Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 506
Filter
1.
Neuropharmacology ; 245: 109818, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38142931

ABSTRACT

Cardiac autonomic neuropathy resulting from human immunodeficiency virus (HIV) infection is common; however, its mechanism remains unknown. The current work attempted to explore the function and mechanism of the P2Y13 receptor in HIV-glycoprotein 120 (gp120)-induced neuropathy in cervical sympathetic ganglion. The superior cervical ganglion (SCG) of the male SD rat was coated with HIV-gp120 to establish a model of autonomic neuropathy. In each group, we measured heart rate, blood pressure, heart rate variability, sympathetic nerve discharge and cardiac function. The expression of P2Y13 mRNA and protein in the SCG was tested by real-time polymerase chain reaction and western blotting. Additionally, this study focused on identifying the protein levels of NOD-like receptor family pyrin domain-containing 3 (NLRP3), Caspase-1, Gasdermin D (GSDMD), interleukin (IL)-1ß and IL-18 in the SCG using western blotting and immunofluorescence. In gp120 rats, increased blood pressure, heart rate, cardiac sympathetic nerve activity, P2Y13 receptor levels and decreased cardiac function could be found. P2Y13 shRNA or MRS2211 inhibited the above mentioned changes induced by gp120, suggesting that the P2Y13 receptor may be engaged in gp120-induced sympathetic nerve injury. Moreover, the levels of NLRP3, Caspase-1, GSDMD, IL-1ß and IL-18 in the gp120 group were increased, while significantly decreased by P2Y13 shRNA or MRS2211. Therefore, the P2Y13 receptor is involved in gp120-induced sympathetic neuropathy, and its molecular mechanism shows an association with the activation of the NLRP3 inflammasome, followed by GSDMD formation along with the release of inflammatory factors including IL-1ß and IL-18. This article is part of the Special Issue on "Purinergic Signaling: 50 years".


Subject(s)
HIV Infections , HIV-1 , Peripheral Nervous System Diseases , Receptors, Purinergic P2 , Animals , Male , Rats , Carrier Proteins , Caspases , Glycoproteins/metabolism , HIV Infections/complications , HIV Infections/metabolism , Inflammasomes/metabolism , Interleukin-18/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peripheral Nervous System Diseases/virology , Rats, Sprague-Dawley , RNA, Small Interfering , Superior Cervical Ganglion/metabolism , HIV Envelope Protein gp120/metabolism , Receptors, Purinergic P2/metabolism
2.
Can J Physiol Pharmacol ; 101(10): 539-547, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37406358

ABSTRACT

Ganglionic long-term potentiation (gLTP) in the rat superior cervical ganglion (SCG) is differentially modulated by neurotrophic factors (Nts): brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). KCNQ/M channels, key regulators of neuronal excitability, and firing pattern are modulated by Nts; therefore, they might contribute to gLTP expression and to the Nts-dependent modulation of gLTP. In the SCG of rats, we characterized the presence of the KCNQ2 isoform and the effects of opposite KCNQ/M channel modulators on gLTP in control condition and under Nts modulation. Immunohistochemical and reverse transcriptase polymerase chain reaction analyses showed the expression of the KCNQ2 isoform. We found that 1 µmol/L XE991, a channel inhibitor, significantly reduced gLTP (∼50%), whereas 5 µmol/L flupirtine, a channel activator, significantly increased gLTP (1.3- to 1.7-fold). Both modulators counterbalanced the effects of the Nts on gLTP. Data suggest that KCNQ/M channels are likely involved in gLTP expression and in the modulation exerted by BDNF and NGF.


Subject(s)
Long-Term Potentiation , Superior Cervical Ganglion , Rats , Animals , Superior Cervical Ganglion/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Nerve Growth Factor/pharmacology , Signal Transduction
3.
Cells ; 12(2)2023 01 05.
Article in English | MEDLINE | ID: mdl-36672160

ABSTRACT

The superior cervical ganglion (SCG) is part of the autonomic nervous system providing sympathetic innervation to the head and neck, and has been regularly used to prepare postnatal neuronal cultures for cell biological studies. We found that during development these neurons change tau expression from the low molecular weight (LMW) isoforms to Big tau, with the potential to affect functions associated with tau such as microtubule dynamic and axonal transport. Big tau contains the large 4a exon that transforms tau from LMW isoforms of 45-60 kDa to 110 kDa. We describe tau expression during postnatal development reporting that the transition from LMW tau to Big tau which started at late embryonic stages is completed by about 4-5 weeks postnatally. We confirmed the presence of Big tau in dissociated postnatal SCG neurons making them an ideal system to study the function of Big tau in neurons. We used SCG explants to examine the response of SCG neurons to lesion and found that Big tau expression returned gradually along the regrowing neurites suggesting that it does not drives regeneration, but facilitates the structure/function of mature SCG neurons. The structural/functional roles of Big tau remain unknown, but it is intriguing that neurons that express Big tau appear less vulnerable to tauopathies.


