Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Drug Des Devel Ther ; 18: 2837-2845, 2024.
Article in English | MEDLINE | ID: mdl-39006192

ABSTRACT

Background: To investigate the effects of nalbuphine on emergency agitation (EA), which affects up to 80% of the children following otolaryngology procedures, in children undergoing cochlear implantation. Methods: A prospective double-blinded randomized controlled clinical trial was conducted between November 2020 and October 2022. Eligible children, aged 6 months to 3 years old, were randomly assigned to either 0.1 mg/kg, 0.15 mg/kg, 0.2 mg/kg nalbuphine or 0.9% saline groups. EA was defined by the Pediatric Anesthesia Emergence Delirium (PAED) score ≥10. Extubation time, post-anesthesia care unit (PACU) length of stay, severe EA (PAED ≥ 15), peak PAED score, the Faces, Legs, Activity, Cry, and Consolability (FLACC) scale, Ramsay sedation score, and adverse events were also recorded. Results: A total of 104 children were enrolled, with 26 children in each group. Nalbuphine significantly reduced the EA occurrence from 73.1% in the saline group to 38.5%, 30.8%, and 26.9% in the 0.1 mg/kg, 0.15 mg/kg, and 0.2 mg/kg nalbuphine groups, respectively (P < 0.001), without affecting the extubation time and PACU length of stay. More children (34.6%) in the 0.9% saline group experienced severe EA. Higher dose nalbuphine (0.15 mg/kg, 0.2 mg/kg) showed lower peak PAED score, better analgesia and sedation effect compared with 0.1 mg/kg nalbuphine and saline groups. However, 0.2mg/kg nalbuphine caused undesired over-sedation in two (7.7%) children. No other adverse events were reported. Conclusion: Young children undergoing cochlear implantation surgery were at a high risk of EA and postoperative pain, while 0.2 mg/kg nalbuphine might be an ideal candidate for EA and pain prevention when used under close monitoring. Trial Registration: ChiCTR2000040407.


Subject(s)
Analgesics, Opioid , Cochlear Implantation , Emergence Delirium , Nalbuphine , Humans , Nalbuphine/administration & dosage , Nalbuphine/therapeutic use , Child, Preschool , Male , Double-Blind Method , Female , Prospective Studies , Infant , Emergence Delirium/prevention & control , Emergence Delirium/drug therapy , Cochlear Implantation/adverse effects , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Dose-Response Relationship, Drug , Psychomotor Agitation/drug therapy , Psychomotor Agitation/prevention & control
2.
Brain Res ; 1829: 148798, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38403038

ABSTRACT

Menopausal women experience neuropathic pain 63% more frequently than men do, which may attribute to the estrogen withdrawal. However, the underlying mechanisms remain unclear. Here, the role of estrogen receptors (ERs) in ovariectomized (OVX) female mice following chronic constriction injury (CCI) was investigated. With 17ß-estradiol (E2) supplemented, aggravated mechanical allodynia in OVX mice could be significantly alleviated, particularly after intra-anterior cingulate cortex (ACC) E2 delivery. Pharmacological interventions further demonstrated that the agonist of G-protein-coupled estrogen receptor 30 (GPR30), rather than ERα or ERß in the ACC, exhibited the similar analgesic effect as E2, whereas antagonist of GPR30 exacerbated allodynia. Furthermore, OVX surgery reduced GPR30 expression in the ACC, which could be restored with estrogen supplementation. Selective downregulation of GPR30 in the ACC of naïve female mice induces mechanical allodynia, whereas GPR30 overexpression in the ACC remarkedly alleviated OVX-exacerbated allodynia. Collectively, estrogen withdrawal could downregulate the ACC GPR30 expression, resulting in exacerbated neuropathic pain. Our findings highlight the importance of GPR30 in the ACC in aggravated neuropathic pain during menopause, and offer a potential therapeutic candidate for neuropathic pain management in menopausal women.


Subject(s)
Hyperalgesia , Neuralgia , Animals , Female , Humans , Male , Mice , Estradiol/pharmacology , Estrogens/pharmacology , Estrogens/metabolism , Gyrus Cinguli/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
CNS Neurosci Ther ; 29(5): 1332-1344, 2023 05.
Article in English | MEDLINE | ID: mdl-36740262

ABSTRACT

AIMS: The circuitry mechanism associated with anesthesia-induced unconsciousness is still largely unknown. It has been reported that orexinergic neurons of the lateral hypothalamus (LHA) facilitate the emergence from anesthesia through their neuronal projections to the arousal-promoting brain areas. However, the lateral habenula (LHb), as one of the orexin downstream targets, is known for its anesthesia-promoting effect. Therefore, the current study aimed to explore whether and how the orexinergic projections from the LHA to the LHb have a regulatory effect on unconsciousness induced by general anesthesia. METHODS: We applied optogenetic, chemogenetic, or pharmacological approaches to regulate the orexinergicLHA-LHb pathway. Fiber photometry was used to assess neuronal activity. Loss or recovery of the righting reflex was used to evaluate the induction or emergence time of general anesthesia. The burst-suppression ratio and electroencephalography spectra were used to measure the anesthetic depth. RESULTS: We found that activation of the orexinergicLHA-LHb pathway promoted emergence and reduced anesthetic depth during sevoflurane anesthesia. Surprisingly, the arousal-promoting effect of the orexinergicLHA-LHb pathway was mediated by excitation of glutamate decarboxylase (GAD2)-expressing neurons, but not glutamatergic neurons in the LHb. CONCLUSION: The orexinergicLHA-LHb pathway facilitates emergence from sevoflurane anesthesia, and this effect was mediated by OxR2 in GAD2-expressing GABA neurons.


