Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 218
Filter
1.
Neuro Oncol ; 25(2): 375-385, 2023 02 14.
Article in English | MEDLINE | ID: mdl-35789275

ABSTRACT

BACKGROUND: Pediatric postoperative cerebellar mutism syndrome (CMS) is a rare but well-known complication of medulloblastoma (Mb) resection with devastating effects on expressive language, mobility, cognition, and emotional regulation that diminishes quality of life for many Mb survivors. The specific anatomical and neuronal basis of CMS remains obscure. We address this issue by identifying patterns of surgical damage and secondary axonal degeneration in Mb survivors with CMS. METHODS: Children with Mb deemed high risk for CMS based on intraventricular location of the tumor had T1 images analyzed for location(s) of surgical damage using a specially developed algorithm. We used three complementary methods of spatial analysis to identify surgical damage linked to CMS diagnosis. Magnetization transfer ratio (MTR) images were analyzed for evidence of demyelination in anatomic regions downstream of the cerebellum, indicating neuronal dysfunction. RESULTS: Spatial analyses highlighted damage to the fastigial nuclei and their associated cerebellar cortices as the strongest predictors of CMS. CMS-related MTR decrease was greatest in the ventral periaqueductal gray (PAG) area and highly consistent in the left red nucleus. CONCLUSION: Our evidence points to disruption of output from the fastigial nuclei as a likely causal trigger for CMS. We propose that core CMS symptoms result from a disruption in the triggering of survival behaviors regulated by the PAG, including the gating of vocalization and volitional movement. The fastigial nuclei provide the densest output to the PAG from the cerebellum, thus sparing these structures may provide a greater likelihood of CMS prevention.


Subject(s)
Cerebellar Diseases , Cerebellar Neoplasms , Medulloblastoma , Mutism , Child , Humans , Periaqueductal Gray/pathology , Mutism/etiology , Quality of Life , Postoperative Complications , Cerebellar Diseases/complications , Cerebellar Diseases/diagnosis , Medulloblastoma/pathology , Cerebellar Neoplasms/surgery , Cerebellar Neoplasms/complications
2.
Neurosci Bull ; 38(12): 1491-1507, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36282466

ABSTRACT

Preoperative sleep loss can amplify post-operative mechanical hyperalgesia. However, the underlying mechanisms are still largely unknown. In the current study, rats were randomly allocated to a control group and an acute sleep deprivation (ASD) group which experienced 6 h ASD before surgery. Then the variations in cerebral function and activity were investigated with multi-modal techniques, such as nuclear magnetic resonance, functional magnetic resonance imaging, c-Fos immunofluorescence, and electrophysiology. The results indicated that ASD induced hyperalgesia, and the metabolic kinetics were remarkably decreased in the striatum and midbrain. The functional connectivity (FC) between the nucleus accumbens (NAc, a subregion of the ventral striatum) and the ventrolateral periaqueductal gray (vLPAG) was significantly reduced, and the c-Fos expression in the NAc and the vLPAG was suppressed. Furthermore, the electrophysiological recordings demonstrated that both the neuronal activity in the NAc and the vLPAG, and the coherence of the NAc-vLPAG were suppressed in both resting and task states. This study showed that neuronal activity in the NAc and the vLPAG were weakened and the FC between the NAc and the vLPAG was also suppressed in rats with ASD-induced hyperalgesia. This study highlights the importance of preoperative sleep management for surgical patients.


Subject(s)
Hyperalgesia , Sleep Deprivation , Rats , Animals , Hyperalgesia/metabolism , Sleep Deprivation/complications , Sleep Deprivation/diagnostic imaging , Sleep Deprivation/metabolism , Rats, Sprague-Dawley , Periaqueductal Gray/metabolism , Periaqueductal Gray/pathology , Proto-Oncogene Proteins c-fos/metabolism , Pain, Postoperative/metabolism , Pain, Postoperative/pathology
3.
Headache ; 61(7): 1136-1139, 2021 07.
Article in English | MEDLINE | ID: mdl-34363407

ABSTRACT

OBJECTIVES/BACKGROUND: Tension-type headache and migraine without aura are the most common primary headaches occurring in people with demyelinating diseases, whereas cluster headache (CH) can be considered exceptional. The location of demyelinating lesions could be strategic in these cases, involving areas interacting with the trigeminovascular system. METHODS AND RESULTS: We report a case of a 54-year-old woman with right-sided CH as the initial manifestation of multiple sclerosis and showing a left dorsal brainstem lesion on magnetic resonance imaging, in the region of the dorsal longitudinal fasciculus (DLF). CONCLUSION: Our case seems to suggest a possible role of the DLF in the process that leads to CH attacks. Because neuroimaging clearly showed a lesion contralateral to CH pain, we hypothesize that some fibers from periaqueductal gray matter project to the contralateral side, besides the known ipsilateral connections.