Subject(s)
Neurons , Superior Cervical Ganglion , Superior Cervical Ganglion/metabolism , Neurons/metabolism , Protein Isoforms/metabolism
4.
Dis Markers ; 2023: 9956950, 2023.
Article in English | MEDLINE | ID: mdl-36660202

ABSTRACT

Diabetic cardiovascular autonomic neuropathy (DCAN) is a common complication of diabetes mellitus which brings about high mortality, high morbidity, and large economic burden to the society. Compensatory tachycardia after myocardial ischemia caused by DCAN can increase myocardial injury and result in more damage to the cardiac function. The inflammation induced by hyperglycemia can increase P2X7 receptor expression in the superior cervical ganglion (SCG), resulting in nerve damage. It is proved that inhibiting the expression of P2X7 receptor at the superior cervical ganglion can ameliorate the nociceptive signaling dysregulation induced by DCAN. However, the effective drug used for decreasing P2X7 receptor expression has not been found. Schisandrin B is a traditional Chinese medicine, which has anti-inflammatory and antioxidant effects. Whether Schisandrin B can decrease the expression of P2X7 receptor in diabetic rats to protect the cardiovascular system was investigated in this study. After diabetic model rats were made, Schisandrin B and shRNA of P2X7 receptor were given to different groups to verify the impact of Schisandrin B on the expression of P2X7 receptor. Pathological blood pressure, heart rate, heart rate variability, and sympathetic nerve discharge were ameliorated after administration of Schisandrin B. Moreover, the upregulated protein level of P2X7 receptor, NLRP3 inflammasomes, and interleukin-1ß in diabetic rats were decreased after treatment, which indicates that Schisandrin B can alleviate the chronic inflammation caused by diabetes and decrease the expression levels of P2X7 via NLRP3. These findings suggest that Schisandrin B can be a potential therapeutical agent for DCAN.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Rats , Animals , Superior Cervical Ganglion/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Rats, Sprague-Dawley , Diabetic Neuropathies/etiology , Diabetic Neuropathies/genetics , Inflammation/metabolism
5.
Auton Neurosci ; 244: 103053, 2023 01.
Article in English | MEDLINE | ID: mdl-36463578

ABSTRACT

BACKGROUND: The superior cervical ganglion (SCG) plays critical roles in the regulation of blood pressure and cardiac output. Metabotropic glutamate receptors (mGluRs) in the SCG are not clearly elucidated yet. Most studies on the expression and functions of mGluRs in the SCG focused on the cultured SCG neurons, and yet little information has been reported in the SCG tissue. Chronic intermittent hypoxia (CIH), one of the major clinical features of obstructive sleep apnea (OSA) patients, is a critical pathological cause of secondary hypertension in OSA patients, but its impact on the level of mGluRs in the SCG is unknown. OBJECTIVE: To explore the expression and localization of mGluR2/3 and the effect of CIH on mGluR2/3 level in rat SCG tissue. METHODS: RT-PCR and immunostaining were conducted to examine the mRNA and protein expression of mGluR2/3 in rat SCG. Immunofluorescence staining was conducted to examine the distribution of mGluR2/3. Rats were divided into control and CIH group which the rats were exposed to CIH for 6 weeks. Western blots were performed to examine the level of mGluR2/3 in rat SCG. RESULTS: mRNAs of mGluR2/3 expressed in rat SCG. mGluR2 distributed in principal neurons and small intensely fluorescent cells but not in satellite glial cells, nerve fibers, and vascular endothelial cells; mGluR3 was detected in nerve fibers rather than in the cells mentioned above. CIH exposure reduced the protein level of mGluR2/3 in rat SCG. CONCLUSION: mGluR2/3 exists in rat SCG with diverse distribution patterns, and may be involved in CIH-induced hypertension.


Subject(s)
Hypertension , Receptors, Metabotropic Glutamate , Sleep Apnea, Obstructive , Superior Cervical Ganglion , Animals , Rats , Endothelial Cells/metabolism , Hypertension/metabolism , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , RNA, Messenger/metabolism , Sleep Apnea, Obstructive/metabolism , Superior Cervical Ganglion/metabolism , Hypoxia/metabolism
6.
Eur J Pharmacol ; 927: 175049, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35644421

