Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-36917193

ABSTRACT

Chronic pain can cause both hyperalgesia and anxiety symptoms. However, how the two components are encoded in the brain remains unclear. The prelimbic cortex (PrL), a critical brain region for both nociceptive and emotional modulations, serves as an ideal medium for comparing how the two components are encoded. We report that PrL neurons projecting to the basolateral amygdala (PrLBLA) and those projecting to the ventrolateral periaqueductal gray (PrLl/vlPAG) were segregated and displayed elevated and reduced neuronal activity, respectively, during pain chronicity. Consistently, optogenetic suppression of the PrL-BLA circuit reversed anxiety-like behaviors, whereas activation of the PrL-l/vlPAG circuit attenuated hyperalgesia in mice with chronic pain. Moreover, mechanistic studies indicated that elevated TNF-α/TNFR1 signaling in the PrL caused increased insertion of GluA1 receptors into PrLBLA neurons and contributed to anxiety-like behaviors in mice with chronic pain. Together, these results provide insights into the circuit and molecular mechanisms in the PrL for controlling pain-related hyperalgesia and anxiety-like behaviors.


Subject(s)
Basolateral Nuclear Complex , Chronic Pain , Mice , Animals , Chronic Pain/genetics , Hyperalgesia , Anxiety/genetics , Cerebral Cortex
2.
Nat Commun ; 14(1): 729, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36759610

ABSTRACT

Alterations in energy metabolism are associated with depression. However, the role of glycolysis in the pathogenesis of depression and the underlying molecular mechanisms remain unexplored. Through an unbiased proteomic screen coupled with biochemical verifications, we show that the levels of glycolysis and lactate dehydrogenase A (LDHA), a glycolytic enzyme that catalyzes L-lactate production, are reduced in the dorsomedial prefrontal cortex (dmPFC) of stress-susceptible mice in chronic social defeat stress (CSDS) model. Conditional knockout of LDHA from the brain promotes depressive-like behaviors in both male and female mice, accompanied with reduced L-lactate levels and decreased neuronal excitability in the dmPFC. Moreover, these phenotypes could be duplicated by knockdown of LDHA in the dmPFC or specifically in astrocytes. In contrast, overexpression of LDHA reverses these phenotypic changes in CSDS-susceptible mice. Mechanistic studies demonstrate that L-lactate promotes neuronal excitability through monocarboxylic acid transporter 2 (MCT2) and by inhibiting large-conductance Ca2+-activated potassium (BK) channel. Together, these results reveal a role of LDHA in maintaining neuronal excitability to prevent depressive-like behaviors.


Subject(s)
Astrocytes , Lactic Acid , Mice , Male , Female , Animals , Lactate Dehydrogenase 5/metabolism , Astrocytes/metabolism , L-Lactate Dehydrogenase/metabolism , Proteomics , Carrier Proteins
3.
Int J Neurosci ; : 1-8, 2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35880488

ABSTRACT

Objective: Depression is a common psychological and physiological disease in the world, which seriously affects the quality of life of patients and families. Exercise is an economic and noninvasive antidepressant measure, which has been widely recognized and applied in daily life and clinical practice, and the related mechanism research has also been paid attention to. In recent years, a new research report pointed out that peripheral administration of L-lactate can reverse depression-like behavior in mice, which suggesting that the lactic acid produced during exercise may be one of the factors leading to antidepressant effect, but the detailed mechanism is not clear. Inflammation is the pathogenic factor of many diseases and a large number of experiments have proved that inflammation is also an important pathogenic factor leading to depression. The purpose of our experiment is to explore whether lactic acid has anti-inflammatory and antidepressant effects.Methods: Based on the LPS induced inflammatory model, animal behavior observation, protein extraction, Western blotting, immunofluorescence and other techniques were used in this experiment.Results: Lactic acid could inhibit the change of some important inflammatory factors, such as TNF-αIL-1ßphospho-NF-κB (p-NF-κB) and NLRP3 inflammasome complex (NLRP3/ASC/caspase-1) induced by LPS.Conclusion: Our current research suggested that lactic acid maybe exert antiinflammatory effect by inhibiting inflammatory factors.