Subject(s)
Cluster Headache/etiology , Multiple Sclerosis/complications , Multiple Sclerosis/pathology , Periaqueductal Gray/pathology , Cluster Headache/diagnosis , Female , Humans , Magnetic Resonance Imaging , Middle Aged , Multiple Sclerosis/diagnostic imaging , Neural Pathways/diagnostic imaging , Neural Pathways/pathology , Periaqueductal Gray/diagnostic imaging
4.
J Histochem Cytochem ; 69(8): 511-522, 2021 08.
Article in English | MEDLINE | ID: mdl-34291686

ABSTRACT

Induction of severe inflammatory arthritis in the collagen antibody-induced arthritis (CAIA) murine model causes extensive joint damage and pain-like behavior compromising analysis. While mild models are less severe, their reduced, variable penetrance makes assessment of treatment efficacy difficult. This study aimed to compare macroscopic and microscopic changes in the paws, along with central nervous system activation between a mild and moderate CAIA model. Balb/c mice (n=18) were allocated to control, mild, and moderate CAIA groups. Paw inflammation, bone volume (BV), and paw volume (PV) were assessed. Histologically, the front paws were assessed for joint inflammation, cartilage damage, and pre/osteoclast-like cells and the lumbar spinal cord and the periaqueductal gray (PAG) region of the brain for glial reactivity. A moderate CAIA dose induced (1) significantly greater local paw inflammation, inflammatory cell infiltration, and PV; (2) significantly more osteoclast-like cells on the bone surface and within the surrounding soft tissue; and (3) significantly greater glial reactivity within the PAG compared with the mild CAIA model. These findings support the use of a moderate CAIA model (higher dose of monoclonal antibodies with low-dose lipopolysaccharide) to induce more consistent histopathological features, without excessive joint destruction.


Subject(s)
Arthritis, Experimental/pathology , Bone Resorption/pathology , Cartilage, Articular/pathology , Disease Models, Animal , Edema/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Arthritis, Experimental/chemically induced , Arthritis, Experimental/diagnosis , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/pathology , Bone Resorption/chemically induced , Bone Resorption/diagnosis , Cartilage, Articular/drug effects , Edema/chemically induced , Edema/diagnosis , Female , Forelimb/drug effects , Forelimb/pathology , Histocytochemistry , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred BALB C , Neuroglia/drug effects , Neuroglia/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Periaqueductal Gray/drug effects , Periaqueductal Gray/pathology , Severity of Illness Index , Spinal Cord/drug effects , Spinal Cord/pathology
5.
Proc Natl Acad Sci U S A ; 118(24)2021 06 15.
Article in English | MEDLINE | ID: mdl-34108238

ABSTRACT

Galanin receptor1 (GalR1) transcript levels are elevated in the rat ventral periaqueductal gray (vPAG) after chronic mild stress (CMS) and are related to depression-like behavior. To explore the mechanisms underlying the elevated GalR1 expression, we carried out molecular biological experiments in vitro and in animal behavioral experiments in vivo. It was found that a restricted upstream region of the GalR1 gene, from -250 to -220, harbors an E-box and plays a negative role in the GalR1 promoter activity. The transcription factor Scratch2 bound to the E-box to down-regulate GalR1 promoter activity and lower expression levels of the GalR1 gene. The expression of Scratch2 was significantly decreased in the vPAG of CMS rats. Importantly, local knockdown of Scratch2 in the vPAG caused elevated expression of GalR1 in the same region, as well as depression-like behaviors. RNAscope analysis revealed that GalR1 mRNA is expressed together with Scratch2 in both GABA and glutamate neurons. Taking these data together, our study further supports the involvement of GalR1 in mood control and suggests a role for Scratch2 as a regulator of depression-like behavior by repressing the GalR1 gene in the vPAG.


Subject(s)
Behavior, Animal , Depression/pathology , Periaqueductal Gray/pathology , Receptor, Galanin, Type 1/metabolism , Transcription Factors/metabolism , Animals , E-Box Elements/genetics , GABAergic Neurons/metabolism , Gene Expression Regulation , Glutamic Acid/metabolism , PC12 Cells , Promoter Regions, Genetic/genetics , Protein Binding , Rats , Receptor, Galanin, Type 1/genetics , Stress, Psychological/complications , Transcription Factors/genetics , Transcription Initiation Site
6.
Neurosci Lett ; 752: 135825, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33727130

ABSTRACT

OBJECTIVE: To investigate the effect of minocycline on morphine withdrawal symptoms. METHODS: We established a rat model of morphine dependence, then injected the animals with naloxone to induce withdrawal symptoms. Minocycline was injected into the midbrain periaqueductal gray and its effect on withdrawal symptoms and Ca2+-dependent protein kinase (CaMKII), Ras, and phospho-extracellular signal-regulated kinase (p-ERK) expression was observed. RESULTS: Minocycline inhibited withdrawal symptoms such as "wet dog" shakes, teeth chatter, and ptosis, perhaps by inhibiting the activation of microglia and the expression of CaMKII, Ras, and p-ERK. Minocycline had no effect on the behavior of control rats or on CaMKII, Ras, or p-ERK expression. CONCLUSION: Minocycline alleviates morphine withdrawal symptoms by inhibiting the activation of microglia and downregulating the expression of CaMKII, Ras, and p-ERK.


