Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.694
Filter
1.
Physiol Behav ; 280: 114564, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38657747

ABSTRACT

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.


Subject(s)
N-Methylaspartate , Parotid Gland , Receptors, N-Methyl-D-Aspartate , Animals , Male , Rats , Parotid Gland/metabolism , Parotid Gland/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , N-Methylaspartate/pharmacology , N-Methylaspartate/metabolism , Rats, Wistar , Salivation/drug effects , Salivation/physiology , Medulla Oblongata/metabolism , Medulla Oblongata/drug effects , Saliva/metabolism , Excitatory Amino Acid Agonists/pharmacology , Atropine/pharmacology , Propranolol/pharmacology , Adrenergic beta-Antagonists/pharmacology , Microinjections , Sialorrhea
2.
Sci Adv ; 10(17): eadj9581, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669335

ABSTRACT

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.


Subject(s)
Analgesics, Opioid , Locus Coeruleus , Medulla Oblongata , Pain , Periaqueductal Gray , Locus Coeruleus/metabolism , Locus Coeruleus/drug effects , Periaqueductal Gray/metabolism , Periaqueductal Gray/drug effects , Animals , Medulla Oblongata/metabolism , Medulla Oblongata/drug effects , Pain/drug therapy , Pain/metabolism , Analgesics, Opioid/pharmacology , Male , Adrenergic Neurons/metabolism , Adrenergic Neurons/drug effects , Mice , Neural Pathways/drug effects
3.
Biochem Pharmacol ; 201: 115102, 2022 07.
Article in English | MEDLINE | ID: mdl-35617998

ABSTRACT

Activation of the rostral ventrolateral medulla (RVLM) cannabinoid receptor-1 (CB1R) causes neuronal nitric oxide synthase (nNOS)-dependent increases in sympathetic activity, blood pressure (BP) and heart rate (HR) in male rats. However, it remains unknown if the CB1R-mediated neurochemical and cardiovascular responses are influenced by the ovarian sex hormones, particularly estrogen (E2). Therefore, we studied the effects of intra-RVLM CB1R activation (WIN 55,212-2) on BP and HR in conscious female rats under the following hormonal states: (1) highest E2 level (proestrus sham-operated, SO); (2) E2-deprivation (ovariectomized, OVX); (3) OVX with E2 replacement (OVXE2). Intra-RVLM WIN55,212-2 elicited dose (100-400 pmol) dependent pressor and tachycardic responses, in OVX rats, which replicated the reported responses in male rats. However, in SO and OVXE2 rats, the CB1R-mediated pressor response was attenuated and the tachycardic response reverted to bradycardic response. The neurochemical findings suggested a key role for the upregulated RVLM sympathoexcitatory molecules phosphorated protein kinase B, phosphorated nNOS and reactive oxygen species in the exaggerated CB1R-mediated BP and HR responses in OVX rats, and an E2-dependent dampening of these responses. The intra-RVLM WIN55212-2-evoked cardiovascular and neurochemical responses were CB1R-mediated because they were attenuated by prior CB1R blockade (AM251). Our findings suggest that attenuation of RVLM neuroexcitation and oxidative stress underlies the protection conferred by E2, in female rats, against the CB1R-mediated adverse cardiovascular effects.


Subject(s)
Estrogens , Medulla Oblongata , Receptor, Cannabinoid, CB1 , Vasoconstrictor Agents , Animals , Benzoxazines/pharmacology , Blood Pressure/drug effects , Estrogens/metabolism , Estrogens/pharmacology , Female , Heart Rate/drug effects , Male , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Morpholines/pharmacology , Naphthalenes/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , Vasoconstrictor Agents/pharmacology
4.
Biomed Pharmacother ; 150: 113018, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35483194

ABSTRACT

This study aims to determine whether toll-like receptor 4 (TLR4)-mediated inflammation in rostral ventrolateral medulla (RVLM) causes sympathetic overactivity leading to preeclampsia (PE) and if TLR4 inhibition with hydrogen sulfide (H2S) would reduce PE severity. Thirty patients with PE and 30 pregnant controls were involved. PE in rats was induced through deoxycorticosterone acetate and normal saline. NaHS (donor of H2S), lipopolysaccharide (LPS) (TLR4 agonist), and TAK-242 (TLR4 inhibitor) were injected in lateral cerebral ventricle to investigate their effect on microglia-mediated inflammation in RVLM, sympathetic activation, and PE symptoms. In patients with PE, plasma levels of NE, TNF-α, and interleukin-1ß were high compared with those of controls, whereas levels of H2S were low. Rats with PE showed an increased amount of renal sympathetic nerve activity and plasma levels of NE, with decreased H2S levels in RVLM. Microglia-mediated inflammation was observed in the RVLM of PE rats. Central infusion of LPS in pregnant rats induced microglia-mediated inflammation, sympathetic nervous tension, and PE-like symptoms, whereas TAK-242 reduced PE symptoms. NaHS treatment lessened microglia-mediated inflammation in the RVLM, sympathetic tension, and symptoms of PE both in PE rats and LPS-treating pregnant rats.These results suggest that inflammation in the RVLM caused by microglial activation might contribute to the progression of PE via an overactive sympathetic system. H2S could reduce PE via inhibiting inflammation in the RVLM. These results might provide a new target for the treatment of PE.