4.
Neuroscience ; 448: 94-106, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32946950

ABSTRACT

Neuroinflammation contributes to neuronal death in cerebral ischemia. Urolithin A (UA), a gut microbial metabolite of ellagic acid, has emerged as a potential anti-inflammatory agent. However, its roles and precise mechanisms in stroke remain unknown. Here we found that UA treatment ameliorated infarction, neurological deficit scores, and spatial memory deficits after cerebral ischemia. Furthermore, UA significantly reduced neuron loss and promoted neurogenesis after ischemic stroke. We also found that UA attenuated apoptosis by regulating apoptotic-related proteins. Meanwhile, UA treatment inhibited glial activation via affecting inflammatory signaling pathways, specifically by enhancing cerebral AMPK and IκBa activation while decreasing the activation of Akt, P65NFκB, ERK, JNK, and P38MAPK. Our findings reveal a key role of UA against ischemic stroke through modulating apoptosis and neuroinflammation in mice.


Subject(s)
Brain Ischemia , Stroke , Animals , Apoptosis , Brain Ischemia/drug therapy , Coumarins/pharmacology , Mice , Signal Transduction , Stroke/drug therapy
5.
Front Neurosci ; 14: 62, 2020.
Article in English | MEDLINE | ID: mdl-32116509

ABSTRACT

Fear extinction remains an unresolved challenge for behavioral exposure therapy in patients with post-traumatic stress disorder (PTSD). Previous reports have suggested that social support from either familiar or unfamiliar same-sex partners is beneficial to attenuating fear responses during fear extinction and renewal. Despite that, few studies have examined the effects of social support in advance on fear extinction and/or retrieval. It is also not clear whether social company by a receptive mating partner in advance facilitates fear extinction. In the present study, we address these questions by introducing a co-housing method, where fear-conditioned male mice are co-housed with or without a receptive mating partner prior to fear extinction. We found that while co-housing with an ovariectomized female mouse showed little effect on fear extinction or retrieval, social company by a receptive mating partner in advance dramatically facilitates fear extinction. In addition, the number of cFos-positive neurons in the basolateral amygdala (BLA) were also found to be reduced in male mice accompanied with receptive mating partner in response to fear extinction and retrieval, indicating diminished neuronal activation. Electrophysiological studies further showed that the excitability of excitatory neurons in BLA was decreased, which is probably due to the attenuated basal level of excitatory synaptic transmission. Together, our observations demonstrate an effect of social company by a receptive mating partner can facilitate fear extinction and afford a possible cellular mechanism.

6.
Brain Behav ; 9(4): e01246, 2019 04.
Article in English | MEDLINE | ID: mdl-30848079

ABSTRACT

INTRODUCTION: Beyond its application as an epilepsy therapy, the ketogenic diet (KD) has been considered a potential treatment for a variety of other neurological and metabolic disorders. However, whether KD promotes functional restoration by reducing the pathological processes underlying individual diseases or through some independent mechanisms is not clear. METHODS: In this study, we evaluated the effect of KD on a series of behaviors and synaptic functions of young adult naive mice. Wild-type C57BL/6J mice at age of 2-3 months were fed with control diet or KD for three months. Body weight and caloric intake were monitored throughout the experiments. We assessed behavioral performance with seizure induction, motor coordination and activity, anxiety level, spatial learning and memory, sociability, and depression. Synaptic transmission and long-term potentiation were also recorded. RESULTS: KD-fed mice performed equivalent to control-diet-fed mice in the behavioral tests and electrophysiological assays except exhibiting slower weight gain and increased seizure threshold. CONCLUSIONS: Our results contribute to the better understanding of effects of the KD on physiological behaviors and synaptic functions.