ABSTRACT

Purinergic 2Y12 (P2Y12) receptor antagonists are used as platelet aggregation inhibitors. Long non-coding RNAs (lncRNAs) play an important role in neuropathological events. Satellite glial cells (SGCs) in the superior cervical ganglia (SCGs) encircle the somata of neurons. This study explored if the upregulated P2Y12 receptor in SCGs was relevant to lncRNA uc.48+ during myocardial ischemia (MI). The results showed that upregulation of P2Y12 receptor was accompanied by increased expression of uc.48+ in the SCGs of MI rats which displayed abnormal changes in cervical sympathetic nerve activity, blood pressure, heart rate, electrocardiograms and cardiac tissue structure. The P2Y12 antagonist clopidogrel improved abnormal alterations in cardiac function and tissue structure in MI rats. Short hairpin RNA (shRNA) against uc.48+ significantly inhibited P2Y12 receptor upregulation and its co-expression with glial fibrillary acidic protein (GFAP) in SCGs, and ameliorated the cardiac dysfunction in MI rats. By contrast, overexpression of uc.48+ increased the expression of P2Y12 in SCGs and enhanced cervical sympathetic nerve activity in control rats. Direct interaction between uc.48+ and the P2Y12 receptor was predicted using the bioinformatic tool CatRAPID and confirmed by RNA immunoprecipitation. Moreover, overexpression of the P2Y12 receptor reversed the protective effect of uc.48+ shRNA on cardiac dysfunction in MI rats. Uc.48 shRNA treatment also inhibited the enhanced rise of intracellular free Ca2+ level ([Ca2+]i) evoked by the P2Y12 agonist 2-methylthio-adenosine-5'-diphosphate (2-MeSADP) in SGCs of SCGs after oxygen-glucose deprivation (OGD) treatment. These data demonstrated that uc.48+ shRNA could counteract the P2Y12 upregulation and improve P2Y12-implicated cardiac dysfunction due to MI.


Subject(s)
Myocardial Ischemia , Receptors, Purinergic P2Y12 , Superior Cervical Ganglion , Animals , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y12/metabolism , Reflex , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/pathology
7.
J Vis Exp ; (163)2020 09 27.
Article in English | MEDLINE | ID: mdl-33044466

ABSTRACT

Sympathetic neurons from the embryonic rat superior cervical ganglia (SCG) have been used as an in vitro model system for peripheral neurons to study axonal growth, axonal trafficking, synaptogenesis, dendritic growth, dendritic plasticity and nerve-target interactions in co-culture systems. This protocol describes the isolation and dissociation of neurons from the superior cervical ganglia of E21 rat embryos, followed by the preparation and maintenance of pure neuronal cultures in serum-free medium. Since neurons do not adhere to uncoated plastic, neurons will be cultured on either 12 mm glass coverslips or 6-well plates coated with poly-D-lysine. Following treatment with an antimitotic agent (Ara-C, cytosine ß-D-arabinofuranoside), this protocol generates healthy neuronal cultures with less than 5% non-neuronal cells, which can be maintained for over a month in vitro. Although embryonic rat SCG neurons are multipolar with 5-8 dendrites in vivo; under serum-free conditions, these neurons extend only a single axon in culture and continue to be unipolar for the duration of the culture. However, these neurons can be induced to extend dendrites in the presence of basement membrane extract, bone morphogenetic proteins (BMPs), or 10% fetal calf serum. These homogenous neuronal cultures can be used for immunocytochemical staining and for biochemical studies. This paper also describes optimized protocol for immunocytochemical staining for microtubule associated protein-2 (MAP-2) in these neurons and for the preparation of neuronal extracts for mass spectrometry.


Subject(s)
Cell Culture Techniques/methods , Neurons/cytology , Proteomics , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/embryology , Animals , Axons/metabolism , Basement Membrane/metabolism , Neurogenesis , Rats , Superior Cervical Ganglion/metabolism
8.
Nat Commun ; 11(1): 4148, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32811834

ABSTRACT

We evaluate gene editing of HSV in a well-established mouse model, using adeno-associated virus (AAV)-delivered meganucleases, as a potentially curative approach to treat latent HSV infection. Here we show that AAV-delivered meganucleases, but not CRISPR/Cas9, mediate highly efficient gene editing of HSV, eliminating over 90% of latent virus from superior cervical ganglia. Single-cell RNA sequencing demonstrates that both HSV and individual AAV serotypes are non-randomly distributed among neuronal subsets in ganglia, implying that improved delivery to all neuronal subsets may lead to even more complete elimination of HSV. As predicted, delivery of meganucleases using a triple AAV serotype combination results in the greatest decrease in ganglionic HSV loads. The levels of HSV elimination observed in these studies, if translated to humans, would likely significantly reduce HSV reactivation, shedding, and lesions. Further optimization of meganuclease delivery and activity is likely possible, and may offer a pathway to a cure for HSV infection.


