Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.829
Filtrer
1.
J Am Heart Assoc ; 13(13): e034965, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38934856

RÉSUMÉ

BACKGROUND: Rostral ventrolateral medulla (RVLM) neuron hyperactivity raises sympathetic outflow, causing hypertension. MicroRNAs (miRNAs) contribute to diverse biological processes, but their influence on RVLM neuronal excitability and blood pressure (BP) remains widely unexplored. METHODS AND RESULTS: The RVLM miRNA profiles in spontaneously hypertensive rats were unveiled using RNA sequencing. Potential effects of these miRNAs in reducing neuronal excitability and BP and underlying mechanisms were investigated through various experiments. Six hundred thirty-seven miRNAs were identified, and reduced levels of miR-193b-3p and miR-346 were observed in the RVLM of spontaneously hypertensive rats. Increased miR-193b-3p and miR-346 expression in RVLM lowered neuronal excitability, sympathetic outflow, and BP in spontaneously hypertensive rats. In contrast, suppressing miR-193b-3p and miR-346 expression in RVLM increased neuronal excitability, sympathetic outflow, and BP in Wistar Kyoto and Sprague-Dawley rats. Cdc42 guanine nucleotide exchange factor (Arhgef9) was recognized as a target of miR-193b-3p. Overexpressing miR-193b-3p caused an evident decrease in Arhgef9 expression, resulting in the inhibition of neuronal apoptosis. By contrast, its downregulation produced the opposite effects. Importantly, the decrease in neuronal excitability, sympathetic outflow, and BP observed in spontaneously hypertensive rats due to miR-193b-3p overexpression was greatly counteracted by Arhgef9 upregulation. CONCLUSIONS: miR-193b-3p and miR-346 are newly identified factors in RVLM that hinder hypertension progression, and the miR-193b-3p/Arhgef9/apoptosis pathway presents a potential mechanism, highlighting the potential of targeting miRNAs for hypertension prevention.


Sujet(s)
Pression sanguine , Hypertension artérielle , Moelle allongée , microARN , Rats de lignée SHR , Rats de lignée WKY , Rat Sprague-Dawley , Animaux , microARN/génétique , microARN/métabolisme , Moelle allongée/métabolisme , Moelle allongée/physiopathologie , Moelle allongée/effets des médicaments et des substances chimiques , Hypertension artérielle/physiopathologie , Hypertension artérielle/génétique , Hypertension artérielle/métabolisme , Pression sanguine/effets des médicaments et des substances chimiques , Pression sanguine/génétique , Mâle , Modèles animaux de maladie humaine , Rats , Rho guanine nucleotide exchange factors/génétique , Rho guanine nucleotide exchange factors/métabolisme , Neurones/métabolisme , Système nerveux sympathique/physiopathologie , Système nerveux sympathique/métabolisme , Apoptose
2.
Sci Adv ; 10(25): eadj0720, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38896627

RÉSUMÉ

Mutations in the transcription factors encoded by PHOX2B or LBX1 correlate with congenital central hypoventilation disorders. These conditions are typically characterized by pronounced hypoventilation, central apnea, and diminished chemoreflexes, particularly to abnormally high levels of arterial PCO2. The dysfunctional neurons causing these respiratory disorders are largely unknown. Here, we show that distinct, and previously undescribed, sets of medullary neurons coexpressing both transcription factors (dB2 neurons) account for specific respiratory functions and phenotypes seen in congenital hypoventilation. By combining intersectional chemogenetics, intersectional labeling, lineage tracing, and conditional mutagenesis, we uncovered subgroups of dB2 neurons with key functions in (i) respiratory tidal volumes, (ii) the hypercarbic reflex, (iii) neonatal respiratory stability, and (iv) neonatal survival. These data provide functional evidence for the critical role of distinct medullary dB2 neurons in neonatal respiratory physiology. In summary, our work identifies distinct subgroups of dB2 neurons regulating breathing homeostasis, dysfunction of which causes respiratory phenotypes associated with congenital hypoventilation.


Sujet(s)
Protéines à homéodomaine , Hypoventilation , Moelle allongée , Neurones , Facteurs de transcription , Hypoventilation/congénital , Hypoventilation/génétique , Animaux , Neurones/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Souris , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Moelle allongée/métabolisme , Apnée centrale du sommeil/génétique , Phénotype , Humains
3.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(5): 960-966, 2024 May 20.
Article de Chinois | MEDLINE | ID: mdl-38862454