Subject(s)
Behavior, Animal/physiology , Brain/physiopathology , Diet, Ketogenic , Long-Term Potentiation/physiology , Seizures/physiopathology , Animals , Body Weight , Male , Mice , Mice, Inbred C57BL , Synaptic Transmission/physiology
7.
J Neurosci ; 38(44): 9600-9613, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30228230

ABSTRACT

Fear learning and memory are vital for livings to survive, dysfunctions in which have been implicated in various neuropsychiatric disorders. Appropriate neuronal activation in amygdala is critical for fear memory. However, the underlying regulatory mechanisms are not well understood. Here we report that Neogenin, a DCC (deleted in colorectal cancer) family receptor, which plays important roles in axon navigation and adult neurogenesis, is enriched in excitatory neurons in BLA (Basolateral amygdala). Fear memory is impaired in male Neogenin mutant mice. The number of cFos+ neurons in response to tone-cued fear training was reduced in mutant mice, indicating aberrant neuronal activation in the absence of Neogenin. Electrophysiological studies show that Neogenin mutation reduced the cortical afferent input to BLA pyramidal neurons and compromised both induction and maintenance of Long-Term Potentiation evoked by stimulating cortical afferent, suggesting a role of Neogenin in synaptic plasticity. Concomitantly, there was a reduction in spine density and in frequency of miniature excitatory postsynaptic currents (mEPSCs), but not miniature inhibitory postsynaptic currents, suggesting a role of Neogenin in forming excitatory synapses. Finally, ablating Neogenin in the BLA in adult male mice impaired fear memory likely by reducing mEPSC frequency in BLA excitatory neurons. These results reveal an unrecognized function of Neogenin in amygdala for information processing by promoting and maintaining neurotransmission and synaptic plasticity and provide insight into molecular mechanisms of neuronal activation in amygdala.SIGNIFICANCE STATEMENT Appropriate neuronal activation in amygdala is critical for information processing. However, the underlying regulatory mechanisms are not well understood. Neogenin is known to regulate axon navigation and adult neurogenesis. Here we show that it is critical for neurotransmission and synaptic plasticity in the amygdala and thus fear memory by using a combination of genetic, electrophysiological, behavioral techniques. Our studies identify a novel function of Neogenin and provide insight into molecular mechanisms of neuronal activation in amygdala for fear processing.


Subject(s)
Basolateral Nuclear Complex/metabolism , Fear/physiology , Learning/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neurons/metabolism , Animals , Excitatory Postsynaptic Potentials/physiology , Fear/psychology , Male , Mice , Mice, Transgenic , Organ Culture Techniques
8.
Am J Physiol Cell Physiol ; 314(3): C278-C288, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29141922

ABSTRACT

receptor potential canonical (TRPC) channels are presently an emerging target for airway disorders. Recent evidence has indicated that TRPC6 as a member of the TRPC family plays an important role in airway inflammation, but its precise function in bronchial epithelial cells remains unclear. The aim of this study was to investigate the role of TRPC6 in Toll-like receptor 4 (TLR4)-mediated inflammation in human bronchial epithelial cells stimulated by endotoxin [lipopolysaccharide (LPS)]. Hyp9 is a simplified phloroglucinol derivative of hyperforin that highly selectively activates TRPC6 channels. The results show that the activation of TRPC6 by Hyp9 induced the production of interleukin (IL)-8 and IL-6. LPS was also able to induce the release of IL-8 and IL-6, which was significantly aggravated by Hyp9 and reduced by knockdown of TRPC6. Treatment with LPS not only chronically induced the expression of TRPC6 mRNA and protein in a TLR4-dependent manner but also acutely increased Ca2+ influx through TRPC6 channels. In addition, LPS-induced overexpression of TRPC6 and Ca2+ influx were associated with the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and Akt. Importantly, TRPC6 was required for the activation of ERK1/2, p38, and NF-κB. In conclusion, these data reveal that LPS induced the overexpression of TRPC6 and TRPC6-dependent Ca2+ influx via the TLR4/PI3K/Akt pathway resulting in Ca2+ mobilization, which subsequently promoted the activation of ERK1/2, p38, and NF-κB and the inflammatory response in bronchial epithelial cells.