Subject(s)
Deoxyribonucleases/genetics , Dependovirus/genetics , Eye Infections/therapy , Gene Editing/methods , Herpes Simplex/therapy , Herpesvirus 1, Human/genetics , Virus Latency/genetics , Animals , CRISPR-Cas Systems/genetics , Cells, Cultured , Chlorocebus aethiops , Eye Infections/genetics , Eye Infections/virology , Female , HEK293 Cells , Herpes Simplex/genetics , Herpesvirus 1, Human/pathogenicity , Humans , Mice , Neurons/metabolism , Neurons/virology , RNA-Seq , Single-Cell Analysis , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/virology , Vero Cells
9.
Auton Neurosci ; 224: 102641, 2020 03.
Article in English | MEDLINE | ID: mdl-32044642

ABSTRACT

Neurons in the superior cervical ganglia (SCG) are classified as rostral and caudal according to their regional locations. Although diverse phenotypes have been reported for these two subpopulations, differences in neuroplasticity, like long-term potentiation (LTP), have not been characterized. Here, we explored possible regional differences of LTP expression in rostral and caudal neurons of the SCG in control rats, Wistar and Wistar Kyoto (WKy), and in the spontaneously hypertensive rats (SHR) as a model of hypertension. We characterized the expression of gLTP evoked by a tetanic train (40 Hz, 3 s) in an in vitro SCG preparation. gLTP was recorded in rostral and caudal neurons at 8-weeks-old (wo) in Wistar rats, 6-wo and 12-wo in SHR and WKy rats. We found that gLTP was differentially expressed; gLTP was larger in caudal neurons in Wistar and adult WKy rats. In adult 12-wo hypertensive SHR, gLTP was expressed in caudal but not in rostral neurons. In contrast, in 6-wo pre-hypertensive SHR, gLTP was expressed in rostral but not in caudal neurons; while in 6-wo WKy, gLTP was expressed in caudal but not in rostral neurons. The lack of gLTP expression in caudal neurons of 6-wo SHR was not due to a GABAergic modulation because several GABA-A receptor antagonists failed to unmask gLTP. Data show that neuroplasticity, particularly gLTP expression, varied according to the ganglionic region. We propose that differential regional expression of gLTP may be correlated with selective innervation on different target organs.


Subject(s)
Ganglia, Sympathetic/drug effects , Long-Term Potentiation/drug effects , Neurons/metabolism , Superior Cervical Ganglion/metabolism , Animals , GABA-A Receptor Antagonists/pharmacology , Ganglia, Sympathetic/metabolism , Male , Neurons/drug effects , Rats, Inbred SHR , Rats, Wistar , Superior Cervical Ganglion/physiopathology
10.
Stem Cells Dev ; 29(4): 198-211, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31701812

ABSTRACT

Traumatic brain injury (TBI) leads to delayed secondary injury events consisting of cellular and molecular cascades that exacerbate the initial injury. Human umbilical cord perivascular cells (HUCPVCs) secrete neurotrophic and prosurvival factors. In this study, we examined the effects of HUCPVC in sympathetic axon and cortical axon survival models and sought to determine whether HUCPVC provide axonal survival cues. We then examined the effects of the HUCPVC in an in vivo fluid percussion injury model of TBI. Our data indicate that HUCPVCs express neurotrophic and neural survival factors. They also express and secrete relevant growth and survival proteins when cultured alone, or in the presence of injured axons. Coculture experiments indicate that HUCPVCs interact preferentially with axons when cocultured with sympathetic neurons and reduce axonal degeneration. Nerve growth factor withdrawal in axonal compartments resulted in 66 ± 3% axon degeneration, whereas HUCPVC coculture rescued axon degeneration to 35 ± 3%. Inhibition of Akt (LY294002) resulted in a significant increase in degeneration compared with HUCPVC cocultures (48 ± 7% degeneration). Under normoxic conditions, control cultures showed 39 ± 5% degeneration. Oxygen glucose deprivation (OGD) resulted in 58 ± 3% degeneration and OGD HUCPVC cocultures reduced degeneration to 34 ± 5% (p < 0.05). In an in vivo model of TBI, immunohistochemical analysis of NF200 showed improved axon morphology in HUCPVC-treated animals compared with injured animals. These data presented in this study indicate an important role for perivascular cells in protecting axons from injury and a potential cell-based therapy to treat secondary injury after TBI.


Subject(s)
Axons/metabolism , Brain Injuries, Traumatic/therapy , Cell- and Tissue-Based Therapy/methods , Neurons/metabolism , Pericytes/transplantation , Animals , Axons/drug effects , Axons/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Chromones/pharmacology , Coculture Techniques , Disease Models, Animal , Embryo, Mammalian , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Glucose/deficiency , Glucose/pharmacology , Humans , Morpholines/pharmacology , Nerve Growth Factor/pharmacology , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/drug effects , Neurons/ultrastructure , Oxygen/pharmacology , Pericytes/drug effects , Pericytes/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism , Umbilical Cord/cytology , Umbilical Cord/metabolism
11.
J Neurophysiol ; 122(6): 2591-2600, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31642403