Subject(s)
Hydrogen Sulfide , Pre-Eclampsia , Toll-Like Receptor 4 , Animals , Blood Pressure/drug effects , Female , Humans , Hydrogen Sulfide/pharmacology , Inflammation/drug therapy , Medulla Oblongata/drug effects , Pre-Eclampsia/drug therapy , Pregnancy , Rats , Toll-Like Receptor 4/antagonists & inhibitors
5.
Life Sci ; 295: 120405, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35181311

ABSTRACT

AIMS: The rostral ventrolateral medulla (RVLM) is the main sympathetic output of the central nervous system to control blood pressure. Reportedly, reactive oxygen species (ROS) can increase arterial pressure, leading to hypertension. As ROS increase the sympathetic tone in RVLM and obese animals present grater oxidative stress, it would be important to note this relationship. MAIN METHODS: Therefore, we evaluated the systemic and central effects (in the RVLM) of vitamin C (vit C, an antioxidant) on the redox balance and cardiovascular and autonomic profiles in hyperadipose male rats. We also evaluated the neurotransmission by L-glutamate (L-glu) and vit C in the RVLM of awake hyperadipose rats. KEY FINDINGS: Our study confirmed that hyperadipose rats were hypertensive and tachycardic, presented increased sympathetic and decreased parasympathetic modulation of the heart, and had increased plasma lipoperoxidation compared with the control rats (CTR). Oral vitamin C treatment reverted cardiovascular, autonomic, and plasma redox dysfunction. Hyperadipose rats presented a higher blood pressure increase after L-glu microinjection and a lower response to vit C in the RVLM compared with the CTR group. Biochemical analysis of redox balance in RVLM punches showed that hyperadipose rats have increased NBT and T-BARS, and after treatment with vit C, the oxidative profile decreased. The antioxidative activity of vit C reduced the amount of ROS in the RVLM area that might have resulted in lowered blood pressure and sympathetic modulation. SIGNIFICANCE: Our data suggest central and peripheral benefits of vit C treatment on cardiovascular, autonomic, and oxidative dysfunctions in hyperadipose animals.


Subject(s)
Ascorbic Acid/pharmacology , Hypertension/drug therapy , Medulla Oblongata/metabolism , Animals , Antioxidants/pharmacology , Autonomic Nervous System/physiopathology , Blood Pressure/drug effects , Cardiovascular System/physiopathology , Heart Rate/drug effects , Hypertension/physiopathology , Male , Medulla Oblongata/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Reactive Oxygen Species/pharmacology , Superoxide Dismutase/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
6.
Respir Physiol Neurobiol ; 296: 103810, 2022 02.
Article in English | MEDLINE | ID: mdl-34728431

ABSTRACT

Systemic 8-OH-DPAT (a 5-HT1A receptor agonist) challenge evokes hyperventilation independent of peripheral 5-HT1A receptors. Though the pre-Botzinger Complex (PBC) is critical in generating respiratory rhythm and activation of local 5-HT1A receptors induces tachypnea via disinhibition of local GABAA neurons, its role in the respiratory response to systemic 8-OH-DPAT challenge is still unclear. In anesthetized rats, 8-OH-DPAT (100 µg/kg, iv) was injected twice to confirm the reproducibility of the evoked responses. The same challenges were performed after bilateral microinjections of (S)-WAY-100135 (a 5-HT1A receptor antagonist) or gabazine (a GABAA receptor antagonist) into the PBC. Our results showed that: 1) 8-OH-DPAT caused reproducible hyperventilation associated with hypotension and bradycardia; 2) microinjections of (S)-WAY-100135 into the PBC attenuated the hyperventilation by ˜60 % without effect on the evoked hypotension and bradycardia; and 3) the same hyperventilatory attenuation was also observed after microinjections of gabazine into the PBC. Our data suggest that PBC 5-HT1A receptors play a key role in the respiratory response to systemic 8-OH-DPAT challenge likely via disinhibiting local GABAergic neurons.


