Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.849
Filter
1.
Nature ; 609(7928): 761-771, 2022 09.
Article in English | MEDLINE | ID: mdl-36071158

ABSTRACT

Infections induce a set of pleiotropic responses in animals, including anorexia, adipsia, lethargy and changes in temperature, collectively termed sickness behaviours1. Although these responses have been shown to be adaptive, the underlying neural mechanisms have not been elucidated2-4. Here we use of a set of unbiased methodologies to show that a specific subpopulation of neurons in the brainstem can control the diverse responses to a bacterial endotoxin (lipopolysaccharide (LPS)) that potently induces sickness behaviour. Whole-brain activity mapping revealed that subsets of neurons in the nucleus of the solitary tract (NTS) and the area postrema (AP) acutely express FOS after LPS treatment, and we found that subsequent reactivation of these specific neurons in FOS2A-iCreERT2 (also known as TRAP2) mice replicates the behavioural and thermal component of sickness. In addition, inhibition of LPS-activated neurons diminished all of the behavioural responses to LPS. Single-nucleus RNA sequencing of the NTS-AP was used to identify LPS-activated neural populations, and we found that activation of ADCYAP1+ neurons in the NTS-AP fully recapitulates the responses elicited by LPS. Furthermore, inhibition of these neurons significantly diminished the anorexia, adipsia and locomotor cessation seen after LPS injection. Together these studies map the pleiotropic effects of LPS to a neural population that is both necessary and sufficient for canonical elements of the sickness response, thus establishing a critical link between the brain and the response to infection.


Subject(s)
Brain Stem , Illness Behavior , Neurons , Animals , Anorexia/complications , Area Postrema/cytology , Area Postrema/metabolism , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/physiology , Illness Behavior/drug effects , Lethargy/complications , Lipopolysaccharides/pharmacology , Mice , Neurons/drug effects , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Solitary Nucleus/cytology , Solitary Nucleus/metabolism
2.
Elife ; 112022 01 26.
Article in English | MEDLINE | ID: mdl-35080494

ABSTRACT

Pain perception is decreased by shifting attentional focus away from a threatening event. This attentional analgesia engages parallel descending control pathways from anterior cingulate (ACC) to locus coeruleus, and ACC to periaqueductal grey (PAG) - rostral ventromedial medulla (RVM), indicating possible roles for noradrenergic or opioidergic neuromodulators. To determine which pathway modulates nociceptive activity in humans, we used simultaneous whole brain-spinal cord pharmacological-fMRI (N = 39) across three sessions. Noxious thermal forearm stimulation generated somatotopic-activation of dorsal horn (DH) whose activity correlated with pain report and mirrored attentional pain modulation. Activity in an adjacent cluster reported the interaction between task and noxious stimulus. Effective connectivity analysis revealed that ACC interacts with PAG and RVM to modulate spinal cord activity. Blocking endogenous opioids with Naltrexone impairs attentional analgesia and disrupts RVM-spinal and ACC-PAG connectivity. Noradrenergic augmentation with Reboxetine did not alter attentional analgesia. Cognitive pain modulation involves opioidergic ACC-PAG-RVM descending control which suppresses spinal nociceptive activity.


Subject(s)
Brain Stem/diagnostic imaging , Brain/diagnostic imaging , Hot Temperature , Magnetic Resonance Imaging/methods , Pain Perception/drug effects , Spinal Cord/diagnostic imaging , Adolescent , Adult , Analgesics, Opioid/administration & dosage , Brain/drug effects , Brain Stem/drug effects , Female , Humans , Male , Middle Aged , Naltrexone/administration & dosage , Pain/drug therapy , Pain Measurement , Reboxetine/administration & dosage , Spinal Cord/drug effects , Young Adult
3.
Environ Sci Pollut Res Int ; 29(1): 936-948, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34345985

ABSTRACT

Fipronil (FIP) is a highly effective insecticide that has been used in agriculture and veterinary medicine. Its neurotoxic effect to insects and to non-target organisms, after nonintentional exposure, was reported. Many studies were conducted to evaluate FIP effects on mammals. However, slight is known about its effect on the brain stem and diencephalon. The current study was designed to investigate the ability of FIP to induce oxidative stress as a molecular mechanism of FIP neurotoxicity that resulted in apoptosis and neural tissue reactivity in these regions. Ten adult male rats received 10 mg/kg of FIP technical grade by oral gavage, daily for 45 days. Brain stem and diencephalon were processed to examine oxidative stress-induced macromolecular alteration (MDA, PCC and DNA fragmentation). Also, the histopathological assessment and immunoreactivity for caspase-3 (active form), iNOS and GFAP were performed on the thalamus, hypothalamus and medulla oblongata. Our results revealed that FIP significantly raised MDA, PCC and DNA fragmentation (p ≤ 0.05). In addition, significantly increased immunoreactivity to GFAP, iNOS and caspase-3 (active form) in the FIP-treated group was noticed (p ≤ 0.05). Moreover, alterations in the histoarchitecture of the neural tissue of these regions were observed. We conclude that FIP can induce oxidative stress, leading to apoptosis and tissue reaction in brain stem and diencephalon.