Subject(s)
MAP Kinase Signaling System/drug effects , Minocycline/pharmacology , Morphine/adverse effects , Substance Withdrawal Syndrome/drug therapy , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Humans , Male , Microglia/drug effects , Microglia/pathology , Minocycline/therapeutic use , Morphine/antagonists & inhibitors , Naloxone/administration & dosage , Periaqueductal Gray/drug effects , Periaqueductal Gray/pathology , Rats , Substance Withdrawal Syndrome/pathology , ras Proteins/metabolism
7.
Brain Res ; 1750: 147171, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33132167

ABSTRACT

The ability to cope with a novel acute stressor in the context of ongoing chronic stress is of critical adaptive value. The hypothalamic-pituitary-adrenal (HPA) axis contributes to the integrated physiological and behavioural responses to stressors. Under conditions of chronic stress, the posterior portion of the paraventricular thalamic nucleus (pPVT) mediates the 'habituation' of HPA-axis responses, and also facilitates HPA-axis reactivation to novel acute stressors amidst this habituation. Since pPVT neurons are sensitive to the inhibitory effects of circulating glucocorticoids, a glucocorticoid-insensitive neural pathway to the pPVT is likely essential for this reactivation process. The pPVT receives substantial inputs from neurons of the periaqueductal gray (PAG) region, which is organised into longitudinal columns critical for processing acute and/or chronic stressors. We investigated the columnar organisation of PAG â†’ pPVT projections and for the first time determined their glucocorticoid sensitivity. Retrograde tracer injections were made into different rostro-caudal regions of the pPVT, and their PAG columnar inputs compared. Glucocorticoid receptor immunoreactivity (GR-ir) was quantified in these projection neurons. We found that the dorsolateral PAG projected most strongly to rostral pPVT and the ventrolateral PAG most strongly to the caudal pPVT. Despite abundant GR-ir in the PAG, we report a striking absence of GR-ir in PAG â†’ pPVT neurons. Our data suggests that these pathways, which are insensitive to the direct actions of circulating glucocorticoids, likely play an important role in both the habituation of HPA-axis to chronic stressors and its facilitation to acute stressors in chronically stressed rats.


Subject(s)
Midline Thalamic Nuclei/physiology , Periaqueductal Gray/metabolism , Periaqueductal Gray/pathology , Afferent Pathways/metabolism , Animals , Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/metabolism , Male , Midline Thalamic Nuclei/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/metabolism , Stress, Physiological/physiology , Thalamus/metabolism
8.
Curr Biol ; 30(23): 4631-4642.e6, 2020 12 07.
Article in English | MEDLINE | ID: mdl-32976802

ABSTRACT

Pain sensation is powerfully modulated by signal processing in the brain, and pain becomes chronic with the dysfunction of the pain modulatory system; however, the underlying mechanisms are unclear. We found that the metabotropic glutamate receptor 5 (mGluR5) in the periaqueductal gray (PAG), the key area of endogenous pain modulation, is persistently active in normal conditions to maintain an appropriate sensory perception. In the neuropathic pain condition, Homer1a, an activity-dependent immediate early gene product, disrupted the persistent mGluR5 activity resulting in chronic pain. Remarkably a single-time blockage of the mGluR5 resulted in chronic neuropathic pain-like symptoms even in the absence of nerve injury. The decline of mGluR5 activity induced the pain modulatory dysfunction with a profound reduction of excitability of PAG neurons. These findings uncover the role of the persistent mGluR5 activity in vivo and provide new insight into how pain becomes chronic with the maladaptive coping of the PAG to pain sensation.


Subject(s)
Chronic Pain/physiopathology , Hyperalgesia/physiopathology , Neuralgia/physiopathology , Periaqueductal Gray/pathology , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Chronic Pain/etiology , Chronic Pain/pathology , Disease Models, Animal , Gene Knockdown Techniques , Homer Scaffolding Proteins/genetics , Homer Scaffolding Proteins/metabolism , Humans , Hyperalgesia/etiology , Hyperalgesia/pathology , Male , Neuralgia/etiology , Neuralgia/pathology , Pain Perception/physiology , Periaqueductal Gray/physiopathology , Rats
9.
J Neurosci ; 40(41): 7837-7854, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32958568