Subject(s)
8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , GABA Antagonists/pharmacology , Hyperventilation/chemically induced , Hyperventilation/drug therapy , Medulla Oblongata/metabolism , Receptor, Serotonin, 5-HT1A/physiology , Respiratory Center/metabolism , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/administration & dosage , Animals , Disease Models, Animal , Male , Medulla Oblongata/drug effects , Piperazines/pharmacology , Pyridazines/pharmacology , Rats , Receptor, Serotonin, 5-HT1A/drug effects , Respiratory Center/drug effects
7.
Brain Res ; 1774: 147726, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34785257

ABSTRACT

Moderate exercise reduces arterial pressure (AP) and heart rate (HR) in spontaneously hypertensive rats (SHR) and changes neurotransmission in medullary areas involved in cardiovascular regulation. We investigated if regularly swimming exercise (SW) affects the cardiovascular adjustments mediated by opioidergic neuromodulation in the RVLM in SHR and Wistar-Kyoto (WKY) rats. Rats were submitted to 6 wks of SW. The day after the last exercise bout, α-chloralose-anesthetized rats underwent a cannulation of the femoral artery for AP and HR recordings, and Doppler flow probes were placed around the lower abdominal aorta and superior mesenteric artery. Bilateral injection of endomorphin-2 (EM-2, 0.4 mmol/L, 60 nL) into the RVLM increased MAP in SW-SHR (20 ± 4 mmHg, N = 6), which was lower than in sedentary (SED)-SHR (35 ± 4 mmHg, N = 6). The increase in MAP in SW-SHR induced by EM-2 into the RVLM was similar in SED- and SW-WKY. Naloxone (0.5 mmol/L, 60 nL) injected into the RVLM evoked an enhanced hypotension in SW-SHR (-66 ± 8 mmHg, N = 6) compared to SED-SHR (-25 ± 3 mmHg, N = 6), which was similar in SED- and SW-WKY. No significant changes were observed in HR after EM-2 or naloxone injections into the RVLM. Changes in hindquarter and mesenteric conductances evoked by EM-2 or naloxone injections into the RVLM in SW- or SED-SHR were not different. Mu Opioid Receptor expression by Western blotting was reduced in SW-SHR than in SED-SHR and SW-WKY. Therefore, regularly SW alters the opioidergic neuromodulation in the RVLM in SHR and modifies the mu opioid receptor expression in this medullary area.


Subject(s)
Analgesics, Opioid/pharmacology , Hypertension/metabolism , Medulla Oblongata/metabolism , Neurons/drug effects , Physical Conditioning, Animal , Receptors, Opioid, mu/metabolism , Animals , Arterial Pressure/drug effects , Arterial Pressure/physiology , Heart Rate/drug effects , Heart Rate/physiology , Medulla Oblongata/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/metabolism , Oligopeptides/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Swimming
8.
J Neurophysiol ; 127(1): 267-278, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34879205

ABSTRACT

Brainstem respiratory neuronal network significantly contributes to cough motor pattern generation. Neuronal populations in the pre-Bötzinger complex (PreBötC) represent a substantial component for respiratory rhythmogenesis. We studied the role of PreBötC neuronal excitation and inhibition on mechanically induced tracheobronchial cough in 15 spontaneously breathing, pentobarbital anesthetized adult cats (35 mg/kg, iv initially). Neuronal excitation by unilateral microinjection of glutamate analog d,l-homocysteic acid resulted in mild reduction of cough abdominal electromyogram (EMG) amplitudes and very limited temporal changes of cough compared with effects on breathing (very high respiratory rate, high amplitude inspiratory bursts with a short inspiratory phase, and tonic inspiratory motor component). Mean arterial blood pressure temporarily decreased. Blocking glutamate-related neuronal excitation by bilateral microinjections of nonspecific glutamate receptor antagonist kynurenic acid reduced cough inspiratory and expiratory EMG amplitude and shortened most cough temporal characteristics similarly to breathing temporal characteristics. Respiratory rate decreased and blood pressure temporarily increased. Limiting active neuronal inhibition by unilateral and bilateral microinjections of GABAA receptor antagonist gabazine resulted in lower cough number, reduced expiratory cough efforts, and prolongation of cough temporal features and breathing phases (with lower respiratory rate). The PreBötC is important for cough motor pattern generation. Excitatory glutamatergic neurotransmission in the PreBötC is involved in control of cough intensity and patterning. GABAA receptor-related inhibition in the PreBötC strongly affects breathing and coughing phase durations in the same manner, as well as cough expiratory efforts. In conclusion, differences in effects on cough and breathing are consistent with separate control of these behaviors.NEW & NOTEWORTHY This study is the first to explore the role of the inspiratory rhythm and pattern generator, the pre-Bötzinger complex (PreBötC), in cough motor pattern formation. In the PreBötC, excitatory glutamatergic neurotransmission affects cough intensity and patterning but not rhythm, and GABAA receptor-related inhibition affects coughing and breathing phase durations similarly to each other. Our data show that the PreBötC is important for cough motor pattern generation, but cough rhythmogenesis appears to be controlled elsewhere.