RÉSUMÉ

OBJECTIVE: To assess the effects of repeated mild traumatic brain injury (rmTBI) in the parietal cortex on neuronal morphology and synaptic plasticity in the medulla oblongata of mice. METHODS: Thirty-two male ICR mice were randomly divided into sham operation group (n=8) and rmTBI group (n=24). The mice in the latter group were subjected to repeated mild impact injury of the parietal cortex by a free-falling object. The mice surviving the injuries were evaluated for neurological deficits using neurological severity scores (NSS), righting reflex test and forced swimming test, and pathological changes of the neuronal cells in the medulla oblongata were observed with HE and Nissl staining. Western blotting and immunofluorescence staining were used to detect the expressions of neuroligin 1(NLG-1) and postsynaptic density protein 95(PSD-95) in the medulla oblongata of the mice that either survived rmTBI or not. RESULTS: None of the mice in the sham-operated group died, while the mortality rate was 41.67% in rmTBI group. The mice surviving rmTBI showed significantly reduced NSS, delayed recovery of righting reflex, increased immobility time in forced swimming test (P < 0.05), and loss of Nissl bodies; swelling and necrosis were observed in a large number of neurons in the medulla oblongata, where the expression levels of NLG-1 and PSD-95 were significantly downregulated (P < 0.05). The mice that did not survive rmTBI showed distorted and swelling nerve fibers and decreased density of neurons in the medulla oblongina with lowered expression levels of NLG-1 and PSD-95 compared with the mice surviving the injuries (P < 0.01). CONCLUSION: The structural and functional anomalies of the synapses in the medulla oblongata may contribute to death and neurological impairment following rmTBI in mice.


Sujet(s)
Molécules d'adhérence cellulaire neuronale , Homologue-4 de la protéine Disks Large , Moelle allongée , Souris de lignée ICR , Lobe pariétal , Animaux , Souris , Moelle allongée/métabolisme , Homologue-4 de la protéine Disks Large/métabolisme , Mâle , Lobe pariétal/métabolisme , Molécules d'adhérence cellulaire neuronale/métabolisme , Neurones/métabolisme , Lésions traumatiques de l'encéphale/métabolisme , Plasticité neuronale
4.
Am J Physiol Regul Integr Comp Physiol ; 327(1): R66-R78, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38708545

RÉSUMÉ

The stress-induced cardiovascular response is based on the defensive reaction in mammals. It has been shown that the sympathetic vasomotor pathway of acute psychological stress is indirectly mediated via neurons in the rostroventral medulla (RVM) from the hypothalamic stress center. In this study, direct projections to the RVM and distribution of neuroexcitatory marker c-Fos-expressed neurons were investigated during social defeat stress (SDS) in conscious rats. The experimental rat that was injected with a neural tracer, FluoroGold (FG) into the unilateral RVM, was exposed to the SDS. Double-positive neurons of both c-Fos and FG were locally distributed in the lateral/ventrolateral periaqueductal gray matter (l/vl PAG) in the midbrain. These results suggest that the neurons in the l/vl PAG contribute to the defensive reaction evoked by acute psychological stress, such as the SDS. During the SDS period, arterial pressure (AP) and heart rate (HR) showed sustained increases in the rat. Therefore, we performed chemical stimulation by excitatory amino acid microinjection within the l/vl PAG and measured cardiovascular response and sympathetic nerve activity in some anesthetized rats. The chemical stimulation of neurons in the l/vl PAG caused significant increases in arterial pressure and renal sympathetic nerve activity. Taken together, our results suggest that neurons in the l/vl PAG are a possible candidate for the cardiovascular descending pathway that modulates sympathetic vascular resistance evoked by acute psychological stress, like the SDS.NEW & NOTEWORTHY The sympathetic vasomotor pathway of an acute psychological stress-induced cardiovascular response is mediated via neurons in the RVM indirectly from the hypothalamus. In this study, we showed the relaying area of the efferent sympathetic vasomotor pathway from the hypothalamus to the RVM. The results suggested that the pressor response during psychological stress is mediated via neurons in the lateral/ventrolateral PAG to the RVM.


Sujet(s)
Moelle allongée , Substance grise centrale du mésencéphale , Défaite sociale , Stress psychologique , Système vasomoteur , Animaux , Stress psychologique/physiopathologie , Mâle , Substance grise centrale du mésencéphale/métabolisme , Substance grise centrale du mésencéphale/physiopathologie , Moelle allongée/physiopathologie , Moelle allongée/métabolisme , Système vasomoteur/physiopathologie , Rats , Rythme cardiaque , Protéines proto-oncogènes c-fos/métabolisme , Rat Wistar , Système nerveux sympathique/physiopathologie , Rat Sprague-Dawley , Pression artérielle , Comportement animal
5.
J Neural Transm (Vienna) ; 131(7): 739-753, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38630191

RÉSUMÉ

Diagnosing and treating chronic orofacial pain is challenging due to its complex structure and limited understanding of its causes and mechanisms. In this study, we used RNA sequencing to identify differentially expressed genes (DEGs) in the rostral ventral medulla (RVM) and thalamus of rats with persistent orofacial pain, aiming to explore its development. DEGs were functionally analyzed using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Results showed a significant association between immune response and pain in this model. Key DEG mRNA expression trends were further validated using real-time quantitative polymerase chain reaction (RT-PCR), confirming their crucial roles in chronic orofacial pain. After injecting complete Freund's adjuvant (CFA) into the bilateral temporomandibular joint cavity for 14 days, we observed 293 upregulated genes and 14 downregulated genes in the RVM, and 1086 upregulated genes and 37 downregulated genes in the thalamus. Furthermore, we identified 27 common DEGs with altered expression (upregulation) in both the thalamus and RVM, including Cd74, C3, Cxcl13, C1qb, Itgal, Fcgr2b, C5ar1, and Tlr2, which are pain-associated genes. Protein-protein interaction (PPI) analysis using Cytoscape revealed the involvement of Toll-like receptors, complement system, differentiation clusters, and antigen presentation-related proteins in the interaction between the thalamus and RVM. The results of this study show that the immune system seems to have a more significant influence on chronic orofacial pain. There may be direct or indirect influence between the thalamus and RVM, which may participate in the regulation of chronic orofacial pain.