Subject(s)
Bronchi/diagnostic imaging , Epithelial Cells/drug effects , Inflammation/chemically induced , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , TRPC6 Cation Channel/agonists , p38 Mitogen-Activated Protein Kinases/metabolism , Bronchi/enzymology , Calcium Signaling/drug effects , Cell Line , Cytokines/metabolism , Epithelial Cells/enzymology , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation Mediators/metabolism , NF-kappa B/metabolism , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism , Terpenes/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism
9.
Int J Radiat Biol ; 93(3): 270-278, 2017 03.
Article in English | MEDLINE | ID: mdl-27809636

ABSTRACT

PURPOSE: Breast cancer is the most common cancer among women and radiotherapy is a conventional therapy following surgery. Previous studies have demonstrated that except the caspase-dependent pathway, caspase-independent pathway is also involved in the cell death responding to irradiation, despite the unclear mechanism. The purpose of the present study was to observe the role of apoptosis-inducing factor (AIF), the first identified caspase-independent molecule, in X-ray-induced breast cancer cell (MCF-7) cell death. MATERIALS AND METHODS: In this study, WST-1 assay, DAPI nuclear staining and clonogenic survival assay were used to test the cell response to different treatments; Western blot was used to detect the protein expression; RT-PCR and plasmid transfection were used to observe the role of AIF. RESULTS: X-ray-induced AIF transferred from the mitochondrion to the nucleus. Inhibition of AIF expression reduced X-ray-induced MCF-7 cell death. Further, AIF nuclear translocation is in a caspase-independent manner in this process, but not caspase-dependent manner. CONCLUSIONS: The present study revealed that AIF nuclear translocation proceeded in X-ray-induced MCF-7 cell death in a caspase-independent manner.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/physiology , Apoptosis/radiation effects , Caspases/metabolism , Cell Nucleus/metabolism , X-Rays , Active Transport, Cell Nucleus/radiation effects , Cell Nucleus/radiation effects , Dose-Response Relationship, Radiation , Humans , MCF-7 Cells , Radiation Dosage
10.
Biochem Biophys Res Commun ; 472(1): 137-43, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26920061

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most common cancer in the world. The aim of radiotherapy is to eradicate cancer cells with ionizing radiation. Except for the caspase-dependent mechanism, several lines of evidence demonstrated that caspase-independent mechanism is directly involved in the cell death responding to irradiation. For this reason, defining the contribution of caspase-independent molecular mechanisms represents the main goal in radiotherapy. In this study, we focused on the role of apoptosis-inducing factor (AIF), the caspase-independent molecular, in ionizing radiation induced hepatocellular carcinoma cell line (HepG2) cell death. We found that ionizing radiation has no function on AIF expression in HepG2 cells, but could induce AIF release from the mitochondria and translocate into nuclei. Inhibition of AIF could reduce ionizing radiation induced HepG2 cell death. These studies strongly support a direct relationship between AIF nuclear translocation and radiation induced cell death. What's more, AIF nuclear translocation is caspase-independent manner, but not caspase-dependent manner, in this process. These new findings add a further attractive point of investigation to better define the complex interplay between caspase-independent cell death and radiation therapy.


Subject(s)
Apoptosis Inducing Factor/metabolism , Carcinoma, Hepatocellular/radiotherapy , Caspases/metabolism , Cell Death/physiology , Cell Death/radiation effects , Liver Neoplasms/radiotherapy , Active Transport, Cell Nucleus/radiation effects , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis Inducing Factor/antagonists & inhibitors , Apoptosis Inducing Factor/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Caspase Inhibitors/pharmacology , Cell Death/drug effects , Dose-Response Relationship, Radiation , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , RNA, Small Interfering/genetics
11.
Neurochem Res ; 38(10): 2216-26, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23974726

