Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Comp Neurol ; 529(13): 3313-3320, 2021 09.
Article in English | MEDLINE | ID: mdl-34008871

ABSTRACT

The retrotrapezoid nucleus (RTN) is a hub for respiratory chemoregulation in the mammal brainstem that integrates chemosensory information from peripheral sites and central relays. Chemosensitive neurons of the RTN express specific genetic and molecular determinants, which have been used to identify RTN precise location within the brainstem of rodents and nonhuman primates. Based on a comparative approach, we hypothesized that among mammals, neurons exhibiting the same specific molecular and genetic signature would have the same function. The co-expression of preprogalanin (PPGAL) and SLC17A6 (VGluT2) mRNAs with duplex in situ hybridization has been studied in formalin fixed paraffin-embedded postmortem human brainstems. Two specimens were processed and analyzed in line with RTN descriptions in adult rats and macaques. Double-labeled PPGAL+/SLC17A6+ neurons were only identified in the parafacial region of the brainstem. These neurons were found surrounding the nucleus of the facial nerve, located ventrally to the nucleus VII on caudal sections, and slightly more dorsally on rostral sections. The expression of neuromedin B (NMB) mRNA as a single marker of chemosensitive RTN neurons has not been confirmed in humans. The location of the RTN in human adults is provided. This should help to develop investigation tools combining anatomic high-resolution imaging and respiratory functional investigations to explore the pathogenic role of the RTN in congenital or acquired neurodegenerative diseases.


Subject(s)
Brain Stem/metabolism , Brain Stem/pathology , Galanin/biosynthesis , Neurons/metabolism , Neurons/pathology , Vesicular Glutamate Transport Protein 2/biosynthesis , Biomarkers/metabolism , Facial Nucleus/metabolism , Facial Nucleus/pathology , Galanin/genetics , Gene Expression , Humans , Trapezoid Body/metabolism , Trapezoid Body/pathology , Vesicular Glutamate Transport Protein 2/genetics
2.
PLoS One ; 15(12): e0241315, 2020.
Article in English | MEDLINE | ID: mdl-33259479

ABSTRACT

Neurotrophic factor prosaposin (PS) is a precursor for saposins A, B, C, and D, which are activators for specific sphingolipid hydrolases in lysosomes. Both saposins and PS are widely contained in various tissues. The brain, skeletal muscle, and heart cells predominantly contain unprocessed PS rather than saposins. PS and PS-derived peptides stimulate neuritogenesis and increase choline acetyltransferase activity in neuroblastoma cells and prevent programmed cell death in neurons. We previously detected increases in PS immunoactivity and its mRNA in the rat facial nucleus following facial nerve transection. PS mRNA expression increased not only in facial motoneurons, but also in microglia during facial nerve regeneration. In the present study, we examined the changes in immunoreactivity of the PS receptors GPR37 and GPR37L1 in the rat facial nucleus following facial nerve transection. Following facial nerve transection, many small Iba1- and glial fibrillary acidic protein (GFAP)-positive cells with strong GPR37L1 immunoreactivity, including microglia and astrocytes, were observed predominately on the operated side. These results indicate that GPR37 mainly works in neurons, whereas GPR37L1 is predominant in microglia or astrocytes, and suggest that increased PS in damaged neurons stimulates microglia or astrocytes via PS receptor GPR37L1 to produce neurotrophic factors for neuronal recovery.