ABSTRACT

NaV1.8 channels play a crucial role in regulating the action potential in nociceptive neurons. A single nucleotide polymorphism in the human NaV1.8 gene SCN10A, A1073V (rs6795970, G>A), has been linked to the diminution of mechanical pain sensation as well as cardiac conduction abnormalities. Furthermore, studies have suggested that this polymorphism may result in a "loss-of-function" phenotype. In the present study, we performed genomic analysis of A1073V polymorphism presence in a cohort of patients undergoing sigmoid colectomy who provided information regarding perioperative pain and analgesic use. Homozygous carriers reported significantly reduced severity in postoperative abdominal pain compared with heterozygous and wild-type carriers. Homozygotes also trended toward using less analgesic/opiates during the postoperative period. We also heterologously expressed the wild-type and A1073V variant in rat superior cervical ganglion neurons. Electrophysiological testing demonstrated that the mutant NaV1.8 channels activated at more depolarized potentials compared with wild-type channels. Our study revealed that postoperative abdominal pain is diminished in homozygous carriers of A1073V and that this is likely due to reduced transmission of action potentials in nociceptive neurons. Our findings reinforce the importance of NaV1.8 and the A1073V polymorphism to pain perception. This information could be used to develop new predictive tools to optimize patient pain experience and analgesic use in the perioperative setting.NEW & NOTEWORTHY We present evidence that in a cohort of patients undergoing sigmoid colectomy, those homozygous for the NaV1.8 polymorphism (rs6795970) reported significantly lower abdominal pain scores than individuals with the homozygous wild-type or heterozygous genotype. In vitro electrophysiological recordings also suggest that the mutant NaV1.8 channel activates at more depolarizing potentials than the wild-type Na+ channel, characteristic of hypoactivity. This is the first report linking the rs6795970 mutation with postoperative abdominal pain in humans.


Subject(s)
Abdominal Pain/genetics , Colectomy , Electrophysiological Phenomena/physiology , Ganglia, Spinal/physiology , NAV1.8 Voltage-Gated Sodium Channel/physiology , Nociception/physiology , Pain, Postoperative/genetics , Superior Cervical Ganglion/metabolism , Sympathetic Nervous System/physiology , Aged , Animals , Female , Humans , Male , Middle Aged , NAV1.8 Voltage-Gated Sodium Channel/genetics , Neurons/physiology , Polymorphism, Genetic , Rats , Retrospective Studies
12.
J Neurochem ; 150(6): 666-677, 2019 09.
Article in English | MEDLINE | ID: mdl-31306490

ABSTRACT

In the sympatho-adrenal system, angiotensin II (Ang II) acts as a key neuromodulatory component. At sympathetic nerve terminals, Ang II influences sympathetic transmission by enhancing norepinephrine (NE) synthesis, facilitating NE release and inhibiting NE uptake. Previously, it was demonstrated that tyrosine hydroxylase (TH) mRNA is trafficked to the distal axons of primary superior cervical ganglia (SCG) neurons, directed by a cis-acting regulatory element (i.e. zipcode) located in the 3'UTR of the transcript. Results of metabolic labeling studies established that the mRNA is locally translated. It was further shown that the axonal trafficking of the mRNA encoding the enzyme plays an important role in mediating dopamine (DA) and NE synthesis and may facilitate the maintenance of axonal catecholamine levels. In the present study, the hypothesis was tested that Ang II induces NE synthesis in rat primary SCG neurons via the modulation of the trafficking of the mRNAs encoding the catecholamine synthesizing enzymes TH and dopamine ß-hydroxylase (DBH). Treatment of SCG neurons with the Ang II receptor type 1 (AT1R) agonist, L-162,313, increases the axonal levels of TH and DBH mRNA and protein and results in elevated NE levels. Conversely, treatment of rat SCG neurons with the AT1R antagonist, Eprosartan, abolished the L-162,313-mediated increase in axonal levels of TH and DBH mRNA and protein. In a first attempt to identify the proteins involved in the Ang II-mediated axonal transport of TH mRNA, we used a biotinylated 50-nucleotide TH RNA zipcode as bait in the affinity purification of TH zipcode-associated proteins. Mass spectrometric analysis of the TH zipcode ribonucleoprotein (RNP) complex immune-purified from SCG neurons led to the identification of 163 somal and 127 axonal proteins functionally involved in binding nucleic acids, the translational machinery or acting as subunits of cytoskeletal and motor proteins. Surprisingly, immune-purification of the TH axonal trafficking complex, results in the acquisition of DBH mRNA, suggesting that these mRNAs maybe transported to the axon together, possibly in the same RNP complex. Taken together, our results point to a novel mechanism by which Ang II participates in the regulation of axonal synthesis of NE by modulating the local trafficking and expression of TH and DBH, two key enzymes involved in the catecholamine biosynthetic pathway.


