Open Access

Hydrogen‑rich saline alleviates early brain injury through inhibition of necroptosis and neuroinflammation via the ROS/HO‑1 signaling pathway after traumatic brain injury

  • Authors:
    • Yun Hu
    • Xiaoyan Feng
    • Junhui Chen
    • Yan Wu
    • Liuyan Shen
  • View Affiliations

  • Published online on: December 9, 2021     https://doi.org/10.3892/etm.2021.11049
  • Article Number: 126
  • Copyright: © Hu 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

Traumatic brain injury (TBI) has been recognized as a serious public health issue and a key contributor to disability and death, with a huge economic burden worldwide. Hydrogen, which is a slight and specific cytotoxic oxygen radical scavenger, has been demonstrated to ameliorate early brain injury (EBI) through reactive oxygen species (ROS), oxidative stress injury, apoptosis and necroptosis. Necroptosis refers to a type of programmed cell death process that has a vital function in neuronal cell death following TBI. The specific function of necroptosis in hydrogen‑mediated neuroprotection after TBI, however, has yet to be determined. The present study aimed to examine the neuroprotective effects and possible molecular basis that underly hydrogen‑rich saline in TBI‑stimulated EBI by examining neural necroptosis in the C57BL/6 mouse model. The brain water content, neurological score, neuroinflammatory cytokines (NF‑κΒ, TNF‑α, IL‑6 and IL‑1β) and ROS were evaluated using flow cytometry. Malondialdehyde, superoxide dismutase (SOD) and glutathione (GSH) levels were evaluated using a biochemical kit. Receptor‑interacting protein kinase (RIP)1, RIP3, Nrf2 and Heme oxygenase‑1 (HO‑1) were evaluated using western blotting. mRNA of Nrf2 and HO‑1 were evaluated using quantitative PCR. Neuronal death was evaluated by TUNEL staining. The outcomes illustrated that hydrogen‑rich saline treatment considerably enhanced the neurological score, increased neuronal survival, decreased the levels of serum MDA and brain ROS, increased the levels of serum GSH and SOD. In addition the protein expression levels of RIP1 and RIP3 and the cytokines NF‑κB, TNF‑α, IL‑1β and IL‑6 were downregulated compared with the TBI group, which demonstrated that hydrogen‑rich saline‑induced inhibition of necroptosis and neuroinflammation ameliorated neuronal death following TBI. The neuroprotective capacity of hydrogen‑rich saline was demonstrated to be partly dependent on the ROS/heme oxygenase‑1 signaling pathway. Taken together, the findings of the present study indicated that hydrogen‑rich saline enhanced neurological outcomes in mice and minimized neuronal death by inducing protective effects against neural necroptosis as well as neuroinflammation.

Introduction

Traumatic brain injury (TBI) has been recognized as a serious public health issue and a key contributor to disability and death, with a huge economic burden worldwide (1,2). High incidence (1/1,000 in China between 1983 and 1985) of TBI is predominantly reported in low- and middle-income countries as well as developing countries, including China and Iran (1-3). The prevalence of TBI has witnessed a rapidly growing trend due to the considerable increase in road accidents, such as motor vehicle collisions (2). Although an increasing number of randomized controlled trials have included intracranial pressure monitoring, therapeutic hypothermia, surgical methods and drug administration in recent years, long-term outcomes have not substantially improved, especially after drug intervention (2-8). Therefore, it is important to further clarify the physiopathological processes of TBI and identify novel efficient pharmacological targets for TBI treatment. It is widely acknowledged that the pathophysiology of TBI encompasses several types of pathological and physiological changes, mainly involving primary brain injury and secondary brain injury, which leads to neuronal death, neurological deficits and mortality after TBI (9). Primary brain injury, which is a direct physical injury to the brain tissue, is difficult to prevent and cannot be usually reversed, and leads to brain tissue disorganization, intracerebral hemorrhage and blood-brain barrier (BBB) damage (1,10,11). Secondary brain injury includes calcium overload, oxidative stress, neuroinflammation, autophagy, lipid peroxidation, apoptosis and necroptosis, and can be reversed (10,11).

Necroptosis has recently been identified as a pathway of modulated necrosis and a mechanism of caspase-independent programmed cell death, requiring the proteins mixed lineage kinase domain-like (MLKL) and receptor-interacting protein kinase-3 (RIP3), and is triggered by death receptors (12). Accumulating evidence indicates that necroptosis performs an important function in central nervous system disorders, such as TBI (13-15), intracerebral hemorrhage (16,17), ischemic stroke (18), Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis (19). The activation of a TNF ligand family member, such as RIP1 and MLKL, is the most upstream signaling activity necessary for necroptosis induction (20,21). The activation of RIP1 contributes to necroptosis via inducting RIP1-RIP3-MLKL complex (22). Necroptosis is common in early brain injury (EBI) and may be a mechanism of TBI.