Subject(s)
Anesthetics, Inhalation , Habenula , Humans , Sevoflurane/pharmacology , Habenula/metabolism , GABAergic Neurons , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/metabolism , Anesthesia, General , Unconsciousness/metabolism
4.
Brain Behav ; 13(1): e2847, 2023 01.
Article in English | MEDLINE | ID: mdl-36495119

ABSTRACT

BACKGROUND: Stroke is the major cause of adult neurocognitive disorders (NCDs), and presents a significant burden on both of the families and society. To improve the cerebral injury, we generated a blood-brain barrier penetrating peptide TAT-LBD-Ngn2, in which Ngn2 (Neurogenin2) is a classical preneural gene that enhances neurogenesis, and neural precursor cells survival and differentiation. We previously demonstrated that it has a short-term protective effect against cerebral ischemia-reperfusion injury. However, it is uncertain if TAT-LBD-Ngn2 could promote neurogenesis to exhibit long-term therapeutic impact. METHODS AND RESULTS: In present study, TAT-LBD-Ngn2 was administered for 14 or 28 days following bilateral common carotid arteries occlusion (BCCAO). After confirming that TAT-LBD-Ngn2 could cross the brain blood barrier and aggregate in the hippocampus, we conducted open field test, Morris water maze and contextual fear conditioning to examine the long-term effect of TAT-LBD-Ngn2 on cognition. We discovered that TAT-LBD-Ngn2 significantly improved the spatial and contextual learning and memory on both days 14 and 28 after BCCAO, while TAT-LBD-Ngn2 exhibited anxiolytic effect only on day 14, but had no effect on locomotion. Using western blot and immunofluorescence, TAT-LBD-Ngn2 was also shown to promote neurogenesis, as evidenced by increased BrdU+ and DCX+ neurons in dentate gyrus. Meanwhile, TAT-LBD-Ngn2 elevated the expression of brain derived neurotrophic factor rather than nerve growth factor compared to the control group. CONCLUSIONS: Our findings revealed that TAT-LBD-Ngn2 could dramatically promote learning and memory in long term by facilitating neurogenesis in the hippocampus after global cerebral ischemia, indicating that TAT-LBD-Ngn2 may be an appealing candidate for treating poststroke NCD.


Subject(s)
Brain Ischemia , Neural Stem Cells , Humans , Brain Ischemia/drug therapy , Neurogenesis/physiology , Hippocampus , Cognition/physiology , Cerebral Infarction
5.
Cereb Cortex ; 33(6): 3026-3042, 2023 03 10.
Article in English | MEDLINE | ID: mdl-35764255

ABSTRACT

Ventromedial prefrontal cortex (vmPFC) processes many critical brain functions, such as decision-making, value-coding, thinking, and emotional arousal/recognition, but whether vmPFC plays a role in sleep-wake promotion circuitry is still unclear. Here, we find that photoactivation of dorsomedial hypothalamus (DMH)-projecting vmPFC neurons, their terminals, or their postsynaptic DMH neurons rapidly switches non-rapid eye movement (NREM) but not rapid eye movement sleep to wakefulness, which is blocked by photoinhibition of DMH outputs in lateral hypothalamus (LHs). Chemoactivation of DMH glutamatergic but not GABAergic neurons innervated by vmPFC promotes wakefulness and suppresses NREM sleep, whereas chemoinhibition of vmPFC projections in DMH produces opposite effects. DMH-projecting vmPFC neurons are inhibited during NREM sleep and activated during wakefulness. Thus, vmPFC neurons innervating DMH likely represent the first identified set of cerebral cortical neurons for promotion of physiological wakefulness and suppression of NREM sleep.


Subject(s)
Sleep, REM , Sleep , Sleep/physiology , Sleep, REM/physiology , Arousal , Wakefulness/physiology , GABAergic Neurons/physiology
6.
Biochem Biophys Res Commun ; 640: 12-20, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36495605