Subject(s)
Angiotensin II/metabolism , Axons/metabolism , Dopamine beta-Hydroxylase/metabolism , Norepinephrine/biosynthesis , Tyrosine 3-Monooxygenase/metabolism , Adrenergic Fibers/metabolism , Animals , Axonal Transport/physiology , Cells, Cultured , Neurons/metabolism , Protein Transport/physiology , RNA, Messenger , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/metabolism
13.
Pharmacol Res Perspect ; 7(3): e00471, 2019 06.
Article in English | MEDLINE | ID: mdl-31065376

ABSTRACT

Metabotropic glutamate receptors (mGluRs) are class C G protein coupled receptors with widespread expression in the central nervous system. There are eight mGluRs in the mammalian genome. Research on mGluRs relies on the availability of selective compounds. While many selective allosteric compounds have been described, selectivity of orthosteric agonists and antagonists has been more difficult due to the similarity of the glutamate binding pocket across the mGluR family. LY341495 has been used for decades as a potent and selective group II mGluR antagonist. The selectivity of LY341495 was investigated here between mGluR2, a group II mGluR, and mGluR4, a group III receptor, heterologously expressed in adult rat sympathetic neurons from the superior cervical ganglion (SCG), which provides a null-mGluR background upon which mGluRs were examined in isolation. The compound does in fact selectively inhibit mGluR2 over mGluR4, but in such a way that it makes signaling of the two receptors more difficult to distinguish. The glutamate potency of mGluR2 is about 10-fold higher than mGluR4. 50 nmol L-1 LY341495 did not alter mGluR4 signaling but shifted the mGluR2 glutamate dose-response about 10-fold, such that it overlapped more closely with that of mGluR4. Increasing the LY341494 dose to 500 nmol L-1 further shifted the glutamate dose-response of mGluR2 by another ~10-fold, but also shifted that of mGluR4 similarly. Thus, while glutamate is a moderately selective agonist of mGluR2 over mGluR4 when applied alone, in the presence of increasing concentrations of LY341495, this selectivity of glutamate is lost.


Subject(s)
Amino Acids/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Superior Cervical Ganglion/metabolism , Xanthenes/pharmacology , Animals , Dose-Response Relationship, Drug , Down-Regulation , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/metabolism , Gene Expression Regulation/drug effects , Male , Rats , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/drug effects , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/drug effects
14.
Auton Neurosci ; 219: 33-41, 2019 07.
Article in English | MEDLINE | ID: mdl-31122599

ABSTRACT

Chronic lead exposure frequently brings about increased blood pressure and other cardiovascular diseases associated with autonomic nervous dysfunction. Purinergic signaling is involved in the development of abnormal sympathoexcitatory response due to myocardial ischemic injury. However, the potential implication of P2X7 receptor in altered sympathoexcitatory response caused by chronic lead exposure and the underlying mechanisms remain unclear. In this study, a rat model of chronic lead exposure was used to explore the changes in sympathoexcitatory response and possible involvement of P2X7 receptor in the superior cervical ganglion (SCG). Rats were divided into three groups called control, low lead and high lead groups according to daily lead exposure levels, i.e. 0, 0.5 and 2 g/L respectively. One year later, changes in P2X7 receptor expression in SCG, sympathetic nerve activity and myocardial function were measured for these rats. Our results showed that increased blood pressure and heart rate, decreased heart rate variability, enhanced cervical sympathetic nerve discharge, higher phosphorylation of ERK1/2, and up-regulated protein and mRNA levels of P2X7 expression in SCG occurred after lead exposure. In addition, double-label immunofluorescence staining of P2X7 receptor and glutamine synthetase (GS) revealed activation of the satellite glial cells of SCG by lead exposure. Moreover, knockdown of P2X7 could effectively relief the effect of lead exposure on enhanced expression of P2X7 receptor and GS. Thus our data suggest that the up-regulation of P2X7 receptor activity in satellite glial cells of SCG may contribute to the raised sympathoexcitatory response due to chronic lead exposure.


Subject(s)
Lead/adverse effects , Receptors, Purinergic P2X7/metabolism , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Dose-Response Relationship, Drug , Female , Heart Rate/drug effects , Heart Rate/physiology , Lead/blood , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , RNA, Messenger/metabolism , Random Allocation , Rats, Sprague-Dawley
15.
Physiol Rep ; 7(6): e14023, 2019 03.
Article in English | MEDLINE | ID: mdl-30891952