ABSTRACT

As one of the thalamic midline nuclei, the thalamic paraventricular nucleus (PVT) is considered to be an important signal integration site for many descending and ascending pathways that modulate a variety of behaviors, including feeding, emotions, and drug-seeking. A recent study has demonstrated that the PVT is implicated in the acute visceral pain response, but it is unclear whether the PVT plays a critical role in the central processing of chronic pain. Here, we report that the neurons in the posterior portion of the PVT (pPVT) and their downstream pathway are involved in descending nociceptive facilitation regarding the development of neuropathic pain conditions in male rats. Lesions or inhibition of pPVT neurons alleviated mechanical allodynia induced by spared nerve injury (SNI). The excitability of pPVT-central amygdala (CeA) projection neurons was significantly increased in SNI rats. Importantly, selective optogenetic activation of the pPVT-CeA pathway induced obvious mechanical hypersensitivity in naive rats. In addition, we used rabies virus (RV)-based and cell-type-specific retrograde transsynaptic tracing techniques to define a novel neuronal circuit in which glutamatergic neurons in the vlPAG were the target of the pPVT-CeA descending facilitation pathway. Our data suggest that this pPVTGlu+-CeA-vlPAGGlu+ circuit mediates central mechanisms of descending pain facilitation underlying persistent pain conditions.SIGNIFICANCE STATEMENT Studies have shown that the interactions between the posterior portion of the thalamic paraventricular nucleus (pPVT) and central amygdala (CeA) play a critical role in pain-related emotional regulation. However, most reports have associated this circuit with fear and anxiety behaviors. Here, an integrative approach of behavioral tests, electrophysiology, and immunohistochemistry was used to advance the novel concept that the pPVT-CeA pathway activation facilitates neuropathic pain processing. Using rabies virus (RV)-based and cell-type-specific retrograde transsynaptic tracing techniques, we found that glutamatergic neurons in the vlPAG were the target of the pPVT-CeA pathway. Thus, this study indicates the involvement of a pPVTGlu+-CeA-vlPAGGlu+ pathway in a descending facilitatory mechanism underlying neuropathic pain.


Subject(s)
Central Amygdaloid Nucleus/pathology , Midline Thalamic Nuclei/pathology , Neural Pathways/pathology , Neuralgia/pathology , Animals , Behavior, Animal , Electrophysiological Phenomena , Hyperalgesia/pathology , Image Processing, Computer-Assisted , Male , Neuralgia/psychology , Neurons/pathology , Nociception , Optogenetics , Periaqueductal Gray/pathology , Rats , Rats, Sprague-Dawley
10.
Neuroimage ; 221: 117186, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32711060

ABSTRACT

Postherpetic Neuralgia (PHN), develops after the resolution of the herpes zoster mucocutaneous eruption, is a debilitating chronic pain. However, there is a lack of knowledge regarding the underlying mechanisms associated with ascending and descending pain modulations in PHN patients. Here, we combined psychophysics with structural and functional magnetic resonance imaging (MRI) techniques to investigate the brain alternations in PHN patients. Psychophysical tests showed that compared with healthy controls, PHN patients had increased state and trait anxiety and depression. Structural MRI data indicated that PHN patients had significantly smaller gray matter volumes of the thalamus and amygdala than healthy controls, and the thalamus volume was negatively correlated with pain intensity (assessed using the Short-form of the McGill pain questionnaire) in PHN patients. When the thalamus and periaqueductal gray matter (PAG) were used as the seeds, resting-state functional MRI data revealed abnormal patterns of functional connectivity within ascending and descending pain pathways in PHN patients, e.g., increased functional connectivity between the thalamus and somatosensory cortices and decreased functional connectivity between the PAG and frontal cortices. In addition, subjective ratings of both Present Pain Index (PPI) and Beck-Depression Inventory (BDI) were negatively correlated with the strength of functional connectivity between the PAG and primary somatosensory cortex (SI), and importantly, the effect of BDI on PPI was mediated by the PAG-SI functional connectivity. Overall, our results provided evidence suggesting deficits in ascending and descending pain modulation pathways, which were highly associated with the intensity of chronic pain and its emotional comorbidities in PHN patients. Therefore, our study deepened our understanding of the pathogenesis of PHN, which would be helpful in determining the optimized treatment for the patients.


Subject(s)
Amygdala , Cerebral Cortex , Connectome , Gray Matter , Magnetic Resonance Imaging , Nerve Net , Neuralgia, Postherpetic/physiopathology , Periaqueductal Gray , Thalamus , Aged , Amygdala/diagnostic imaging , Amygdala/pathology , Amygdala/physiopathology , Anxiety/physiopathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Depression/physiopathology , Female , Gray Matter/diagnostic imaging , Gray Matter/pathology , Gray Matter/physiopathology , Humans , Male , Middle Aged , Nerve Net/diagnostic imaging , Nerve Net/pathology , Nerve Net/physiopathology , Neural Pathways/diagnostic imaging , Neural Pathways/physiopathology , Neuralgia, Postherpetic/diagnostic imaging , Neuralgia, Postherpetic/pathology , Periaqueductal Gray/diagnostic imaging , Periaqueductal Gray/pathology , Periaqueductal Gray/physiopathology , Thalamus/diagnostic imaging , Thalamus/pathology , Thalamus/physiopathology
11.
Eur Rev Med Pharmacol Sci ; 24(9): 5053-5061, 2020 05.
Article in English | MEDLINE | ID: mdl-32432769