Subject(s)
Apoptosis , Brain Stem/pathology , Diencephalon/pathology , Oxidative Stress , Pyrazoles/toxicity , Animals , Apoptosis/drug effects , Brain Stem/drug effects , Diencephalon/drug effects , Insecticides/toxicity , Male , Oxidative Stress/drug effects , Rats
4.
J Toxicol Environ Health A ; 85(5): 175-183, 2022 03 04.
Article in English | MEDLINE | ID: mdl-34913848

ABSTRACT

Degenerate neural circuits exhibit "different" circuit properties yet produce similar circuit outcomes (many-to-one) which ensures circuit robustness and complexity. However, neuropathies may hijack degeneracy to yield robust and complex pathological circuits. The aim of the current study was to test the hypothesis that physiochemical exposure to combined jet fuel and noise might induce degeneracy in the brainstem. The auditory brainstem of pigmented rats was used as a model system. The animals were randomized into the following experimental groups: Fuel+Noise, fuel-only, noise-only, and control. Ascending volume conductance from various auditory brainstem regions were evaluated simultaneously with peripheral nervous system (PNS) input to brainstem circuitry. Data demonstrated normal PNS inputs for all groups. However, the Fuel+Noise exposure group produced different caudal brainstem circuit properties while rostral brainstem circuitry initiated outputs that were similar to that of control. This degenerative effect was specific to Fuel+Noise exposure, since neither noise-alone or fuel-alone produced the same result. Degeneracy in the auditory brainstem is consistent with perceptual abnormalities, such as poor speech discrimination (hear but not understand), tinnitus (ringing in the ear), hyperacusis (hypersensitivity to even low-level sound), and loudness intolerance. Therefore, a potential consequence of Fuel+Noise exposure among military and civilian populations may be evidenced as increased rates of super-threshold auditory perceptual abnormalities. This is particularly important because to date, the ototoxic profile of Fuel+Noise exposure has remained unresolved.


Subject(s)
Auditory Perception/drug effects , Brain Stem/drug effects , Hydrocarbons/toxicity , Noise/adverse effects , Animals , Male , Peripheral Nervous System/physiopathology , Rats, Long-Evans
5.
Mol Biol Rep ; 49(2): 1601-1608, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34797492

ABSTRACT

BACKGROUND: Neurosteroids are involved in several important brain functions and have recently been considered novel players in the mechanic actions of neuropsychiatric drugs. There are no reports of murine studies focusing on the effect of chronic neurosteroid treatment in parallel with antipsychotics on key steroidogenic enzyme expression and we therefore focused on steroidogenic enzyme gene expression in the brainstem of rats chronically treated with olanzapine and haloperidol. METHODS AND RESULTS: Studies were carried out on adult, male Sprague-Dawley rats which were divided into 3 groups: control and experimental animals treated with olanzapine or haloperidol. Total mRNA was isolated from homogenized brainstem samples for RealTime-PCR to estimate gene expression of related aromatase, 3ß-HSD and P450scc. Long-term treatment with the selected antipsychotics was reflected in the modulation of steroidogenic enzyme gene expression in the examined brainstem region; with both olanzapine and haloperidol increasing aromatase, 3ß-HSD and P450scc gene expression. CONCLUSIONS: The present findings shed new light on the pharmacology of antipsychotics and suggest the existence of possible regulatory interplay between neuroleptic action and steroidogenesis at the level of brainstem neuronal centres.


Subject(s)
Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacology , Brain Stem/metabolism , Neurosteroids/metabolism , Animals , Brain Stem/chemistry , Brain Stem/drug effects , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Gene Expression/drug effects , Male , Neurons/metabolism , Olanzapine/pharmacology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
6.
Eur Rev Med Pharmacol Sci ; 25(22): 7051-7057, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34859869