Sujet(s)
Douleur chronique , Algie faciale , Moelle allongée , Rat Sprague-Dawley , Thalamus , Animaux , Algie faciale/génétique , Algie faciale/métabolisme , Algie faciale/physiopathologie , Moelle allongée/métabolisme , Mâle , Rats , Douleur chronique/génétique , Douleur chronique/métabolisme , Thalamus/métabolisme , Analyse de séquence d'ARN , Modèles animaux de maladie humaine , Transcriptome
6.
Physiol Behav ; 280: 114564, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38657747

RÉSUMÉ

Although salivation is essential during eating behavior, little is known about the brainstem centers that directly control the salivary glands. With regard to the inferior salivatory nucleus (ISN), the site of origin of the parasympathetic preganglionic cell bodies that innervate the parotid glands, previous anatomical studies have located it within the rostrodorsal medullary reticular formation. However, to date there is no functional data that shows the secretory nature of the somas grouped in this region. To activate only the somas and rule out the activation of the efferent fibers from and the afferent fibers to the ISN, in exp. 1, NMDA neurotoxin was administered to the rostrodorsal medullary region and the secretion of saliva was recorded during the following hour. Results showed an increased secretion of parotid saliva but a total absence of submandibular-sublingual secretion. In exp. 2, results showed that the hypersecretion of parotid saliva after NMDA microinjection was completely blocked by the administration of atropine (a cholinergic blocker) but not after administration of dihydroergotamine plus propranolol (α and ß-adrenergic blockers, respectively). These findings suggest that the somata of the rostrodorsal medulla are secretory in nature, controlling parotid secretion via a cholinergic pathway. The data thus functionally supports the idea that these cells constitute the ISN.


Sujet(s)
N-Méthyl-aspartate , Glande parotide , Récepteurs du N-méthyl-D-aspartate , Salivation , Animaux , Mâle , Rats , Antagonistes bêta-adrénergiques/pharmacologie , Atropine/pharmacologie , Agonistes des acides aminés excitateurs/pharmacologie , Moelle allongée/métabolisme , Moelle allongée/effets des médicaments et des substances chimiques , Microinjections , N-Méthyl-aspartate/pharmacologie , N-Méthyl-aspartate/métabolisme , Glande parotide/métabolisme , Glande parotide/effets des médicaments et des substances chimiques , Propranolol/pharmacologie , Rat Wistar , Récepteurs du N-méthyl-D-aspartate/métabolisme , Salive/métabolisme , Salivation/effets des médicaments et des substances chimiques , Salivation/physiologie , Ptyalisme
7.
Auton Neurosci ; 253: 103177, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636284

RÉSUMÉ

BACKGROUND: Many esophageal striated muscles of mammals are dually innervated by the vagal and enteric nerves. Recently, substance P (SP)-sensory nerve terminals with calcitonin gene-related peptide (CGRP) were found on a few striated muscle fibers in the rat esophagus, implying that these muscle fibers are triply innervated. In this study, we examined the localization and origin of CGRP-nerve endings in striated muscles to consider their possible roles in the esophagus regarding triple innervation. METHODS: Wholemounts of the rat esophagus were immunolabeled to detect CGRP-nerve endings in striated muscles. Also, retrograde tracing was performed by injecting Fast Blue (FB) into the esophagus, and cryostat sections of the medulla oblongata, nodose ganglion (NG), and the tenth thoracic (T10) dorsal root ganglion (DRG) were immunostained to identify the origin of the CGRP-nerve endings. RESULTS: CGRP-fine, varicose nerve endings were localized in motor endplates on a few esophageal striated muscle fibers (4 %), most of which received nitric oxide (NO) synthase nerve terminals, and most of the CGRP nerve endings were SP- and transient receptor potential vanilloid member 1 (TRPV1)-positive. Retrograde tracing showed many FB-labeled CGRP-neurons positive for SP and TRPV1 in the NG and T10 DGR. CONCLUSIONS: This study suggests that the CGRP-varicose nerve endings containing SP and TRPV1 in motor endplates are sensory, and a few esophageal striated muscle fibers are triply innervated. The nerve endings may detect acetylcholine-derived acetic acid from the vagal motor nerve endings and NO from esophageal intrinsic nerve terminals in the motor endplates to regulate esophageal motility.


