Open Access

Tanshinone IIA improves hypoxic ischemic encephalopathy through TLR‑4‑mediated NF‑κB signal pathway

  • Authors:
    • Chengzhi Fang
    • Lili Xie
    • Chunmei Liu
    • Chunhua Fu
    • Wei Ye
    • Hong Liu
    • Binghong Zhang
  • View Affiliations

  • Published online on: June 26, 2018     https://doi.org/10.3892/mmr.2018.9227
  • Pages: 1899-1908
  • Copyright: © Fang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hypoxic ischemic encephalopathy (HIE) is the most common brain injury following hypoxia and/or ischemia caused by various factors during the perinatal period, resulting in detrimental neurological deficits in the nervous system. Tanshinone IIA (Tan‑IIA) is a potential agent for the treatment of cardiovascular and cerebrovascular diseases. In this study, the efficacy of Tan‑IIA was investigated in a newborn mouse model of HIE. The dynamic mechanism of Tan‑IIA was also investigated in the central nervous system of neonate mice. Intravenous injection of Tan‑IIA (5 mg/kg) was administered and changes in oxidative stress, inflammation and apoptosis‑associated proteins in neurons. Histology and immunohistochemistry was used to determine infarct volume and the number of damaged neurons by Fluoro‑Jade C staining. The effects of Tan‑IIA on mice with HIE were evaluated by body weight, brain water content, neurobehavioral tests and blood‑brain barrier permeability. The results demonstrated that the apoptosis rate was decreased following Tan‑IIA administration. Expression levels of pro‑apoptotic proteins, caspase‑3 and caspase‑9 and P53 were downregulated. Expression of Bcl‑2 anti‑apoptotic proteins was upregulated by Tan‑IIA treatment in neuro. Results also found that Tan‑IIA treatment decreased production of inflammatory cytokines such as interleukin‑1, tumor necrosis factor‑α, C‑X‑C motif chemokine 10, and chemokine (C‑C motif) ligand 12. Oxidative stress was also reduced by Tan‑IIA in neurons, as determined by the expression levels of superoxide dismutase, glutathione and catalase, and the production of reactive oxygen species. The results demonstrated that Tan‑IIA treatment reduced the infarct volume and the number of damaged neurons. Furthermore, body weight, brain water content and blood‑brain barrier permeability were markedly improved by Tan‑IIA treatment of newborn mice following HIE. Furthermore, the results indicated that Tan‑IIA decreased Toll‑like receptor‑4 (TLR‑4) and nuclear factor‑κB (NF‑κB) expression in neurons. TLR‑4 treatment of neuronal cell in vitro addition stimulated NF‑κB activity, and further enhanced the production of inflammatory cytokines and oxidative stress levels in neurons. In conclusion, these results suggest that Tan‑IIA treatment is beneficial for improvement of HIE through TLR‑4‑mediated NF‑κB signaling.

Introduction

Hypoxic ischemic encephalopathy (HIE) is the most common brain injury caused by hypoxia and/or ischemia during the perinatal period, which is a leading cause of perinatal mortality in obstetrics and gynecology departments (1). HIE frequently leads to neurological sequelae, including seizures, learning impairment, mental retardation, epilepsy, visual impairment and cerebral palsy, unconsciousness and muscle weakness (2,3). Research has indicated that neuroprotection has a crucial role in the progression of perinatal HIE in pediatric patients in low-income countries (4). Additionally, reports have suggested that apoptosis rate, inflammatory cytokines and oxidative stress levels may regulate neuroprotection in neurons during the progression of HIE. Evidence has suggested that Tanshinone IIA (Tan-IIA) is a potential agent for the treatment of cardiovascular and cerebrovascular diseases (5). Therefore, the therapeutic effects of Tan-IIA were investigated in a HIE mouse model.

Tan-IIA is a traditional Chinese medicine, extracted from danshen that has been clinically used for treatment of various human diseases (5,6). Research has indicated that Tan-IIA administration may improve biochemical changes associated with cardiac functions and increase fetal systolic pressure (7). In addition, Tan-IIA exhibits therapeutic potential with protective effects on cardiovascular functions (8). Furthermore, Tan-IIA is reported to regulate mitogen-activated protein kinase signaling pathway to enhance neuron regeneration (9). In this study, the beneficial effects of Tan-IIA on apoptosis, inflammation and oxidative stress were investigated in a mouse model of HIE. The potential neuroprotective effects of Tan-IIA were primarily examined.

Currently, apoptosis of neurons has an essential role in progression of HIE, with potential markers of apoptosis-associated proteins are upregulated during neonatal HIE injury (10). The role of inflammatory cytokines in neuronal apoptosis of neonatal rats with HIE has been reported previously and the results indicated that inhibition of inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), suppresses HIE-induced neuronal apoptosis (11). In addition, endoplasmic reticulum stress also was associated with the activation of activating transcription factor-6 (ATF6) and caspase-12 induced by apoptosis in neonatal HIE during the perinatal period, and results indicated that apoptosis promotes HIE brain injury, mediated by upregulation of endoplasmic reticulum stress (12). Furthermore, Johnson et al (13) demonstrated that perinatal inflammation and infection caused by ischemia is associated with correction of metabolic acidosis in HIE. Furthermore, Chapados and Cheung (14) suggested that oxidative stress is increased in rat pups following HIE and decreasing oxidative stress is beneficial for improving HIE of the newborn rats. These reports suggest that inflammation, oxidative stress and apoptosis are associated with the progression of HIE during the perinatal period.

