2.Naegleria fowleri Lysate Induces Strong Cytopathic Effects and Pro-inflammatory Cytokine Release in Rat Microglial Cells.
Yang Jin LEE ; Chang Eun PARK ; Jong Hyun KIM ; Hae Jin SOHN ; Jinyoung LEE ; Suk Yul JUNG ; Ho Joon SHIN
The Korean Journal of Parasitology 2011;49(3):285-290
Naegleria fowleri, a ubiquitous free-living ameba, causes fatal primary amebic meningoencephalitis in humans. N. fowleri trophozoites are known to induce cytopathic changes upon contact with microglial cells, including necrotic and apoptotic cell death and pro-inflammatory cytokine release. In this study, we treated rat microglial cells with amebic lysate to probe contact-independent mechanisms for cytotoxicity, determining through a combination of light microscopy and scanning and transmission electron microscopy whether N. fowleri lysate could effect on both necrosis and apoptosis on microglia in a time- as well as dose-dependent fashion. A 51Cr release assay demonstrated pronounced lysate induction of cytotoxicity (71.5%) toward microglial cells by 24 hr after its addition to cultures. In an assay of pro-inflammatory cytokine release, microglial cells treated with N. fowleri lysate produced TNF-alpha, IL-6, and IL-1beta, though generation of the former 2 cytokines was reduced with time, and that of the last increased throughout the experimental period. In summary, N. fowleri lysate exerted strong cytopathic effects on microglial cells, and elicited pro-inflammatory cytokine release as a primary immune response.
Animals
;
*Cell Death
;
Chromium Radioisotopes/metabolism
;
Cytokines/*secretion
;
Humans
;
Microglia/cytology/immunology/*physiology
;
Microscopy
;
Naegleria fowleri/*pathogenicity
;
Rats
;
Staining and Labeling
3.Effects of electromagnetic pulse exposure on the morphological change and excretion function of BV-2 cells and possible mechanism.
Long-long YANG ; Yan ZHOU ; Hai-juan LI ; Juan GUO ; Yan-jun ZHANG ; Gui-rong DING ; Guo-zhen GUO
Chinese Journal of Industrial Hygiene and Occupational Diseases 2012;30(3):163-167
OBJECTIVETo study the effects of electromagnetic pulse (EMP) exposure on the morphological change and excretion functions of mouse microglia (BV-2) cells and possible mechanism.
METHODSBV-2 cells were divided into two groups: the group exposed to EMP at 200 kV/m for 200 pulses and sham exposure group. At 1, 6, 12 and 24 hour after exposure the cells and culture supernatant were collected. Cellular morphological change was observed under invert microscope, the levels of TNF-α, IL-1β and IL-10 in culture supernatant were determined by enzyme-linked immunosorbent assay (ELISA), nitric oxide (NO) and reactive oxygen species (ROS) were detected by nitrate reductase method and DCFH-DA probe, respectively. The protein and phosphorylation levels of ERK, JNK and p38 were measured by Western Blot method. After the cells pre-treated with the inhibitor of p38 (SB203580) were exposed to EMP, the levels of NO and ROS in culture supernatant were detected.
RESULTSIt was found that the large ameboid shape appeared in some microglia cells exposed to EMP for 1, 6 and 12 h. Moreover, the number of microglia cells with ameboid shape increased significantly at 1 h, 6 h and 12 h after EMP exposure compared with sham group (P < 0.05). The levels of cytokines, such as TNF-α, IL-1β and IL-10, in culture supernatant did not change obviously after EMP exposure. The levels of NO and ROS increased significantly at 1h after EMP exposure, reached the peak at 6 h, began to recover at 12 h and recovered to sham group level at 24 h (P < 0.05). Western blot results showed that the protein and protein phosphorylation levels of ERK and JNK did not change significantly after EMP exposure, however, the protein and protein phosphorylation levels of p38 increased obviously at 1 h and 6 h after EMP exposure, compared with sham group (P < 0.05). In addition, the pretreatment of p38 inhibitor (SB203580) significantly decreased NO and ROS production induced by EMP.
CONCLUSIONEMP exposure may activate microglia cells and promote the production of NO and ROS in mouse microglia cells, and p38 pathway is involved in this process.