Sujet(s)
Peptide relié au gène de la calcitonine , Oesophage , Ganglion inférieur du nerf vague , Cellules réceptrices sensorielles , Animaux , Peptide relié au gène de la calcitonine/métabolisme , Peptide relié au gène de la calcitonine/analyse , Oesophage/innervation , Oesophage/métabolisme , Mâle , Cellules réceptrices sensorielles/métabolisme , Ganglion inférieur du nerf vague/métabolisme , Plaque terminale motrice/métabolisme , Rats , Ganglions sensitifs des nerfs spinaux/métabolisme , Moelle allongée/métabolisme , Substance P/métabolisme , Muscle strié/innervation , Muscle strié/métabolisme , Nerf vague/métabolisme , Rat Wistar , Rat Sprague-Dawley , Fibres musculaires squelettiques/métabolisme , Canaux cationiques TRPV/métabolisme , Amidines
8.
Sci Adv ; 10(17): eadj9581, 2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38669335

RÉSUMÉ

The supraspinal descending pain modulatory system (DPMS) shapes pain perception via monoaminergic modulation of sensory information in the spinal cord. However, the role and synaptic mechanisms of descending noradrenergic signaling remain unclear. Here, we establish that noradrenergic neurons of the locus coeruleus (LC) are essential for supraspinal opioid antinociception. While much previous work has emphasized the role of descending serotonergic pathways, we find that opioid antinociception is primarily driven by excitatory output from the ventrolateral periaqueductal gray (vlPAG) to the LC. Furthermore, we identify a previously unknown opioid-sensitive inhibitory input from the rostroventromedial medulla (RVM), the suppression of which disinhibits LC neurons to drive spinal noradrenergic antinociception. We describe pain-related activity throughout this circuit and report the presence of prominent bifurcating outputs from the vlPAG to the LC and the RVM. Our findings substantially revise current models of the DPMS and establish a supraspinal antinociceptive pathway that may contribute to multiple forms of descending pain modulation.


Sujet(s)
Analgésiques morphiniques , Locus ceruleus , Moelle allongée , Douleur , Substance grise centrale du mésencéphale , Locus ceruleus/métabolisme , Locus ceruleus/effets des médicaments et des substances chimiques , Substance grise centrale du mésencéphale/métabolisme , Substance grise centrale du mésencéphale/effets des médicaments et des substances chimiques , Animaux , Moelle allongée/métabolisme , Moelle allongée/effets des médicaments et des substances chimiques , Douleur/traitement médicamenteux , Douleur/métabolisme , Analgésiques morphiniques/pharmacologie , Mâle , Neurones adrénergiques/métabolisme , Neurones adrénergiques/effets des médicaments et des substances chimiques , Souris , Voies nerveuses/effets des médicaments et des substances chimiques
9.
J Neuroinflammation ; 21(1): 101, 2024 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-38632579

RÉSUMÉ

BACKGROUND: Increased neuroinflammation in brain regions regulating sympathetic nerves is associated with hypertension. Emerging evidence from both human and animal studies suggests a link between hypertension and gut microbiota, as well as microbiota-derived metabolites short-chain fatty acids (SCFAs). However, the precise mechanisms underlying this gut-brain axis remain unclear. METHODS: The levels of microbiota-derived SCFAs in spontaneously hypertensive rats (SHRs) were determined by gas chromatography-mass spectrometry. To observe the effect of acetate on arterial blood pressure (ABP) in rats, sodium acetate was supplemented via drinking water for continuous 7 days. ABP was recorded by radio telemetry. The inflammatory factors, morphology of microglia and astrocytes in rostral ventrolateral medulla (RVLM) were detected. In addition, blood-brain barrier (BBB) permeability, composition and metabolomics of the gut microbiome, and intestinal pathological manifestations were also measured. RESULTS: The serum acetate levels in SHRs are lower than in normotensive control rats. Supplementation with acetate reduces ABP, inhibits sympathetic nerve activity in SHRs. Furthermore, acetate suppresses RVLM neuroinflammation in SHRs, increases microglia and astrocyte morphologic complexity, decreases BBB permeability, modulates intestinal flora, increases fecal flora metabolites, and inhibits intestinal fibrosis. CONCLUSIONS: Microbiota-derived acetate exerts antihypertensive effects by modulating microglia and astrocytes and inhibiting neuroinflammation and sympathetic output.


Sujet(s)
Hypertension artérielle , Microbiote , Humains , Rats , Animaux , Rats de lignée SHR , Maladies neuro-inflammatoires , Hypertension artérielle/métabolisme , Pression sanguine , Moelle allongée/métabolisme , Acétates/pharmacologie
10.
J Neuropathol Exp Neurol ; 83(3): 144-160, 2024 02 21.
Article de Anglais | MEDLINE | ID: mdl-38323418

RÉSUMÉ

The failure of chemoreflexes, arousal, and/or autoresuscitation to asphyxia may underlie some sudden infant death syndrome (SIDS) cases. In Part I, we showed that some SIDS infants had altered 5-hydroxytryptamine (5-HT)2A/C receptor binding in medullary nuclei supporting chemoreflexes, arousal, and autoresuscitation. Here, using the same dataset, we tested the hypotheses that the prevalence of low 5-HT1A and/or 5-HT2A/C receptor binding (defined as levels below the 95% confidence interval of controls-a new approach), and the percentages of nuclei affected are greater in SIDS versus controls, and that the distribution of low binding varied with age of death. The prevalence and percentage of nuclei with low 5-HT1A and 5-HT2A/C binding in SIDS were twice that of controls. The percentage of nuclei with low 5-HT2A/C binding was greater in older SIDS infants. In >80% of older SIDS infants, low 5-HT2A/C binding characterized the hypoglossal nucleus, vagal dorsal nucleus, nucleus of solitary tract, and nuclei of the olivocerebellar subnetwork (important for blood pressure regulation). Together, our findings from SIDS infants and from animal models of serotonergic dysfunction suggest that some SIDS cases represent a serotonopathy. We present new hypotheses, yet to be tested, about how defects within serotonergic subnetworks may lead to SIDS.