In the current study, the regulatory effects of Tan-IIA on the progression of HIE during perinatal period were investigated. The inhibitory effects of Tan-IIA on inflammation, oxidative stress and apoptosis was analyzed in a mouse model of HIE. Body weight, brain water content and blood-brain barrier permeability were also determined in newborn mice with HIE following Tan-IIA treatment.

Materials and methods

Ethics statement

This preclinical investigation was conducted by accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of Renmin Hospital of Wuhan University (Wuhan, China). This study was approved by the ethics committee of Renmin Hospital of Wuhan University. All surgery and experiments were performed under anesthetic.

Cell culture

Neurons were isolated from experimental mice and cultured in Dulbecco's modified Eagle's medium with 10% fetal bovine serum and incubated overnight at 37°C humidified atmosphere of 5% CO2. Neurons were treated with 10 mg/ml Toll-like receptor-4 (TLR-4) and/or Tan-IIA with PBS as control for 24 h for further analysis in vitro.

Animal study

A HIE mouse model was conducted as previously described (15). Postnatal day 10 C57BL/6J mice were purchased from Slac Shang Experimental Co., Ltd. (Shanghai, China) and anesthetized to make a unilateral right common carotid artery ligation by performing a 5-0 surgical silk suture. Mice were then divided into two groups: One treated with Tan-IIA (5 mg/kg) and the other treated with PBS daily for 15 days by oral administration.

Counting of lymphocytes and monocytes

The percentage of lymphocytes and monocytes was calculated according to electrical impedance method (16) and laser scattering method by blood cytometer (Vi-cell XR, Beckman Coulter, Inc., Brea, CA, USA). The normal reference values of lymphocytes and monocytes in rats were lymphocytes (62.76 to 90.19%): Mononuclear cells (0–3%).

ELISA

In the protein detection assay, mouse IL-1 (cat. no. MLB00C; R&D Systems, Inc., Minneapolis, MN, USA), mouse IFN-γ (cat. no. MIF00; R&D Systems, Inc., Minneapolis, MN, USA), TNF-α (cat. no. P06804; R&D Systems, Inc.), C-X-C motif chemokine 10 (CXCL10; cat. no. 15945; R&D Systems, Inc.) and chemokine (C-C motif) ligand 12 (CCL12; cat. no. ABIN924766; antibodies-online GmbH, Aachen, Germany) ELISA kits were used to analyze serum levels of inflammatory factors in mice treated with Tan-IIA or PBS. The procedures were conducted according to the manufacturer's instructions. The final results were recorded at 450 nm using an ELISA plate reader.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from hippocampus using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Total RNA was used to produce cDNA by one step RT-PCR kit (RR037A, Takara Biotechnology, Co., Inc., Tokyo, Japan) and the protocol is 37°C for 15 min, 85°C for 5 sec The prepared cDNA was used to analyze reactive oxygen species (ROS), superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), SRY-box 2 (Sox2) and nerve growth factor (NGF) with β-actin as an endogenous control. The amplified PCR products were quantified by measuring the calculated quantitation cycle (Cq) of sample mRNA using SYBR® qPCR: Premix Ex Taq II (TIi RNase H Plus) kit and the thermocycling conditions were: Preheating 92°C (1 min 30 sec), followed by 28 cycles of 92°C (30 sec) and 65°C (1 min) (RR420A, Takara Biotechnology, Co., Inc.). All primers were synthesized by Invitrogen (Thermo Fisher Scientific, Inc.; Table I). Relative changes in mRNA expression were calculated by 2−ΔΔCq (17). The results are expressed as the n-fold difference relative to β-actin.

Table I.

Primer sequences used for reverse transcription-quantitative polymerase chain reaction.

Table I.

Primer sequences used for reverse transcription-quantitative polymerase chain reaction.

Target geneForward primerReverse primer
SOD 5′-AATGTGTCCATTGAAGATCGTGTGA-3′ 5′-GCTTCCAGCATTTCCAGTCTTTGTA-3′
GSH 5′-AATCCTGCTTGGGTATCAGG-3′ 5′-GAGACCCAGTCTCAGGGAAA-3′
CAT 5′-GAGCCTCCTAGAAAGATCTAC-3′ 5′-GCCAGCCTAGGGCTGAGCTG-3′
Sox-2 5′-GGTCGAGGTAGTAGACCTTACA-3′ 5′-GTTCGTCTCTGTGGTCAGATTC-3′
NGF 5′-TTTGTCTAACCCTAACTGAGAAGG-3′ 5′-CTCTAGAATGAACGGTGGAAGG-3′
β-actin 5′-GTGGGCGCCCAGGCACCA-3′ 5′-CTCCTTAATGTCACGCACGATTT-3′

[i] SOD, superoxide dismutase; GSH, glutathione; CAT, catalase; Sox2, SRY-box 2; NGF, nerve growth factor.