Animals ; Cell Line ; Cytokines ; secretion ; Electromagnetic Fields ; Mice ; Microglia ; cytology ; metabolism ; secretion ; Nitric Oxide ; metabolism ; Reactive Oxygen Species ; metabolism ; p38 Mitogen-Activated Protein Kinases ; metabolism
4.Intrathecal Lamotrigine Attenuates Antinociceptive Morphine Tolerance and Suppresses Spinal Glial Cell Activation in Morphine-Tolerant Rats.
In Gu JUN ; Sung Hoon KIM ; Yang In YOON ; Jong Yeon PARK
Journal of Korean Medical Science 2013;28(2):300-307
Glial cells play a critical role in morphine tolerance, resulting from repeated administration of morphine. Both the development and the expression of tolerance are suppressed by the analgesic lamotrigine. This study investigated the relationship between the ability of lamotrigine to maintain the antinociceptive effect of morphine during tolerance development and glial cell activation in the spinal cord. In a rat model, morphine (15 microg) was intrathecally injected once daily for 7 days to induce morphine tolerance. Lamotrigine (200 microg) was co-administered with morphine either for 7 days or the first or last 3 days of this 7 day period. Thermal nociception was measured. OX-42 and GFAP immunoreactivity, indicating spinal microglial and astrocytic activation were evaluated on day 8. Tolerance developed after 7 days of intrathecal morphine administration; however, this was completely blocked and reversed by co-administration of lamotrigine. When lamotrigine was coinjected with morphine on days 5-7, the morphine effect was partially restored. Glial cell activation increased with the development of morphine tolerance but was clearly inhibited in the presence of lamotrigine. These results suggest that, in association with the suppression of spinal glial cell activity, intrathecally coadministered lamotrigine attenuates antinociceptive tolerance to morphine.
Analgesics/*pharmacology
;
Animals
;
Antigens, CD11b/metabolism
;
Astrocytes/cytology
;
Drug Tolerance
;
Immunohistochemistry
;
Male
;
Microglia/cytology
;
Morphine/*pharmacology
;
Nerve Tissue Proteins/metabolism
;
Neuroglia/cytology/*metabolism
;
Rats
;
Rats, Sprague-Dawley
;
Spinal Cord/*cytology
;
Triazines/*pharmacology
5.Ganoderma lucidum extract protects dopaminergic neurons through inhibiting the production of inflammatory mediators by activated microglia.
Hui DING ; Ming ZHOU ; Rui-Ping ZHANG ; Sheng-Li XU
Acta Physiologica Sinica 2010;62(6):547-554
Abundant evidence has suggested that neuroinflammation participates in the pathogenesis of Parkinson's disease (PD). The emerging evidence has supported that microglia may play key roles in the progressive neurodegeneration in PD and might be a promising therapeutic target. Ganoderma lucidum (GL), a traditional Chinese medicinal herb, has been shown potential neuroprotective effect in our clinical trials that lead us to speculate that it might possess potent anti-inflammatory and immunomodulating properties. To test this hypothesis, the present study investigated the potential neuroprotective effect of GL and underlying mechanism through inhibiting microglial activation using co-cultures of dopaminergic neurons and microglia. The cultures of microglia or MES23.5 cells alone or together were treated for 24 h with lipopolysaccharide (LPS, 0.25 μg/mL) as a positive control, GL extracts (50-400 μg/mL) or MES23.5 cell membrane fragments (150 μg/mL) were used in treatment groups. Microglia activation, microglia-derived harmful factors and [(3)H]dopamine ([(3)H]DA) uptake of MES23.5 cells were analyzed. The results showed that microglia were activated by LPS and MPP(+)-treated MES23.5 cell membrane fragments, respectively. Meanwhile, GL extracts significantly prevented the production of microglia-derived proinflammatory and cytotoxic factors, including nitric oxide, tumor necrosis factor-α (TNF-α) and interleukin 1β (IL-1β), in a dose-dependent manner and down-regulated the TNF-α and IL-1β expressions on mRNA level. In addition, GL extracts antagonized the reduction of [(3)H]DA uptake induced by MPP(+) and microglial activation. In conclusion, these results suggest that GL may be a promising agent for the treatment of PD through anti-inflammation.