ABSTRACT

Our previous immunoprecipitation analysis of nicotinic acetylcholine receptors (nAChRs) in the mouse superior cervical ganglion (SCG) revealed that approximately 55%, 24%, and 21% of receptors are comprised of α3ß4, α3ß4α5, and α3ß4ß2 subunits, respectively. Moreover, mice lacking ß4 subunits do not express α5-containing receptors but still express a small number of α3ß2 receptors. Here, we investigated how synaptic transmission is affected in the SCG of α5ß4-KO and α5ß2-KO mice. Using an ex vivo SCG preparation, we stimulated the preganglionic cervical sympathetic trunk and measured compound action potentials (CAPs) in the postganglionic internal carotid nerve. We found that CAP amplitude was unaffected in α5ß4-KO and α5ß2-KO ganglia, whereas the stimulation threshold for eliciting CAPs was significantly higher in α5ß4-KO ganglia. Moreover, intracellular recordings in SCG neurons revealed no difference in EPSP amplitude. We also found that the ganglionic blocking agent hexamethonium was the most potent in α5ß4-KO ganglia (IC50 : 22.1 µmol/L), followed by α5ß2-KO (IC50 : 126.7 µmol/L) and WT ganglia (IC50 : 389.2 µmol/L). Based on these data, we estimated an IC50 of 568.6 µmol/L for a receptor population consisting solely of α3ß4α5 receptors; and we estimated that α3ß4α5 receptors comprise 72% of nAChRs expressed in the mouse SCG. Similarly, by measuring the effects of hexamethonium on ACh-induced currents in cultured SCG neurons, we found that α3ß4α5 receptors comprise 63% of nAChRs. Thus, in contrast to our results obtained using immunoprecipitation, these data indicate that the majority of receptors at the cell surface of SCG neurons consist of α3ß4α5.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Nicotinic/metabolism , Superior Cervical Ganglion/metabolism , Synaptic Transmission , Animals , Cells, Cultured , Ganglionic Blockers/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Superior Cervical Ganglion/drug effects , Synaptic Potentials , Synaptic Transmission/drug effects
16.
Sci Rep ; 9(1): 2353, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30787395

ABSTRACT

We have reported the gateway reflex, which describes specific neural activations that regulate immune cell gateways at specific blood vessels in the central nervous system (CNS). Four types of gateway reflexes exist, all of which induce alterations in endothelial cells at specific vessels of the blood-brain barrier followed by inflammation in the CNS in the presence of CNS-autoreactive T cells. Here we report a new gateway reflex that suppresses the development of retinal inflammation by using an autoreactive T cell-mediated ocular inflammation model. Exposure to photopic light down-regulated the adrenoceptor pathway to attenuate ocular inflammation by suppressing breaching of the blood-retina barrier. Mechanistic analysis showed that exposure to photopic light down-regulates the expression of α1A-adrenoceptor (α1AAR) due to high levels of norepinephrine and epinephrine, subsequently suppressing inflammation. Surgical ablation of the superior cervical ganglion (SCG) did not negate the protective effect of photopic light, suggesting the involvement of retinal noradrenergic neurons rather than sympathetic neurons from the SCG. Blockade of α1AAR signaling under mesopic light recapitulated the protective effect of photopic light. Thus, targeting regional adrenoceptor signaling might represent a novel therapeutic strategy for autoimmune diseases including those that affect organs separated by barriers such as the CNS and eyes.


Subject(s)
Color Vision/physiology , Receptors, Adrenergic, alpha-1/metabolism , Retinitis/physiopathology , Adrenergic Agents/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmunity/genetics , Autoimmunity/physiology , Blood-Brain Barrier/metabolism , Blood-Retinal Barrier/metabolism , Central Nervous System/metabolism , Endothelial Cells/metabolism , Epinephrine/metabolism , Female , Inflammation/metabolism , Light , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurons/metabolism , Norepinephrine/metabolism , Receptors, Adrenergic/metabolism , Superior Cervical Ganglion/metabolism , T-Lymphocytes/immunology
17.
Dev Neurobiol ; 79(11-12): 949-962, 2019 11.
Article in English | MEDLINE | ID: mdl-32077240

ABSTRACT

CD40-activated CD40L reverse signaling is a major physiological regulator of the growth of neural processes in the developing nervous system. Previous work on superior cervical ganglion (SCG) neurons of the paravertebral sympathetic chain has shown that CD40L reverse signaling enhances NGF-promoted axon growth and tissue innervation. Here we show that CD40L reverse signaling has the opposite function in prevertebral ganglion (PVG) sympathetic neurons. During a circumscribed perinatal window of development, PVG neurons cultured from Cd40-/- mice had substantially larger, more exuberant axon arbors in the presence of NGF than PVG neurons cultured from wild-type mice. Tissues that receive their sympathetic innervation from PVG neurons were markedly hyperinnervated in Cd40-/- mice compared with wild-type mice. The exuberant axonal growth phenotype of cultured CD40-deficient perinatal PVG neurons was pared back to wild-type levels by activating CD40L reverse signaling with a CD40-Fc chimeric protein, but not by activating CD40 forward signaling with CD40L. The co-expression of CD40 and CD40L in PVG neurons suggests that these proteins engage in an autocrine signaling loop in these neurons. Our work shows that CD40L reverse signaling is a physiological regulator of NGF-promoted sympathetic axon growth and tissue innervation with opposite effects in paravertebral and prevertebral neurons.