ABSTRACT

The destruction of calcium homeostasis is an important factor leading to neurological diseases. Store-operated Ca(2+) (SOC) channels are essential for Ca(2+) homeostasis in many cell types. However, whether SOC channels are involved in astrocyte activation induced by lipopolysaccharide (LPS) still remains unknown. In this study, we used LPS as an exogenous stimulation to investigate the role of SOC channels in astrocyte activation. Using calcium imaging technology, we first found that SOC channels blockers, 1-[h-[3-(4-methoxyphenyl)propoxy]-4-methoxyphenethyl]-1H-imidazole (SKF-96365) and 2-aminoethyldiphenyl borate (2-APB), inhibited LPS induced [Ca(2+)]i increase, which prompted us to speculate that SOC channels may be involved in LPS induced astrocyte activation. Further experiments confirmed our speculation shown as SOC channels blockers inhibited LPS induced astrocyte activation characterized as cell proliferation by MTS and BrdU assay, raise in glial fibrillary acidic protein expression by immunofluorescence and Western Blot and secretion of interleukin 6 (IL-6) and interleukin 1ß (IL-1ß) by ELISA. So, our studies showed that SOC channels are involved in LPS-induced astrocyte activation.


Subject(s)
Astrocytes/physiology , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Animals , Astrocytes/drug effects , Boron Compounds/pharmacology , Calcium Channels/drug effects , Calcium Signaling/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Glial Fibrillary Acidic Protein/biosynthesis , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Rats , Rats, Wistar
12.
Neurochem Int ; 63(8): 826-31, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23770272

ABSTRACT

Hydrogen sulfide (H2S), an endogenous gaseous mediator, has been shown to have protective effects against neuronal damage caused by brain ischemia. In this study, we explored the potential effects of H2S on oxygen-glucose deprivation/reoxygenation (OGD/R)-induced neuronal apoptosis and the possible mechanisms. We find that sodium hydrosulfide (NaHS, a donator of H2S) prevents OGD/R-induced intracellular reactive oxygen species (ROS) elevation and activation of caspase-3 in cultured mouse cortical neurons. The pretreatment of N-acetyl-l-cysteine (NAC, an ROS scavenger) also prevents OGD/R-induced activation of caspase-3. Both NaHS and NAC counteract OGD/R-induced decline in mitochondria membrane potential (MMP). Additionally, NaHS, NAC or N-Acetyl-Asp-Glu-Val-Asp-CHO (DEVD-CHO, a caspase-3 inhibitor), is shown to significantly inhibit OGD/R-induced neuronal apoptosis. These data suggest that H2S can protect against OGD/R-induced neuronal apoptosis through improving mitochondria dysfunction and suppressing an ROS-activated caspase-3 signaling pathway.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Cerebral Cortex/drug effects , Hydrogen Sulfide/pharmacology , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Animals , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Matrix Metalloproteinases/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism
13.
Neurochem Res ; 38(3): 581-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23277413

ABSTRACT

Humanin (HN) has been proved to be an extensive neuroprotective peptide against AD-related and unrelated insults, but little is know about the effect of HN in inflammation response. Current studies indicated the receptors of HN have a close relationship with immune system, which led us to hypothesize HN might have a role in inflammatory response. In this study, we used lipopolysaccharide (LPS) to induce astrocyte inflammation response. This model in vitro allowed us to study the effect of HN on the pure response of astrocyte without the exogenous influence between cells in vivo. Our results showed that 1.0 µg/ml LPS induced a significant activation of astrocyte, shown as the marked increase in the glial fibrillary acidic protein (GFAP) expression, the cell viability and the number of 5-bromo-2'-deoxyuridine (BrdU)-positive living cells. Pretreatment with HN (5, 10, 20 µM) led to a significant inhibition in astrocyte overactivation in a concentration dependent manner. We also found pretreatment with HN decreased the level of proinflammatory cytokines, interleukin (IL)-6, IL-1ß and tumor necrosis factor α (TNFα) induced by LPS. Furthermore, we noticed HN couldn't completely reverse the above inflammatory injury. Our findings imply that HN partly antagonizes inflammation injury induced by LPS and the protective effect of HN on astrocyte is concentration-dependent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Astrocytes/drug effects , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Lipopolysaccharides/pharmacology , Neuroprotective Agents/pharmacology , Animals , Bromodeoxyuridine/metabolism , Female , Glial Fibrillary Acidic Protein/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-6/metabolism , Male , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/biosynthesis
14.
Neurochem Res ; 37(1): 153-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21935731