Sujet(s)
Mort subite du nourrisson , Nourrisson , Animaux , Humains , Sujet âgé , Moelle allongée/métabolisme , Sérotonine/métabolisme , Récepteurs sérotoninergiques/métabolisme
11.
Sci Rep ; 14(1): 4069, 2024 02 19.
Article de Anglais | MEDLINE | ID: mdl-38374419

RÉSUMÉ

We investigated the participation of the nucleus of the tractus solitarius (NTS) in tonic‒clonic seizures and postictal antinociception control mediated by NMDA receptors, the role of NTS GABAergic interneurons and noradrenergic pathways from the locus coeruleus (LC) in these phenomena. The NTS-lateral nucleus reticularis paragigantocellularis (lPGi)-LC pathway was studied by evaluating neural tract tracer deposits in the lPGi. NMDA and GABAergic receptors agonists and antagonists were microinjected into the NTS, followed by pharmacologically induced seizures. The effects of LC neurotoxic lesions caused by DSP-4, followed by NTS-NMDA receptor activation, on both tonic‒clonic seizures and postictal antinociception were also investigated. The NTS is connected to lPGi neurons that send outputs to the LC. Glutamatergic vesicles were found on dendrites and perikarya of GABAergic interneurons in the NTS. Both tonic‒clonic seizures and postictal antinociception are partially dependent on glutamatergic-mediated neurotransmission in the NTS of seizing rats in addition to the integrity of the noradrenergic system since NMDA receptor blockade in the NTS and intrathecal administration of DSP-4 decrease the postictal antinociception. The GABAA receptor activation in the NTS decreases both seizure severity and postictal antinociception. These findings suggest that glutamatergic inputs to NTS-GABAergic interneurons, in addition to ascending and descending noradrenergic pathways from the LC, are critical for the control of both seizures and postictal antinociception.


Sujet(s)
Benzylamines , Locus ceruleus , Récepteurs du N-méthyl-D-aspartate , Rats , Animaux , Locus ceruleus/physiologie , Récepteurs du N-méthyl-D-aspartate/métabolisme , Moelle allongée/métabolisme , Noyau du tractus solitaire/métabolisme , Norépinéphrine/métabolisme , Crises épileptiques/métabolisme
12.
Prog Neurobiol ; 232: 102561, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38142769

RÉSUMÉ

Chronic craniofacial pain is intractable and its mechanisms remain unclarified. The rostral ventromedial medulla (RVM) plays a crucial role in descending pain facilitation and inhibition. It is unclear how the descending circuits from the RVM to spinal trigeminal nucleus (Sp5) are organized to bidirectionally modulate craniofacial nociception. We used viral tracing, in vivo optogenetics, calcium signaling recording, and chemogenetic manipulations to investigate the structure and function of RVM-Sp5 circuits. We found that most RVM neurons projecting to Sp5 were GABAergic or glutamatergic and facilitated or inhibited craniofacial nociception, respectively. Both GABAergic interneurons and glutamatergic projection neurons in Sp5 received RVM inputs: the former were antinociceptive, whereas the latter were pronociceptive. Furthermore, we demonstrated activation of both GABAergic and glutamatergic Sp5 neurons receiving RVM inputs in inflammation- or dysfunction-induced masseter hyperalgesia. Activating GABAergic Sp5 neurons or inhibiting glutamatergic Sp5 neurons that receive RVM projections reversed masseter hyperalgesia. Our study identifies specific cell types and projections of RVM-Sp5 circuits involved in facilitating or inhibiting craniofacial nociception respectively. Selective manipulation of RVM-Sp5 circuits can be used as potential treatment strategy to relieve chronic craniofacial muscle pain.


Sujet(s)
Hyperalgésie , Noyau spinal du nerf trijumeau , Humains , Hyperalgésie/métabolisme , Noyau spinal du nerf trijumeau/métabolisme , Douleur , Moelle allongée/métabolisme , Neurones GABAergiques/métabolisme
13.
Article de Anglais | MEDLINE | ID: mdl-38147959

RÉSUMÉ

Zymosan is a fungi-derived pathogen-associated molecular pattern. It activates the immune system and induces the reduction of feed passage rate in the gastrointestinal tract of vertebrates including birds. However, the mechanism mediating the zymosan-induced inhibition of feed passage in the gastrointestinal tract remains unknown. Since the medulla oblongata regulates the digestive function, it is plausible that the medulla oblongata is involved in the zymosan-induced inhibition of feed passage. The present study was performed to identify the genes that were affected by zymosan within the medulla oblongata of chicks (Gallus gallus) using an RNA sequencing approach. We found that mRNAs of several bioactive molecules including neuropeptide Y (NPY) were increased with an intraperitoneal (IP) injection of zymosan. The increase of mRNA expression of NPY in the medulla oblongata was also observed after the IP injection of lipopolysaccharide, derived from gram-negative bacteria. These results suggest that medullary NPY is associated with physiological changes during fungal and bacterial infection. Furthermore, we found that intracerebroventricular injection of NPY and its receptor agonists reduced the feed passage from the crop. Additionally, the injection of NPY reduced the feed passage from the proventriculus to lower digestive tract. NPY also suppressed the activity of duodenal activities of amylase and trypsin. The present study suggests that fungi- and bacteria-induced activation of the immune system may activate the NPY neurons in the medulla oblongata and thereby reduce the digestive function in chicks.