Western blot analysis

Neurons were isolated from experimental mice and homogenized in lysate buffer (cat. no. 3096; Bio-Rad Laboratories, Inc., Hercules, CA, USA). The supernatant was acquired by centrifugation at 13,400 × g for 25 min at 4°C; total protein was quantified using a Bicinchoninic Acid Protein Assay kit (cat. no. 23228; Beyotime Institute of Biotechnology, Nanjing, China). Samples (20 µg) were electrophoresed via 15% SDS-PAGE and transferred to a polyvinylidene fluoride membrane (EMD Millipore, Billerica, MA, USA).

Proteins of caspase-3 (cat. no. ab2172; Abcam, Cambridge, UK), caspase-9 (cat. no. ab32539; Abcam), Bcl-2 apoptosis regulator (Bcl-2; cat. no. ab692; Abcam), P53 (cat. no. ab61241; Abcam), TLR-4 (cat. no. ab22048; Abcam), nuclear factor-κB (NF-κB; cat. no. ab28849; Abcam), ATF-6 (cat. no. ab11909; Abcam), the transcription factor C/EBP homologous protein (CHOP; cat. no. ab171894; Abcam) and apoptotic peptidase activating factor-1 (Apaf-1; cat. no. ab32372; Abcam) were detected. Transmembrane proteins were extracted using Transmembrane Protein Extraction kit (Qiagen Sciences, Inc., Gaithersburg, MD, USA) according to the manufacturer's instructions. For western blotting, primary antibodies were added after blocking (5% skimmed milk) and were incubated for 1 h at 37°C and washed with PBS three times. Subsequently, the nitrocellulose membrane was incubated with secondary antibodies, for 24 h at 4°C and washed with PBS three times. The results were visualized by using browser-based Digital Darkroom software (Darkroom Software LLC, Plano, TX, USA) of FluorChem R chemi-luminescence detection system (ProteinSimple, San Jose, CA, USA).

Flow cytometry analysis

Apoptosis rates of neurons were evaluated using Annexin V-fluorescein isothiocyanate (Annexin V-FITC) and propidium iodide (PI) apoptosis detection kit (BD Biosciences, San Jose, CA, USA). Neurons were collected and suspended with Annexin V-FITC and PI according to the manufacturer's instructions. Fluorescence was measured with a FACS scan flow cytometer (BD Biosciences).

Immunohistochemistry

Brains tissues were fixed in formalin fluid for 24 h at room temperature and embedded in paraffin; 8 µm sections were used for further analysis. The paraffin sections were incubated in hydrogen peroxide (4.0%) for 10–15 min at 37°C and subsequently rinsed three times with PBS, 5 min per wash. The sections were blocked with blocking solution (pH 7.2–7.4, NaCl 137 mmol/l, KCl 2.7 mmol/l, Na2HPO4 10 mmol/l, KH2PO4 2 mmol/l) at room temperature for 30 min and stained with 0.1% cresyl violet at 4°C for 12 h. All sections were washed three times with PBS. Cerebral infarction area was measured under a microscope, with Stereo Investigator software (MBF Bioscience, Williston, VT, USA) as previously described (18).

Detection of NF-κB activity

The activity of NF-κB in experimental mice rats was detected according to the manufacturer' protocols using an NF-κB Activation-Nuclear Translocation Assay kit (cat. no. SN368, Beyotime Institute of Biotechnology).

Brain water content

The experimental mice were sacrificed under isoflurane anesthesia at 15 days post-treatment with Tan-IIA. Brain hemispheres were weighed prior to and following drying. Brain water content (%) was calculated as (wet weight-dry weight)/wet weight ×100.

Evan's blue dye extravasation assay

Evan's Blue dye extravasation was performed according to a previously described report (19). Evan's Blue dye was administered by intraperitoneal injection. Experimental mice were sacrificed to measure blood-brain barrier permeability 24 h after administration. The brain hemispheres were homogenized (50% tri-chloroacetic acid; 3:1) and analyzed at 620 nm by spectrophotometry.

ROS productions assay

Reactive Oxygen Species Assay Kit (University of Rochester Medical Center, Rochester, NY, United States) was used to evaluate the changes of ROS productions in neurons isolated from experimental mice. Briefly, neurons were cultured in for 24 h at 37°C. Supernatants were removed and incubated with dichlorofluorescin (10 µmol/l) for 20 min at 37°C. Cells were washed with serum-free medium and were measured at 480 nm using an ELISA plate reader.

Transfection of small interfering RNA (Si-RNA)

All siRNAs were synthesized by Invitrogen (Thermo Fisher Scientific, Inc.) including Si-RNA-TLR-4 (Si-TLR-4) or Si-RNA-vector (Si-vector). A total of 120 pmol Si-TLR-4 (sense, 5′-UCCCCAAGUCAAUCUCUCUTT-3′ and anti-sense-5′-AGAGAGAUUGACUUGGGGATT-3′) or Si-vector (sense, 5′-UUCUCCGAACGUGUCACGUTT-3′ and anti-sense-5′-ACGUGACACGUUCGGAGAATT-3′) was delivered into the neurons cells by electroporation. Standard electroporation was performed using an Amaxa Cell line Nucleofector Kit L (Lonza Group, Ltd., Basel, Switzerland) according to the manufacturer's protocols. Briefly, 5×106 cells were suspended in 100 ìl of nucleofector solution, Si-RNA-TLR-4 or negative control (Si-RNA-vector) was added to the suspension, and then electroporation was conducted at 140 V (250 ms/pulse, eight pulses; 10 µsec).