In previous years, it has become widely acknowledged that hydrogen-rich saline or hydrogen gas can protect the human body from numerous diseases, such as neurodegenerative disorders, spontaneous subarachnoid hemorrhage, stroke and ischemia-reperfusion damage, by modulating neuronal apoptosis, inflammatory response and oxidative stress (23-26). A previous study has indicated that hydrogen may selectively minimize peroxynitrites and hydroxyl radicals, and subsequently have a crucial function exhibiting cytoprotective, anti-inflammatory, anti-apoptotic and antioxidant properties (27). However, the neuroprotective benefits of hydrogen-rich saline treatment on TBI are controversial. Heme oxygenase-1 (HO-1) is an essential element of the cellular defense system that is activated by oxidant-stimulated damage and acts against it (28,29). In the central nervous system (CNS), HO-1 exerts anti-necroptotic, anti-neuroinflammatory and neuroprotective functions (27,30). In our previous study, HO-1 was demonstrated to regulate neuronal death in acute CNS disease (31). Therefore, treatments that target HO-1 may be potentially effective at preventing necroptosis, oxidative stress and inflammation following TBI. Nonetheless, the specific mechanisms regarding the potential neuroprotective benefits of hydrogen-rich saline treatment are unknown. The present study examined whether neuroprotective benefits of hydrogen-rich saline treatment in a mouse model of TBI occur via effects on neuroinflammation and necroptosis, and whether neuroprotection is dependent on the reactive oxygen species (ROS)/HO-1 pathway.

Materials and methods

Animals

All the animal experiments conducted in the present study were completed in compliance with guidelines for the appropriate handling of laboratory animals formulated by the National Institutes of Health (32). Approval of the experiments was provided by the Ethics Committee of Wuxi Clinical College of Anhui Medical University (Wuxi, China). A total of 36 healthy adult male C57BL/6J mice (age, 8-10 weeks; Anhui Medical University, Hefei, China) weighing between 22-25 g were used when conducting all the experiments for the current study. The mice were maintained in animal care facilities (temperature, 25±2˚C; humidity, 55±5%) with a 12/12 h dark/light cycle and an unrestricted supply of water and food.

Animal TBI model

The Feeney weight-drop model of focal injury was strictly followed when developing the TBI model (33,34). Briefly, anesthetization of the mice was performed using 1% sodium pentobarbital (40 mg/kg) injected intraperitoneally. The mice were subsequently placed in a brain stereotaxic apparatus. While being operated, a heating pad was utilized to ensure that the rectal temperature remained at 37±0.5˚C. The coordinates utilized when making a burr hole in the left hemisphere were set as follows: 0.2 mm posterior, 1 mm lateral and 2.2 mm ventral of bregma's horizontal plane. To visualize the dura mater, the bone fap was removed. The dura was subsequently placed under weight-drop equipment that included an impact sensor pedal; (Anhui Zhenghua Biological Instrument Equipment Co., Ltd.). Metal with a tip diameter of 3-mm and weight of 240 g was dropped onto the dura mater from a distance of 1 cm above the dura via a catheter. The scalp was subsequently closed and the mice were removed from the stereotaxic apparatus. Subsequently, a medical bone wax was utilized to cover the hole. The animals in the sham cohort received comparable surgical treatments but without the weight-drop procedure. At 72 h following TBI, the mice were sacrificed with 100 mg/kg sodium pentobarbital via an intraperitoneal injection. Brain tissue samples were collected after the mice were sacrificed. Fresh specimens (cerebral cortex) and serum were stored in liquid nitrogen (-196˚C) or hippocampus was stored in 4% formalin-fixed (4˚C) for ≥48 h.

Drug preparation and administration

After the TBI model was established successfully, the mice received intraperitoneal injections every day for 72 h that contained either plain saline (control) or hydrogen-rich (5 ml/kg; experimental). Hydrogen-rich saline was prepared in accordance with the procedure described in previous studies (35,36). Briefly, purified hydrogen was dissolved in normal saline for 2 h at an elevated pressure of 0.4 MPa. The physiological concentration was maintained at 1.73 ml hydrogen per 100 ml saline (average, >6 mmol/l). Hydrogen-rich saline was stored at 4˚C in an aluminum bag without dead volume under atmospheric pressure. Every week, fresh hydrogen-rich saline was synthesized to guarantee a consistent concentration. The content of hydrogen in saline was evaluated and detected by gas chromatography as reported in a previous study (37).