Sujet(s)
Lipopolysaccharides , Neuropeptide Y , Animaux , Neuropeptide Y/génétique , Neuropeptide Y/métabolisme , Lipopolysaccharides/pharmacologie , Zymosan/pharmacologie , Poulets/métabolisme , Moelle allongée/métabolisme , Tube digestif/métabolisme
14.
Neurosci Lett ; 818: 137568, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38008350

RÉSUMÉ

Alamandine, a peptide known to interact with Mas-related G protein-coupled receptor subtype D (MrgD), has been implicated in moderating inflammatory signals. MrgD receptors are abundantly found in pain transmission pathways, but the role of alamandine/MrgD in pain modulation has not been thoroughly explored. This study aimed to investigate the effects of alamandine (10, 40, and 100 pmol) in a rat model of allodynia induced by sciatic nerve ligation, with a specific focus on examining the involvement of MrgD receptors, NMDAR1, and serotonin transporter (SERT) in the ventrolateral periaqueductal gray (vlPAG) and rostral ventromedial medulla (RVM). Microinjection of alamandine into the vlPAG at a dose of 100 pmol and into the RVM at doses of 40 and 100 pmol resulted in a significant increase in paw withdrawal threshold (PWT). Additionally, co-administration of D-Pro7-Ang-(1-7) at 50 pmol, an MrgD receptor antagonist, effectively blocked the analgesic effects of alamandine. Immunofluorescence analysis confirmed the presence of MrgD receptors in both the vlPAG and RVM regions. Importantly, an upregulation of MrgD receptor expression was observed following allodynia induction, suggesting a potential compensatory mechanism in response to pain. Our findings support the co-localization of MrgD receptors with NMDAR1 in vlPAG neurons, suggesting their ability to initiate analgesic pathways similar to those activated by NMDA receptors in the vlPAG. Furthermore, our results underscore a significant co-localization of MrgD receptors with the SERT in the RVM, underscoring their potential impact on serotonergic neurons involved in promoting analgesic effects.


Sujet(s)
Hyperalgésie , Substance grise centrale du mésencéphale , Rats , Animaux , Hyperalgésie/traitement médicamenteux , Hyperalgésie/métabolisme , Substance grise centrale du mésencéphale/métabolisme , Douleur/traitement médicamenteux , Analgésiques/pharmacologie , Nerf ischiatique/métabolisme , Moelle allongée/métabolisme
15.
Chin J Physiol ; 66(5): 326-334, 2023.
Article de Anglais | MEDLINE | ID: mdl-37929343

RÉSUMÉ

Post-traumatic stress disorder (PTSD) is a serious psychiatric disorder, and there is an association between it and the development of cardiovascular disease. The aim of this study was to explore whether there is a glutamatergic pathway connecting the medial habenula (MHb) with the rostral ventrolateral medulla (RVLM) that is involved in the regulation of cardiovascular function in a rat model of PTSD. Vesicular glutamate transporter 2 (VGLUT2)-positive neurons in the MHb region were retrogradely labeled with FluoroGold (FG) by the double-labeling technique of VGLUT2 immunofluorescence and FG retrograde tracing. Rats belonging to the PTSD model group were microinjected with artificial cerebrospinal fluid (ACSF) or kynurenic acid (KYN; a nonselective glutamate receptor blocker) into their RVLM. Subsequently, with electrical stimulation of MHb, the discharge frequency of the RVLM neurons, heart rate, and blood pressure were found to be significantly increased after microinjection of ACSF using an in vivo multichannel synchronous recording technology; however, this effect was inhibited by injection of KYN. The expression of N-methyl-D-aspartic acid (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits was significantly increased in RVLM of PTSD model rats analyzed by the Western blotting technique. These findings suggest that there may be a glutamatergic pathway connection between MHb and RVLM and that this pathway may be involved in the regulation of cardiovascular function in the PTSD model rats, by acting on NMDA and AMPA receptors in the RVLM.


