transient receptor potential vanilloid 1 (TRPV1), promotes release of calcitonin gene-related peptide (CGRP) and substance P (SP) from trigeminal nerve terminals. CGRP and SP contribute to the development of peripheral hyperalgesia. The expression of SP and CGRP by primary afferent neurons is rapidly increased in response to peripheral inflammation. CGRP receptor activation promotes activation of AMPA receptors, leading to increased firing of neurons which is reflected as central sensitization. In this study we investigated whether inferior alveolar nerve (IAN) injury influences AMPA receptors, CGRP, SP and TRPV1 expression in the trigeminal ganglion (TG). The relative expression of the protein of interest from naive rats was compared to those from injured rats and animals that received low level laser therapy (LLLT). IAN-injury did not change expression of GluA1, GluA2 and CGRP, but increased the expression of TRPV1 and SP. LLLT increases GluA1 and GluA2 expression and decreases TVPV1, SP and CGRP. These results, together with previous behavioral data, suggest that IAN-injury induced changes in the proteins analyzed, which could impact on nociceptive threshold. These data may help to understand the molecular mechanisms of pain sensitization in the TG.


Subject(s)
Facial Nerve Injuries/radiotherapy , Gene Expression Regulation/radiation effects , Low-Level Light Therapy , Mandibular Nerve/radiation effects , Trigeminal Ganglion/radiation effects , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Facial Nerve Injuries/genetics , Facial Nerve Injuries/metabolism , Facial Nerve Injuries/pathology , Male , Mandibular Nerve/metabolism , Mandibular Nerve/pathology , Neurons, Afferent/metabolism , Neurons, Afferent/pathology , Neurons, Afferent/radiation effects , Photic Stimulation/methods , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Signal Transduction , Substance P/genetics , Substance P/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Trigeminal Ganglion/injuries , Trigeminal Ganglion/metabolism
9.
J Neuroinflammation ; 9: 78, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22537405

ABSTRACT

BACKGROUND: The transcription factor SRF (serum response factor) mediates neuronal survival in vitro. However, data available so far suggest that SRF is largely dispensable for neuron survival during physiological brain function. FINDINGS: Here, we demonstrate that upon neuronal injury, that is facial nerve transection, constitutively-active SRF-VP16 enhances motorneuron survival. SRF-VP16 suppressed active caspase 3 abundance in vitro and enhanced neuron survival upon camptothecin induced apoptosis. Following nerve fiber injury in vitro, SRF-VP16 improved survival of neurons and re-growth of severed neurites. Further, SRF-VP16 enhanced immune responses (that is microglia and T cell activation) associated with neuronal injury in vivo. Genome-wide transcriptomics identified target genes associated with axonal injury and modulated by SRF-VP16. CONCLUSION: In sum, this is a first report describing a neuronal injury-related survival function for SRF.


Subject(s)
Axons/pathology , Facial Nerve Injuries/pathology , Neurons/pathology , Peripheral Nerve Injuries/pathology , Serum Response Factor/physiology , Animals , Axons/physiology , Cell Survival/genetics , Disease Models, Animal , Facial Nerve Injuries/genetics , Mice , Mice, Knockout , Neurons/physiology , Peripheral Nerve Injuries/genetics , Serum Response Factor/deficiency , Serum Response Factor/genetics
10.
Glia ; 60(5): 782-93, 2012 May.
Article in English | MEDLINE | ID: mdl-22344792