ABSTRACT

OBJECTIVE: Complex regional pain syndrome (CRPS) is caused by injuries from fracture after trauma and orthopaedic surgical procedures in the hind limbs. The symptoms of CRPS include warmth, pain, allodynia, and hyperalgesia. It is known that 5-hydroxytryptamine 3 (5-HT3) receptors contribute to hyperalgesia, but their role has not yet been fully elucidated. This study investigated the mechanism of pain relief when a 5-HT3 receptor antagonist was administered in a CRPS animal model. MATERIALS AND METHODS: To establish a CRPS animal model, 10-week-old Sprague-Dawley rats were used in the experiment. On the fourth week post tibial fracture surgery, we performed the von Frey test to measure mechanical allodynia. After performing behavioural tests, we collected blood and tissue samples after sacrificing the animals. Enzyme-linked immunosorbent assay and western blot were also performed. RESULTS: The experimental tibia fracture model-induced CRPS animals elicited increased 5-HT3 receptor expression, and the 5-HT transporter was decreased in the brain stem after 4 weeks of surgical intervention. Additionally, in CRPS-induced animals, both the concentration of substance P and the level of interleukin 6 were increased peripherally and centrally. Treatment with the 5-HT3 receptor antagonist, ramosetron, exerted an analgesic effect in the paw withdrawal test and was dependent on the attenuation of the 5-HT3 receptor population with inflammatory pain mediators. CONCLUSIONS: These data suggest that treatment with the 5-HT3 receptor antagonist, ramosetron, in experimental CRPS animal models alleviated pain-related behaviours and may be a new therapeutic option or potential therapeutic agent for patients with CRPS.


Subject(s)
Analgesics/therapeutic use , Benzimidazoles/therapeutic use , Complex Regional Pain Syndromes/drug therapy , Pain/drug therapy , Serotonin 5-HT3 Receptor Antagonists/therapeutic use , Tibial Fractures/drug therapy , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Complex Regional Pain Syndromes/etiology , Cytokines/metabolism , Disease Models, Animal , Pain/etiology , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT3/metabolism , Substance P/metabolism , Tibial Fractures/complications
7.
Nat Commun ; 12(1): 5962, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34645823

ABSTRACT

Pitt-Hopkins syndrome (PTHS) is a rare autism spectrum-like disorder characterized by intellectual disability, developmental delays, and breathing problems involving episodes of hyperventilation followed by apnea. PTHS is caused by functional haploinsufficiency of the gene encoding transcription factor 4 (Tcf4). Despite the severity of this disease, mechanisms contributing to PTHS behavioral abnormalities are not well understood. Here, we show that a Tcf4 truncation (Tcf4tr/+) mouse model of PTHS exhibits breathing problems similar to PTHS patients. This behavioral deficit is associated with selective loss of putative expiratory parafacial neurons and compromised function of neurons in the retrotrapezoid nucleus that regulate breathing in response to tissue CO2/H+. We also show that central Nav1.8 channels can be targeted pharmacologically to improve respiratory function at the cellular and behavioral levels in Tcf4tr/+ mice, thus establishing Nav1.8 as a high priority target with therapeutic potential in PTHS.


Subject(s)
Haploinsufficiency , Homeodomain Proteins/genetics , Hyperventilation/genetics , Intellectual Disability/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , Neurons/metabolism , Transcription Factor 4/genetics , Transcription Factors/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzimidazoles/pharmacology , Brain Stem/drug effects , Brain Stem/metabolism , Brain Stem/pathology , Carbon Dioxide/metabolism , Carbon Dioxide/pharmacology , Disease Models, Animal , Facies , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , Hyperventilation/drug therapy , Hyperventilation/metabolism , Hyperventilation/pathology , Intellectual Disability/drug therapy , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mice , Mice, Knockout , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neurons/drug effects , Neurons/pathology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Pyrazoles/pharmacology , Respiration/drug effects , Transcription Factor 4/deficiency , Transcription Factors/metabolism
8.
Respir Physiol Neurobiol ; 294: 103743, 2021 12.
Article in English | MEDLINE | ID: mdl-34273553

ABSTRACT

Neuroplasticity is a fundamental property of the respiratory control system, enabling critical adaptations in breathing to meet the challenges, but little is known whether neonates express neuroplasticity similar to adults. We tested the hypothesis that, similar to adults, tyrosine receptor kinase B (TrkB) or adenosine A2a receptor activation in neonates are independently sufficient to elicit respiratory motor facilitation, and that co-induction of TrkB and A2a receptor-dependent plasticity undermines respiratory motor facilitation. TrkB receptor activation with 7,8-dihydroxyflavone (DHF) in neonatal brainstem-spinal cord preparations induced a long-lasting increase in respiratory motor output in 55 % of preparations, whereas adenosine A2a receptor activation with CGS21680 only sporadically induced respiratory motor plasticity. CGS21680 and DHF co-application prevented DHF-dependent respiratory motor facilitation, whereas co-application of MSX-3 (adenosine A2a receptor antagonist) and DHF more rapidly induced respiratory motor plasticity. Collectively, these data suggest that mechanisms underlying respiratory neuroplasticity may be only partially operational in early neonatal life, and that adenosine A2a receptor activation undermines TrkB-induced respiratory plasticity.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Flavones/pharmacology , Neuronal Plasticity/physiology , Receptor, Adenosine A2A/metabolism , Receptor, trkB/agonists , Receptor, trkB/metabolism , Respiratory Physiological Phenomena , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Animals, Newborn , Brain Stem/drug effects , Disease Models, Animal , Neuronal Plasticity/drug effects , Phenethylamines/pharmacology , Rats , Respiratory Physiological Phenomena/drug effects , Spinal Cord/drug effects
9.
Respir Physiol Neurobiol ; 293: 103737, 2021 11.
Article in English | MEDLINE | ID: mdl-34229065