ABSTRACT

OBJECTIVE: Neuropathic pain is directly developed from lesions or somatosensory nervous system diseases that are associated with emotion regulation. In general population, the incidence of neuropathic pain ranges from 7% to 10%, but the underlying mechanism remains largely unknown. Neuropathic pain is often associated with structural and functional abnormalities in multiple brain regions, and its regulation has been shown to correspond with the forebrain, including nucleus accumbens (NAc), medial prefrontal cortex (mPFC) and periaqueductal gray (PAG). MATERIALS AND METHODS: To investigate the molecular mechanism of neuropathic pain across different brain regions, we identified the differentially expressed genes (DEGs) between the spared nerve injury model (SNI) mice suffering neuropathic pain and the control Sham mice in NAc, mPFC and PAG three brain regions, and mapped these genes onto a comprehensively functional association network. Thereafter, novel neuropathic pain genes in these three regions were identified using With Random Walk with Restart (RWR) analysis, such as Asic3, Cd200r1 and MT2, besides well-known Capn11 and CYP2E1. RESULTS: Interactions or cross talks among DEGs in NAc, mPFC and PAG three brain regions were discovered. CONCLUSIONS: Our results provide novel insights into neuropathic pain and help to explore therapeutic targets in the treatment.


Subject(s)
Disease Models, Animal , Gene Regulatory Networks/genetics , Neuralgia/genetics , Nucleus Accumbens/metabolism , Periaqueductal Gray/metabolism , Prefrontal Cortex/metabolism , Animals , Mice , Neuralgia/metabolism , Neuralgia/pathology , Nucleus Accumbens/pathology , Periaqueductal Gray/pathology , Prefrontal Cortex/pathology
12.
J Clin Neurosci ; 78: 422-425, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32360161

ABSTRACT

A 22-year old male with a history of B-cell acute lymphoblastic leukemia with recent bone marrow transplantation and on immunosuppressive therapy presented with painless, subacute vision loss of two weeks duration. He exhibited a horizontal gaze palsy, nystagmus, and mildly swollen and hyperemic optic discs with peripapillary flame hemorrhage on retinal exam. He had bilateral cecocentral scotomas on visual field exam, and MRI of his brain/orbits demonstrated hyperintensities in the hypothalamus, periaqueductal gray, and dorsal rostral medullary regions. After continued progression of symptoms despite discontinuation of the patient's tacrolimus, an empiric trial of IV thiamine treatment was started before the patient's lab vitamin levels were available, given strong clinical suspicion for a nutritional etiology. The patient's clinical presentation improved dramatically, and he achieved a final visual acuity of 20/20, full visual fields bilaterally, and resolution of nystagmus. A final diagnosis of Wernicke's encephalopathy was supported by his clinical course, imaging findings, and further confirmation with blood thiamine levels. This case presents unique ocular manifestations of Wernicke's encephalopathy and highlights the importance of early diagnosis in this potentially reversible condition.


Subject(s)
Leukemia, B-Cell/pathology , Nystagmus, Pathologic/etiology , Thiamine/blood , Vision Disorders/drug therapy , Wernicke Encephalopathy/etiology , Brain/diagnostic imaging , Humans , Immunosuppressive Agents/therapeutic use , Leukemia, B-Cell/complications , Leukemia, B-Cell/therapy , Magnetic Resonance Imaging/adverse effects , Male , Nystagmus, Pathologic/diagnosis , Nystagmus, Pathologic/drug therapy , Ophthalmoplegia, Chronic Progressive External/etiology , Periaqueductal Gray/pathology , Scoliosis/etiology , Thiamine/administration & dosage , Thiamine/therapeutic use , Vision Disorders/etiology , Wernicke Encephalopathy/diagnosis , Young Adult
13.
Behav Brain Res ; 377: 112236, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31536735

ABSTRACT

CO2 inhalation can provoke panic attacks in humans, and the likelihood is increased in patients with panic disorder. Identifying brain sites involved could provide important mechanistic insight into the illness. In mice, the amygdala has been suggested to promote CO2 responses; however, recent studies in humans with amygdala damage indicate the amygdala is not required for CO2-induced fear and panic and might actually oppose these responses. To clarify the role of the amygdala, we produced lesions in mice paralleling the human lesions, and characterized behavioral responses to CO2. Compared to sham controls, we found that amygdala-lesioned mice froze less to 10% CO2, and unlike shams they also began to jump frenetically. At 20% CO2, controls also exhibited jumping, suggesting it is a normal response to more extreme CO2 concentrations. The effect of amygdala lesions was specific to CO2 as amygdala-lesioned mice did not jump in response to a predator odor or to an auditory conditioned stimulus. In amygdala-lesioned mice, jumping evoked by 10% CO2 was eliminated by co-lesioning the dorsal periaqueductal gray, a structure implicated in panic and escape-related behaviors. Together, these observations suggest a dual role for the amygdala in the CO2 response: promoting CO2-induced freezing, and opposing CO2-induced jumping, which may help explain the exaggerated CO2 responses in humans with amygdala lesions.