ABSTRACT

Activated microglia are observed in various neurodegenerative diseases and are thought to be involved in the processes of neuronal cell death. Motoneuron damage in the facial nuclei after facial nerve avulsion is accelerated in presymptomatic transgenic rats expressing human mutant Cu(2+) /Zn(2+) superoxide dismutase 1 (SOD1), compared with that in wild-type rats. To reveal the functional role of microglia in motoneuronal death, we investigated the microglial response after facial nerve avulsion in presymptomatic mutant SOD1(H46R) (mSOD1(H46R) ) rats. At 3 days after avulsion, microglial clusters were observed in the facial nuclei of both wild-type and mSOD1(H46R) rats. The numbers of microglial clusters, proliferating microglia, and microglial attachments to motoneurons were significantly higher in mSOD1(H46R) rats, compared with those in wild-type rats. Immunopositive signals for the phagocytic marker ED1 were significantly stronger in mSOD1(H46R) rats, compared with that in wild-type rats, at 2 weeks after avulsion. Furthermore, primary microglia prepared from mSOD1(H46R) rats showed enhanced phagocytic activity, compared with that in wild-type rats. The expression of P2Y(12) mRNA was higher in the facial nuclei of mSOD1(H46R) rats, compared with that in wild-type rats. A laser microdissection system revealed that the expression of ATF3 mRNA was higher in the motoneurons of mSOD1(H46R) rats, compared with that in wild-type rats, at 2 days after avulsion. These results indicate that microglial activation in response to early neuronal damage increased in mSOD1(H46R) rats and suggest that the enhanced activation of microglia may lead to an increase in the vulnerability of motoneurons after avulsion in mSOD1(H46R) rats.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Disease Models, Animal , Facial Nerve Injuries/metabolism , Microglia/metabolism , Motor Neurons/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Newborn , Cells, Cultured , Facial Nerve Injuries/genetics , Facial Nerve Injuries/pathology , Humans , Microglia/pathology , Motor Neurons/pathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Superoxide Dismutase-1
11.
J Comp Neurol ; 519(17): 3488-506, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21800301

ABSTRACT

Previously, we compared molecular profiles of one population of wild-type (WT) mouse facial motoneurons (FMNs) surviving with FMNs undergoing significant cell death after axotomy. Regardless of their ultimate fate, injured FMNs respond with a vigorous pro-survival/regenerative molecular response. In contrast, the neuropil surrounding the two different injured FMN populations contained distinct molecular differences that support a causative role for glial and/or immune-derived molecules in directing contrasting responses of the same cell types to the same injury. In the current investigation, we utilized the facial nerve axotomy model and a presymptomatic amyotrophic lateral sclerosis (ALS) mouse (SOD1) model to experimentally mimic the axonal die-back process observed in ALS pathogenesis without the confounding variable of disease onset. Presymptomatic SOD1 mice had a significant decrease in FMN survival compared with WT, which suggests an increased susceptibility to axotomy. Laser microdissection was used to accurately collect uninjured and axotomized facial motor nuclei of WT and presymptomatic SOD1 mice for mRNA expression pattern analyses of pro-survival/pro-regeneration genes, neuropil-specific genes, and genes involved in or responsive to the interaction of FMNs and non-neuronal cells. Axotomized presymptomatic SOD1 FMNs displayed a dynamic pro-survival/regenerative response to axotomy, similar to WT, despite increased cell death. However, significant differences were revealed when the axotomy-induced gene expression response of presymptomatic SOD1 neuropil was compared with WT. We propose that the increased susceptibility of presymptomatic SOD1 FMNs to axotomy-induced cell death and, by extrapolation, disease progression, is not intrinsic to the motoneuron, but rather involves a dysregulated response by non-neuronal cells in the surrounding neuropil.


Subject(s)
Gene Expression Regulation , Motor Neurons/metabolism , Superoxide Dismutase/biosynthesis , Animals , Axotomy , Facial Nerve/metabolism , Facial Nerve Injuries/genetics , Facial Nerve Injuries/metabolism , Female , Genetic Predisposition to Disease , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superoxide Dismutase/genetics , Superoxide Dismutase-1
12.
Neuroscience ; 182: 241-7, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21440044

ABSTRACT

Functional recovery following facial nerve injury is poor. Adjacent neuromuscular junctions (NMJs) are "bridged" by terminal Schwann cells and numerous regenerating axonal sprouts. We have recently shown that manual stimulation (MS) restores whisking function and reduces polyinnervation of NMJs. Furthermore, MS requires both insulin-like growth factor-1 (IGF-1) and brain-derived neurotrophic factor (BDNF). Here, we investigated whether fibroblast growth factor-2 (FGF-2) was also required for the beneficial effects of MS. Following transection and suture of the facial nerve (facial-facial anastomisis, FFA) in homozygous mice lacking FGF-2 (FGF-2(-/-)), vibrissal motor performance and the percentage of poly-innervated NMJ were quantified. In intact FGF-2(-/-) mice and their wildtype (WT) counterparts, there were no differences in amplitude of vibrissal whisking (about 50°) or in the percentage of polyinnervated NMJ (0%). After 2 months FFA and handling alone (i.e. no MS), the amplitude of vibrissal whisking in WT-mice decreased to 22±3°. In the FGF-2(-/-) mice, the amplitude was reduced further to 15±4°, that is, function was significantly poorer. Functional deficits were mirrored by increased polyinnervation of NMJ in WT mice (40.33±2.16%) with polyinnervation being increased further in FGF-2(-/-) mice (50.33±4.33%). However, regardless of the genotype, MS increased vibrissal whisking amplitude (WT: 33.9°±7.7; FGF-2(-/-): 33.4°±8.1) and concomitantly reduced polyinnervation (WT: 33.9%±7.7; FGF-2(-/-): 33.4%±8.1) to a similar extent. We conclude that, whereas lack of FGF-2 leads to poor functional recovery and target reinnervation, MS can nevertheless confer some functional benefit in its absence.