Sujet(s)
Habénula , Troubles de stress post-traumatique , Humains , Rats , Animaux , Troubles de stress post-traumatique/métabolisme , N-Méthyl-aspartate/métabolisme , N-Méthyl-aspartate/pharmacologie , Habénula/métabolisme , Moelle allongée/métabolisme , Pression sanguine , Acide glutamique/métabolisme , Acide glutamique/pharmacologie
16.
Am J Physiol Regul Integr Comp Physiol ; 325(3): R229-R237, 2023 09 01.
Article de Anglais | MEDLINE | ID: mdl-37424401

RÉSUMÉ

To investigate the role of glial cells in the regulation of glucoprivic responses in rats, a chemogenetic approach was used to activate astrocytes neighboring catecholamine (CA) neurons in the ventromedial medulla (VLM) where A1 and C1 CA cell groups overlap (A1/C1). Previous results indicate that activation of CA neurons in this region is necessary and sufficient for feeding and corticosterone release in response to glucoprivation. However, it is not known whether astrocyte neighbors of CA neurons contribute to glucoregulatory responses. Hence, we made nanoinjections of AAV5-GFAP-hM3D(Gq)-mCherry to selectively transfect astrocytes in the A1/C1 region with the excitatory designer receptor exclusively activated by designer drugs (DREADDs), hM3D(Gq). After allowing time for DREADD expression, we evaluated the rats for increased food intake and corticosterone release in response to low systemic doses of the antiglycolytic agent, 2-deoxy-d-glucose (2DG), alone and in combination with the hM3D(Gq) activator clozapine-n-oxide (CNO). We found that DREADD-transfected rats ate significantly more food when 2DG and CNO were coadministered than when either 2DG or CNO was injected alone. We also found that CNO significantly enhanced 2DG-induced FOS expression in the A1/C1 CA neurons, and that corticosterone release also was enhanced when CNO and 2DG were administered together. Importantly, CNO-induced activation of astrocytes in the absence of 2DG did not trigger food intake or corticosterone release. Our results indicate that during glucoprivation, activation of VLM astrocytes cells markedly increases the sensitivity or responsiveness of neighboring A1/C1 CA neurons to glucose deficit, suggesting a potentially important role for VLM astrocytes in glucoregulation.


Sujet(s)
Astrocytes , Corticostérone , Rats , Animaux , Astrocytes/métabolisme , Désoxyglucose/pharmacologie , Rat Sprague-Dawley , Moelle allongée/métabolisme , Glucose/métabolisme , Catécholamines/métabolisme
17.
Nat Biomed Eng ; 7(11): 1350-1373, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37414976

RÉSUMÉ

The mechanisms by which physical exercise benefits brain functions are not fully understood. Here, we show that vertically oscillating head motions mimicking mechanical accelerations experienced during fast walking, light jogging or treadmill running at a moderate velocity reduce the blood pressure of rats and human adults with hypertension. In hypertensive rats, shear stresses of less than 1 Pa resulting from interstitial-fluid flow induced by such passive head motions reduced the expression of the angiotensin II type-1 receptor in astrocytes in the rostral ventrolateral medulla, and the resulting antihypertensive effects were abrogated by hydrogel introduction that inhibited interstitial-fluid movement in the medulla. Our findings suggest that oscillatory mechanical interventions could be used to elicit antihypertensive effects.


Sujet(s)
Antihypertenseurs , Hypertension artérielle , Adulte , Rats , Humains , Animaux , Pression sanguine , Antihypertenseurs/métabolisme , Antihypertenseurs/pharmacologie , Hypertension artérielle/thérapie , Hypertension artérielle/métabolisme , Moelle allongée/métabolisme
18.
J Neuroinflammation ; 20(1): 137, 2023 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-37264405

RÉSUMÉ

BACKGROUND: Neuroinflammation in the rostral ventrolateral medulla (RVLM) has been associated with the pathogenesis of stress-induced hypertension (SIH). Neuronal mitochondrial dysfunction is involved in many pathological and physiological processes. However, the impact of neuroinflammation on neuronal mitochondrial homeostasis and the involved signaling pathway in the RVLM during SIH are largely unknown. METHODS: The morphology and phenotype of microglia and the neuronal mitochondrial injury in vivo were analyzed by immunofluorescence, Western blot, RT-qPCR, transmission electron microscopy, and kit detection. The underlying mechanisms of microglia-derived tumor necrosis factor-α (TNF-α) on neuronal mitochondrial function were investigated through in vitro and in vivo experiments such as immunofluorescence and Western blot. The effect of TNF-α on blood pressure (BP) regulation was determined in vivo via intra-RVLM microinjection of TNF-α receptor antagonist R7050. RESULTS: The results demonstrated that BP, heart rate (HR), renal sympathetic nerve activity (RSNA), plasma norepinephrine (NE), and electroencephalogram (EEG) power increased in SIH rats. Furthermore, the branching complexity of microglia in the RVLM of SIH rats decreased and polarized into M1 phenotype, accompanied by upregulation of TNF-α. Increased neuronal mitochondria injury was observed in the RVLM of SIH rats. Mechanistically, Sirtuin 3 (Sirt3) and p-AMPK expression were markedly downregulated in both SIH rats and TNF-α-treated N2a cells. AMPK activator A769662 upregulated AMPK-Sirt3 signaling pathway and consequently reversed TNF-α-induced mitochondrial dysfunction. Microinjection of TNF-α receptor antagonist R7050 into the RVLM of SIH rats significantly inhibited the biological activities of TNF-α, increased p-AMPK and Sirt3 levels, and alleviated neuronal mitochondrial injury, thereby reducing c-FOS expression, RSNA, plasma NE, and BP. CONCLUSIONS: This study revealed that microglia-derived TNF-α in the RVLM impairs neuronal mitochondrial function in SIH possibly through inhibiting the AMPK-Sirt3 pathway. Therefore, microglia-derived TNF-α in the RVLM may be a possible therapeutic target for the intervention of SIH.