ABSTRACT

The neuroprotective effects of superoxide dismutase (SOD) against hypoxia/reperfusion (I/R) injury and of humanin (HN) against toxicity by familial amyotrophic lateral sclerosis (ALS)-related mutant SOD led us to hypothesize that HN might have a role to increase the activity of SOD, which might be involved in the protective effects of HN on neuron against Alzheimer's disease-unrelated neurotoxicities. In the present study, we found that 4 h ischemia and 24 h reperfusion induced a significant increase in lactate dehydrogenase (LDH) release, malondialdehyde (MDA) formation and the number of karyopyknotic nuclei (4',6-diamidino-2-phenylindole dihydrochloride nuclear dyeing) and a decrease in the number of Calcein-AM-positive living cells and cell viability. Pretreatment of the cells with HN led to a significant decrease in LDH release, MDA formation and the number of karyopyknotic nuclei, and an increase in the number of Calcein-AM-positive living cells and cell viability in neurons treated with I/R. We also found a significant decrease in SOD activity in neurons treated with I/R only, while pre-treatment with HN before I/R induced a significant increase in the activity of SOD as compared with the I/R group. Our findings implied that HN protects cortical neurons from I/R injury by the increased SOD activity and that the protective effect of HN on neurons against I/R is concentration-dependent.


Subject(s)
Cerebral Cortex/cytology , Intracellular Signaling Peptides and Proteins/physiology , Neurons/cytology , Reperfusion Injury/pathology , Superoxide Dismutase/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Neurons/enzymology , Neurons/metabolism
15.
BMC Neurosci ; 10: 113, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19737385

ABSTRACT

BACKGROUND: Caspase-independent apoptotic pathways are suggested as a mechanism for the delayed neuronal death following ischemic insult. However, the underlying signalling mechanisms are largely unknown. Recent studies imply the involvement of several mitochondrial proteins, including endonuclease G (EndoG) and Bcl-2/adenovirus E1B 19 kDa-interacting protein (BNIP3), in the pathway of non-neuronal cells. RESULTS: In this report, using western blot analysis and immunocytochemistry, we found that EndoG upregulates and translocates from mitochondria to nucleus in a time-dependent manner in cultured hippocampal neurons following oxygen-glucose deprivation (OGD). Moreover, the translocation of EndoG occurs hours before the observable nuclear pyknosis. Importantly, the mitochondrial upregulation of BNIP3 precedes the translocation of EndoG. Forced expression of BNIP3 increases the nuclear translocation of EndoG and neuronal death while knockdown of BNIP3 decreases the OGD-induced nuclear translocation of EndoG and neuronal death. CONCLUSION: These results suggest that BNIP3 and EndoG play important roles in hippocampal neuronal apoptosis following ischemia, and mitochondrial BNIP3 is a signal protein upstream of EndoG that can induce neuronal death.


Subject(s)
Apoptosis/physiology , Endodeoxyribonucleases/physiology , Hippocampus/physiology , Membrane Proteins/physiology , Mitochondrial Proteins/physiology , Neurons/physiology , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/genetics , Blotting, Western , Cell Hypoxia , Cells, Cultured , Endodeoxyribonucleases/metabolism , Glucose/deficiency , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Protein Transport/genetics , Protein Transport/physiology , RNA Interference , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Subcellular Fractions/physiology , Transfection , Up-Regulation/genetics , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...