Subject(s)
Facial Muscles/innervation , Facial Nerve Injuries/genetics , Facial Nerve Injuries/therapy , Fibroblast Growth Factor 2/deficiency , Musculoskeletal Manipulations/methods , Neuronal Plasticity/genetics , Recovery of Function/genetics , Animals , Disease Models, Animal , Facial Muscles/physiopathology , Facial Nerve Injuries/physiopathology , Fibroblast Growth Factor 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Denervation/methods , Nerve Regeneration/genetics , Vibrissae/innervation
13.
Arch Facial Plast Surg ; 12(5): 315-20, 2010.
Article in English | MEDLINE | ID: mdl-20855773

ABSTRACT

OBJECTIVE: To introduce a Thy1-GFP transgenic rat model, whose axons constitutively express green fluorescent protein (GFP), in order to study facial nerve regeneration. Facial nerve injury can cause devastating physical and social sequelae. The functional recovery of the facial nerve can result in synkinesis and permanent axonal misrouting. Facial nerve research has been hindered by the lack of available animal models and reliable outcome measures. METHODS: Transgenic Thy1-GFP rats underwent a proximal facial nerve crush injury and were imaged at 0, 1, 2, 4, and 8 weeks after injury. Nerve regeneration was assessed via confocal imaging and fluorescence microscopy. RESULTS: Uninjured animals reliably demonstrated facial nerve fluorescence and had predictable anatomical landmarks. Fluorescence microscopy demonstrated the loss and reappearance of fluorescence with regeneration of axons following injury. This was confirmed with the visualization of denervation and reinnervation of zygomaticus muscle motor end plates using confocal microscopy. CONCLUSIONS: The Thy1-GFP rat is a novel transgenic tool that enables direct visualization of facial nerve regeneration after injury. The utility of this model extends to a variety of clinical facial nerve injury paradigms.


Subject(s)
Disease Models, Animal , Facial Nerve Injuries/genetics , Facial Nerve/physiology , Gene Expression/genetics , Green Fluorescent Proteins/genetics , Nerve Regeneration/genetics , Rats, Transgenic/genetics , Thy-1 Antigens/genetics , Animals , Axons/physiology , Facial Nerve Injuries/physiopathology , Microscopy, Confocal , Microscopy, Fluorescence , Nerve Crush , Rats , Rats, Sprague-Dawley
14.
Electromagn Biol Med ; 28(4): 383-90, 2009.
Article in English | MEDLINE | ID: mdl-20017629

ABSTRACT

Rats were exposed to cell phone radiation for 6 hours per day for 18 weeks. The buccal and mandibular branches of the facial nerve were evaluated for this study. The mRNA levels of four proteins that are usually up regulated when an injury has occurred were investigated; included were Calcium ATP-ase, Endothelin, Neural Cell Adhesion Molecule, and Neural Growth Factor. These isolated mRNAs were subjected to RT-PCR and all four were up regulated. The mandibular nerve showed a higher and broader level of up regulation than the buccal nerve. All four mRNA up regulations for the mandibular nerve and two for the buccal nerve were also statistically significant. These specific injury-related findings were mild. As the use of these cell phones continues, there most likely will be permanent damage to these tissues over the years and the likelihood of tumors, cancers, and system failures will potentially increase.