Fluoro-Jade C staining assay

Fluoro-Jade C staining was used to label degenerating neurons using Fluoro-Jade C Ready-to-Dilute Staining kit (cat. no. TR-100-FJ; Biosensis Pty Ltd., Thebarton, Australia) to identify degenerating neurons. Neurons were isolated from experimental mice and cultured in 6-well plates for 12 h at 37°C. Cell culture medium was removed and labeled with 1 mg/ml DAPI for 15 min or 0.001% Fluoro-Jade C for 20 min according to manufacturer's instructions. Fluoro-Jade C-labeled degenerating neurons were visualized with blue light excitation while DAPI counter stained cell nuclei were visualized with ultra-violet illumination.

Short-term neurobehavioral analysis

The geotactic reflex test was performed as previously described (20). Neurobehavioral analysis of mice was performed at baseline and the 72-h after treatment with Tan-IIA or PBS.

Statistical analysis

SPSS 12.0 statistical analysis software (SPSS Inc., Chicago, IL, USA) was used for statistical analysis. All data are presented as the mean ± standard error from three or more experimental repeats. Unpaired data were analyzed by Student's t-test. Comparisons between multiple groups were analyzed by one-way ANOVA. P<0.05 was considered to indicate a statistically significant difference.

Results

Tan-IIA inhibits inflammatory cytokines in peripheral blood of mice with perinatal HIE

As presented in Fig. 1A, Tan-IIA treatment decreased production of inflammatory cytokine IL-1 in the peripheral blood of mice with perinatal HIE. Additionally, TNF-α plasma concentration levels were decreased by Tan-IIA (Fig. 1B). Results also demonstrated that serum levels of CXCL10 and CCL12 were downregulated in mice with perinatal HIE following treatment with Tan-IIA (Fig. 1C and D). Further, counting with a blood cytometer revealed that the percentage of lymphocytes and monocytes in peripheral blood were decreased by Tan-IIA in mice with perinatal HIE compared with control treatment (Fig. 1E and F). These data indicate that Tan-IIA significantly reduces the level of inflammatory cytokines in peripheral blood of mice with perinatal HIE.

Tan-IIA decreases endoplasmic reticulum stress and oxidative stress in mice with perinatal HIE

Following analysis of Tan-IIA-mediated inflammatory cytokines, endoplasmic reticulum and oxidative stress were examined in neurons from Tan-IIA-treated HIE mice. The results demonstrated that ROS productions and GSH mRNA expression levels were downregulated by Tan-IIA in serum in experimental mice (Fig. 2A and B). In addition, the mRNA expression levels of SOD and CAT were also decreased by Tan-IIA in the serum of experimental mice compared with the control group (Fig. 2C and D). Furthermore, protein expression levels of ATF-6 and CHOP were decreased in neurons following treatment with Tan-IIA (Fig. 2E and F). These data indicate that Tan-IIA treatment decreases endoplasmic reticulum stress and oxidative stress in mice with perinatal HIE, which may contribute to reduced apoptosis of neurons in the central nervous system following HIE.

Tan-IIA suppresses apoptosis of neurons in mice with perinatal HIE

Subsequently, the anti-apoptosis effects of Tan-IIA on neurons in mice model of HIE. As presented in Fig. 3A, the apoptosis rate of neurons was decreased by 15 days of Tan-IIA treatment. The expression levels of pro-apoptosis protein, caspase-3 and caspase-9, were suppressed by Tan-IIA compared with control in neurons from HIE model mice (Fig. 3B and C). However, the expression levels of anti-apoptosis proteins, Bcl-2 were upregulated by Tan-IIA treatment in neurons from HIE mice compared with the control (Fig. 3D and E). Notably, serum levels of IFN-γ anti-cytokine were upregulated by Tan-IIA compared with the control group (Fig. 3F). The data demonstrate that Tan-IIA can markedly inhibit apoptosis of neurons in mice with perinatal HIE.

Tan-IIA regulates neuroactive factors through inhibition of TLR-4-mediated NF-κB pathway

The molecular mechanisms of Tan-IIA-mediated neuroprotective effects in neurons isolated from experimental mice were investigated. TLR-4 expression levels were downregulated by Tan-IIA treatment (Fig. 4A). NF-κB expression was also decreased by Tan-IIA treatment in neurons isolated from experimental mice compared with control (Fig. 4B). Tan-IIA treatment promoted the mRNA expression of neuroactive factors Sox2 and NGF in neurons (Fig. 4C and D). In vitro experiments using cells used extracted from the HIE model control demonstrated that TLR-4 addition abolished the inhibitory effects of Tan-IIA on NF-κB activity (Fig. 4E). In addition, TLR-4 addition inhibited production of Sox2 and NGF promoted by Tan-IIA (Fig. 4F). Furthermore, TLR-4 addition upregulated Apaf-1 expression, and downregulated Bcl-2 expression in neurons (Fig. 4G and H). These results suggest that Tan-IIA regulates neuroactive factors and apoptosis of neurons through inhibition of TLR-4-mediated pathways.