Neurobehavioral assessment

By employing a previously published neurological grading system (38,39), the degree of brain injury was assessed 72 h following TBI via the determination of neurological function. The scoring system consisted of reflex, sensory, balance and motor tests. The neurological scores ranged between 0 and 18 and were calculated by summing the individual scores (Table SI). A behavioral assessment was conducted on all mice in each cohort, with a higher score indicating impaired neurological function.

Brain water-content measurement

As reported previously, the degree of brain edema was examined by quantifying the brain water content utilizing the standard wet-dry technique (39-41). A total of 72 h following TBI the mice were euthanized, and their whole brain was extracted and divided into the contralateral and ipsilateral cortex, contralateral and ipsilateral basal ganglia and cerebellum (wet weight). The dry weight of each cohort's brain specimen was then determined after dehydrating each part at 105˚C for 24 h. The proportion of brain water content was equal to (wet weight-dry weight)/wet weight x100%.

Cytokine measurements

ELISA was utilized to measure cerebral cortex NF-κΒ (cat. no. ab176663), cerebral cortex TNF-α (cat. no. ab208348), cerebral cortex IL-6 (cat. no. ab222503) and cerebral cortex IL-1β (cat. no. ab197742; all from Abcam). The cerebral cortex was extracted from mice brain. The procedure was conducted in accordance with the manufacturer's protocols.

Analysis of ROS

The brain tissue intracellular ROS synthesis was evaluated utilizing the non-fluorescent diacetylated 2',7'-dichlorofluorescein diacetate (DCFH-DA) probe (Sigma-Aldrich; Merck KGaA) that becomes highly fluorescent upon oxidation and was used according to the guidelines provided by the manufacturer. Fresh cerebral cortex was used to form a single cell suspension. The cells were harvested at a concentration of 2x106 cells/ml, then DCFH-DA (10 µM final concentration) was added and the mixture was incubated at 37˚C for 15 min. The DCFH results were examined via flow cytometry (CytoFLEX; Beckman Coulter, Inc.). CytExpert (version 2.4; Beckman Coulter, Inc.) was used to analyze the data.

Analysis of MDA, superoxide dismutase (SOD) and glutathione (GSH)

The malondialdehyde (MDA) Assay Kit (excitation/emission, 532/553 nm; cat. no. ab118970; Abcam) was utilized to detect serum MDA levels in compliance with the instructions provided by the manufacturer. The serum was extracted from mice venous blood. The SOD Assay Kit (cat. no. A001-3-2; Nanjing Jiancheng Bioengineering Institute) and GSH (42) Assay Kit (cat. no. A005-1-2; Nanjing Jiancheng Bioengineering Institute) were utilized to detect serum SOD and serum GSH levels in compliance with the manufacturer's instructions (43,44).

TUNEL staining

To evaluate neuronal death in the brain cortex, TUNEL staining was utilized. The procedure was performed with a TUNEL staining kit (cat. no. 11684817910; Roche Diagnostics GmbH) according to the protocols provided by the manufacturer. In each 4% formalin-fixed (4˚C; duration, ≥48 h) for specimen, paraffin-embedded sections (10-µm) were cut from formalin-fixed tissue. Subsequently, 50 µl TUNEL reaction mixture was added. The negative control used did not include the TUNEL reaction mixture. Subsequently, the slides were subjected to incubation in a humidified dark chamber at a temperature of 37˚C for 60 min. Then, a primary antibody against the neuronal marker NeuN (1:200; rabbit polyclonal; cat. no. ab128886; Abcam) diluted in PBS was added, followed by incubation overnight at 4˚C. Subsequently, incubated with FITC goat anti-mouse IgG secondary antibodies (1:100; cat. no. BA1101; Boster Biological Technology Co., Ltd.) at temperature of 37˚C for 1.5 h. Next, the slides were incubated at ambient temperature for 5 min in the dark with DAPI for staining of the nuclei, followed by imaging using a fluorescence microscope (magnification, x200). The validation of the cell count was performed using four high-power fields randomly selected, and the data obtained from each field were averaged.