ABSTRACT

The general anesthesia associated with long-term cognitive impairment has been causing the concern of the whole society. In particular, repeated anesthetic exposures may affect executive function, processing speed, and fine motor skills, which all directly depended on the functions of oligodendrocytes, myelin, and axons. However, the underlying mechanisms are still largely unknown. To investigate the spatial and temporal alterations in oligodendrocytes in the corpus callosum (CC) and hippocampus following repeated sevoflurane exposures (3%, for 2 h) from postnatal day 6 (P6) to P8, we used immunofluorescence, Western blot, and a battery of behavioral tests. As previously stated, we confirmed that early anesthetic exposures hampered both cognitive and motor performance during puberty in the rotarod and banes tests. Intriguingly, we discovered that the proliferation of oligodendrocyte progenitor cells (OPCs) was immediately enhanced after general anesthesia in the CC and hippocampus from P8 to P32. From P8 through P15, the overall oligodendrocyte population remained constant. However, along with the structural myelin abnormalities, the matured oligodendrocytes statistically reduced in the CC (from P15) and hippocampus (from P32). Administration of clemastine, which could induce OPC differentiation and myelin formation, significantly increased matured oligodendrocytes and promoted myelination and cognition. Collectively, we first demonstrated the bi-directional influence of early sevoflurane exposures on oligodendrocyte maturation and proliferation, which contributes to the cognitive impairment induced by general anesthesia. These findings illustrated the dynamic changes in oligodendrocytes in the developing brain following anesthetic exposures, as well as possible therapeutic strategies for multiple general anesthesia associated cognitive impairment.


Subject(s)
Oligodendroglia , Sexual Maturation , Animals , Mice , Sevoflurane/adverse effects , Animals, Newborn , Myelin Sheath
7.
Front Aging Neurosci ; 14: 1036428, 2022.
Article in English | MEDLINE | ID: mdl-36533179

ABSTRACT

Background: Post-operative delirium (POD), a common post-operative complication that affects up to 73. 5% of surgical patients, could prolong hospital stays, triple mortality rates, cause long-term cognitive decline and dementia, and boost medical expenses. However, the underlying mechanisms, especially the circuit mechanisms of POD remain largely unclear. Previous studies demonstrated that cannabis use might cause delirium-like behavior through the endocannabinoid system (eCBs), a widely distributed retrograde presynaptic neuromodulator system. We also found that the prelimbic (PrL) and intralimbic (IL) prefrontal cortex, a crucial hub for cognition and emotion, was involved in the eCBs-associated general anesthesia recovery. Objectives: The present study aimed to investigate the role of eCBs in POD development, and further clarify its neuronal specificity and circuit specificity attributed to POD. Methods: According to a previous study, 2 h of 1.4% isoflurane anesthesia and simple laparotomy were conducted to establish the POD model in C57/BL6 mice aged 8-12 weeks. A battery of behavioral tests, including the buried food, open field, and Y maze tests, were performed at 24 h before anesthesia and surgery (AS) and 6 and 9 h after AS. The behavioral results were calculated as a composite Z score for the POD assessment. To explore the dynamics of eCBs and their effect on POD regulation, an endocannabinoid (eCB) sensor was microinjected into the PrL, and the antagonists (AM281 and hemopressin) and agonist (nabilone) of type 1 cannabinoid receptor (CB1R), were administered systemically or locally (into PrL). Chemogenetics, combined Cre-loxP and Flp-FRT system, were employed in mutant mice for neuronal specificity and circuit specificity observation. Results: After AS, the composite Z score significantly increased at 6 and 9 but not at 24 h, whereas blockade of CB1R systemically and intra-PrL could specifically decrease the composite Z score at 6 and 9 h after AS. Results of fiber photometry further confirmed that the activity of eCB in the PrL was enhanced by AS, especially in the Y maze test at 6 h post-operatively. Moreover, the activation of glutamatergic neurons in the PrL could reduce the composite Z score, which could be significantly reversed by exogenous cannabinoid (nabilone) at 6 and 9 h post-operatively. However, activation of GABAergic neurons only decreased composite Z score at 9 h post-operatively, with no response to nabilone application. Further study revealed the glutamatergic projection from mediodorsal thalamus (MD) to PrL glutamatergic neurons, but not hippocampus (HIP)-PrL circuit, was in charge of the effect of eCBs on POD. Conclusion: Our study firstly demonstrated the involvement of eCBs in the POD pathogenesis and further revealed that the eCBs may regulate POD through the specific MDglu-PrLglu circuit. These findings not only partly revealed the molecular and circuit mechanisms of POD, but also provided an applicable candidate for the clinical prevention and treatment of POD.