Knockdown of TLR-4 blocks Tan-IIA-improved inflammation, stress and apoptosis in neurons in HIE

The effects of TLR-4 knockdown were analyzed using Si-TLR-4 to determine the effects on inflammation, stress and apoptosis in Tan-IIA treated neurons following HIE. As presented in Fig. 5A, IL-1 and TNF-α expression levels were increased by TLR-4 knockdown in Tan-IIA-treated neurons isolated from HIE mice. TLR-4 knockdown also abrogated expression levels of CAT, SOD and GSH in neurons isolated from Tan-IIA-treated HIE mice (Fig. 5B). The results demonstrated that TLR-4 knockdown abolished Tan-IIA-inhibited apoptosis of neurons isolated from Tan-IIA-treated HIE mice (Fig. 5C). NF-κB, ATF-6 and CHOP expression levels were upregulated by Si-TLR-4 neurons isolated from Tan-IIA-treated HIE mice (Fig. 5D). TLR-4 knockdown increased ROS production in neurons isolated from Tan-IIA-treated HIE mice (Fig. 5E). These results suggest that TLR-4 knockdown can block Tan-IIA-induced effects on inflammation, stress and apoptosis in neurons from HIE model mice.

Tan-IIA improves infarct volume and neuronal degeneration in mice with perinatal HIE

Finally, the effect of Tan-IIA on infarct volume and neuronal degeneration in mice with perinatal HIE was investigated. As presented in Fig. 6A, Tan-IIA treatment decreased infarct volume caused by perinatal HIE. In order to analyze the efficacy of Tan-IIA on apoptosis of hippocampal cells, Fluoro-Jade C staining was performed. Immunohistochemistry demonstrated that Tan-IIA treatment decreased the number of damaged neurons in the hippocampal region of the brain of HIE mice (Fig. 6B). In addition, neurobehavioral tests demonstrated that Tan-IIA treatment markedly improved physical dysfunction of mice with perinatal HIE determined by short-term neurobehavioral analysis (Fig. 6C). Furthermore, Tan-IIA treatment markedly reduced brain water content and blood barrier permeability in brain in mice with perinatal HIE (Fig. 6D and E). Furthermore, body weight of mice with perinatal HIE was also increased by Tan-IIA treatment compared with the control (Fig. 6F). These results suggest that Tan-IIA is beneficial for improving the infarct volume and neuronal degeneration in mice with perinatal HIE.

Discussion

HIE is severe brain disease caused by brain hypoxia and/or ischemic damage, which leads to a series of brain dysfunction, such as perinatal asphyxia and various neurological sequelae for newborns (21). Clinical investigation has indicated that HIE often results in mental retardation, epilepsy and cerebral palsy (22). Currently, although more and more therapeutic methods have been proposed, the therapeutic efficacy is limited for patients with HIE following neuroprotection pharmacotherapy, regarding improvement of inflammation, apoptosis and oxidative stress in the central nervous system (23). Tan-IIA is a traditional Chinese medicine that is used as drug for the treatment of cardiovascular and cerebrovascular diseases (5). The current study investigated the therapeutic effects of Tan-IIA and analyzed the molecular mechanism of Tan-IIA-mediated benefits for HIE. The data indicated that Tan-IIA improved inflammation, oxidative stress and apoptosis in a mouse model of HIE.

Inflammation is an important feature of patients with HIE, and HIE may also lead to other conditions, including cranial nerve palsy (24). A previous study reported that inflammation is associated with metabolic acidosis in HIE, which may aggravate neurological outcomes (13). Girard et al (25) reported that administration of IL-1 receptor antagonist exerts neuroprotective effects during perinatal inflammation and/or hypoxic-ischemic injuries. In addition, the role of TNF-α in neuronal apoptosis in neonatal rats with HIE has been investigated and may presents an important role in the progression of HIE (11,26). Furthermore, chemokines CXCL10 and CCL12 have been identified to be involved in intracranial inflammation and contribute to nerve injury (27,28). The results of the current study demonstrated that Tan-IIA reduced the plasma concentration of IL-1, TNF-α, CXCL10 and CCL12 in the mouse model of HIE. Inflammatory responses were also downregulated by Tan-IIA treatment.