Cell Line
;
Dopaminergic Neurons
;
cytology
;
drug effects
;
Down-Regulation
;
drug effects
;
Humans
;
Interleukin-1beta
;
metabolism
;
Materia Medica
;
pharmacology
;
Microglia
;
cytology
;
metabolism
;
Neuroprotective Agents
;
pharmacology
;
Nitric Oxide
;
metabolism
;
Parkinson Disease
;
physiopathology
;
Reishi
;
chemistry
;
Tumor Necrosis Factor-alpha
;
metabolism
6.Attenuation of Spinal Cord Injury-Induced Astroglial and Microglial Activation by Repetitive Transcranial Magnetic Stimulation in Rats.
Ji Young KIM ; Gyu Sik CHOI ; Yun Woo CHO ; Heekyung CHO ; Se Jin HWANG ; Sang Ho AHN
Journal of Korean Medical Science 2013;28(2):295-299
Spinal cord injury (SCI) causes not only loss of sensory and motor function below the level of injury but also chronic pain, which is difficult and challenging of the treatment. Repetitive transcranial magnetic stimulation (rTMS) to the motor cortex, of non-invasive therapeutic methods, has the motor and sensory consequences and modulates pain in SCI-patients. In the present study, we studied the effectiveness of rTMS and the relationship between the modulation of pain and the changes of neuroglial expression in the spinal cord using a rat SCI-induced pain model. Elevated expressions of Iba1 and GFAP, specific microglial and astrocyte markers, was respectively observed in dorsal and ventral horns at the L4 and L5 levels in SCI rats. But in SCI rats treated with 25 Hz rTMS for 8 weeks, these expressions were significantly reduced by about 30%. Our finding suggests that this attenuation of activation by rTMS is related to pain modulation after SCI. Therefore, rTMS might provide an alternative means of attenuating neuropathic pain below the level of SCI.
Animals
;
Astrocytes/*cytology
;
Calcium-Binding Proteins/metabolism
;
Disease Models, Animal
;
Immunohistochemistry
;
Male
;
Microfilament Proteins/metabolism
;
Microglia/*cytology
;
Nerve Tissue Proteins/metabolism
;
Neuralgia/etiology
;
Rats
;
Rats, Sprague-Dawley
;
Spinal Cord Injuries/complications/pathology/*therapy
;
*Transcranial Magnetic Stimulation
7.Minocycline attenuates microglial response and reduces neuronal death after cardiac arrest and cardiopulmonary resuscitation in mice.
Qian-yan WANG ; Peng SUN ; Qing ZHANG ; Shang-long YAO
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(2):225-229
The possible role of minocycline in microglial activation and neuronal death after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) in mice was investigated in this study. The mice were given potassium chloride to stop the heart beating for 8 min to achieve CA, and they were subsequently resuscitated with epinephrine and chest compressions. Forty adult C57BL/6 male mice were divided into 4 groups (n=10 each): sham-operated group, CA/CPR group, CA/CPR+minocycline group, and CA/CPR+vehicle group. Animals in the latter two groups were intraperitoneally injected with minocycline (50 mg/kg) or vehicle (normal saline) 30 min after recovery of spontaneous circulation (ROSC). Twenty-four h after CA/CPR, the brains were removed for histological evaluation of the hippocampus. Microglial activation was evaluated by detecting the expression of ionized calcium-binding adapter molecule-1 (Iba1) by immunohistochemistry. Neuronal death was analyzed by hematoxylin and eosin (H&E) staining and the levels of tumor necrosis factor-alpha (TNF-α) in the hippocampus were measured by enzyme-linked immunosorbent assay (ELISA). The results showed that the neuronal death was aggravated, most microglia were activated and TNF-α levels were enhanced in the hippocampus CA1 region of mice subjected to CA/CPR as compared with those in the sham-operated group (P<0.05). Administration with minocycline 30 min after ROSC could significantly decrease the microglial response, TNF-α levels and neuronal death (P<0.05). It was concluded that early administration with minocycline has a strong therapeutic potential for CA/CPR-induced brain injury.
Animals
;
Cardiopulmonary Resuscitation
;
Cell Death
;
drug effects
;
Enzyme-Linked Immunosorbent Assay
;
Heart Arrest
;
pathology
;
Hippocampus
;
cytology
;
drug effects
;
metabolism
;
Male
;
Mice
;
Mice, Inbred C57BL
;
Microglia
;
cytology
;
drug effects
;
Minocycline
;
pharmacology
;
Neurons
;
drug effects
;
Tumor Necrosis Factor-alpha
;
metabolism
9.Effect of hyperoxia exposure on the function of N9 microglia in vitro.