Subject(s)
Central Pattern Generators , Cough , Excitatory Amino Acid Antagonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Glutamic Acid/pharmacology , Inhalation , Medulla Oblongata , Reflex , Respiratory Rate , Abdominal Muscles/drug effects , Abdominal Muscles/physiopathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cats , Central Pattern Generators/drug effects , Central Pattern Generators/metabolism , Central Pattern Generators/physiopathology , Cough/drug therapy , Cough/metabolism , Cough/physiopathology , Electromyography , Excitatory Amino Acid Antagonists/administration & dosage , Female , GABA-A Receptor Antagonists/administration & dosage , Glutamic Acid/administration & dosage , Glutamic Acid/analysis , Homocysteine/analogs & derivatives , Homocysteine/pharmacology , Inhalation/drug effects , Inhalation/physiology , Kynurenic Acid/pharmacology , Male , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Medulla Oblongata/physiopathology , Pyridazines/pharmacology , Reflex/drug effects , Reflex/physiology , Respiratory Rate/drug effects , Respiratory Rate/physiology
9.
J Biochem Mol Toxicol ; 35(11): e22888, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34392583

ABSTRACT

Although doxorubicin (DOX) is used in many cancer treatments, it causes neurotoxicity. In this study, the effect of thymoquinone (THQ), a powerful antioxidant, on DOX-induced neurotoxicity was evaluated. In total, 40 rats were used and 5 groups were formed. Group I: control group (n = 8); Group II: olive oil group (n = 8); Group III: the THQ group (n = 8); THQ 10 mg/kg per day was given intraperitoneally (i.p.) throughout the experiment; group IV: DOX group (n = 8); On Day 7 of the experiment, a single dose of 15 mg/kg intraperitoneally DOX injected; group V: DOX + THQ group (n = 8); Throughout the experiment, 10 mg/kg THQ per day and intraperitoneally 15 mg/kg DOX on Day 7 were injected. Immunohistochemically, tumor necrosis factor-α (TNF-α), interleukin-17 (IL-17), hypoxia-inducible factor 1α (HIF1-α), glucose regulatory protein 78 (GRP78), and the gene inducible by growth arrest and DNA damage 153 (GADD153) proteins were evaluated in the brain cortex, medulla, and hippocampus regions. Total oxidant status (TOS) levels and total antioxidant status (TAS) in the brain tissue were measured. TNF-α, IL-17, HIF1-α, GRP78, and GADD153 immunoreactivities significantly increased in the DOX group in the study. THQ significantly reduced these values. THQ increased the TAS level significantly and decreased the TOS level significantly compared to the DOX group. THQ may play a role as a neuroprotective agent in DOX-induced neurotoxicity in the cortex, medulla, and hippocampus regions of the brain.


Subject(s)
Benzoquinones/pharmacology , Cerebral Cortex/drug effects , Doxorubicin/toxicity , Endoplasmic Reticulum Stress/drug effects , Hippocampus/drug effects , Inflammation/prevention & control , Medulla Oblongata/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Antibiotics, Antineoplastic/toxicity , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Hippocampus/metabolism , Hippocampus/pathology , Medulla Oblongata/metabolism , Medulla Oblongata/pathology , Rats , Rats, Wistar
10.
Respir Physiol Neurobiol ; 293: 103736, 2021 11.
Article in English | MEDLINE | ID: mdl-34224867

ABSTRACT

The preBötzinger complex (preBötC) is a medullary area essential for normal breathing and widely recognized as necessary and sufficient to generate the inspiratory phase of respiration. It has been studied mainly in rodents. Here we report the main results of our studies revealing the characteristics of the rabbit preBötC identified by means of neuronal recordings, D,L-homocysteic acid microinjections and histological controls. A crucial role in the respiratory rhythmogenesis within this neural substrate is played by excitatory amino acids, but also GABA and glycine display important contributions. Increases in respiratory frequency are induced by microinjections of neurokinins, somatostatin as well by serotonin (5-HT) through an action on 5-HT1A and 5-HT3 receptors or the disinhibition of a GABAergic circuit. Respiratory depression is observed in response to microinjections of the µ-opioid receptor agonist DAMGO. Our results show similarities and differences with the rodent preBötC and emphasize the importance of comparative studies on the mechanisms underlying respiratory rhythmogenesis in different animal species.


Subject(s)
Central Pattern Generators/physiology , Medulla Oblongata/physiology , Neurotransmitter Agents/pharmacology , Respiratory Center/physiology , Respiratory Physiological Phenomena , Animals , Central Pattern Generators/drug effects , Medulla Oblongata/drug effects , Rabbits , Respiratory Center/drug effects , Respiratory Physiological Phenomena/drug effects
11.
CNS Neurosci Ther ; 27(11): 1313-1326, 2021 11.
Article in English | MEDLINE | ID: mdl-34255932