Oxidative and reductive stress are essential for dynamic phases experienced by cells undergoing adaptation towards endogenous or exogenous noxious stimuli (29). Mitochondrial malfunction is the common denominator arising from the aberrant functioning of the rheostat that maintains the homeostasis between oxidative and reductive stress in neurons (30). Maladaptation during oxidative stress may have a pivotal role in the pathophysiology of HIE (14,31). Demarest et al (32) have analyzed the correlation of mitochondrial respiratory impairment and oxidative stress in a rat model of neonatal HIE and the results demonstrated that attenuation of oxidative stress can protect neurons against the loss of mitochondrial glutathione peroxidase activity. Evidence has also indicated that endoplasmic reticulum stress is a crucial risk factor in the pathogenic progression of HIE. Wang et al (33) reported that notoginsenoside R1 protects neurons against neonatal cerebral HIE through estrogen receptor-dependent activation of endoplasmic reticulum stress pathways. In the current study, Tan-IIA-mediated endoplasmic reticulum stress and oxidative stress were investigated in HIE mice. The data demonstrated that Tan-IIA may be an efficient agent for inhibition of endoplasmic reticulum stress and oxidative stress in the central nervous system.

Apoptosis of neurons has a crucial role in the initiation and development of HIE. In the current study, the effect of Tan-IIA on apoptosis of neurons in mice with HIE was analyzed. The results provided potential mechanisms of Tan-IIA-mediated apoptosis resistance. A previous study reported that inhibition of apoptosis can protect neurons against hypoxic-ischemic injury by inhibiting parthanatos and necroptosis (34). In addition, Yan et al (35) also reported that upregulation of Bax and Bcl-2 expression inhibited neural apoptosis in neonatal rats with hypoxic-ischemic brain damage. Furthermore, research has indicated that NF-κB contributes to 6-hydroxydopamine-induced apoptosis of nigral dopaminergic neurons through downregulation of P53 (36). Furthermore, blocking of the NF-κB pathway inhibited apoptosis of neurons in transgenic X-linked inhibitor of apoptotic protein mice and hippocampal rat (37). Notably, the present study demonstrated that Tan-IIA decreases TLR-4 expression, which has been reported to be positively associated with apoptosis of neuroblastoma cells following ischemia-reperfusion injury in the rat retina in vivo (38). The findings of the current study illustrated that Tan-IIA regulates apoptosis of neurons through inhibition of TLR-4-mediated NF-κB signaling.

In conclusion, intravenous injection of Tan-IIA treatment reduced neuronal apoptosis through TLR-4-mediated NF-κB signaling in a neonatal HIE mouse model. Tan-IIA treatment reduced the plasma levels inflammatory factors, IL-1, TNF-α, CXCL10 and CCL12, and also reduced endoplasmic reticulum stress and oxidative stress in the central nervous system. Notably, Tan-IIA treatment markedly suppresses apoptosis of neurons and reduced the physical dysfunction in mice following perinatal HIE. Tan-IIA treatment improved the infarct volume and neuronal degeneration in mice with perinatal HIE. These investigations suggest that Tan-IIA treatment may be a potential agent for the treatment of HIE; however, a sham group was not employed in the present study, which may pose a limitation. Further preclinical and clinical investigations should be performed to validate the efficacy and safety of Tan-IIA.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

BZ made substantial contributions to the design of the present study. CZF was responsible for cell culture, detection of NF-κB activity and flow cytometry analysis. LX was responsible for establishment of the rat model preparation and immunohistochemistry. CL and HL analyzed the data and drafted the manuscript. CHF performed the statistical analysis. WY conducted the data collection. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the ethics committee of Renmin Hospital of Wuhan University (Wuhan, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Hochwald O, Jabr M, Osiovich H, Miller SP, McNamara PJ and Lavoie PM: Preferential cephalic redistribution of left ventricular cardiac output during therapeutic hypothermia for perinatal hypoxic-ischemic encephalopathy. J Pediatr. 164:999–1004.e1. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Looney AM, Ahearne C, Boylan GB and Murray DM: Glial Fibrillary acidic protein is not an early marker of injury in perinatal asphyxia and Hypoxic-ischemic encephalopathy. Front Neurol. 6:2642015. View Article : Google Scholar : PubMed/NCBI

3 

Khan RH, Islam MS, Haque SA, Hossain MA, Islam MN, Khaleque MA, Chowdhury B and Chowdhury MA: Correlation between grades of intraventricular hemorrhage and severity of hypoxic ischemic encephalopathy in perinatal asphyxia. Mymensingh Med J. 23:7–12. 2014.PubMed/NCBI

4 

Tagin M, Abdel-Hady H, Rahman Ur S, Azzopardi DV and Gunn AJ: Neuroprotection for perinatal hypoxic ischemic encephalopathy in low-and Middle-income countries. J Pediatr. 167:25–28. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Li H, Han W, Wang H, Ding F, Xiao L, Shi R, Ai L and Huang Z: Tanshinone IIA inhibits Glutamate-induced oxidative toxicity through prevention of mitochondrial dysfunction and suppression of MAPK activation in SH-SY5Y human neuroblastoma cells. Oxid Med Cell Longev. 2017:45174862017. View Article : Google Scholar : PubMed/NCBI