8.
Anesth Analg ; 133(3): 781-793, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34403389

ABSTRACT

BACKGROUND: Orexin, a neuropeptide derived from the perifornical area of the hypothalamus (PeFLH), promotes the recovery of propofol, isoflurane, and sevoflurane anesthesias, without influencing the induction time. However, whether the orexinergic system also plays a similar role in desflurane anesthesia, which is widely applied in clinical practice owing to its most rapid onset and offset time among all volatile anesthetics, has not yet been studied. In the present study, we explored the effect of the orexinergic system on the consciousness state induced by desflurane anesthesia. METHODS: The c-Fos staining was used to observe the activity changes of orexinergic neurons in the PeFLH and their efferent projection regions under desflurane anesthesia. Chemogenetic and optogenetic techniques were applied to compare the effect of PeFLH orexinergic neurons on the induction, emergence, and maintenance states between desflurane and isoflurane anesthesias. Orexinergic terminals in the paraventricular thalamic nucleus (PVT) were manipulated with pharmacologic, chemogenetic, and optogenetic techniques to assess the effect of orexinergic circuitry on desflurane anesthesia. RESULTS: Desflurane anesthesia inhibited the activity of orexinergic neurons in the PeFLH, as well as the neuronal activity in PVT, basal forebrain, dorsal raphe nucleus, and ventral tegmental area, as demonstrated by c-Fos staining. Activation of PeFLH orexinergic neurons prolonged the induction time and accelerated emergence from desflurane anesthesia but only influenced the emergence in isoflurane anesthesia, as demonstrated by chemogenetic and pharmacologic techniques. Meanwhile, optical activation of orexinergic neurons exhibited a long-lasting inhibitory effect on burst-suppression ratio (BSR) under desflurane anesthesia, and the effect may be contributed by the orexinergic PeFLH-PVT circuitry. The orexin-2 receptor (OX2R), but not orexin-1 receptor (OX1R), in the PVT, which had been inhibited most significantly by desflurane, mediated the proemergence effect of desflurane anesthesia. CONCLUSIONS: We discovered, for the first time, that orexinergic neurons in the PeFLH could not only influence the maintenance and emergence from isoflurane and desflurane anesthesias but also affect the induction under desflurane anesthesia. Furthermore, this specific effect is probably mediated by orexinergic PeFLH-PVT circuitry, especially OX2Rs in the PVT.


Subject(s)
Anesthesia Recovery Period , Anesthesia, Inhalation , Anesthetics, Inhalation/pharmacology , Consciousness/drug effects , Desflurane/pharmacology , Isoflurane/pharmacology , Midline Thalamic Nuclei/drug effects , Neurons/drug effects , Orexins/pharmacology , Action Potentials , Animals , Electroencephalography , Male , Midline Thalamic Nuclei/metabolism , Neurons/metabolism , Optogenetics , Orexin Receptors/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Time Factors
9.
Neurosci Bull ; 37(7): 934-946, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33847915

ABSTRACT

The lateral hypothalamic area (LHA) plays a pivotal role in regulating consciousness transition, in which orexinergic neurons, GABAergic neurons, and melanin-concentrating hormone neurons are involved. Glutamatergic neurons have a large population in the LHA, but their anesthesia-related effect has not been explored. Here, we found that genetic ablation of LHA glutamatergic neurons shortened the induction time and prolonged the recovery time of isoflurane anesthesia in mice. In contrast, chemogenetic activation of LHA glutamatergic neurons increased the time to anesthesia and decreased the time to recovery. Optogenetic activation of LHA glutamatergic neurons during the maintenance of anesthesia reduced the burst suppression pattern of the electroencephalogram (EEG) and shifted EEG features to an arousal pattern. Photostimulation of LHA glutamatergic projections to the lateral habenula (LHb) also facilitated the emergence from anesthesia and the transition of anesthesia depth to a lighter level. Collectively, LHA glutamatergic neurons and their projections to the LHb regulate anesthetic potency and EEG features.


Subject(s)
Anesthetics , Habenula , Isoflurane , Animals , GABAergic Neurons , Hypothalamic Area, Lateral , Isoflurane/pharmacology , Mice
10.
Brain Res ; 1757: 147336, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33548269

ABSTRACT

Rosmarinic acid (RA), a natural polyphenol, possesses potent antioxidant and anti-inflammatory activities. To evaluate the ability of RA to cure ischemic stroke and post-stroke depression (PSD), rats were treated with various doses of RA after cerebral ischemia. Neurological deficits and infarct volume of the brain were measured. The activities of superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) were examined at different time points. In addition, a forced swimming test and sucrose preference test were performed to detect the anti-depressive effects of RA. Our results revealed RA administration significantly alleviated neurological deficits and reduced infarct volumes. RA attenuated the decrease of SOD, CAT activities and GSH levels in the ischemic penumbra of the brain. Most importantly, RA treatment alleviated the depression behaviors. Increased expression of Nrf2 was also induced by RA, while down regulation Nrf2 by Nrf2-short-hairpin RNA sequences reversed the increasing activity of SOD and CAT induced by RA, as well as the protection against PSD. The present study indicates that RA exerts a potent neuroprotective effect against stroke and PSD, which could be a promising therapeutic intervention for stroke.


Subject(s)
Antioxidants/pharmacology , Brain Ischemia/drug therapy , Cinnamates/pharmacology , Depression/drug therapy , Depsides/pharmacology , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Ischemia/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Male , Neuroprotective Agents/pharmacology , Rats, Sprague-Dawley , Rosmarinic Acid
11.
Front Neural Circuits ; 13: 73, 2019.
Article in English | MEDLINE | ID: mdl-31798420