Sujet(s)
Hypertension artérielle , Sirtuine-3 , Rats , Animaux , Facteur de nécrose tumorale alpha/métabolisme , AMP-Activated Protein Kinases/métabolisme , Maladies neuro-inflammatoires , Microglie/métabolisme , Hypertension artérielle/métabolisme , Pression sanguine , Mitochondries/anatomopathologie , Moelle allongée/métabolisme
19.
Neurobiol Dis ; 183: 106173, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37247681

RÉSUMÉ

Neuronal hyperexcitation in the rostral ventrolateral medulla (RVLM) drives heightened sympathetic nerve activity and contributes to the etiology of stress-induced hypertension (SIH). Maintenance of mitochondrial functions is central to neuronal homeostasis. PDZD8, an endoplasmic reticulum (ER) transmembrane protein, tethers ER to mitochondria. However, the mechanisms of PDZD8-mediated ER-mitochondria associations regulating neuronal mitochondrial functions and thereby mediating blood pressure (BP) in the RVLM of SIH were largely unknown. SIH rats were subjected to intermittent electric foot shocks plus noise for 2 h twice daily for 15 consecutive days. The underlying mechanisms of PDZD8 were investigated through in vitro experiments by using small interfering RNA and through in vivo experiments, such as intra-RVLM microinjection and Western blot analysis. The function of PDZD8 on BP regulation in the RVLM was determined in vivo via the intra-RVLM microinjection of adeno-associated virus (AAV)2-r-Pdzd8. We found that the c-Fos-positive RVLM tyrosine hydroxylase (TH) neurons, renal sympathetic nerve activity (RSNA), plasma norepinephrine (NE) level, BP, and heart rate (HR) were elevated in SIH rats. ER-mitochondria associations in RVLM neurons were significantly reduced in SIH rats. PDZD8 was mainly expressed in RVLM neurons, and mRNA and protein levels were markedly decreased in SIH rats. In N2a cells, PDZD8 knockdown disrupted ER-mitochondria associations and mitochondrial structure, decreased mitochondrial membrane potential (MMP) and respiratory metabolism, enhanced ROS levels, and reduced catalase (CAT) activity. These effects suggested that PDZD8 dysregulation induced mitochondrial malfunction. By contrast, PDZD8 upregulation in the RVLM of SIH rats could rescue neuronal mitochondrial function, thereby suppressing c-Fos expression in TH neurons and decreasing RSNA, plasma NE, BP, and HR. Our results indicated that the dysregulation of PDZD8-mediated ER-mitochondria associations led to the loss of the activity homeostasis of RVLM neurons by disrupting mitochondrial functions, thereby participating in the regulation of SIH pathology.


Sujet(s)
Hypertension artérielle , Rats , Animaux , Pression sanguine , Hypertension artérielle/étiologie , Hypertension artérielle/métabolisme , Mitochondries/métabolisme , Antioxydants/pharmacologie , Neurones/métabolisme , Homéostasie , Réticulum endoplasmique/métabolisme , Moelle allongée/métabolisme
20.
Sci Rep ; 13(1): 6550, 2023 04 21.
Article de Anglais | MEDLINE | ID: mdl-37085567

RÉSUMÉ

High fat diet (HFD) promotes cardiovascular disease and blunted cardiac vagal regulation. Temporal onset of loss of cardiac vagal control and its underlying mechanism are presently unclear. We tested our hypothesis that reduced central vagal regulation occurs early after HFD and contributes to poor cardiac regulation using cardiovascular testing paired with pharmacology in mice, molecular biology, and a novel bi-transgenic mouse line. Results show HFD, compared to normal fat diet (NFD), significantly blunted cardio/pulmonary chemoreflex bradycardic responses after 15 days, extending as far as tested (> 30 days). HFD produced resting tachycardia by day 3, reflected significant loss of parasympathetic tone. No differences in bradycardic responses to graded electrical stimulation of the distal cut end of the cervical vagus indicated diet-induced differences in vagal activity were centrally mediated. In nucleus ambiguus (NA), surface expression of δ-subunit containing type A gamma-aminobutyric acid receptors (GABAA(δ)R) increased at day 15 of HFD. Novel mice lacking δ-subunit expression in vagal motor neurons (ChAT-δnull) failed to exhibit blunted reflex bradycardia or resting tachycardia after two weeks of HFD. Thus, reduced parasympathetic output contributes to early HFD-induced HR dysregulation, likely through increased GABAA(δ)Rs. Results underscore need for research on mechanisms of early onset increases in GABAA(δ)R expression and parasympathetic dysfunction after HFD.


Sujet(s)
Alimentation riche en graisse , Moelle allongée , Souris , Animaux , Modèles animaux de maladie humaine , Alimentation riche en graisse/effets indésirables , Moelle allongée/métabolisme , Nerf vague/physiologie , Bradycardie , Acide gamma-amino-butyrique/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...