6 

Kim EO, Kang SE, Im CR, Lee JH, Ahn KS, Yang WM, Um JY, Lee SG and Yun M: Tanshinone IIA induces TRAIL sensitization of human lung cancer cells through selective ER stress induction. Int J Oncol. 48:2205–2212. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Mao C, Zhang Y, Zhang Y, Cao L, Shao H, Wang L, Zhu L and Xu Z: The effect of tanshinone IIA on the cardiovascular system in ovine fetus in utero. Am J Chin Med. 37:1031–1044. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Gao S, Liu Z, Li H, Little PJ, Liu P and Xu S: Cardiovascular actions and therapeutic potential of tanshinone IIA. Atherosclerosis. 220:3–10. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Shen JL, Chen YS, Lin JY, Tien YC, Peng WH, Kuo CH, Tzang BS, Wang HL, Tsai FJ, Chou MC, et al: Neuron regeneration and proliferation effects of Danshen and Tanshinone IIA. Evid Based Complement Alternat Med. 2011:3789072011. View Article : Google Scholar : PubMed/NCBI

10 

Hernandez-Jimenez M, Sacristan S, Morales C, García-Villanueva M, García-Fernández E, Alcázar A, González VM and Martín ME: Apoptosis-related proteins are potential markers of neonatal hypoxic-ischemic encephalopathy (HIE) injury. Neurosci Lett. 558:143–148. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Li SJ, Liu W, Wang JL, Zhang Y, Zhao DJ, Wang TJ and Li YY: The role of TNF-α, IL-6, IL-10, and GDNF in neuronal apoptosis in neonatal rat with hypoxic-ischemic encephalopathy. Eur Rev Med Pharmacol Sci. 18:905–909. 2014.PubMed/NCBI

12 

Liu L, Liu C, Lu Y and Jiang Y: ER stress related factor ATF6 and caspase-12 trigger apoptosis in neonatal hypoxic-ischemic encephalopathy. Int J Clin Exp Pathol. 8:6960–6966. 2015.PubMed/NCBI

13 

Johnson CT, Burd I, Raghunathan R, Northington FJ and Graham EM: Perinatal inflammation/infection and its association with correction of metabolic acidosis in hypoxic-ischemic encephalopathy. J Perinatol. 36:448–452. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Chapados I and Cheung PY: Not all models are created equal: Animal models to study hypoxic-ischemic encephalopathy of the newborn. Commentary on Gelfand SL et al: A new model of oxidative stress in rat pups (Neonatology 2008;94:293-299). Neonatology. 94:300–303. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Cikla U, Chanana V, Kintner DB, Udho E, Eickhoff J, Sun W, Marquez S, Covert L, Otles A, Shapiro RA, et al: ERα signaling is required for TrkB-mediated hippocampal neuroprotection in female neonatal mice after hypoxic ischemic encephalopathy(1,2,3). eNeuro. 3:pii2016. View Article : Google Scholar

16 

Can MM, Tanboğa IH, Türkyilmaz E, Karabay CY, Akgun T, Koca F, Tokgoz HC, Keles N, Ozkan A, Bezgin T, et al: The risk of false results in the assessment of platelet function in the absence of antiplatelet medication: Comparison of the PFA-100, multiplate electrical impedance aggregometry and verify now assays. Thromb Res. 125:e132–e137. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time. quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Hashiguchi A, Yano S, Nitta K, Ide W, Hashimoto I, Kamada H and Kuratsu J: Hemisplenial-accompanied by internal border-zone infarction: Clinical relevance of the splenium of the corpus callosum as a border-zone area between anterior and posterior cerebral arteries. J Neurol Neurosurg Psychiatry. 81:704–706. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Bechet S, Hill F, Gilheaney Ó and Walshe M: Diagnostic accuracy of the modified Evan's blue dye test in detecting aspiration in patients with tracheostomy: A systematic review of the evidence. Dysphagia. 31:721–729. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Farahat FM, Rohlman DS, Storzbach D, Ammerman T and Anger WK: Measures of short-term test-retest reliability of computerized neurobehavioral tests. Neurotoxicology. 24:513–521. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Sant'Anna G, Laptook AR, Shankaran S, Bara R, McDonald SA, Higgins RD, Tyson JE, Ehrenkranz RA, Das A, Goldberg RN, et al: Phenobarbital and temperature profile during hypothermia for hypoxic-ischemic encephalopathy. J Child Neurol. 27:451–457. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Shankaran S: Hypoxic-ischemic encephalopathy and novel strategies for neuroprotection. Clin Perinatol. 39:919–929. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Peliowski-Davidovich A: Hypothermia for newborns with hypoxic ischemic encephalopathy. Paediat Child Health. 17:41–43. 2012. View Article : Google Scholar

24 

Barks JD, Liu YQ, Shangguan Y, Li J, Pfau J and Silverstein FS: Impact of indolent inflammation on neonatal hypoxic-ischemic brain injury in mice. Int J Dev Neurosci. 26:57–65. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Girard S, Sebire H, Brochu ME, Briota S, Sarret P and Sébire G: Postnatal administration of IL-1Ra exerts neuroprotective effects following perinatal inflammation and/or hypoxic-ischemic injuries. Brain Behav Immun. 26:1331–1339. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Aly H, Khashaba MT, El-Ayouty M, El-Sayed O and Hasanein BM: IL-1beta, IL-6 and TNF-alpha and outcomes of neonatal hypoxic ischemic encephalopathy. Brain Dev. 28:178–182. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Gotsch F, Romero R, Friel L, Kusanovic JP, Espinoza J, Erez O, Than NG, Mittal P, Edwin S, Yoon BH, et al: CXCL10/IP-10: A missing link between inflammation and anti-angiogenesis in preeclampsia? J Matern Fetal Neonatal Med. 20:777–792. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Tomita K, Freeman BL, Bronk SF, LeBrasseur NK, White TA, Hirsova P and Ibrahim SH: CXCL10-mediates macrophage, but not other innate immune cells-associated inflammation in murine nonalcoholic steatohepatitis. Sci Rep. 6:287862016. View Article : Google Scholar : PubMed/NCBI