ABSTRACT

AIMS: Chronification of postoperative pain is a common clinical phenomenon following surgical operation, and it perplexes a great number of patients. Estrogen and its membrane receptor (G protein-coupled estrogen receptor, GPER) play a crucial role in pain regulation. Here, we explored the role of GPER in the rostral ventromedial medulla (RVM) during chronic postoperative pain and search for the possible mechanism. METHODS AND RESULTS: Postoperative pain was induced in mice or rats via a plantar incision surgery. Behavioral tests were conducted to detect both thermal and mechanical pain, showing a small part (16.2%) of mice developed into pain persisting state with consistent low pain threshold on 14 days after incision surgery compared with the pain recovery mice. Immunofluorescent staining assay revealed that the GPER-positive neurons in the RVM were significantly activated in pain persisting rats. In addition, RT-PCR and immunoblot analyses showed that the levels of GPER and phosphorylated µ-type opioid receptor (p-MOR) in the RVM of pain persisting mice were apparently increased on 14 days after incision surgery. Furthermore, chemogenetic activation of GPER-positive neurons in the RVM of Gper-Cre mice could reverse the pain threshold of pain recovery mice. Conversely, chemogenetic inhibition of GPER-positive neurons in the RVM could prevent mice from being in the pain persistent state. CONCLUSION: Our findings demonstrated that the GPER in the RVM was responsible for the chronification of postoperative pain and the downstream pathway might be involved in MOR phosphorylation.


Subject(s)
Chronic Pain/genetics , Medulla Oblongata/drug effects , Pain, Postoperative/genetics , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Chronic Pain/physiopathology , Hyperalgesia/psychology , Male , Mice , Mice, Inbred C57BL , Pain Measurement , Pain, Postoperative/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics
12.
Behav Brain Res ; 414: 113450, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34265318

ABSTRACT

Investigations have shown that the circadian rhythm can affect the mechanisms associated with drug dependence. In this regard, we sought to assess the negative consequence of morphine withdrawal syndrome on conditioned place aversion (CPA) and lateral paragigantocellularis (LPGi) neuronal activity in morphine-dependent rats during light (8:00-12:00) and dark (20:00-24:00) cycles. Male Wistar rats (250-300 g) were received 10 mg/kg morphine or its vehicle (Saline, 2 mL/kg/12 h, s.c.) in 13 consecutive days for behavioral assessment tests. Then, naloxone-induced conditioned place aversion and physical signs of withdrawal syndrome were evaluated during light and dark cycles. In contrast to the behavioral part, we performed in vivo extracellular single-unit recording for investigating the neural response of LPGi to naloxone in morphine-dependent rats on day 10 of morphine/saline exposure. Results showed that naloxone induced conditioned place aversion in both light and dark cycles, but the CPA score during the light cycle was larger. Moreover, the intensity of physical signs of morphine withdrawal syndrome was more severe during the light cycle (rest phase) compare to the dark one. In electrophysiological experiments, results indicated that naloxone evoked both excitatory and inhibitory responses in LPGi neurons and the incremental effect of naloxone on LPGi activity was stronger in the light cycle. Also, the neurons with the excitatory response exhibited higher baseline activity in the dark cycle, but the neurons with the inhibitory response showed higher baseline activity in the light cycle. Interestingly, the baseline firing rate of neurons recorded in the light cycle was significantly different in response (excitatory/inhibitory) -dependent manner. We concluded that naloxone-induced changes in LPGi cellular activity and behaviors of morphine-dependent rats can be affected by circadian rhythm and the internal clock.


Subject(s)
Behavior, Animal/physiology , Circadian Rhythm/physiology , Conditioning, Classical/physiology , Electrophysiological Phenomena/physiology , Medulla Oblongata/physiopathology , Morphine Dependence/physiopathology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Substance Withdrawal Syndrome/physiopathology , Animals , Behavior, Animal/drug effects , Conditioning, Classical/drug effects , Disease Models, Animal , Electrophysiological Phenomena/drug effects , Male , Medulla Oblongata/drug effects , Neurons/physiology , Rats , Rats, Wistar
13.
PLoS One ; 16(5): e0251495, 2021.
Article in English | MEDLINE | ID: mdl-34010316

ABSTRACT

Baroreflex dysfunction is partly implicated in hypertension and one responsible region is the dorsal medulla oblongata including the nucleus tractus solitarius (NTS). NTS neurons receive and project glutamatergic inputs to subsequently regulate blood pressure, while G-protein-coupled metabotropic glutamate receptors (mGluRs) play a modulatory role for glutamatergic transmission in baroreflex pathways. Stimulating group II mGluR subtype 2 and 3 (mGluR2/3) in the brainstem can decrease blood pressure and sympathetic nervous activity. Here, we hypothesized that the chronic stimulation of mGluR2/3 in the dorsal medulla oblongata can alleviate hypertensive development via the modulation of autonomic nervous activity in young, spontaneously hypertensive rats (SHRs). Compared with that in the sham control group, chronic LY379268 application (mGluR2/3 agonist; 0.40 µg/day) to the dorsal medulla oblongata for 6 weeks reduced the progression of hypertension in 6-week-old SHRs as indicated by the 40 mmHg reduction in systolic blood pressure and promoted their parasympathetic nervous activity as evidenced by the heart rate variability. No differences in blood catecholamine levels or any echocardiographic indices were found between the two groups. The improvement of reflex bradycardia, a baroreflex function, appeared after chronic LY379268 application. The mRNA expression level of mGluR2, but not mGluR3, in the dorsal medulla oblongata was substantially reduced in SHRs compared to that of the control strain. In conclusion, mGluR2/3 signaling might be responsible for hypertension development in SHRs, and modulating mGluR2/3 expression/stimulation in the dorsal brainstem could be a novel therapeutic strategy for hypertension via increasing the parasympathetic activity.