Subject(s)
Amygdala/physiology , Behavior, Animal , Carbon Dioxide/pharmacology , Fear/drug effects , Locomotion , Amygdala/pathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Carbon Dioxide/administration & dosage , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Locomotion/drug effects , Locomotion/physiology , Mice , Mice, Inbred C57BL , Periaqueductal Gray/pathology
14.
J Ethnopharmacol ; 249: 112437, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31794788

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Migraine is a disabling neurovascular disorder, which increases risk of cardiovascular events and is a social burden worldwide. The present first-line anti-migraine medications can cause overwhelming side-effects, of which one includes the onset of cardiovascular disease. As one of the marketed Tibetan drugs, Ru-yi-Zhen-bao Pills (RYZBP) have been clinically used to treat cardiovascular disorders and as anti-migraine medication. However, there is currently no research exploring the anti-migraine actions of RYZBP. AIM OF THE STUDY: The current research was designed to assess the anti-migraine roles of RYZBP and explore the underlying mechanisms in a nitroglycerin (NTG)-induced migraine rat model trial. MATERIALS AND METHODS: 120 rats were randomly divided into the following six groups of 20 rats each: normal control group, model control group, positive control group, and RYZBP high/medium/low-dose groups (Ru-yi-Zhen-bao Pills; TH 1.00 g/kg, TM 0.50 g/kg and TL 0.25 g/kg). All rats were administered intragastrically for 7 consecutive days, which were subcutaneously injected with the NTG (10 mg/kg) after the last gavage (except in the normal control group). 3min after NTG treatment, 30 rats (5 rats from each group) were anesthetized and devoted to electroencephalogram(EEG) testing, which was used to evaluate the analgesic effect of RYZBP. One hour after NTG treatment, the rest of the 90 rats (15 rats from each group) were anesthetized and midbrain tissue sample was dissected. The dissection was then washed with physiological saline and collected. The histopathological changes in the periaqueductal gray(PAG) of 5 tissue samples were determined by aematoxylin-eosin (H&E) staining, as well as an estimation of substance P (SP) and neurokinin 1 receptor (NK1R) expression through immunohistochemically staining(IHC). Another 5 midbrain preparations were carried out to evaluate calcitonin gene-related peptide (CGRP), proenkephalin (PENK), SP, and cholecystokinin (CCK) expressions by real-time quantitative polymerase chain reaction (RT-qPCR). The rest of the 5 brainstem tissues were then used to measure CCK, CGRP, and opioid peptide receptor (DORR) levels by western blotting(WB). RESULTS: In the EEG test, RYZBP (TM 0.50 g / kg) treatment transformed the EEG pain-wave of the NTG-induced migraine model rats in different time period. In the mechanism assay, compared with the model control group, RYZBP pretreatment reduced inflammatory cell infiltration, fibrosis and vacuolation of neuronal cells of PAG tissue seen by HE staining. IHC experiments further showed that RYZBPTM up-regulated SP expression levels and enhanced NK1R levels in the NTG-induced migraine rats (P < 0.05). Therapeutic administration of RYZBP also increased PENK mRNA expression and DORR protein level. Both RT-qPCR and western blotting trials indicated that RYZBP treatment significantly decreased CCK and CGRP expression levels (P < 0.01 or P < 0.05) in the NTG-induced migraine rats. CONCLUSIONS: RYZBP has the potential to be an effective anti-migraine treatment through suppressing the EEG pain-wave, increasing the levels of SP, PENK, DORR and reducing expression of CCK and CGRP. Mediating the PAG anti-nociceptive channel and inhibiting central sensitization were the two potential mechanisms, which offers further evidence for clinical therapy.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Medicine, Tibetan Traditional/methods , Migraine Disorders/drug therapy , Nociception/drug effects , Periaqueductal Gray/drug effects , Animals , Calcitonin Gene-Related Peptide/metabolism , Cholecystokinin/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Electroencephalography , Enkephalins/metabolism , Humans , Male , Migraine Disorders/chemically induced , Migraine Disorders/diagnosis , Migraine Disorders/pathology , Nitroglycerin/toxicity , Periaqueductal Gray/pathology , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid/metabolism
15.
Nat Neurosci ; 22(10): 1659-1668, 2019 10.
Article in English | MEDLINE | ID: mdl-31501573

ABSTRACT

Neuropathic pain can be a debilitating condition with both sensory and affective components, the underlying brain circuitry of which remains poorly understood. In the present study, a basolateral amygdala (BLA)-prefrontal cortex (PFC)-periaqueductal gray (PAG)-spinal cord pathway was identified that is critical for the development of mechanical and thermal hypersensitivity after peripheral nerve injury. It was shown that nerve injury strengthens synaptic input from the BLA onto inhibitory interneurons located in the prelimbic medial PFC, by virtue of reduced endocannabinoid modulation. These augmented synaptic connections mediate a feedforward inhibition of projections from the PFC to the ventrolateral PAG region and its downstream targets. Optogenetic approaches combined with in vivo pharmacology reveal that these BLA-PFC-PAG connections alter pain behaviors by reducing descending noradrenergic and serotoninergic modulation of spinal pain signals. Thus, a long-range brain circuit was identified that is crucial for pain processing and that can potentially be exploited toward targeting neuropathic pain.


Subject(s)
Neural Pathways/pathology , Neuralgia/pathology , Neurons/pathology , Amygdala/pathology , Animals , Behavior, Animal , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Motor Activity , Neuralgia/psychology , Optogenetics , Periaqueductal Gray/pathology , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/psychology , Physical Stimulation , Prefrontal Cortex/pathology , Spinal Cord/pathology , Synapses/pathology
16.
Balkan Med J ; 36(5): 263-269, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31218879

ABSTRACT

Background: The receptors of salmon calcitonin, located on certain areas of the brain such as the periaqueductal gray matter, are responsible for pain modulation. Aims: The effects of intracerebroventricular injection of salmon calcitonin on the behavioral response to pain and on the levels of monoamines in the periaqueductal gray were explored using a biphasic animal model of pain. Study Design: Animal experiment. Methods: A total of 45 male rats were divided into four groups (n=6). Salmon calcitonin was injected into the lateral ventricle of the brain (1.5 nmol, with a volume of 5 µL). After 20 min, 2.5% formalin was subcutaneously injected into the right leg claw, and pain behavior was recorded on a numerical basis. At the time of the formalin test, the periaqueductal gray area was microdialized. High-performance liquid chromatography method was used to gauge the levels of monoamines and their metabolites. Results: Intracerebroventricular injections of salmon calcitonin resulted in pain reduction in the formalin test (p<0.05). The dialysate concentrations of serotonin, dopamine, norepinephrine, 5-hydroxyindoleacetic acid, 3,4-dihydroxyphenylacetic, and 4-hydroxy-3-methoxyphenylglycol increased in the periaqueductal gray area in different phases of the formalin pain test (p<0.05). Conclusion: Salmon calcitonin reduced pain by increasing the concentrations of monoamines and the metabolites derived from them in the periaqueductal gray area.


Subject(s)
Biogenic Monoamines/physiology , Calcitonin/administration & dosage , Periaqueductal Gray/chemistry , Salmon/blood , Analysis of Variance , Animals , Biogenic Monoamines/analysis , Calcitonin/pharmacology , Pain Measurement/methods , Periaqueductal Gray/pathology , Rats , Rats, Sprague-Dawley/metabolism , Rats, Sprague-Dawley/physiology , Salmon/physiology
17.
Trends Neurosci ; 42(5): 349-360, 2019 05.
Article in English | MEDLINE | ID: mdl-30955857

ABSTRACT

Neurons in the periaqueductal gray (PAG) integrate negative emotions with the autonomic, neuroendocrine, and immune systems to facilitate responses to threat. Modern functional track tracing in animals and optogenetic and chemogenetic techniques show that the PAG is a rich substrate for the integration of active and passive responses to threat. In humans, the same regions of the PAG that give rise to adaptive anger/fight, fear/panic, depression/shutdown, pain, and predatory behaviors in response to challenging situations or overwhelming threats can become activated pathologically, resulting in symptoms that resemble those of psychiatric disorders. This review coalesces human and animal studies to link PAG neuropathways to specific elements of psychiatric diagnoses. The insights gained from this overview may eventually lead to new therapeutic interventions.


Subject(s)
Fear/psychology , Mental Disorders/metabolism , Mental Disorders/psychology , Pain/metabolism , Pain/psychology , Periaqueductal Gray/metabolism , Animals , Fear/physiology , Humans , Mental Disorders/pathology , Optogenetics/methods , Pain/pathology , Periaqueductal Gray/chemistry , Periaqueductal Gray/pathology , Psychopathology
18.
Mol Pain ; 15: 1744806919831909, 2019.
Article in English | MEDLINE | ID: mdl-30700204

ABSTRACT

Descending nociceptive modulation from the supraspinal structures has an important role in cancer-induced bone pain (CIBP). Midbrain ventrolateral periaqueductal gray (vlPAG) is a critical component of descending nociceptive circuits; nevertheless, its precise cellular and molecular mechanisms involved in descending facilitation remain elusive. Our previous study has shown that the activation of p38 MAPK in vlPAG microglia is essential for the neuropathic pain sensitization. However, the existence of potential connection between astrocytes and c-Jun N-terminal kinase (JNK) pathway in CIBP has not yet been elucidated. The following study examines the involvement of astrocyte activation and upregulation of p-JNK in vlPAG, using a CIBP rat model. Briefly, CIBP was mimicked by an intramedullary injection of Walker 256 mammary gland carcinoma cells into the animal tibia. A significant increase in expression levels of astrocytes in the vlPAG of CIBP rats was observed. Furthermore, stereotaxic microinjection of the astrocytic cytotoxin L-α-aminoadipic acid decreased the mechanical allodynia as well as established and reversed the astrocyte activation in CIBP rats. A significant increase in expression levels of p-JNK in astrocytes in vlPAG of CIBP rats was also observed. Moreover, the intrathecal administration of JNK inhibitors SP600125 reduced the expression of glial fibrillary acidic protein, while microinjection of the SP600125 decreased the mechanical allodynia of CIBP rats. These results suggested that CIBP is associated with astrocyte activation in the vlPAG that probably participates in driving descending pain facilitation through the JNK MAPK signaling pathway. To sum up, these findings reveal a novel site of astrocytes modulation of CIBP.