Subject(s)
Facial Nerve/metabolism , Nerve Regeneration/genetics , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Saposins/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Facial Nerve/surgery , Facial Nucleus/metabolism , Facial Nucleus/pathology , Gene Expression Regulation/genetics , Humans , Microglia/metabolism , Microglia/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , RNA, Messenger/genetics , Rats
3.
World Neurosurg ; 109: e252-e257, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28987828

ABSTRACT

OBJECTIVE: To research the early acute response events of facial nerve injury. METHODS: Sixty male Sprague-Dawley rats were randomly divided into 2 groups. Facial nerve anastomosis was performed for rats in study group. Rats in control group underwent the same surgical procedure, but without cutting off the facial nerve. Before nerve anastomosis and at days 1, 3, 7, 14, and 28 after nerve anastomosis, 5 rats of each group were selected and right side brainstem tissue samples containing the facial nerve nucleus were obtained. Hematoxylin and eosin (H&E) staining and TUNEL detection was performed to observe facial neurons changes. Facial neurons mortality and apoptosis were studied. Expression of caspase-3, LC3, and Beclin1 was detected with Western blot assay. RESULTS: In study group on day 7 day after nerve anastomosis, Nissl body dissolution and apoptotic facial neurons were significantly increased, the typical polygonal shape and swollen cells disappeared, the number of facial neurons was significantly lower, and the number of apoptotic facial neurons was significantly higher (P < 0.01). In addition, facial neuron mortality rate was significantly increased at day 7, reaching the peak at day 14. Expression of caspase-3, LC3, and Beclin1 was also significantly up-regulated after nerve anastomosis. CONCLUSION: Nissl body dissolution, typical polygonal shape disappearing, cell swelling, facial neuron mortality and apoptosis, and up-regulated expression of caspase-3, LC3, and Beclin1 are the early events of cell death after facial nerve injury, which are the important precursors to facial nerve injury.


Subject(s)
Anastomosis, Surgical , Cell Death/physiology , Facial Nerve Injuries/pathology , Facial Nerve Injuries/surgery , Facial Nerve/pathology , Facial Nucleus/pathology , Neurons/pathology , Retrograde Degeneration/pathology , Animals , Beclin-1/analysis , Caspase 3/analysis , Male , Rats , Rats, Sprague-Dawley
4.
J Chem Neuroanat ; 79: 38-50, 2017 01.
Article in English | MEDLINE | ID: mdl-27865907

ABSTRACT

We previously reported that perineuronal astrocytic and microglial reactions are drastically upregulated in the facial nucleus after facial axotomy at the brain stem surface or the stylomastoid foramen. Furthermore, periaxonal astrocytic and microglial reactions develop retrogradely in the central facial tract which contains proximal facial axons in the brain stem. Because reconnection of interrupted peripheral nerve by microsurgical suture is a common clinical practice, the aim of this study was to investigate the spatiotemporal patterns of glial reactions in the central facial tract and the facial nucleus after facial neurorrhaphy. Here, we show immunofluorescent and immunohistochemical evidence that facial neurorrhaphy at the stylomastoid foramen largely prevented axotomy-induced astrocytic and microglial activation in the central facial tract. In contrast, glial reactions in the facial nucleus were still highly elevated after facial neurorrhaphy. Microglial and astrocytic processes were observed to ensheath the facial motoneurons in the facial nucleus. Nevertheless, the transformation of ramified to amoeboid shape of microglia, occurring at 10 weeks after facial axotomy, was not seen after neurorrhaphy. We further examined the effect of N-nitro-l-arginine methyl ester (L-NAME), an inhibitor of nitric oxide synthase (NOS), on glial reactions after neurorrhaphy. Western blot analyses demonstrate that inhibition of nitric oxide (NO) production significantly reduced microglial but not astrocytic reaction in the facial nucleus after neurorrhaphy. Taken together, these results indicate that in contrast to the intense glial reactions in both the central facial tract and the facial nucleus after facial axotomy, glial reactions are differentially regulated in these two compartments after facial neurorrhaphy. NO is involved in the activation of microglia in the facial nucleus after facial neurorrhaphy.