Subject(s)
Amino Acids/therapeutic use , Antihypertensive Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Hypertension/drug therapy , Medulla Oblongata/drug effects , Receptors, Metabotropic Glutamate/agonists , Amino Acids/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Hypertension/physiopathology , Male , Medulla Oblongata/physiopathology , Rats, Inbred SHR
14.
J Neuroendocrinol ; 33(5): e12977, 2021 05.
Article in English | MEDLINE | ID: mdl-33942389

ABSTRACT

A neural circuit between the paraventricular nucleus of the hypothalamus (PVN) and the dorsal motor nucleus of the vagus (DMNV) constitutes part of an important parasympathetic autonomic pathway that controls hepatic glucose production. Intracerebroventricular injection of insulin activates oxytocinergic neurones in the PVN and elicits the release of oxytocin into the circulation, which plays an important role in the metabolism of glucose. Moreover, the central action of insulin can reduce the concentration of glucose in blood taken from the hepatic vein of Wistar rats via activation of vagal efferent nerves to the liver. This mechanism is impaired in sedentary spontaneously hypertensive rats (SHR). Because aerobic exercise increases vagal tone, partly mediated by increasing the oxytocinergic connections between the PVN and DMNV, we hypothesised that oxytocin (OT) might alter the excitability of liver-projecting DMNV neurones. Thus, we investigated the effects of OT on electrical properties of the liver-projecting DMNV neurones from Wistar, SHR subjected to 4 weeks of exercise training, as well sedentary controls, using whole cell patch-clamping. The results show that OT increased the resting membrane potential of DMNV neurones in Wistar rats, as well as the firing frequency of these cells, but not in sedentary SHR. However, in SHR subjected to 4 weeks of exercise training, the effects of OT on liver-projecting DMNV neurones of were similar to those seen in Wistar rats. These findings show that OT elicits similar changes in the electrophysiological properties of liver-projecting DMNV neurones of Wistar and exercise-trained but not sedentary SHR. These results indicate that exercise training can restore the sensitivity of liver-projecting DMNV neurones of exercise-trained SHR to OT.


Subject(s)
Brain Stem/drug effects , Liver/innervation , Neurons/drug effects , Oxytocin/pharmacology , Physical Conditioning, Animal , Animals , Blood Glucose , Brain Stem/metabolism , Male , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Neurons/metabolism , Rats , Rats, Inbred SHR , Rats, Wistar
15.
Metab Brain Dis ; 36(6): 1305-1314, 2021 08.
Article in English | MEDLINE | ID: mdl-33914222

ABSTRACT

Exposure to high fat diet during perinatal period (PHFD) leads to neuroplastic changes in autonomic circuits, however, the role of gender has been incompletely understood. This study aims to investigate (i) short, and (ii) long-term effects of PHFD on autonomic outflow, and (iii) sexual dimorphic variations emerge at adulthood. Male and female rats were fed a control diet (13.5 % kcal from fat) or PHFD (60 % kcal from fat) from embryonic day-14 to postnatal day-21. To assess changes in autonomic outflow, heart rate variability (HRV) was analyzed at 10- and 20-week-old ages. Expressions of tyrosine hydroxylase (TH), metabotropic glutamate2/3 receptor (mGlu2/3R), N-methyl-D-aspartate1 receptor (NMDA1R), and gamma aminobutyric acidA receptor (GABAAR) were evaluated by immunohistochemistry. PHFD did not affect the body weight of 4-, 10-or 20-week-old male or female offsprings. PHFD significantly increased the sympathetic marker low frequency (LF) component, and sympatho-vagal balance (LF:HF) only in 10-week-old PHFD males. Compared with control, the propranolol-induced (4 mg·kg- 1, ip) decline in LF was observed more prominently in PHFD rats, however, these changes were found to be restored at the age of 20 weeks. In caudal ventrolateral medulla and nucleus tractus solitarius, expression of mGlu2/3R was downregulated in PHFD males, whereas no change was detected in NMDA1R. The number of GABAAR-expressing TH-immunoreactive cells was decreased in rostral ventrolateral medulla of PHFD males. The findings of this study suggest that exposure to maternal high-fat diet could lead to autonomic imbalance with increased sympathetic tone in the early adulthood of male offspring rats without developing obesity.