ABSTRACT

The ventral tegmental area (VTA) reportedly regulates sleep and wakefulness through communication with the lateral hypothalamus (LH). It has also been suggested that adequate anesthesia produced by administration of chloral hydrate, ketamine, or halothane significantly reduces the GABAergic neuronal firing rate within the VTA. However, the exact effects on GABAergic neurons in the VTA and the mechanisms through which these neurons modulate anesthesia through associated neural circuits is still unclear. Here, we used optogenetic and chemogenetic methods to specifically activate or inhibit GABAergic neuronal perikarya in the VTA or their projections to the LH in Vgat-Cre mice. Electroencephalogram (EEG) spectral analyses and burst suppression ratio (BSR) calculations were conducted following administration of 0.8 or 1.0% isoflurane, respectively; and loss of righting reflex (LORR), recovery of righting reflex (RORR), and anesthesia sensitivity were assessed under 1.4% isoflurane anesthesia. The results showed that activation of GABAergic neurons in the VTA increased delta wave power from 40.0 to 46.4% (P = 0.006) and decreased gamma wave power from 15.2 to 11.5% (P = 0.017) during anesthesia maintenance. BSR was increased from 51.8 to 68.3% (P = 0.017). Induction time (LORR) was reduced from 333 to 290 s (P = 0.019), whereas arousal time (RORR) was prolonged from 498 to 661 s (P = 0.007). Conversely, inhibition of VTA GABAergic neurons led to opposite effects. In contrast, optical activation of VTA-LH GABAergic projection neurons increased power of slow delta waves from 44.2 to 48.8% (P = 0.014) and decreased that of gamma oscillations from 10.2 to 8.0%. BSR was increased from 39.9 to 60.2% (P = 0.0002). LORR was reduced from 330 to 232 s (P = 0.002), and RORR increased from 396 to 565 s (P = 0.007). Optical inhibition of the projection neurons caused opposite effects in terms of both the EEG spectrum and the BSR, except that inhibition of this projection did not accelerate arousal time. These results indicate that VTA GABAergic neurons could facilitate the anesthetic effects of isoflurane during induction and maintenance while postponing anesthetic recovery, at least partially, through modulation of their projections to the LH.


Subject(s)
Anesthesia, General , Anesthetics, Inhalation/administration & dosage , GABAergic Neurons/physiology , Hypothalamic Area, Lateral/physiology , Isoflurane/administration & dosage , Ventral Tegmental Area/physiology , Animals , Electroencephalography , GABAergic Neurons/drug effects , Hypothalamic Area, Lateral/drug effects , Mice , Neural Pathways/drug effects , Neural Pathways/physiology , Optogenetics , Ventral Tegmental Area/drug effects
12.
J Clin Invest ; 127(6): 2295-2309, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28463228

ABSTRACT

Consciousness can be defined by two major attributes: awareness of environment and self, and arousal, which reflects the level of awareness. The return of arousal after general anesthesia presents an experimental tool for probing the neural mechanisms that control consciousness. Here we have identified that systemic or intracerebral injection of the cannabinoid CB1 receptor (CB1R) antagonist AM281 into the dorsomedial nucleus of the hypothalamus (DMH) - but not the adjacent perifornical area (Pef) or the ventrolateral preoptic nucleus of the hypothalamus (VLPO) - accelerates arousal in mice recovering from general anesthesia. Anesthetics selectively activated endocannabinoid (eCB) signaling at DMH glutamatergic but not GABAergic synapses, leading to suppression of both glutamatergic DMH-Pef and GABAergic DMH-VLPO projections. Deletion of CB1R from widespread cerebral cortical or prefrontal cortical (PFC) glutamatergic neurons, including those innervating the DMH, mimicked the arousal-accelerating effects of AM281. In contrast, CB1R deletion from brain GABAergic neurons or hypothalamic glutamatergic neurons did not affect recovery time from anesthesia. Inactivation of PFC-DMH, DMH-VLPO, or DMH-Pef projections blocked AM281-accelerated arousal, whereas activation of these projections mimicked the effects of AM281. We propose that decreased eCB signaling at glutamatergic terminals of the PFC-DMH projection accelerates arousal from general anesthesia through enhancement of the excitatory DMH-Pef projection, the inhibitory DMH-VLPO projection, or both.


Subject(s)
Endocannabinoids/physiology , Hypothalamus/physiology , Receptor, Cannabinoid, CB1/metabolism , Synaptic Transmission , Anesthesia, General , Animals , Arousal , GABAergic Neurons/physiology , Hypothalamus/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic , Morpholines/pharmacology , Nerve Net/drug effects , Nerve Net/physiology , Pyrazoles/pharmacology , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors
13.
Mol Neurobiol ; 54(2): 1240-1253, 2017 03.
Article in English | MEDLINE | ID: mdl-26820679