ABSTRACT

Effects of acetylcholine (ACh) on respiratory activity have been an intriguing theme especially in relation to central chemoreception and the control of hypoglossal nerve activity. We studied the effects of ACh on hypoglossal and phrenic (C4) nerve activities and inspiratory and pre-inspiratory neurons in the rostral ventrolateral medulla in brainstem-spinal cord preparations from newborn rats. ACh application increased respiratory rhythm, decreased inspiratory hypoglossal and C4 nerve burst amplitude, and enhanced pre-inspiratory hypoglossal activity. ACh induced membrane depolarization of pre-inspiratory neurons that might be involved in facilitation of respiratory rhythm by ACh. Effects of ACh on hypoglossal and C4 nerve activity were partially reversed by a nicotinic receptor blocker, mecamylamine. Further application of a muscarinic receptor antagonist, oxybutynin, resulted in slight increase of hypoglossal (but not C4) burst amplitude. Thus, ACh induced different effects on hypoglossal and C4 nerve activity in the brainstem-spinal cord preparation.


Subject(s)
Acetylcholine/pharmacology , Brain Stem/drug effects , Hypoglossal Nerve/drug effects , Phrenic Nerve/drug effects , Respiratory Physiological Phenomena/drug effects , Spinal Cord/drug effects , Animals , Animals, Newborn , Chemoreceptor Cells/drug effects , Intralaminar Thalamic Nuclei/drug effects , Motor Neurons/drug effects , Rats , Rats, Wistar
10.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34301891

ABSTRACT

Clinical research into consciousness has long focused on cortical macroscopic networks and their disruption in pathological or pharmacological consciousness perturbation. Despite demonstrating diagnostic utility in disorders of consciousness (DoC) and monitoring anesthetic depth, these cortico-centric approaches have been unable to characterize which neurochemical systems may underpin consciousness alterations. Instead, preclinical experiments have long implicated the dopaminergic ventral tegmental area (VTA) in the brainstem. Despite dopaminergic agonist efficacy in DoC patients equally pointing to dopamine, the VTA has not been studied in human perturbed consciousness. To bridge this translational gap between preclinical subcortical and clinical cortico-centric perspectives, we assessed functional connectivity changes of a histologically characterized VTA using functional MRI recordings of pharmacologically (propofol sedation) and pathologically perturbed consciousness (DoC patients). Both cohorts demonstrated VTA disconnection from the precuneus and posterior cingulate (PCu/PCC), a main default mode network node widely implicated in consciousness. Strikingly, the stronger VTA-PCu/PCC connectivity was, the more the PCu/PCC functional connectome resembled its awake configuration, suggesting a possible neuromodulatory relationship. VTA-PCu/PCC connectivity increased toward healthy control levels only in DoC patients who behaviorally improved at follow-up assessment. To test whether VTA-PCu/PCC connectivity can be affected by a dopaminergic agonist, we demonstrated in a separate set of traumatic brain injury patients without DoC that methylphenidate significantly increased this connectivity. Together, our results characterize an in vivo dopaminergic connectivity deficit common to reversible and chronic consciousness perturbation. This noninvasive assessment of the dopaminergic system bridges preclinical and clinical work, associating dopaminergic VTA function with macroscopic network alterations, thereby elucidating a critical aspect of brainstem-cortical interplay for consciousness.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Stem/pathology , Connectome , Consciousness Disorders/pathology , Dopamine/metabolism , Propofol/pharmacology , Ventral Tegmental Area/pathology , Wakefulness/drug effects , Adolescent , Adult , Aged , Brain Stem/drug effects , Case-Control Studies , Consciousness Disorders/etiology , Consciousness Disorders/metabolism , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Ventral Tegmental Area/drug effects , Young Adult
11.
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
12.
Exp Neurol ; 343: 113760, 2021 09.
Article in English | MEDLINE | ID: mdl-34000248