Subject(s)
Diet, High-Fat , Maternal Exposure , Medulla Oblongata/drug effects , Nerve Net/drug effects , Sex Characteristics , Sympathetic Nervous System/drug effects , Adrenergic beta-Antagonists/pharmacology , Animals , Animals, Newborn , Body Weight , Female , Heart Rate/drug effects , Male , Pregnancy , Prenatal Exposure Delayed Effects , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism
16.
J Neurophysiol ; 125(5): 1899-1919, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33826874

ABSTRACT

Opioid-induced respiratory depression (OIRD) represents the primary cause of death associated with therapeutic and recreational opioid use. Within the United States, the rate of death from opioid abuse since the early 1990s has grown disproportionally, prompting the classification as a nationwide "epidemic." Since this time, we have begun to unravel many fundamental cellular and systems-level mechanisms associated with opioid-related death. However, factors such as individual vulnerability, neuromodulatory compensation, and redundancy of opioid effects across central and peripheral nervous systems have created a barrier to a concise, integrative view of OIRD. Within this review, we bring together multiple perspectives in the field of OIRD to create an overarching viewpoint of what we know, and where we view this essential topic of research going forward into the future.


Subject(s)
Analgesics, Opioid/pharmacology , Central Pattern Generators/drug effects , Medulla Oblongata/drug effects , Opioid-Related Disorders/complications , Respiratory Insufficiency/chemically induced , Analgesics, Opioid/adverse effects , Animals , Humans
17.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33764905

ABSTRACT

Acute high-fat diet (aHFD) exposure induces a brief period of hyperphagia before caloric balance is restored. Previous studies have demonstrated that this period of regulation is associated with activation of synaptic N-methyl-D-aspartate (NMDA) receptors on dorsal motor nucleus of the vagus (DMV) neurons, which increases vagal control of gastric functions. Our aim was to test the hypothesis that activation of DMV synaptic NMDA receptors occurs subsequent to activation of extrasynaptic NMDA receptors. Sprague-Dawley rats were fed a control or high-fat diet for 3-5 days prior to experimentation. Whole-cell patch-clamp recordings from gastric-projecting DMV neurons; in vivo recordings of gastric motility, tone, compliance, and emptying; and food intake studies were used to assess the effects of NMDA receptor antagonism on caloric regulation. After aHFD exposure, inhibition of extrasynaptic NMDA receptors prevented the synaptic NMDA receptor-mediated increase in glutamatergic transmission to DMV neurons, as well as the increase in gastric tone and motility, while chronic extrasynaptic NMDA receptor inhibition attenuated the regulation of caloric intake. After aHFD exposure, the regulation of food intake involved synaptic NMDA receptor-mediated currents, which occurred in response to extrasynaptic NMDA receptor activation. Understanding these events may provide a mechanistic basis for hyperphagia and may identify novel therapeutic targets for the treatment of obesity.


Subject(s)
Appetite Regulation/physiology , Diet, High-Fat , Energy Intake/physiology , Gastric Emptying/physiology , Medulla Oblongata/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Vagus Nerve/metabolism , Animals , Appetite Regulation/drug effects , Eating/drug effects , Eating/physiology , Energy Intake/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Gastric Emptying/drug effects , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Medulla Oblongata/drug effects , Medulla Oblongata/physiology , Memantine/pharmacology , Neurons/drug effects , Patch-Clamp Techniques , Piperidines/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Stomach , Vagus Nerve/drug effects , Vagus Nerve/physiology
18.
Eur J Pharmacol ; 899: 174033, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33727058

ABSTRACT

The dorsomedial hypothalamus (DMH) receives dense orexinergic innervation. Intra-DMH application of orexins increases arterial pressure and heart rate in rats. We studied the effects of orexin-A on DMH neurons, including those innervating the medullary cardiovascular center, the rostral ventrolateral medulla (RVLM), by using whole-cell recordings in brain slices. In the presence of tetrodotoxin, orexin-A (30-1000 nM) depolarized 56% of DMH neurons (EC50 82.4 ± 4.4 nM). Under voltage-clamp recording, orexin-A (300 nM) induced three types of responses characterized by different current-voltage relationships, namely unchanged, increased, and decreased slope conductance in 68%, 14%, and 18% of orexin-A-responsive neurons, respectively. The reversal potential of the decreased-conductance response was near the equilibrium potential of K+ and became more positive in a high-K+ solution, suggesting that K+ conductance blockade is the underlying mechanism. In a low-Na+ solution, unchanged-, increased-, and decreased-conductance responses were observed in 56%, 11%, and 33% of orexin-A-responsive neurons, respectively, implying that a non-selective cation current (NSCC) underlies orexin-A-induced responses in a small population of DMH neurons. KBR-7943 (70 µM), an inhibitor of Na+-Ca2+ exchanger (NCX), suppressed orexin-A-induced depolarization in 7 of 10 neurons. In the presence of KBR-7943, the majority of orexin-A-responsive neurons exhibited decreased-conductance responses. These findings suggest that NCX activation may underlie orexin-A-induced depolarization in the majority of orexin-responsive DMH neurons. Of 19 RVLM-projecting DMH neurons identified by retrograde labeling, 17 (90%) were orexin-A responsive. In conclusion, orexin-A directly excited over half of DMH neurons, including those innervating the RVLM, through decreasing K+ conductance, activating NCX, and/or increasing NSCC.