ABSTRACT

Arachidonyl-2-chloroethylamide (ACEA), a highly selective agonist of cannabinoid receptor 1 (CB1R), has been reported to protect neurons in ischemic injury. We sought to investigate whether mitochondrial biogenesis was involved in the therapeutic effect of ACEA in cerebral ischemia. Focal cerebral ischemic injury was induced in adult male Sprague Dawley rats. Intraperitoneal injection of 1 mg/kg ACEA improved neurological behavior, reduced infarct volume, and inhibited apoptosis. The volume and numbers of mitochondria were significantly increased after ACEA administration. Expression of mitochondrial transcription factor A (Tfam), nuclear transcription factor-1 (Nrf-1), and cytochrome C oxidase subunit IV (COX IV) were also significantly up-regulated in animals administered ACEA. One thousand nanomoles of ACEA inhibited mitochondrial dysfunction in primary rat cortical neurons exposed to oxygen-glucose deprivation (OGD). Furthermore, ACEA administration increased phosphorylation of glycogen synthase kinase-3ß (GSK-3ß) after reperfusion. Phosphorylation of GSK-3ß induced mitochondrial biogenesis and preserved mitochondrial function whereas inhibition of phosphatidylinositol 3-kinase (PI3K) dampened phosphorylation of GSK-3ß and reversed induction of mitochondrial biogenesis and function following ACEA administration. In conclusion, ACEA could induce mitochondrial biogenesis and improve mitochondrial function at the beginning of cerebral ischemia, thus alleviating cerebral ischemia injury. Phosphorylation of GSK-3ß might be involved in the regulation of mitochondrial biogenesis induced by ACEA.


Subject(s)
Arachidonic Acids/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Mitochondria/metabolism , Organelle Biogenesis , Animals , Arachidonic Acids/pharmacology , Brain Ischemia/pathology , Male , Mitochondria/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists
14.
J Drug Target ; 24(1): 13-23, 2016.
Article in English | MEDLINE | ID: mdl-26061295

ABSTRACT

Therapeutics targeting the Nogo-A signal pathway hold promise to promote recovery following brain injury. Based on the temporal characteristics of Nogo-A expression in the process of cerebral ischemia and reperfusion, we tested a novel asynchronous treatment, in which TAT-M9 was used in the early stage to decrease neuronal loss, and TAT-NEP1-40 was used in the delayed stage to promote neurite outgrowth after bilateral common carotid artery occlusion (BCCAO) in mice. Both TAT-M9 and TAT-NEP1-40 were efficiently delivered into the brains of mice by intraperitoneal injection. TAT-M9 treatment promoted neuron survival and inhibited neuronal apoptosis. Asynchronous therapy with TAT-M9 and TAT-NEP1-40 increased the expression of Tau, GAP43 and MAP-2 proteins, and enhanced short-term and long-term cognitive functions. In conclusion, the asynchronous treatment had a long-term neuroprotective effect, which reduced neurologic injury and apoptosis, promoted neurite outgrowth and enhanced functional recovery after ischemia. It suggests that this asynchronous treatment could be a promising therapy for cerebral ischemia in humans.


Subject(s)
Brain Ischemia/physiopathology , Myelin Proteins/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Animals , Apoptosis/drug effects , Behavior Rating Scale , Cell Survival/drug effects , Disease Models, Animal , Drug Administration Schedule , GAP-43 Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Myelin Proteins/administration & dosage , Myelin Proteins/pharmacology , Neurites/drug effects , Nogo Proteins , Peptide Fragments/administration & dosage , Random Allocation , Reperfusion Injury/physiopathology , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
15.
Sci Rep ; 5: 17310, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26611106

ABSTRACT

Recent studies have demonstrated that adiponectin (APN) attenuates cerebral ischemic/reperfusion via globular adiponectin (gAD). However, the therapeutic role of gAD in cerebral ischemic injury in type 1 diabetes mellitus (T1DM) remains unclear. Our results showed that gAD improved neurological scores and reduced the infarct volumes in the 8-week T1DM (T1DM-8W) mice, but not in the 2-week T1DM (T1DM-2W) mice. Moreover, the ischemic penumbra APN levels increased and peaked in T1DM-2W mice, and reduced to normal in T1DM-8W mice, while the APN receptor 1 (AdipoR1) expression change was the opposite. Administration of rosiglitazone in T1DM-2W mice up-regulated the expression of AdipoR1 and restored the neuroprotection of gAD, while intracerebroventricular injection of AdipoR1 small interfering RNA (siRNA) in T1DM-8W mice reversed it. Furthermore, the expression of p-PERK, p-IRE1 and GRP78 were increased whereas the expressions of CHOP and cleaved caspase-12 as well as the number of apoptotic neurons were decreased after gAD treatment in T1DM-8W mice. These beneficial effects of gAD were reversed by pretreatment with AdipoR1 siRNA. These results demonstrated a dynamic dysfunction of APN/AdipoR1 accompanying T1DM progression. Interventions bolstering AdipoR1 expression during early stages and gAD supplementation during advanced stages may potentially reduce the cerebral ischemic injury in diabetic patients.