Subject(s)
Facial Nerve Injuries/pathology , Facial Nerve/pathology , Facial Nucleus/pathology , Neuroglia/pathology , Animals , Facial Nerve/metabolism , Facial Nerve Injuries/metabolism , Facial Nucleus/metabolism , Male , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley
5.
J Integr Neurosci ; 15(2): 175-90, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27345027

ABSTRACT

It was suggested that muscarinic, and nicotinic receptors increase free Ca[Formula: see text] levels in the facial nerve nucleus via various channels following facial nerve injury. However, intracellular Ca[Formula: see text] overload can trigger either necrotic or apoptotic cell death. It is assumed that, following facial nerve injury, the interactions of nicotinic and muscarinic acetylcholine receptors in facial nerve nucleus may negatively regulate free Ca[Formula: see text] concentrations in the facial nerve nucleus, which provide important information for the repair and regeneration of the facial nerve. The present study investigated the regulatory effects of nicotine on muscarinic receptor-mediated free calcium ion level changes in the facial nucleus in a rat model of facial nerve injury at 7, 30, and 90 days following facial nerve injury using laser confocal microscopy. The dose-dependent regulation of nicotine on muscarinic receptor-mediated free calcium ion level changes in the facial nucleus may decrease the range of free Ca[Formula: see text] increases following facial nerve injury, which is important for nerve cell regeneration. It is concluded that the negative effects of nicotine on muscarinic receptors are related to the [Formula: see text] subtype of nicotinic receptors.


Subject(s)
Calcium/metabolism , Facial Nerve Injuries/drug therapy , Facial Nucleus/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Muscarinic/metabolism , Animals , Cations, Divalent/metabolism , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Facial Nerve Injuries/metabolism , Facial Nerve Injuries/pathology , Facial Nucleus/metabolism , Facial Nucleus/pathology , Female , Male , Nerve Regeneration/drug effects , Random Allocation , Rats, Sprague-Dawley , Tissue Culture Techniques
6.
BMC Neurosci ; 16: 39, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26156498

ABSTRACT

BACKGROUND: Axons from the visceral motor neurons (vMNs) project from nuclei in the hindbrain to innervate autonomic ganglia and branchial arch-derived muscles. Although much is known about the events that govern specification of somatic motor neurons, the genetic pathways responsible for the development of vMNs are less well characterized. We know that vMNs, like all motor neurons, depend on sonic hedgehog signaling for their generation. Similarly, the paired-like homeobox 2b (Phox2b) gene, which is expressed in both proliferating progenitors and post-mitotic motor neurons, is essential for the development of vMNs. Given that our previous study identified a novel role for the short stature homeobox 2 (Shox2) gene in the hindbrain, and since SHOX2 has been shown to regulate transcription of islet 1 (Isl1), an important regulator of vMN development, we sought to determine whether Shox2 is required for the proper development of the facial motor nucleus. RESULTS: Using a Nestin-Cre driver, we show that elimination of Shox2 throughout the brain results in elevated cell death in the facial motor nucleus at embryonic day 12.5 (E12.5) and E14.5, which correlates with impaired axonal projection properties of vMNs. We also observed changes in the spatial expression of the vMN cell fate factors Isl1 and Phox2b, and concomitant defects in Shh and Ptch1 expression in Shox2 mutants. Furthermore, we demonstrate that elimination of Shox2 results in the loss of dorsomedial and ventromedial subnuclei by postnatal day 0 (P0), which may explain the changes in physical activity and impaired feeding/nursing behavior in Shox2 mutants. CONCLUSIONS: Combined, our data show that Shox2 is required for development of the facial motor nucleus and its associated facial (VII) nerves, and serves as a new molecular tool to probe the genetic programs of this understudied hindbrain region.