Subject(s)
Cell Phone/instrumentation , Facial Nerve Injuries/genetics , Facial Nerve/radiation effects , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Radio Waves/adverse effects , Animals , Calcium-Transporting ATPases/metabolism , Dose-Response Relationship, Radiation , Endothelins/metabolism , Facial Nerve/metabolism , Facial Nerve Injuries/etiology , Male , Mandibular Nerve/metabolism , Mandibular Nerve/radiation effects , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trigeminal Nerve Injuries
15.
J Neurochem ; 108(6): 1453-63, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19284475

ABSTRACT

Activation of Ras into the GTP-binding, 'ON' state is a key switch in the neurotrophin-mediated neuronal survival and neurite outgrowth, in vitro as well as in vivo. In the current study we explored changes in GTP-Ras levels following facial nerve injury and the ensuing regeneration and the effects of perturbing these changes in vivo using synapsin-promoter mediated neuronal expression of constitutively active Val12H-Ras (synRas). Quantification of GTP-Ras and total Ras revealed a precipitous drop in the relative GTP-Ras levels in the axotomized facial motor nucleus, to 40% of normal levels at 2 days after cut, followed by a partial recovery to 50-65% at 4-28 days. On western blots, control and axotomized nuclei from synRas mutants showed a 2.2- and 2.5-fold elevation in GTP-Ras, respectively, compared with their wild type littermate controls (p < 5%, anova, TUKEY post-hoc), with the levels in the axotomized synRas nucleus slightly but not significantly above that in the uninjured littermate control (p = 9.9%). Similar increase was also observed in the pERK but not pAKT targets of the Ras cascade. This moderate elevation of GTP-Ras strongly curtailed post-traumatic neuronal cell death (-65%), the influx of T-cells (-48%) as well as other parameters of neuroinflammatory response. Although synRas did not affect the speed of axonal regeneration or functional recovery it caused a very pronounced increase in central axonal sprouting. These current data emphasize the role of reduced active Ras, and by extension, the reduced overall level of retrograde neurotrophin signalling after axotomy, in mediating post-traumatic cell death and inflammation and in restricting the sprouting response. Moreover, the neuroprotective and central sprouting-enhancing effects of neuronal Val12H-Ras could help promote recovery in CNS injury.


Subject(s)
Facial Nerve Injuries/physiopathology , GTP-Binding Proteins/physiology , Nerve Regeneration/physiology , Neurons/metabolism , Recovery of Function/physiology , Analysis of Variance , Animals , Axotomy/methods , CD3 Complex/metabolism , Calcitonin Gene-Related Peptide/metabolism , Disease Models, Animal , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Facial Nerve Injuries/genetics , GTP-Binding Proteins/genetics , Galanin/metabolism , Histidine/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nerve Regeneration/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recovery of Function/genetics , Time Factors , Valine/genetics , ras Proteins/genetics , ras Proteins/physiology
16.
Neurosci Res ; 60(1): 82-94, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18022721

ABSTRACT

Prosaposin acts as a neurotrophic factor, in addition to its role as the precursor protein for saposins A, B, C, and D, which are activators for specific sphingolipid hydrolases in lysosomes. In rats, the prosaposin gene generates two alternative splicing forms of mRNA: Pro+9 containing a 9-base insertion and Pro+0 without. The expression of these mRNAs changes after brain injury. We examined the expression patterns of the alternative splicing forms of prosaposin mRNA in the rat facial nerve nucleus for 52 days following facial nerve transection. Pro+0 mRNA increased within 3 days of transection, peaked after 5-10 days, and remained significantly elevated for 21 days. In contrast, the expression of Pro+9 mRNA was constant throughout the regenerative period. Prosaposin mRNA expression increased not only in facial motoneurons, but also in microglia during facial nerve regeneration. Our findings indicate that the saposin B domain of prosaposin, which is the domain affected by alternative splicing, plays an important role in both neurons and microglia during neuroregeneration.


Subject(s)
Alternative Splicing/genetics , Facial Nerve Injuries/metabolism , Facial Nerve/metabolism , Motor Neurons/metabolism , Rhombencephalon/metabolism , Saposins/genetics , Animals , Denervation , Facial Nerve/physiopathology , Facial Nerve Injuries/genetics , Facial Nerve Injuries/physiopathology , Gene Expression Regulation/physiology , Male , Microglia/metabolism , Nerve Regeneration/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/physiology , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recovery of Function/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rhombencephalon/physiopathology , Saposins/biosynthesis , Saposins/chemistry , Up-Regulation/genetics
17.
Ital J Anat Embryol ; 112(3): 157-68, 2007.
Article in English | MEDLINE | ID: mdl-18078237