29 

Kurian GA, Rajagopal R, Vedantham S and Rajesh M: The role of oxidative stress in myocardial ischemia and reperfusion injury and remodeling: Revisited. Oxid Med Cell Longev. 2016:16564502016. View Article : Google Scholar : PubMed/NCBI

30 

Perrone S, Szabo M, Bellieni CV, Longini M, Bangó M, Kelen D, Treszl A, Negro S, Tataranno ML and Buonocore G: Whole body hypothermia and oxidative stress in babies with hypoxic-ischemic brain injury. Pediatr Neurol. 43:236–240. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Ten VS, Yao J, Ratner V, Sosunov S, Fraser DA, Botto M, Sivasankar B, Morgan BP, Silverstein S, Stark R, et al: Complement component c1q mediates mitochondria-driven oxidative stress in neonatal hypoxic-ischemic brain injury. J Neurosci. 30:2077–2087. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Demarest TG, Schuh RA, Waddell J, McKenna MC and Fiskum G: Sex-dependent mitochondrial respiratory impairment and oxidative stress in a rat model of neonatal hypoxic-ischemic encephalopathy. J Neurochem. 137:714–729. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Wang Y, Tu L, Li Y, Chen D and Wang S: Notoginsenoside R1 protects against neonatal cerebral Hypoxic-ischemic injury through estrogen Receptor-dependent activation of endoplasmic reticulum stress pathways. J Pharmacol Exp Ther. 357:591–605. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Kong D, Zhu J, Liu Q, Jiang Y, Xu L, Luo N, Zhao Z, Zhai Q, Zhang H, Zhu M and Liu X: Mesenchymal stem cells protect neurons against hypoxic-ischemic injury via inhibiting parthanatos, necroptosis, and apoptosis, but not autophagy. Cell Mol Neurobiol. 37:303–313. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Yan SZ, Wang XL, Wang HY, Dong P and Zhao YS: Effects of umbilical cord blood mononuclear cells transplantation via lateral ventricle on the neural apoptosis and the expression of Bax and Bcl-2 proteins in neonatal rats with hypoxic-ischemic brain damage. Zhongguo Dang Dai Er Ke Za Zhi. 18:862–866. 2016.(In Chinese). PubMed/NCBI

36 

Liang ZQ, Li YL, Zhao XL, Han R, Wang XX, Wang Y, Chase TN, Bennett MC and Qin ZH: NF-kappaB contributes to 6-hydroxydopamine-induced apoptosis of nigral dopaminergic neurons through p53. Brain Res. 1145:190–203. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Kairisalo M, Korhonen L, Sepp M, Pruunsild P, Kukkonen JP, Kivinen J, Timmusk T, Blomgren K and Lindholm D: NF-kappaB-dependent regulation of brain-derived neurotrophic factor in hippocampal neurons by X-linked inhibitor of apoptosis protein. Eur J Neurosci. 30:958–966. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Ulbrich F, Lerach T, Biermann J, Kaufmann KB, Lagreze WA, Buerkle H, Loop T and Goebel U: Argon mediates protection by interleukin-8 suppression via a TLR2/TLR4/STAT3/NF-κB pathway in a model of apoptosis in neuroblastoma cells in vitro and following ischemia-reperfusion injury in rat retina in vivo. J Neurochem. 138:859–873. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 18 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fang C, Xie L, Liu C, Fu C, Ye W, Liu H and Zhang B: Tanshinone IIA improves hypoxic ischemic encephalopathy through TLR‑4‑mediated NF‑κB signal pathway. Mol Med Rep 18: 1899-1908, 2018
APA
Fang, C., Xie, L., Liu, C., Fu, C., Ye, W., Liu, H., & Zhang, B. (2018). Tanshinone IIA improves hypoxic ischemic encephalopathy through TLR‑4‑mediated NF‑κB signal pathway. Molecular Medicine Reports, 18, 1899-1908. https://doi.org/10.3892/mmr.2018.9227
MLA
Fang, C., Xie, L., Liu, C., Fu, C., Ye, W., Liu, H., Zhang, B."Tanshinone IIA improves hypoxic ischemic encephalopathy through TLR‑4‑mediated NF‑κB signal pathway". Molecular Medicine Reports 18.2 (2018): 1899-1908.
Chicago
Fang, C., Xie, L., Liu, C., Fu, C., Ye, W., Liu, H., Zhang, B."Tanshinone IIA improves hypoxic ischemic encephalopathy through TLR‑4‑mediated NF‑κB signal pathway". Molecular Medicine Reports 18, no. 2 (2018): 1899-1908. https://doi.org/10.3892/mmr.2018.9227