Subject(s)
Facial Nerve/embryology , Facial Nucleus/embryology , Homeodomain Proteins/metabolism , Motor Neurons/metabolism , Animals , Animals, Newborn , Apoptosis/physiology , Facial Nerve/metabolism , Facial Nerve/pathology , Facial Nucleus/metabolism , Facial Nucleus/pathology , Feeding Behavior/physiology , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Intermediate Filaments/metabolism , LIM-Homeodomain Proteins/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/pathology , Patched Receptors , Patched-1 Receptor , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism
7.
J Neurosci Res ; 93(6): 913-21, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25581872

ABSTRACT

This study biochemically determined glycogen content in the axotomized facial nucleus of adult rats up to 35 days postinsult. The amounts of glycogen in the transected facial nucleus were significantly increased at 5 days postinsult, peaked at 7 days postinsult, and declined to the control levels at 21-35 days postinsult. Immunohistochemical analysis with antiglycogen antibody revealed that the quantity of glycogen granules in the axotomized facial nucleus was greater than that in the control nucleus at 7 days postinjury. Dual staining methods with antiglycogen antibody and a motoneuron marker clarified that the glycogen was localized mainly in motoneurons. Immunoblotting and quantification analysis revealed that the ratio of inactive glycogen synthase (GS) to total GS was significantly decreased in the injured nucleus at about 1-3 days postinsult and significantly increased from 7 to 14 days postinsult, suggesting that glycogen is actively synthesized in the early period postinjury but suppressed after 7 days postinsult. The enhanced glycogen at about 5-7 days postinsult is suggested to be responsible for the decrease in inactive GS levels, and the decrease of glycogen after 7 days postinsult is considered to be caused by increased inactive GS levels and possibly the increase in active glycogen phosphorylase.


Subject(s)
Facial Nucleus/injuries , Facial Nucleus/pathology , Glycogen/metabolism , Motor Neurons/metabolism , Animals , Axotomy , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , Glycogen Synthase/metabolism , Male , Motor Neurons/classification , Rats , Rats, Wistar , Time Factors
8.
Brain Behav Immun ; 40: 55-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24911596

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease involving motoneuron (MN) axonal withdrawal and cell death. Previously, we established that facial MN (FMN) survival levels in the SOD1(G93A) transgenic mouse model of ALS are reduced and nerve regeneration is delayed, similar to immunodeficient RAG2(-/-) mice, after facial nerve axotomy. The objective of this study was to examine the functionality of SOD1(G93A) splenic microenvironment, focusing on CD4(+) T cells, with regard to defects in immune-mediated neuroprotection of injured MN. We utilized the RAG2(-/-) and SOD1(G93A) mouse models, along with the facial nerve axotomy paradigm and a variety of cellular adoptive transfers, to assess immune-mediated neuroprotection of FMN survival levels. We determined that adoptively transferred SOD1(G93A) unfractionated splenocytes into RAG2(-/-) mice were unable to support FMN survival after axotomy, but that adoptive transfer of isolated SOD1(G93A) CD4(+) T cells could. Although WT unfractionated splenocytes adoptively transferred into SOD1(G93A) mice were able to maintain FMN survival levels, WT CD4(+) T cells alone could not. Importantly, these results suggest that SOD1(G93A) CD4(+) T cells retain neuroprotective functionality when removed from a dysfunctional SOD1(G93A) peripheral splenic microenvironment. These results also indicate that the SOD1(G93A) central nervous system microenvironment is able to re-activate CD4(+) T cells for immune-mediated neuroprotection when a permissive peripheral microenvironment exists. We hypothesize that a suppressive SOD1(G93A) peripheral splenic microenvironment may compromise neuroprotective CD4(+) T cell activation and/or differentiation, which, in turn, results in impaired immune-mediated neuroprotection for MN survival after peripheral axotomy in SOD1(G93A) mice.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , CD4-Positive T-Lymphocytes/immunology , Facial Nucleus/immunology , Motor Neurons/immunology , Superoxide Dismutase/genetics , Adoptive Transfer , Amyotrophic Lateral Sclerosis/pathology , Animals , Axotomy , CD4-Positive T-Lymphocytes/transplantation , DNA-Binding Proteins/genetics , Facial Nerve Injuries , Facial Nucleus/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/pathology , Superoxide Dismutase-1
9.
J Comp Neurol ; 522(10): 2349-76, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24424947