ABSTRACT

The current work has documented the expression of the mRNAs for serine protease inhibitor 3 (SPI-3) in the facial and hypoglossal nuclei following peripheral nerve transection by using the in situ hybridization method. The signals appeared 6 hour after nerve injury; they became stronger on day 1 of injury and persisted for 21 days. SPI-3 may be involved during early events of modulating the activities of serine proteases following nerve injury. Such activities may include synaptic stripping and re-organization and facilitation of glial cell reaction to nerve injury. In the later stages of nerve injury SPI-3 may enhance neuronal survival, growth of neurites and re-establishment of synaptic contacts in the facial and hypoglossal nuclei. Hypoglossal but not facial nerve transection caused the expression of mRNAs for SPI-3 in the pineal gland. The signals appeared 6 hours after nerve injury and persisted for 21 days. The significance of this observation is not known but it indicates that the pineal gland senses injury to some peripheral nerves including the hypoglossal nerve. The study has also showed that axotomy of the sciatic nerve did not give rise to the up-regulation of the mRNAs for SPI-3 in the spinal cord. There was no hybridization signals in the lumbar segments; an indication that SPI-3 may not form part of molecules that are released during sciatic nerve transaction by the neural and non-neural cells of the spinal cord. At the moment there are no antibodies for SPI-3 and therefore future studies are needed to verify the findings. It will be interesting also to study on the role of pineal gland during peripheral nerve injuries.


Subject(s)
Acute-Phase Proteins/genetics , Facial Nerve Injuries/metabolism , Hypoglossal Nerve Diseases/metabolism , Motor Neurons/metabolism , Peripheral Nervous System/injuries , RNA, Messenger/metabolism , Serpins/genetics , Animals , Facial Nerve/metabolism , Facial Nerve/pathology , Facial Nerve Injuries/genetics , Facial Nerve Injuries/pathology , Growth Cones/metabolism , Growth Cones/ultrastructure , Hypoglossal Nerve/metabolism , Hypoglossal Nerve/pathology , Hypoglossal Nerve Diseases/genetics , Hypoglossal Nerve Diseases/pathology , In Situ Hybridization , Male , Motor Neurons/pathology , Nerve Regeneration/genetics , Neural Pathways/metabolism , Neural Pathways/pathology , Peripheral Nerves/immunology , Peripheral Nerves/pathology , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Pineal Gland/metabolism , Pineal Gland/pathology , Rats , Rats, Wistar , Recovery of Function , Rhombencephalon/metabolism , Rhombencephalon/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Synapses/metabolism , Synapses/ultrastructure
18.
Neurosci Lett ; 417(2): 160-4, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17418948

ABSTRACT

IL-15 is a potent T cell chemoattractant, and this cytokine and its unique alpha subunits, IL-15R alpha, can modify immune cell expression of several T cell chemokines and their receptors. Facial nerve axotomy in mice leads to T cell migration across an intact blood-brain-barrier (BBB), and under certain conditions T cells can provide neuroprotection to injured neurons in the facial motor nucleus (FMN). Although chemokines and chemoattractant cytokines are thought to be responsible for T cell migration to the injured cell bodies, data addressing this question are lacking. This study tested the hypothesis that T cell homing to the axotomized FMN would be impaired in knockout (KO) mice with the IL-15 and IL-15R alpha genes deleted, and sought to determine if microglial responsiveness and motoneuron death are affected. Both IL-15KO and IL-15R alpha KO mice exhibited a marked reduction in CD3(+) T cells and had fewer MHC2(+) activated microglia in the injured FMN than their respective WT controls at day 14 post-axotomy. Although there was a relative absence of T cell recruitment into the axotomized FMN in both knockout strains, IL-15R alpha KO mice had five times more motoneuron death (characterized by perineuronal microglial clusters engulfing dead motoneurons) than their WT controls, whereas dead neurons in IL-15KO did not differ from their WT controls. Further studies are needed to dissect the mechanisms that underlie these observations (e.g., central vs. peripheral immune contributions).