ABSTRACT

General anesthetic agents are thought to induce loss-of-consciousness (LOC) and enable pain-free surgery by acting on the endogenous brain circuitry responsible for sleep-wake cycling. In clinical use, the entire CNS is exposed to anesthetic molecules with LOC and amnesia usually attributed to synaptic suppression in the cerebral cortex and immobility and analgesia to agent action in the spinal cord and brainstem. This model of patch-wise suppression has been challenged, however, by the observation that all functional components of anesthesia can be induced by focal delivery of minute quantities of GABAergic agonists to the brainstem mesopontine tegmental anesthesia area (MPTA). We compared spectral features of the cortical electroencephalogram (EEG) in rats during systemic anesthesia and anesthesia induced by MPTA microinjection. Systemic administration of (GABAergic) pentobarbital yielded the sustained, δ-band dominant EEG signature familiar in clinical anesthesia. In contrast, anesthesia induced by MPTA microinjection (pentobarbital or muscimol) featured epochs of δ-band EEG alternating with the wake-like EEG, the pattern typical of natural non-rapid-eye-movement (NREM) and REM sleep. The rats were not sleeping, however, as they remained immobile, atonic and unresponsive to noxious pinch. Recalling the paradoxical wake-like quality the EEG during REM sleep, we refer to this state as "paradoxical anesthesia". GABAergic anesthetics appear to co-opt both cortical and spinal components of the sleep network via dedicated axonal pathways driven by MPTA neurons. Direct drug exposure of cortical and spinal neurons is not necessary, and is probably responsible for off-target side-effects of systemic administration including monotonous δ-band EEG, hypothermia and respiratory depression. SIGNIFICANCE STATEMENT: The concept that GABAergic general anesthetic agents induce loss-of-consciousness by substituting for an endogenous neurotransmitter, thereby co-opting neural circuitry responsible for sleep-wake transitions, has gained considerable traction. However, the electroencephalographic (EEG) signatures of sleep and anesthesia differ fundamentally. We show that when the anesthetic state is generated by focal delivery of GABAergics into the mesopontine tegmental anesthesia area (MPTA) the resulting EEG repeatedly transitions between delta-wave-dominant and wake-like patterns much as in REM-NREM sleep. This suggests that systemic (clinical) anesthetic delivery, which indiscriminately floods the entire cerebrum with powerful inhibitory agents, obscures the sleep-like EEG signature associated with the less adulterated form of anesthesia obtained when the drugs are applied selectively to loci where the effective neurotransmitter substitution actually occurs.


Subject(s)
Anesthesia/methods , Brain Stem/drug effects , Electroencephalography/drug effects , GABA Agents/administration & dosage , Microinjections/methods , Sleep Stages/drug effects , Animals , Brain Stem/physiology , Electroencephalography/methods , Female , Male , Rats , Rats, Wistar , Reflex, Righting/drug effects , Reflex, Righting/physiology , Sleep Stages/physiology
13.
Neurosci Lett ; 755: 135904, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33894332

ABSTRACT

Cranial parasympathetic activation produces vasodilation in the head and neck region, but little is known about its central and peripheral mechanisms. This study was conducted to examine whether external and internal carotid-vasodilation origin sites triggered by chemical stimulation are distributed topographically in the parasympathetic brainstems of anesthetized rats, and to examine the effects of peripheral receptors on vasodilation. Microinjection of the neuromodulator candidate l-cysteine revealed that external and internal carotid vasodilation-triggering sites were distributed non-topographically along the full extent of the parasympathetic parvocellular reticular formation (PcRt). Intravenous injection of a muscarinic blocker and a nitric oxide synthase inhibitor abolished external carotid vasodilation, suggesting the peripheral involvement of muscarinic and nitric oxide receptors. Further work is needed to fully understand the PcRt mechanisms underlying timely and appropriate vasodilation to support various cranial functions.


Subject(s)
Brain Stem/physiology , Carotid Artery, External/physiology , Carotid Artery, Internal/physiology , Parasympathetic Nervous System/physiology , Regional Blood Flow/physiology , Vasodilation/physiology , Animals , Brain Stem/blood supply , Brain Stem/drug effects , Carotid Artery, External/drug effects , Carotid Artery, Internal/drug effects , Cysteine/administration & dosage , Male , Microinjections , Parasympathetic Nervous System/drug effects , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Vasodilation/drug effects
14.
Pharmacol Rep ; 73(4): 1188-1194, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33928538

ABSTRACT

BACKGROUND: Phoenixin, spexin and nesfatin-1 belong to a family of newly discovered multifunctional neuropeptides that play regulatory roles in several brain structures and modulate the activity of important neural networks. However, little is known about their expression and action at the level of brainstem. The present work was, therefore, focused on gene expression of the aforementioned peptides in the brainstem of rats chronically treated with olanzapine, a second generation antipsychotic drug. METHODS: Studies were carried out on adult, male Sprague-Dawley rats that were divided into 2 groups: control and experimental animals treated with olanzapine (28-day-long intraperitoneal injection, at dose 5 mg/kg daily). All individuals were killed under anesthesia and the brainstem excised. Total mRNA was isolated from homogenized samples of both structures and the RT-PCR method was used for estimation of related SMIM20/phoenixin, NPQ/spexin and NUCB2/nesfatin-1 gene expression. RESULTS: Long-term treatment with olanzapine is reflected in qualitatively different changes in expression of examined neuropeptides mRNA in the rat brainstem. Olanzapine significantly decreased NPQ/spexin mRNA expression, but increased SMIM20/phoenixin mRNA level in the rat brainstem; while NUCB2/nesfatin-1 mRNA expression remained unchanged. CONCLUSIONS: Olanzapine can affect novel peptidergic signaling in the rat brainstem. This may cautiously suggest the presence of an alternative mode of its action.


Subject(s)
Brain Stem/drug effects , Gene Expression/drug effects , Membrane Proteins/metabolism , Nucleobindins/metabolism , Olanzapine/pharmacology , Peptide Hormones/metabolism , Animals , Antipsychotic Agents/pharmacology , Brain Stem/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
15.
J Cardiovasc Pharmacol ; 77(5): 632-641, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33852527

ABSTRACT

ABSTRACT: Inconsistent reports are available on the role of testosterone in end-organ damage caused by endotoxemia. Here, pharmacologic, surgical, and molecular studies were employed to assess the testosterone modulation of cardiovascular, autonomic, and peripheral and central inflammatory derangements caused by endotoxemia. Studies were performed in conscious male rats preinstrumented with femoral indwelling catheters for the measurement of blood pressure and subjected to castration or pharmacologic interventions that interrupt the biosynthetic cascade of testosterone. Compared with the effects of lipopolysaccharide (10 mg/kg intravenously) in sham operated rats, 2-week castration reduced the lipopolysaccharide-evoked (1) falls in blood pressure, (2) decreases in time- and frequency-domain indices of heart rate variability, (3) shifts in spectral measures of cardiac sympathovagal balance toward parasympathetic dominance, and (4) increases in protein expressions of toll-like receptor-4 and monocyte chemoattractant protein-1 in heart and medullary neurons of the nucleus tractus solitarius and rostral ventrolateral medulla. While the ameliorating actions of castration on endotoxic cardiovascular manifestations were maintained after testosterone replacement, the concomitant inflammatory signals were restored to near-sham levels. The favorable influences of castration on inflammatory and cardiovascular abnormalities of endotoxemia were replicated in intact rats pretreated with degarelix (gonadotropin-releasing hormone receptor blocker) or finasteride (5α-reductase inhibitor) but not formestane (aromatase inhibitor). The data signifies the importance of androgens and its biosynthetic enzymes in cardiovascular and autonomic insults induced by the endotoxic inflammatory response. Clinically, the interruption of testosterone biosynthesis could offer a potential strategy for endotoxemia management.


Subject(s)
Autonomic Nervous System/physiopathology , Brain Stem/physiopathology , Encephalitis/etiology , Endotoxemia/complications , Heart Diseases/etiology , Heart/innervation , Testosterone/blood , 5-alpha Reductase Inhibitors/pharmacology , Androstenedione/analogs & derivatives , Androstenedione/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Autonomic Nervous System/drug effects , Blood Pressure , Brain Stem/drug effects , Brain Stem/metabolism , Disease Models, Animal , Encephalitis/blood , Encephalitis/physiopathology , Encephalitis/prevention & control , Endotoxemia/blood , Endotoxemia/drug therapy , Endotoxemia/physiopathology , Finasteride/pharmacology , Heart Diseases/blood , Heart Diseases/physiopathology , Heart Diseases/prevention & control , Heart Rate , Inflammation Mediators/metabolism , Male , Oligopeptides/pharmacology , Orchiectomy , Rats, Wistar , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/metabolism
16.
Neurotoxicol Teratol ; 87: 106980, 2021.
Article in English | MEDLINE | ID: mdl-33838245

ABSTRACT

Using drugs of abuse while pregnant has tremendous negative consequences for the offspring, including an enhanced risk for substance use disorder (SUD). This vulnerability suggests that gestational exposure to drugs alters the developmental trajectory of neurons important in SUD processes, which could lead to later life changes in responsiveness to motivationally salient stimuli. The laterodorsal tegmentum (LDT) gates the behaviorally relevant firing pattern signaling stimuli saliency in mesoaccumbal circuits. Accordingly, any alterations in LDT functionality could alter output, and play a role in negative outcomes on motivated behavior associated with early-life nicotine exposure. Therefore, we investigated whether prenatal exposure to nicotine (PNE), which is a known teratogen, altered responsiveness of LDT neurons to alcohol by conducting electrophysiology in brain slices. Alcohol induced an outward current in control LDT cells, which was not seen in PNE LDT neurons. The frequency of mEPSCs was significantly decreased by alcohol in LDT PNE cells and accompanied by a decrease in action potential frequency, which were actions not seen in controls. Changes in baseline activity of PNE LDT cells were also observed. In summary, PNE LDT neurons showed alterations in baseline activity and membrane and synaptic responses to postnatal exposures to alcohol. The differences in PNE baseline activity and alcohol responses likely lead to differential output from the LDT to mesoaccumbal targets that could play a role in biasing coding of relevant stimuli, which could participate in the enhanced proclivity for development of SUD in those exposed during gestation to nicotine.


Subject(s)
Brain Stem/drug effects , Brain Stem/growth & development , Neurons/drug effects , Nicotine/pharmacology , Prenatal Exposure Delayed Effects/pathology , Animals , Female , Mice , Nicotinic Agonists/pharmacology , Pregnancy , Signal Transduction/drug effects , Tegmentum Mesencephali/drug effects
17.
Molecules ; 26(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805709

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterised by selective neuronal death in the brain stem and spinal cord. The cause is unknown, but an increasing amount of evidence has firmly certified that neuroinflammation plays a key role in ALS pathogenesis. Neuroinflammation is a pathological hallmark of several neurodegenerative disorders and has been implicated as driver of disease progression. Here, we describe a treatment study demonstrating the therapeutic potential of a tandem version of the well-known all-d-peptide RD2 (RD2RD2) in a transgenic mouse model of ALS (SOD1*G93A). Mice were treated intraperitoneally for four weeks with RD2RD2 vs. placebo. SOD1*G93A mice were tested longitudinally during treatment in various behavioural and motor coordination tests. Brain and spinal cord samples were investigated immunohistochemically for gliosis and neurodegeneration. RD2RD2 treatment in SOD1*G93A mice resulted not only in a reduction of activated astrocytes and microglia in both the brain stem and lumbar spinal cord, but also in a rescue of neurons in the motor cortex. RD2RD2 treatment was able to slow progression of the disease phenotype, especially the motor deficits, to an extent that during the four weeks treatment duration, no significant progression was observed in any of the motor experiments. Based on the presented results, we conclude that RD2RD2 is a potential therapeutic candidate against ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Anti-Inflammatory Agents/therapeutic use , Oligopeptides/therapeutic use , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Anti-Inflammatory Agents/chemistry , Brain Stem/drug effects , Brain Stem/pathology , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Mice, Transgenic , Motor Neurons/drug effects , Motor Neurons/pathology , Motor Skills/drug effects , Motor Skills/physiology , Mutant Proteins/genetics , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Oligopeptides/chemistry , Phenotype , Spinal Cord/drug effects , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1/genetics
18.
Eur J Pharmacol ; 900: 174065, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-33775646

ABSTRACT

Akt (protein kinase B) signaling is frequently activated in diverse cancers. Akt inhibitors such as perifosine and MK-2206 have been evaluated as potential cancer chemotherapeutics. Although both drugs are generally well tolerated, among their most common side-effects vomiting is a major concern. Here we investigated whether these Akt inhibitors evoke emesis in the least shrew model of vomiting. Indeed, both perifosine and MK-2206 induced vomiting with maximal efficacies of 90% at 50 mg/kg (i.p.) and 100% at 10 mg/kg (i.p.), respectively. MK-2206 (10 mg/kg, i.p.) increased c-Fos immunoreactivity both centrally in the shrew brainstem dorsal vagal complex (DVC) emetic nuclei, and peripherally in the jejunum. MK-2206 also evoked phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) in both the DVC emetic nuclei and the enteric nervous system in the jejunum. The ERK1/2 inhibitor U0126 suppressed MK-2206-induced emesis dose-dependently. We then evaluated the suppressive efficacy of diverse antiemetics against MK-2206-evoked vomiting including antagonists/inhibitors of the: L-type Ca2+ channel (nifedipine at 2.5 mg/kg, subcutaneously (s.c.)); glycogen synthase kinase 3 (GSK-3) (AR-A014418 at 10 mg/kg and SB216763 at 0.25 mg/kg, i.p.); 5-hydroxytryptamine 5-HT3 receptor (palonosetron at 0.5 mg/kg, s.c.); substance P neurokinin NK1 receptor (netupitant at 10 mg/kg, i.p.) and dopamine D2/3 receptor (sulpride at 8 mg/kg, s.c.). All tested antagonists/blockers attenuated emetic parameters to varying degrees. In sum, this is the first study to demonstrate how pharmacological inhibition of Akt evokes vomiting via both central and peripheral mechanisms, a process which involves multiple emetic receptors.


Subject(s)
Antiemetics/pharmacology , Central Nervous System/drug effects , Heterocyclic Compounds, 3-Ring , Oncogene Protein v-akt/antagonists & inhibitors , Peripheral Nervous System/drug effects , Shrews/physiology , Vomiting/chemically induced , Vomiting/physiopathology , Animals , Antiemetics/therapeutic use , Brain Stem/drug effects , Dose-Response Relationship, Drug , Emetics/pharmacology , Enteric Nervous System/drug effects , Heterocyclic Compounds, 3-Ring/antagonists & inhibitors , Jejunum/drug effects , MAP Kinase Signaling System/drug effects , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , Vomiting/drug therapy
19.
Sci Rep ; 11(1): 6521, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33753753

ABSTRACT

Drug delivery in diffuse intrinsic pontine glioma is significantly limited by the blood-brain barrier (BBB). Focused ultrasound (FUS), when combined with the administration of microbubbles can effectively open the BBB permitting the entry of drugs across the cerebrovasculature into the brainstem. Given that the utility of FUS in brainstem malignancies remains unknown, the purpose of our study was to determine the safety and feasibility of this technique in a murine pontine glioma model. A syngeneic orthotopic model was developed by stereotactic injection of PDGF-B+PTEN-/-p53-/- murine glioma cells into the pons of B6 mice. A single-element, spherical-segment 1.5 MHz ultrasound transducer driven by a function generator through a power amplifier was used with concurrent intravenous microbubble injection for tumor sonication. Mice were randomly assigned to control, FUS and double-FUS groups. Pulse and respiratory rates were continuously monitored during treatment. BBB opening was confirmed with gadolinium-enhanced MRI and Evans blue. Kondziela inverted screen testing and sequential weight lifting measured motor function before and after sonication. A subset of animals were treated with etoposide following ultrasound. Mice were either sacrificed for tissue analysis or serially monitored for survival with daily weights. FUS successfully caused BBB opening while preserving normal cardiorespiratory and motor function. Furthermore, the degree of intra-tumoral hemorrhage and inflammation on H&E in control and treated mice was similar. There was also no difference in weight loss and survival between the groups (p > 0.05). Lastly, FUS increased intra-tumoral etoposide concentration by more than fivefold. FUS is a safe and feasible technique for repeated BBB opening and etoposide delivery in a preclinical pontine glioma model.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Stem Neoplasms/drug therapy , Drug Delivery Systems , Glioma/drug therapy , Animals , Biological Transport/drug effects , Brain Stem/diagnostic imaging , Brain Stem/drug effects , Brain Stem Neoplasms/diagnostic imaging , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Disease Models, Animal , Etoposide/pharmacology , Evans Blue/pharmacology , Gadolinium/pharmacology , Glioma/diagnostic imaging , Glioma/genetics , Glioma/pathology , Humans , Magnetic Resonance Imaging , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/pharmacology , Pons/diagnostic imaging , Pons/drug effects , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/pharmacology , Ultrasonography
20.
Microcirculation ; 28(4): e12686, 2021 05.
Article in English | MEDLINE | ID: mdl-33595915

ABSTRACT

Methyl palmitate (MP) is a fatty acid methyl ester. Our recent study indicated that adrenergic nerve-dependent functional sympathetic-sensory nerve interactions were abolished by MP in mesenteric arteries. However, the effect of MP on perivascular nerves and cerebral blood flow remains unclear. In this study, the increase in basilar arterial blood flow (BABF) after the topical application of nicotinic acetylcholine receptor agonists was measured using laser Doppler flowmetry in anesthetized rats. The choline (a selective α7-nicotinic acetylcholine receptor agonist)-induced increase in BABF was abolished by tetrodotoxin (a neurotoxin), NG -nitro-L-arginine (a nonselective NO synthase inhibitor), α-bungarotoxin (a selective α7-nicotinic acetylcholine receptor inhibitor), and chronic sympathetic denervation. In addition, the nicotine (a nicotinic acetylcholine receptor agonist)-induced increase in BABF was inhibited by MP in a concentration-dependent manner. The acetylcholine-induced increase in BABF was not affected by MP. The myography results revealed that nicotine-induced vasorelaxation was significantly inhibited by MP, but was reversed by chelerythrine (a protein kinase C inhibitor). MP-induced vasodilation was significantly greater in BA rings without endothelium compared to those with endothelium. Meanwhile, MP did not affect baseline BABF. Our results indicate that MP acts as a neuromodulator in the cerebral circulation where it activates the PKC pathway and causes a diminished nicotine-induced increase in blood flow in the brainstem, and that the vasorelaxation effect of MP may play a minor role.


Subject(s)
Basilar Artery , Brain Stem , Neurotransmitter Agents , Nicotine , Palmitates , Receptors, Nicotinic , Animals , Basilar Artery/diagnostic imaging , Basilar Artery/drug effects , Basilar Artery/physiology , Brain Stem/blood supply , Brain Stem/diagnostic imaging , Brain Stem/drug effects , Laser-Doppler Flowmetry , Male , Neurotransmitter Agents/pharmacology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Palmitates/pharmacology , Rats , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/physiology , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...