Subject(s)
Adiponectin/pharmacology , Brain Ischemia/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Receptors, Adiponectin/genetics , Reperfusion Injury/drug therapy , Adiponectin/genetics , Adiponectin/metabolism , Animals , Apoptosis/drug effects , Brain Ischemia/complications , Brain Ischemia/genetics , Brain Ischemia/metabolism , Caspase 12/genetics , Caspase 12/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Hypoglycemic Agents/pharmacology , Injections, Intraventricular , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Adiponectin/agonists , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/metabolism , Reperfusion Injury/complications , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Rosiglitazone , Signal Transduction , Thiazolidinediones/pharmacology , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
16.
Brain Res ; 1573: 84-91, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-24854123

ABSTRACT

We investigated the protective effect of electroacupuncture (EA) on cerebral ischemic injury in diabetic mice, and explored the role of NADPH oxidase-mediated oxidative stress. Male C57BL/6 mice were injected streptozotocin to induce diabetes. The mice were pretreated with EA at acupoint "Baihui" for 30 min. Two hours after the end of EA pretreatment, focal cerebral ischemia was induced following 24h reperfusion. The neurobehavioral scores and infarction volumes, malondialdehyde (MDA), reactive oxygen species (ROS), and activation of NADPH oxidase were determined in the presence or absence of the NADPH oxidase inhibitor apocynin or activator tetrabromocinnamic acid (TBCA). EA pretreatment reduced infarct size and improved neurological outcomes 24h after reperfusion in the diabetic mice. EA also decreased cerebral MDA and ROS levels compared with the control group, and inhibited the NADPH oxidase activation. The beneficial effects were abolished by TBCA while pretreatment with apocynin mimicked the neuroprotective and anti-oxidative effects of EA. Our results demonstrated that EA attenuated cerebral ischemic injury by inhibiting NAPDH oxidase-mediated oxidative damage in diabetic mice. These results suggest a novel mechanism of EA pretreatment-induced tolerance in diabetic cerebral ischemia.


Subject(s)
Brain Ischemia/therapy , Diabetes Mellitus, Experimental/complications , Electroacupuncture/methods , NADPH Oxidases/metabolism , Oxidative Stress/physiology , Acetophenones/pharmacology , Animals , Brain/drug effects , Brain/enzymology , Brain/pathology , Brain Ischemia/complications , Brain Ischemia/enzymology , Brain Ischemia/pathology , Cinnamates/pharmacology , Enzyme Activators/pharmacology , Male , Malondialdehyde/metabolism , Mice, Inbred C57BL , NADPH Oxidases/antagonists & inhibitors , Oxidative Stress/drug effects , Random Allocation , Reactive Oxygen Species/metabolism , Reperfusion Injury/complications , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control
17.
Mol Neurobiol ; 50(2): 305-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24390566

ABSTRACT

Electroacupuncture (EA) has demonstrated therapeutic potential for the treatment of Alzheimer's disease (AD). A previous study reported that N-myc downstream-regulated gene 2 (NDRG2) was upregulated in the brain of patients with AD. In the present study, we investigated the effects of repeated EA administration on reference memory impairment and the role of NDRG2 in an amyloid precursor protein (APP)/presenilin-1 (PS1) double transgenic mouse model. Age-matched wild-type and transgenic mice were treated with EA (once per day for 30 min) for 4 weeks (four courses of 5 days EA administration and 2 days rest) beginning at 10 months of age. At seven and ten postnatal months of age and following a 4-week EA treatment regime, mice received training in the Morris water maze (MWM) and a probe test. Brain tissue was analyzed via Western blot and double-label immunofluorescence. Beginning at 7 months of age, APP/PS1 mice began to exhibit deficits in reference memory in the MWM test, an impairment associated with upregulation of glial fibrillary acidic protein (GFAP) and NDRG2. Four weeks of EA administration significantly ameliorated cognitive impairments and suppressed GFAP and NDRG2 upregulation. In conclusion, our findings demonstrated that EA administration can alleviate reference memory deficits and suppress NDRG2 upregulation in an AD transgenic mouse model. This study provides supportive evidence for EA as an effective therapeutic intervention for AD, as well as NDRG2 as a novel target for AD treatment.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Electroacupuncture , Memory Disorders/therapy , Presenilin-1/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/physiology , Disease Models, Animal , Memory/physiology , Mice , Mice, Transgenic
18.
PLoS One ; 8(11): e81709, 2013.
Article in English | MEDLINE | ID: mdl-24312338

ABSTRACT

RhoE, a novel member of the Rho protein family, is a key regulator of the cytoskeleton and cell migration. Our group has previously shown that RhoE as a direct target for HIF-1α and mediates hypoxia-induced epithelial to mesenchymal transition in gastric cancer cells. Therefore, we assumed that RhoE might play an important role in gastric cancer metastasis. In the present study, we have explored the role of RhoE expression in gastric cancer, cell invasion and metastasis, and the influence of RhoE on regulating the potential expression of down-stream genes. RhoE expression was elevated in gastric cancer tissues as compared with normal gastric tissues. We also found a close correlation between the histological grade and the diagnosis of the patient. Up-regulation of RhoE significantly enhanced the migratory and invasive abilities of gastric cancer cells both in vitro and in vivo. Moreover, down-regulation of RhoE diminished the metastatic potential of cancer cells. PCR array and subsequent transwell assay showed that the regulation of gastric cancer metastasis by RhoE was partially mediated by CXCR4. This observation suggested that CXCR4 might be a downstream effector for RhoE. In summary, our study identified RhoE as a novel prognostic biomarker and metastatic-promoting gene of gastric cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Receptors, CXCR4/genetics , Stomach Neoplasms/pathology , rho GTP-Binding Proteins/metabolism , Cell Line, Tumor , Cell Movement , Humans , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , rho GTP-Binding Proteins/genetics
19.
Biomaterials ; 34(34): 8786-97, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23942209