Subject(s)
Chemotaxis, Leukocyte/immunology , Facial Nerve Injuries/immunology , Gliosis/immunology , Interleukin-15/immunology , Motor Neurons/immunology , Nerve Degeneration/immunology , Animals , Axotomy , Chemotaxis, Leukocyte/genetics , Facial Nerve/immunology , Facial Nerve/metabolism , Facial Nerve Injuries/genetics , Facial Nerve Injuries/metabolism , Female , Gliosis/genetics , Interleukin-15/genetics , Interleukin-15 Receptor alpha Subunit/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Neuroscience ; 141(4): 1827-34, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16843609

ABSTRACT

The normal function of the cellular prion protein, PrP(c), remains largely unknown. Recently, PrP(c) has been implicated in the regulation of neuronal survival and was shown to confer neuroprotection in the brain. To pursue investigation of the role of PrP(c) in the CNS, we used the facial nerve section, a well-established experimental model of motoneuronal stress. Nerve sections were performed in 2- and 7-day-old newborn mice and in 2 month-old adult mice expressing different levels of PrP(c). We observed no differences in motoneuronal death triggered by facial nerve section between Prnp-/- and wild-type mice, whether in neonatal or adult mice. By contrast, overexpression of PrP(c) in Tga20 newborn mice was correlated with a better survival of motoneurons in the few days following axotomy. The protection was, however transient since motoneuron number in lesioned facial nuclei of Tga20 mice became identical to that of wild-type mice 7 days and 14 days following the lesion when performed in 2- and 7-day-old mice respectively. In Tga20 adult mice, no protection was observed 2 months after the lesion, a time with a significant degree of motoneuron death in adult control mice. These results, while providing further evidence that PrP(c) is endowed with neuroprotective capacity in vivo, also suggest that PrP(c) does not play a physiological role in the regulation of motoneuronal survival.


Subject(s)
Axotomy/adverse effects , Facial Nerve Injuries/pathology , Gene Expression Regulation, Developmental/physiology , Motor Neurons/physiology , Prions/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Axotomy/methods , Blotting, Western/methods , Caspase 3/metabolism , Cell Count/methods , Cell Death/physiology , Facial Nerve Injuries/etiology , Facial Nerve Injuries/genetics , Immunohistochemistry/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prions/genetics , Time Factors
20.
Exp Neurol ; 201(1): 212-24, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16806176

ABSTRACT

The CD4(+) T lymphocyte has recently been found to promote facial motoneuron (FMN) survival after nerve injury. Signal Transducer and Activator of Transcription (STAT)4 and STAT6 are key proteins involved in the CD4(+) T cell differentiation pathways leading to T helper type (Th)1 and Th2 cell development, respectively. To determine which CD4(+) T cell subset mediates FMN survival, the facial nerve axotomy paradigm was applied to STAT4-deficient (-/-) and STAT6-/- mice. A significant decrease in FMN survival 4 weeks after axotomy was observed in STAT6-/- mice compared to wild-type (WT) or STAT4-/- mice. Reconstituting STAT6-/- mice with CD4(+) T cells obtained from WT mice promoted WT levels of FMN survival after injury. Furthermore, rescue of FMN from axotomy-induced cell death in recombination activating gene (RAG)-2-/- mice (lacking T and B cells) could be achieved only by reconstitution with CD4(+) T cells expressing functional STAT6 protein. To determine if either the Th1 cytokine, interferon-gamma (IFN-gamma) or the Th2 cytokine IL-4 is involved in mediating FMN survival, facial nerve axotomy was applied to IFN-gamma-/- and IL-4-/- mice. A significant decrease in FMN survival after axotomy occurred in IL-4-/- but not in IFN-gamma-/- mice compared to WT mice, indicating that IL-4 but not IFN-gamma is important for FMN survival after nerve injury. In WT mice, intracellular IFN-gamma vs. IL-4 expression was examined in CD4(+) T cells from draining cervical lymph nodes 14 days after axotomy, and substantial increase in the production of both CD4(+) effector T cell subsets was found. Collectively, these data suggest that STAT6-mediated CD4(+) T cell differentiation into the Th2 subset is necessary for FMN survival. A hypothesis relevant to motoneuron disease progression is presented.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Facial Nerve/physiopathology , Interleukin-4/physiology , Motor Neurons/physiology , STAT6 Transcription Factor/physiology , Animals , Axotomy , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Survival/immunology , Cell Survival/physiology , Facial Nerve/immunology , Facial Nerve/surgery , Facial Nerve Injuries/genetics , Facial Nerve Injuries/immunology , Facial Nerve Injuries/physiopathology , Genotype , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/physiology , Interleukin-4/biosynthesis , Interleukin-4/genetics , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Motor Neurons/cytology , Motor Neurons/immunology , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/physiology , STAT6 Transcription Factor/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL