Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37847562

ABSTRACT

Tumor burden, considered a common chronic stressor, can cause widespread anxiety. Evidence suggests that cancer-induced anxiety can promote tumor progression, but the underlying neural mechanism remains unclear. Here, we used neuroscience and cancer tools to investigate how the brain contributes to tumor progression via nerve-tumor crosstalk in a mouse model of breast cancer. We show that tumor-bearing mice exhibited significant anxiety-like behaviors and that corticotropin-releasing hormone (CRH) neurons in the central medial amygdala (CeM) were activated. Moreover, we detected newly formed sympathetic nerves in tumors, which established a polysynaptic connection to the brain. Pharmacogenetic or optogenetic inhibition of CeMCRH neurons and the CeMCRH→lateral paragigantocellular nucleus (LPGi) circuit significantly alleviated anxiety-like behaviors and slowed tumor growth. Conversely, artificial activation of CeMCRH neurons and the CeMCRH→LPGi circuit increased anxiety and tumor growth. Importantly, we found alprazolam, an antianxiety drug, to be a promising agent for slowing tumor progression. Furthermore, we show that manipulation of the CeMCRH→LPGi circuit directly regulated the activity of the intratumoral sympathetic nerves and peripheral nerve-derived norepinephrine, which affected tumor progression by modulating antitumor immunity. Together, these findings reveal a brain-tumor neural circuit that contributes to breast cancer progression and provide therapeutic insights for breast cancer.


Subject(s)
Corticotropin-Releasing Hormone , Neoplasms , Mice , Animals , Corticotropin-Releasing Hormone/metabolism , Neurons/metabolism , Anxiety , Brain/metabolism
2.
Chin J Integr Med ; 29(10): 875-884, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36843056

ABSTRACT

OBJECTIVE: To investigate protective effect of Cordyceps sinensis (CS) through autophagy-associated adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling pathway in acute kidney injury (AKI)-induced acute lung injury (ALI). METHODS: Forty-eight male Sprague-Dawley rats were divided into 4 groups according to a random number table, including the normal saline (NS)-treated sham group (sham group), NS-treated ischemia reperfusion injury (IRI) group (IRI group), and low- (5 g/kg·d) and high-dose (10 g/kg·d) CS-treated IRI groups (CS1 and CS2 groups), 12 rats in each group. Nephrectomy of the right kidney was performed on the IRI rat model that was subjected to 60 min of left renal pedicle occlusion followed by 12, 24, 48, and 72 h of reperfusion. The wet-to-dry (W/D) ratio of lung, levels of serum creatinine (Scr), blood urea nitrogen (BUN), inflammatory cytokines such as interleukin- ß and tumor necrosis factor- α, and biomarkers of oxidative stress such as superoxide dismutase, malonaldehyde (MDA) and myeloperoxidase (MPO), were assayed. Histological examinations were conducted to determine damage of tissues in the kidney and lung. The protein expressions of light chain 3 II/light chain 3 I (LC3-II/LC3-I), uncoordinated-51-like kinase 1 (ULK1), P62, AMPK and mTOR were measured by Western blot and immunohistochemistry, respectively. RESULTS: The renal IRI induced pulmonary injury following AKI, resulting in significant increases in W/D ratio of lung, and the levels of Scr, BUN, inflammatory cytokines, MDA and MPO (P<0.01); all of these were reduced in the CS groups (P<0.05 or P<0.01). Compared with the IRI groups, the expression levels of P62 and mTOR were significantly lower (P<0.05 or P<0.01), while those of LC3-II/LC3-I, ULK1, and AMPK were significantly higher in the CS2 group (P<0.05 or P<0.01). CONCLUSION: CS had a potential in treating lung injury following renal IRI through activation of the autophagy-related AMPK/mTOR signaling pathway in AKI-induced ALI.


Subject(s)
Acute Kidney Injury , Acute Lung Injury , Cordyceps , Reperfusion Injury , Rats , Male , Animals , AMP-Activated Protein Kinases/metabolism , Cordyceps/metabolism , Rats, Sprague-Dawley , Kidney/pathology , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Reperfusion Injury/metabolism , Cytokines/metabolism , Acute Lung Injury/drug therapy , Mammals/metabolism
3.
Neurosci Bull ; 39(6): 962-972, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36629979

ABSTRACT

The anterior auditory field (AAF) is a core region of the auditory cortex and plays a vital role in discrimination tasks. However, the role of the AAF corticostriatal neurons in frequency discrimination remains unclear. Here, we used c-Fos staining, fiber photometry recording, and pharmacogenetic manipulation to investigate the function of the AAF corticostriatal neurons in a frequency discrimination task. c-Fos staining and fiber photometry recording revealed that the activity of AAF pyramidal neurons was significantly elevated during the frequency discrimination task. Pharmacogenetic inhibition of AAF pyramidal neurons significantly impaired frequency discrimination. In addition, histological results revealed that AAF pyramidal neurons send strong projections to the striatum. Moreover, pharmacogenetic suppression of the striatal projections from pyramidal neurons in the AAF significantly disrupted the frequency discrimination. Collectively, our findings show that AAF pyramidal neurons, particularly the AAF-striatum projections, play a crucial role in frequency discrimination behavior.


Subject(s)
Auditory Cortex , Neurons , Acoustic Stimulation/methods , Neurons/physiology , Auditory Cortex/physiology , Auditory Perception , Pyramidal Cells
4.
Sci Adv ; 8(30): eabn4408, 2022 Jul 29.
Article in English | MEDLINE | ID: mdl-35905177

ABSTRACT

Itch is a cutaneous sensation that is critical in driving scratching behavior. The long-standing question of whether there are specific neurons for itch modulation inside the brain remains unanswered. Here, we report a subpopulation of itch-specific neurons in the ventrolateral orbital cortex (VLO) that is distinct from the pain-related neurons. Using a Tet-Off cellular labeling system, we showed that local inhibition or activation of these itch-specific neurons in the VLO significantly suppressed or enhanced itch-induced scratching, respectively, whereas the intervention did not significantly affect pain. Conversely, suppression or activation of pain-specific neurons in the VLO significantly affected pain but not itch. Moreover, fiber photometry and immunofluorescence verified that these itch- and pain-specific neurons are distinct in their functional activity and histological location. In addition, the downstream targets of itch- and pain-specific neurons were different. Together, the present study uncovers an important subpopulation of neurons in the VLO that specifically modulates itch processing.

5.
Neurobiol Dis ; 140: 104862, 2020 07.
Article in English | MEDLINE | ID: mdl-32251841

ABSTRACT

Although the anterior cingulate cortex (ACC) plays a vital role in neuropathic pain-related aversion, the underlying mechanisms haven't been fully studied. The mesolimbic dopamine system encodes reward and aversion, and participates in the exacerbation of chronic pain. Therefore, we investigated whether the ACC modulates aversion to neuropathic pain via control of the mesolimbic dopamine system, in a rat model of chronic constriction injury (CCI) to the sciatic nerve. Using anterograde and retrograde tracings, we confirmed that a subgroup of ACC neurons projected to the nucleus accumbens (NAc) and ventral tegmental area (VTA), which are two crucial nodes of the mesolimbic dopamine system. Combining electrophysiology in juvenile rats 7 days post-CCI, we found that the NAc/VTA-projecting neurons were hyperexcitable after CCI. Chemogenetic inhibition of these projections induced conditioned place preference in young adult rats 10-14 days post-CCI, without modulating the evoked pain threshold, whereas activation of these projections in sham rats mimicked aversive behavior. Furthermore, the function of the ACC projections was probably mediated by NAc D2-type medium spiny neurons and VTA GABAergic neurons. Taken together, our findings suggest that projections from the ACC to the NAc and VTA mediate neuropathic pain-related aversive behavior.


Subject(s)
Neuralgia/physiopathology , Nucleus Accumbens/physiopathology , Pain Threshold/physiology , Ventral Tegmental Area/physiopathology , Animals , Chronic Pain , Conditioning, Classical , Dopaminergic Neurons , Gyrus Cinguli/physiopathology , Male , Rats , Reward
7.
Neurosci Lett ; 516(1): 5-8, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22402191

ABSTRACT

Perinatal hypoxic-ischemic (H-I) is a major cause of brain injury in the newborn. The hippocampus is more sensitive to H-I injury than the other brain regions. It is believed that H-I brain damage causes a loss of neurons in the central nervous system. The patterns of neuronal death include apoptosis and necrosis. With regard to the responses of neurons, the neural functional changes should be earlier than the morphologic changes. The aim of the present study is to evaluate the electrophysiological characteristics and the synaptic transmission functions. Seven-day-old Sprague-Dawley rat pups were randomly divided into sham operation and H-I groups. The patch clamp, immunohistochemistry and Western blotting techniques were used to achieve this objective. The results of the study showed a decrease in neuronal excitability and a significant increase in the frequency of spontaneous excitatory postsynaptic currents and the duration of EPSCs in the CA1 pyramidal cells of H-I brain damage rats. The glutamate transporter subtype 1 (GLT-1) expression level of the hippocampal CA1 area in the H-I group was decreased compared with the control. There was no difference in the amplitude of excitatory postsynaptic currents and should be no difference in the expression of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR), N-methyl-D-aspartate receptor (NMDAR) and synaptophysin between the control and H-I brain injury group. These results revealed that changes of electrophysiological characteristics and synaptic functions occur instantly after H-I brain damage in the hippocampal pyramidal cells of neonatal rats. The failure to eliminate glutamate should be one of the important factors of excitotoxicity injury on hippocampal CA1 pyramidal cells, while neuronal excitation was not increased in the H-I brain injury model.


Subject(s)
Action Potentials , Hypoxia-Ischemia, Brain/physiopathology , Nerve Net/physiopathology , Pyramidal Cells , Synaptic Transmission , Animals , Animals, Newborn , Rats , Rats, Sprague-Dawley
8.
Glia ; 57(3): 307-19, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18942747

ABSTRACT

Oligodendrocyte precursor cells (OPCs) are the predominant oligodendrocyte-lineage stage in the cerebral hemispheres of neonatal rat. Prior studies have shown that OPCs are highly vulnerable to hypoxic-ischemic injury, yet the mechanisms are not well understood. P2X(7) receptor (P2X(7)R) is an ATP-gated ion channel that has unusual properties and plays very complex roles in a variety of neuropathologic conditions. However, little is known about the involvement of P2X(7)R in OPCs development and injury. The present study was aimed at examining the presence of P2X(7)R in OPCs and evaluating the change of the receptor expression after hypoxia ischemia. Using Immunofluorescence, RT-PCR, and western blot analysis, we demonstrated that OPCs expressed P2X(7)R in vitro and in vivo. Activation of P2X(7)R in OPCs in response to 3'-O-(4-benzoyl) benzoyl-ATP (BzATP) led to an increased mobilization of intracellular calcium [Ca(2+)]i, formation of large pores and cell death. These functional responses were sensitive to pretreatment of cells with the P2X(7)R antagonist, Brilliant Blue G (BBG, 100 nM), which was a selective antagonist for P2X(7)R in nanomole range. A decrease in P2X(7)R expression was observed in cultured OPCs after exposure to oxygen-glucose deprivation (OGD) for 2 h in vitro. Using a neonatal hypoxic-ischemic injury model in postnatal 3 rats, the similar downregulation was also detected in ischemic cerebral cortex, subcortical white matter and hippocampus compared with sham operation controls. In conclusion, the present data demonstrated that OPCs expressed functional P2X(7)R. The post-ischemic downregulation of P2X(7)R suggested a role for this receptor in the pathophysiology of hypoxic-ischemic brain injury.


Subject(s)
Adult Stem Cells/physiology , Down-Regulation/physiology , Hypoxia-Ischemia, Brain/pathology , Oligodendroglia/physiology , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Benzenesulfonates/pharmacology , Calcium/metabolism , Cell Death/physiology , Cells, Cultured , Glucose/deficiency , L-Lactate Dehydrogenase/metabolism , Oligodendroglia/drug effects , Platelet Aggregation Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7
9.
Brain Res ; 1243: 27-37, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-18824157

ABSTRACT

A widespread population of cells in CNS is identified by specific expression of the NG2 chondroitin sulphate proteoglycan and named as oligodendrocyte precursor cell (OPC). OPCs may possess stem cell-like characteristics, including multipotentiality in vitro and in vivo. It was proposed that OPCs in the CNS parenchyma comprise a unique population of glia, distinct from oligodendrocytes and astrocytes. This study confirmed that NG2 immunoreactive OPCs were continuously distributed in cerebral cortex and hippocampus during different postnatal developmental stages. These cells rapidly increased in number over the postnatal 7 days and migrate extensively to populate with abundant processes both in developing cortex and hippocampus. The morphology of OPCs exhibited extremely complex changes with the distribution of long distance primary process gradually increased from neonatal to adult CNS. Immunohistochemical studies showed that OPCs exhibited the morphological properties that can be distinguished from astrocytes. The electrophysiological properties showed that OPCs expressed a small amount of inward Na(+) currents which was distinguished from Na(+) currents in neurons owing to their lower Na-to-K conductance ratio and higher command voltage step depolarized maximum Na(+) current amplitude. These observations suggest that OPCs can be identified as the third type of macroglia because of their distribution in the CNS, the morphological development in process diversity and the electrophysiological difference from astrocyte.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Hippocampus/cytology , Hippocampus/growth & development , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Antigens/analysis , Antigens/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation , Cell Shape/physiology , Electrophysiology , Glial Fibrillary Acidic Protein/analysis , Glial Fibrillary Acidic Protein/metabolism , Image Cytometry , Membrane Potentials/physiology , Microscopy, Confocal , Oligodendroglia/cytology , Proteoglycans/analysis , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channels/metabolism , Stem Cells/cytology
10.
Pharmacology ; 82(2): 164-70, 2008.
Article in English | MEDLINE | ID: mdl-18679041

ABSTRACT

The effects of corticosterone (CORT), a natural glucocorticoid hormone, on ATP-induced currents in rat dorsal root ganglion (DRG) neurons and the underlying signaling mechanism were studied by using patch-clamp techniques. Three types of currents (fast, slow and mixed) were evoked by ATP in cultured DRG neurons. Pretreatment with CORT (0.01-10 mumol/l) for 30 s could inhibit the fast current and the fast component of the mixed current. In contrast, CORT had no significant effect on the slow current evoked by ATP. The inhibitory effects were concentration dependent, reversible and could be blocked by glucocorticoid receptor antagonist RU38486 (10 micromol/l), but not by GDP-beta-S (0.2 mmol/l), a blocker of G protein activation. Membrane-impermeable bovine serum albumin-conjugated corticosterone failed to mimic the effects of CORT. The inhibitory effects of CORT on ATP-induced currents diminished after adding protein kinase A inhibitor H89 (10 micromol/l), but were not influenced by protein kinase C inhibitor chelerythrine chloride (10 micromol/l). These results suggest that glucocorticoid hormones might participate in the control of pain by modulating P2X(3) receptor-mediated events in sensory neurons, and the effect is mediated by glucocorticoid receptors and the downstream activation of protein kinase A.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Corticosterone/pharmacology , Receptors, Purinergic P2/drug effects , Adenosine Triphosphate/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Cattle , Cells, Cultured , Corticosterone/administration & dosage , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Pain/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X3 , Serum Albumin, Bovine , Signal Transduction/drug effects
11.
Sheng Li Xue Bao ; 57(2): 181-7, 2005 Apr 25.
Article in English | MEDLINE | ID: mdl-15830102

ABSTRACT

To determine the characteristics of spontaneous discharges of hippocampal pyramidal cells (PCs), extracellular neuronal firing in CA1 and CA3 regions of dorsal hippocampus was recorded, the firing modes and interspike interval (ISI) were analyzed with the conventional and nonlinear methods. PCs were discriminated from interneurons using the measurement of action-potential duration and firing rate in this study. There was no significant difference in duration, mean firing frequency, complexity and firing mode between the neurons in CA1 and CA3 regions both in anesthetized and awake animals. The complexity of neurons was higher in awake group than that in anesthetized group, though no difference was found in firing rate. There were differences in the type of pyramidal cells and the coefficient of variance of ISI of neurons. The results obtained from the spontaneous discharges of dorsal hippocampal pyramidal cells reveal some nonlinear and linear aspects in anesthetized and awake states. It seems likely that the combination of conventional and non-linear measurements of the hippocampal pyramidal cells encoding may reflect genuine characteristics of the hippocampal pyramidal cells.


Subject(s)
Action Potentials/physiology , Hippocampus/physiology , Pyramidal Cells/physiology , Animals , Female , Guinea Pigs , Male , Microelectrodes , Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...