Pu JIANG ; Ying XU ; Liangan HU ; Yang LIU ; Shixiong DENG
Journal of Southern Medical University 2012;32(1):71-74
OBJECTIVETo observe the effect of normobaric hyperoxia exposure on the functions of N9 microglia and explore the underlying mechanism of hyperoxia-induced immature brain injury.
METHODSN9 microglial cells were exposed to 900 ml/L O(2) for 2, 6, 12, 24 or 48 h, and the cell apoptotic rate was assessed using flow cytometry. The intracellular oxidative stress was measured using a fluorescent DCFH-DA probe, and the expression of Toll-like receptor 4 (TLR4) mRNA was detected using RT-PCR. Interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) concentrations in the supernatant of the cell cultures were tested with ELISA following the exposures. TLR4 protein expression was observed using immunofluorescence staining.
RESULTSSignificant cell apoptosis was detected after oxygen exposures for 12-24 h. Accumulation of reactive oxygen species (ROS) were detected after a 2-h exposure. After prolonged hyperoxia exposure, TLR4 expression and IL-1β and TNF-α levels significantly increased in the cells.
CONCLUSIONHyperoxia exposure activates TLR4 signaling pathway in N9 microglial cells in vitro, leading to massive production of ROS, IL-1β, and TNF-α and thus triggering cell apoptosis.
Animals ; Apoptosis ; drug effects ; Cell Hypoxia ; Cells, Cultured ; Interleukin-1beta ; metabolism ; Mice ; Mice, Inbred ICR ; Microglia ; cytology ; drug effects ; physiology ; Oxygen ; pharmacology ; RNA, Messenger ; genetics ; metabolism ; Reactive Oxygen Species ; metabolism ; Toll-Like Receptor 4 ; genetics ; metabolism
10.Leukotriene D4 activates BV2 microglia in vitro.
Zhuang ZHANG ; Jiangyun LUO ; Jing HUANG ; Zhixian LIU ; Sanhua FANG ; Wei-Ping ZHANG ; Erqing WEI ; Yunbi LU
Journal of Zhejiang University. Medical sciences 2013;42(3):253-260
OBJECTIVETo investigate the effects of CysLT receptor agonist leukotriene D4(LTD4) and antagonists on activation of microglia BV2 cells.
METHODSThe expression of CysLT1 and CysLT2 protein was determined by Western blotting and immunostaining in microglia BV2 cells. BV2 cells were pretreated with or without CysLT1 receptor selective antagonist montelukast, CysLT2 receptor selective antagonist HAMI 3379, or CysLT1/CysLT2 receptor dual antagonist BAY u9773 for 30 min, then the cells were treated with LTD4 for 24 h. Cell viability was detected by MTT reduction assay. Phagocytosis and mRNA expression of IL-6 were determined by fluorescent bead tracking and RT-PCR, respectively.
RESULTSIn BV2 cells, LTD4 did not affect proliferation but significantly enhanced phagocytosis and increased IL-6 mRNA expression in a concentration-dependent manner. LTD4 at 100 nmol/L induced a 1.4-fold increase of phagocytic index and a 2-fold up-regulation of IL-6 mRNA expression (P<0.01). HAMI 3379 and BAY u9773 (100 nmol/L) further increased LTD4-induced phagocytosis; BAY u9773 and montelukast decreased LTD4-induced IL-6 mRNA expression, while HAMI 3379 had no effect on that.
CONCLUSIONLTD4 activates BV2 cells in vitro and enhances IL-6 mRNA expression mediated by CysLT1 receptor, LTD4 induces phagocytosis which might be negatively regulated by CysLT2 receptor in BV2 cells.
Acetates ; pharmacology ; Cell Line ; Cell Proliferation ; Cyclohexanecarboxylic Acids ; pharmacology ; Humans ; Interleukin-6 ; metabolism ; Leukotriene Antagonists ; pharmacology ; Leukotriene D4 ; pharmacology ; Microglia ; cytology ; metabolism ; Phagocytosis ; Phthalic Acids ; pharmacology ; Quinolines ; pharmacology ; Receptors, Leukotriene ; metabolism ; SRS-A ; analogs & derivatives ; pharmacology