ABSTRACT

The target disconnection theory of amyotrophic lateral sclerosis (ALS) pathogenesis suggests that disease onset is initiated by a peripheral pathological event resulting in neuromuscular junction loss and motoneuron (MN) degeneration. Presymptomatic mSOD1(G93A) mouse facial MN (FMN) are more susceptible to axotomy-induced cell death than wild-type (WT) FMN, which suggests additional CNS pathology. We have previously determined that the mSOD1 molecular response to facial nerve axotomy is phenotypically regenerative and indistinguishable from WT, whereas the surrounding microenvironment shows significant dysregulation in the mSOD1 facial nucleus. To elucidate the mechanisms underlying the enhanced mSOD1 FMN loss after axotomy, we superimposed the facial nerve axotomy model on presymptomatic mSOD1 mice and investigated gene expression for death receptor pathways after target disconnection by axotomy vs. disease progression. We determined that the TNFR1 death receptor pathway is involved in axotomy-induced FMN death in WT and is partially responsible for the mSOD1 FMN death. In contrast, an inherent mSOD1 CNS pathology resulted in a suppressed glial reaction and an upregulation in the Fas death pathway after target disconnection. We propose that the dysregulated mSOD1 glia fail to provide support the injured MN, leading to Fas-induced FMN death. Finally, we demonstrate that, during disease progression, the mSOD1 facial nucleus displays target disconnection-induced gene expression changes that mirror those induced by axotomy. This validates the use of axotomy as an investigative tool in understanding the role of peripheral target disconnection in the pathogenesis of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Cell Death/physiology , Facial Nerve/physiopathology , Motor Neurons/physiology , Nerve Degeneration/physiopathology , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axotomy , Disease Progression , Facial Nerve/pathology , Facial Nucleus/pathology , Facial Nucleus/physiopathology , Female , Gene Expression , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Motor Neurons/pathology , Nerve Degeneration/pathology , Neuroglia/pathology , Neuroglia/physiology , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , fas Receptor/metabolism
10.
Neuroscience ; 248: 307-18, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23806716

ABSTRACT

Crush injuries of peripheral nerves typically lead to axonotmesis, axonal damage without disruption of connective tissue sheaths. Generally, human patients and experimental animals recover well after axonotmesis and the favorable outcome has been attributed to precise axonal reinnervation of the original peripheral targets. Here we assessed functionally and morphologically the long-term consequences of facial nerve axonotmesis in rats. Expectedly, we found that 5 months after crush or cryogenic nerve lesion, the numbers of motoneurons with regenerated axons and their projection pattern into the main branches of the facial nerve were similar to those in control animals suggesting precise target reinnervation. Unexpectedly, however, we found that functional recovery, estimated by vibrissal motion analysis, was incomplete at 2 months after injury and did not improve thereafter. The maximum amplitude of whisking remained substantially, by more than 30% lower than control values even 5 months after axonotmesis. Morphological analyses showed that the facial motoneurons ipsilateral to injury were innervated by lower numbers of glutamatergic terminals (-15%) and cholinergic perisomatic boutons (-26%) compared with the contralateral non-injured motoneurons. The structural deficits were correlated with functional performance of individual animals and associated with microgliosis in the facial nucleus but not with polyinnervation of muscle fibers. These results support the idea that restricted CNS plasticity and insufficient afferent inputs to motoneurons may substantially contribute to functional deficits after facial nerve injuries, possibly including pathologic conditions in humans like axonotmesis in idiopathic facial nerve (Bell's) palsy.


Subject(s)
Facial Nerve Injuries/rehabilitation , Facial Nerve/physiopathology , Facial Nucleus/cytology , Nerve Regeneration , Presynaptic Terminals/pathology , Recovery of Function , Animals , Disease Models, Animal , Facial Nerve Injuries/pathology , Facial Nerve Injuries/physiopathology , Facial Nucleus/pathology , Facial Nucleus/physiopathology , Male , Motor Neurons/cytology , Motor Neurons/pathology , Nerve Crush/methods , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...