ABSTRACT

Neurogenin-2 (Ngn2), as a proneural gene that promotes the survival and differentiation of neural precursor cells, is an attractive candidate for therapy against cerebral ischemia-reperfusion injury. However, the delivery approach limits its clinical application. To deliver Ngn2 protein into the cerebral ischemic region and exert a therapeutic effect on injured neurons after ischemia, we here reported that the fusion protein TAT-LBD-Ngn2 was constructed by fusing a transactivator of transcription (TAT) domain and a laminin-binding domain (LBD) to Ngn2. TAT-LBD-Ngn2 promoted the outgrowth of neuronal neurite, increased the survival rate and alleviated apoptosis of hippocampal neurons exposed to oxygen glucose deprivation in vitro. Furthermore, a focal cerebral ischemia model in C57BL/6 mice showed that TAT-LBD-Ngn2 efficiently crossed the blood brain barrier, aggregated in the ischemic zone and was consistently incorporated into neurons. Moreover, TAT-LBD-Ngn2 transduced into brains attenuated neuronal degeneration and apoptosis in the ischemic zone. TAT-LBD-Ngn2 treatment resulted in a reduction of infarct volume that was associated with a parallel improvement in neurological functional outcomes after reperfusion. In conclusion, the targeted delivery of TAT-LBD-Ngn2 into the ischemic zone attenuated cerebral ischemia-reperfusion injury through the inhibition of neuronal degeneration and apoptosis, suggesting that TAT-LBD-Ngn2 is a promising target candidate for the treatment of ischemic stroke.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/therapeutic use , Brain Ischemia/therapy , Gene Transfer Techniques , Nerve Tissue Proteins/therapeutic use , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Blood-Brain Barrier/metabolism , Brain Ischemia/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Differentiation/physiology , Disease Models, Animal , Gene Products, tat/genetics , Gene Products, tat/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/pathology , Nerve Degeneration/therapy , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Recombinant Fusion Proteins/therapeutic use , Stroke/pathology , Stroke/therapy , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
20.
Paediatr Anaesth ; 23(8): 726-33, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23668330

ABSTRACT

BACKGROUND: Remote ischemic postconditioning (RPostC) is a noninvasive intervention that has demonstrated cardioprotection and neuroprotection in animal studies. OBJECTIVE: Our goal was to investigate the cardio-cerebral protective effects of RPostC on children undergoing open-heart surgery for repair of congenital heart defects (CHD). METHODS: Children undergoing open-heart repair of CHD were randomly assigned to a RPostC or control group. RPostC was induced by three 5-min cycles of lower limb ischemia and reperfusion using a blood pressure cuff (200 mmHg) at the onset of aortic unclamping. Serum cardiac troponin I (cTnI), creatine kinase-MB (CK-MB), neuron-specific enolase (NSE), S100ß, cytokines, and clinical outcomes were assessed. RESULTS: There were 35 children in the control group and 34 in the RPostC group. The mean age (3.64 ± 1.95 years vs. 3.45 ± 3.02 years, P = 0.80), weight (15.11 ± 6.91 kg vs. 13.40 ± 6.33 kg, P = 0.37), surgical time (144.82 ± 38.51 min vs. 129.92 ± 30.76 min, P = 0.15), and bypass time (78.01 ± 27.22 min vs. 72.52 ± 26.05 min, P = 0.49) were not different. Compared with the control group, the postoperative levels of cTnI (P = 0.037) and CK-MB (P = 0.046) were significantly reduced in the RPostC group. Furthermore, the MAP was higher (P = 0.008), and ICU stay (36.87 ± 3.30 h vs. 60.57 ± 7.35 h, P = 0.006) and postoperative hospital stay (8.56 ± 1.50 days vs. 10.06 ± 2.41 days, P = 0.048) were shorter in the RPostC group than in the control group. However, the postoperative CVP and the concentrations of NSE, S100ß, CRP, TNF-α, IL-1ß, IL-6, and IL-10 were not significantly different. CONCLUSION: RPostC significantly alleviates cardiac injury in children undergoing open-heart repair of CHD and may also reduce cerebral injury.


Subject(s)
Cardiac Surgical Procedures/methods , Heart Defects, Congenital/surgery , Heart Diseases/prevention & control , Intraoperative Complications/prevention & control , Ischemic Postconditioning/methods , Postoperative Complications/prevention & control , Anesthesia , Biomarkers/blood , Cerebrovascular Disorders/prevention & control , Child, Preschool , Creatine Kinase, MB Form/blood , Cytokines/blood , Data Interpretation, Statistical , Female , Humans , Infant , Male , Phosphopyruvate Hydratase/blood , S100 Calcium Binding Protein beta Subunit/blood , Sample Size , Treatment Outcome , Troponin I/blood
SELECTION OF CITATIONS
SEARCH DETAIL