Subject(s)
Astrocytes/pathology , Cancer Pain/pathology , Gene Expression Regulation, Neoplastic/physiology , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , Periaqueductal Gray/pathology , Animals , Anthracenes/pharmacology , Body Weight/drug effects , Bone Neoplasms/complications , Bone Neoplasms/pathology , CD11b Antigen/metabolism , Cancer Pain/etiology , Carcinoma/complications , Carcinoma/pathology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/etiology , MAP Kinase Signaling System/drug effects , Periaqueductal Gray/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley
19.
Neuropharmacology ; 139: 52-60, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29928886

ABSTRACT

Painful conditions and sleep disturbances are major public health problems worldwide and one directly affects the other. Sleep loss increases pain prevalence and severity; while pain disturbs sleep. However, the underlying mechanisms are largely unknown. Here we asked whether chronic sleep restriction for 6 h daily progressively increases pain sensitivity and if this increase is reversed after two days of free sleep. Also, whether the pronociceptive effect of chronic sleep restriction depends on the periaqueductal grey and on the nucleus accumbens, two key regions involved in the modulation of pain and sleep-wake cycle. We showed that sleep restriction induces a pronociceptive effect characterized by a significant decrease in the mechanical paw withdrawal threshold in rats. Such effect increases progressively from day 3 to day 12 remaining stable thereafter until day 26. Two consecutive days of free sleep were not enough to reverse the effect, not even to attenuate it. This pronociceptive effect depends on the periaqueductal grey and on the nucleus accumbens, since it was prevented by their excitotoxic lesion. Complementarily, chronic sleep restriction significantly increased c-Fos protein expression within the periaqueductal grey and the nucleus accumbens and this correlates with the intensity of the pronociceptive effect, suggesting that the greater the neural activity in this regions, the greater the effect. These findings may contribute not only to understand why painful conditions are more prevalent and severe among people who sleep poorly, but also to develop therapeutic strategies to prevent this, increasing the effectiveness of pain management in this population.


Subject(s)
Nucleus Accumbens/physiopathology , Pain Perception/physiology , Pain Threshold/physiology , Periaqueductal Gray/physiopathology , Sleep Deprivation/physiopathology , Animals , Male , N-Methylaspartate/toxicity , Nociceptive Pain/pathology , Nociceptive Pain/physiopathology , Nucleus Accumbens/pathology , Periaqueductal Gray/pathology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Wistar , Sleep Deprivation/pathology , Time Factors , Touch
20.
Mol Pain ; 14: 1744806918783535, 2018.
Article in English | MEDLINE | ID: mdl-29900804

ABSTRACT

Background Neuropathic pain is observed in patients as chemotherapeutic oxaliplatin is used to treat metastatic digestive tumors; however, the mechanisms responsible for hyperalgesia are not well understood. Chronic neuroinflammation is one of the hallmarks of pathophysiology of neuropathic pain. Since the midbrain periaqueductal gray is an important component of the descending inhibitory pathway controlling on central pain transmission, we examined the role for pro-inflammatory cytokines system of the periaqueductal gray in regulating mechanical hyperalgesia and cold hypersensitivity evoked by oxaliplatin. Methods Neuropathic pain was induced by intraperitoneal injection of oxaliplatin in rats. ELISA and western blot analysis were used to examine pro-inflammatory cytokine levels and their receptors expression. Results IL-1ß, IL-6, and TNF-α were elevated within the periaqueductal gray of oxaliplatin rats. Protein expression of IL-1ß, IL-6, and TNF-α receptors (namely, IL-1R, IL-6R, and TNFR subtype TNFR1) in the plasma membrane periaqueductal gray of oxaliplatin rats was upregulated, whereas the total expression of pro-inflammatory cytokine receptors was not altered. In oxaliplatin rats, impaired inhibitory gamma-aminobutyric acid within the periaqueductal gray was accompanied with decreases in withdrawal thresholds to mechanical stimulus and % time spent on the cold plate. Our data further showed that the concentrations of gamma-aminobutyric acid were largely restored by blocking those pro-inflammatory cytokine receptors in periaqueductal gray of oxaliplatin rats; and mechanical hyperalgesia and cold hypersensitivity evoked by oxaliplatin were attenuated. Stimulation of gamma-aminobutyric acid receptors in the periaqueductal gray also blunted neuropathic pain in oxaliplatin rats. Conclusions Our data suggest that the upregulation of pro-inflammatory cytokines and membrane pro-inflammatory cytokine receptor in the periaqueductal gray of oxaliplatin rats is likely to impair the descending inhibitory pathways in regulating pain transmission and thereby contributes to the development of neuropathic pain after application of chemotherapeutic oxaliplatin.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Neuralgia/chemically induced , Neuralgia/metabolism , Organoplatinum Compounds/adverse effects , Signal Transduction , gamma-Aminobutyric Acid/metabolism , Animals , Cryopyrin-Associated Periodic Syndromes/metabolism , Cryopyrin-Associated Periodic Syndromes/pathology , Male , Oxaliplatin , Periaqueductal Gray/metabolism , Periaqueductal Gray/pathology , Rats, Sprague-Dawley , Receptors, Cytokine/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...