Subject(s)
Dorsomedial Hypothalamic Nucleus/drug effects , Medulla Oblongata/drug effects , Neurons/drug effects , Orexins/pharmacology , Animals , Dorsomedial Hypothalamic Nucleus/cytology , Dorsomedial Hypothalamic Nucleus/metabolism , Female , In Vitro Techniques , Male , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Membrane Potentials , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuroanatomical Tract-Tracing Techniques , Neurons/metabolism , Potassium/metabolism , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/metabolism
19.
J Neurosci ; 41(14): 3192-3203, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33608383

ABSTRACT

Behavioral and internal-state modulation of sensory processing has been described in several organisms. In insects, visual neurons in the optic lobe are modulated by locomotion, but the degree to which visual-motor feedback modulates these neurons remains unclear. Moreover, it also remains unknown whether self-generated and externally generated visual motion are processed differently. Here, we implemented a virtual reality system that allowed fine-scale control over visual stimulation in relation to animal motion, in combination with multichannel recording of neural activity in the medulla of a female honeybee (Apis mellifera). We found that this activity was modulated by locomotion, although, in most cases, only when the bee had behavioral control over the visual stimulus (i.e., in a closed-loop system). Moreover, closed-loop control modulated a third of the recorded neurons, and the application of octopamine (OA) evoked similar changes in neural responses that were observed in a closed loop. Additionally, in a subset of modulated neurons, fixation on a visual stimulus was preceded by an increase in firing rate. To further explore the relationship between neuromodulation and adaptive control of the visual environment of the bee, we modified motor gain sensitivity while locally injecting an OA receptor antagonist into the medulla. Whereas female honeybees were tuned to a motor gain of -2 to 2 (between the heading of the bee and its visual feedback), local disruption of the OA pathway in the medulla abolished this tuning, resulting in similar low levels of response across levels of motor gain. Our results show that behavioral control modulates neural activity in the medulla and ultimately impacts behavior.SIGNIFICANCE STATEMENT When moving, an animal generates the motion of the visual scene over its retina. We asked whether self-generated and externally generated optic flow are processed differently in the insect medulla. Our results show that closed-loop control of the visual stimulus modulates neural activity as early as the medulla and ultimately impacts behavior. Moreover, blocking octopaminergic modulation further disrupted object-tracking responses. Our results suggest that the medulla is an important site for context-dependent processing of visual information and that placing the animal in a closed-loop environment may be essential to understanding its visual cognition and processing.


Subject(s)
Feedback, Sensory/physiology , Locomotion/physiology , Medulla Oblongata/physiology , Photic Stimulation/methods , Psychomotor Performance/physiology , Animals , Bees , Feedback, Sensory/drug effects , Female , Locomotion/drug effects , Medulla Oblongata/drug effects , Octopamine/agonists , Octopamine/antagonists & inhibitors , Octopamine/pharmacology , Psychomotor Performance/drug effects
20.
Mol Pain ; 17: 1744806921992187, 2021.
Article in English | MEDLINE | ID: mdl-33573476

ABSTRACT

Neuropeptide W (NPW) messenger ribonucleic acid (mRNA) and NPBW1 and/or NPBW2 mRNA are expressed in the descending pain inhibitory system. In the present study, we examined whether NPW microinjected into the descending pain inhibitory system, such as the periaqueductal gray (PAG), locus coeruleus (LC), and rostral ventromedial medulla (RVM), produces an analgesic effect using a rat formalin test. Microinjections of NPW into the PAG ipsilateral and contralateral to the formalin-injected side, LC ipsilateral and contralateral to the formalin-injected side, and RVM produced an analgesic effect. In the RVM study, the analgesic effect was antagonized by WAY100135, a 5-HT1A antagonist, and enhanced by prazosin, an α1 antagonist, and SB269970, a 5-HT7 antagonist. Naloxone, an opioid antagonist, also antagonized the effect of NPW in the RVM study. In the ipsilateral LC study, the analgesic effect was antagonized by WAY100135, idazoxan, an α2 antagonist, and naloxone and was enhanced by prazosin and SB269970. In the contralateral LC study, the analgesic effect was antagonized by prazosin, idazoxan, SB269970, and naloxone. The analgesic effect was antagonized by WAY100135, SB269970, idazoxan, and naloxone in the ipsilateral and contralateral PAG studies. These findings strongly suggest that NPBW1/W2 activation by NPW microinjection into the RVM, LC, and PAG affect the descending pain modulatory system and produce anti-nociceptive and pro-nociceptive effects in the rat formalin test.


Subject(s)
Analgesics/pharmacology , Neuropeptides/pharmacology , Pain/pathology , Receptors, Neuropeptide/metabolism , Analgesics/administration & dosage , Animals , Formaldehyde , Injections , Ligands , Locus Coeruleus/drug effects , Male , Medulla Oblongata/drug effects , Neuropeptides/administration & dosage , Periaqueductal Gray/drug effects , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...