Subject(s)
Axons/metabolism , CD40 Ligand/metabolism , Neurons/metabolism , Superior Cervical Ganglion/metabolism , Animals , Ganglia, Sympathetic/metabolism , Mice, Transgenic , Nerve Growth Factor/metabolism
18.
Eur J Histochem ; 62(4)2018 Nov 22.
Article in English | MEDLINE | ID: mdl-30465595

ABSTRACT

The neuropeptide calcitonin gene-related peptide (CGRP) mediates inflammation and head pain by influencing the functional vascular blood supply. CGRP is a well-characterized mediator of receptor-regulated neurotransmitter release. However, knowledge regarding the role of CGRP during the development of the superior cervical ganglion (SCG) is limited. In the present study, we observed the localization of CGRP and vascular endothelial growth factor (VEGF-A) mRNAs during prenatal development at embryonic day 14.5 (E14.5), E17.5 and postnatal day 1 (P1) using in situ hybridization. The antisense probe for CGRP was detected by in situ hybridization at E14.5, E17.5, and P1, and the highest levels were detected at E17.5. In contrast, the antisense probe for VEGF-A was detected by in situ hybridization in gradually increasing intensity from E14.5 to P1. The differences in the expression of these two markers revealed specific characteristics related to CGRP concentration and release compared to those of VEGF-A during development. The correlation between CGRP and VEGF-A may influence functional stress and the vascular blood supply during prenatal and postnatal development.


Subject(s)
Superior Cervical Ganglion/metabolism , Vascular Endothelial Growth Factors/metabolism , Animals , Cell Count , Mice , RNA, Messenger/metabolism , Staining and Labeling , Superior Cervical Ganglion/embryology , Superior Cervical Ganglion/growth & development , Vascular Endothelial Growth Factors/genetics
19.
Development ; 145(22)2018 11 19.
Article in English | MEDLINE | ID: mdl-30337376

ABSTRACT

TWE-PRIL is a naturally occurring fusion protein of components of two TNF superfamily members: the extracellular domain of APRIL; and the intracellular and transmembrane domains of TWEAK with no known function. Here, we show that April-/- mice (which lack APRIL and TWE-PRIL) exhibited overgrowth of sympathetic fibres in vivo, and sympathetic neurons cultured from these mice had significantly longer axons than neurons cultured from wild-type littermates. Enhanced axon growth from sympathetic neurons cultured from April-/- mice was prevented by expressing full-length TWE-PRIL in these neurons but not by treating them with soluble APRIL. Soluble APRIL, however, enhanced axon growth from the sympathetic neurons of wild-type mice. siRNA knockdown of TWE-PRIL but not siRNA knockdown of APRIL alone also enhanced axon growth from wild-type sympathetic neurons. Our work reveals the first and physiologically relevant role for TWE-PRIL and suggests that it mediates reverse signalling.


Subject(s)
Axons/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , B-Cell Maturation Antigen/genetics , B-Cell Maturation Antigen/metabolism , Cells, Cultured , Cytokine TWEAK/genetics , Cytokine TWEAK/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Mice , Models, Biological , Nerve Growth Factor/pharmacology , Phenotype , RNA, Small Interfering/metabolism , Solubility , Superior Cervical Ganglion/metabolism , Sympathetic Nervous System/growth & development , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
20.
Neurochem Int ; 120: 206-212, 2018 11.
Article in English | MEDLINE | ID: mdl-30196147

ABSTRACT

Diabetic cardiac autonomic neuropathy (DCAN) is a common and serious complication of diabetes mellitus (DM), is manifested by nerve fiber injury in the sympathetic and parasympathetic nerve of the autonomic nervous system, and causes hypertension, cardiac arrhythmias, silent myocardial infarction, and sudden death. Our previous study observed that P2X3 receptor in superior cervical ganglia in rat was associated with sympathetic neuropathy caused by myocardial ischemia. However, whether the P2X3 receptor is involved in the diabetic cardiac autonomic neuropathy and the underlying mechanisms remain unclear. The aim of this research was explored the effect of P2X3 short hairpin RNA (shRNA) on information transmission of sympathetic nerve induced by DCAN. Sprague-Dawley (SD) male rats were randomly divided into four groups: Control, DM, DM treated with P2X3 shRNA and DM treated with scramble shRNA. Blood pressure, heart rate and heart rate variability were measured in each group. The expression of P2X3 in stellate ganglion (SG) was detected by immunohistochemistry, western blotting and QPCR. Results showed that P2X3 shRNA alleviated blood pressure and heart rate, improved heart rate variability, decreased the up-regulated expression levels of P2X3, interleukin-1beta and tumor necrosis factor alpha in stellate ganglion (SG) of diabetic rats. P2X3 shRNA also reduced the incremental concentration of serum epinephrine and the phosphorylation level of extracellular regulated protein kinases1/2 in diabetic rats. These results indicated that P2X3 shRNA could decrease sympathetic activity via inhibiting P2X3 receptor in the SG to alleviate DCAN.


Subject(s)
Diabetic Neuropathies/genetics , Gene Knockdown Techniques , Receptors, Purinergic P2X3/genetics , Stellate Ganglion/metabolism , Animals , Blood Pressure/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/metabolism , Gene Knockdown Techniques/methods , Interleukin-1beta/metabolism , Male , Myocardial Ischemia/metabolism , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , Receptors, Purinergic P2X3/metabolism , Superior Cervical Ganglion/metabolism , Sympathetic Nervous System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...