Western blot analysis

Western blot analysis was conducted as indicated previously (40). Cerebral cortex specimens were collected and extracted. Cerebral cortex samples were collected, homogenized and total protein was extracted using RIPA buffer (CoWin Biosciences). The protein concentrations were quantified utilizing a BCA Protein Assay kit (Beyotime Institute of Biotechnology) and proteins (40 µg/lane) were separated using 10% SDS-PAGE. Then, the proteins were transferred onto Immobilon nitrocellulose membranes. Blocking of the membranes was performed with 5% non-fat milk for 1 h at room temperature. The membranes were then subjected to incubation using the following primary antibodies overnight at 4˚C: Anti-β-actin (1:1,000; rabbit polyclonal; cat. no. ab8227; Abcam), anti-RIP1 (1:1,000; rabbit polyclonal; cat. no. ab106393; Abcam), anti-nuclear factor erythroid 2-related factor 2 (Nrf2; 1:1,000; rabbit polyclonal; cat. no. ab31163; Abcam), anti-HO-1 (1:1,000; rabbit polyclonal; cat. no. ab13243; Abcam) and anti-RIP3 (1:1,000; rabbit polyclonal; cat. no. ab62344; Abcam). After washing the membranes using 0.5% TBS-Tween-20 three times, the membranes were incubated with HRP-conjugated goat anti-rabbit IgG secondary antibodies (1:2,000; cat. no. 7074s; Cell Signaling Technology, Inc.) at room temperature for 1.5 h. The signals were developed using an enhanced chemiluminescence reagent (MilliporeSigma) according to the manufacturer's instructions. A Bio-Rad imaging system (Bio-Rad Laboratories, Inc.) was utilized to identify protein bands that were then measured using ImageJ software (version 1.52; National Institutes of Health).

RNA extraction and reverse transcription-quantitative PCR (RT-qPCR)

TRIzol® reagent (Thermo Fisher Scientific, Inc.) was utilized to isolate total RNA from the cerebral cortex of brain specimens as per the guidelines provided by the manufacturer, which was then quantified utilizing a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Inc.). The RevertAid First Strand cDNA Synthesis Kit (cat. no. K1622; Thermo Fisher Scientific Inc.) was subsequently utilized to perform reverse transcription of RNA to cDNA according to the manufacturer's protocol. Each specimen's mRNA levels were quantified via qPCR utilizing SYBR Green Master Mix (Toyobo Life Science). The expression levels of all genes were normalized to that of β-actin. The following qPCR thermocycling criteria were used: 45˚C (2 min) and 95˚C (10 min), followed by 40 cycles of denaturation at 95˚C (15 sec), annealing at 60˚C (1 min) and extension at 72˚C (1 min). The analysis of all specimens was performed in triplicate. The primers used are listed as follows: Nrf2 forward, 5'-CAGTGCTCCTATGCGTGAA-3' and reverse, 5'-GCGGCTTGAATGTTTGTCT-3'; HO-1 forward, 5'-TGACAGAAGAGGCTAAGACCG-3' and reverse, 5'-AGTGAGGACCCACTGGAGGA-3'; and GAPDH forward, 5'-ATGGGTGTGAACCACGAGA-3' and reverse, 5'-CAGGGATGATGTTCTGGGCA-3' (45).

Statistical analysis

All experiments were performed with at least three experimental repeats, and the data are presented as the mean ± SEM. Neurological scores are presented as the median and interquartile range. Statistical analyses were implemented using GraphPad Prism 6 (GraphPad Software, Inc.) and SPSS 14.0 (SPSS, Inc.). Differences between multiple groups were analyzed using one-way ANOVA followed by Tukey's post hoc test. Neurological scores were analyzed using a non-parametric test (Kruskal-Wallis followed by Dunn's post hoc test). P<0.05 was considered to indicate a statistically significant difference.

Results

Hydrogen-rich saline alleviates neurological deficits and brain edema after TBI

Modified neurological severity scores were employed to assess neurological impairments to understand the neuroprotection of hydrogen-rich saline following TBI. To assess brain damage, the wet-dry technique was employed to measure brain water content 72 h after TBI. The findings illustrated that TBI was associated with a considerable increase in the brain water content, which was alleviated after hydrogen-rich saline treatment (Fig. 1B). Comparable findings in neurological scores indicated that the scores were considerably higher in animals suffering from hydrogen-rich saline treatment compared with the TBI group, and that hydrogen-rich saline treatment substantially improved the neurological function (Fig. 1A).

Hydrogen-rich saline alleviates neuronal necroptosis after TBI

Neuronal necroptosis is the main factor resulting in EBI after TBI (46). Therefore, a TUNEL assay was conducted to examine the degree of cell death in TBI mice treated or non-treated with hydrogen-rich saline 72 h following model establishment. Neuronal death in the hippocampus decreased upon hydrogen-rich saline treatment compared with the TBI group (Fig. 2A). The expression levels of necroptosis-related proteins were additionally detected via western blotting (Fig. 2B). The results of western blotting also demonstrated that hydrogen-rich saline significantly reduced the expression levels of the necroptosis-related proteins RIP1 and RIP3 compared with the TBI group (Fig. 2C and D). These findings indicated that hydrogen-rich saline exhibited neuroprotective benefits after TBI.

Hydrogen-rich saline alleviates neuroinflammation after TBI

Previous studies have demonstrated that neuroinflammation exhibits a vital function in EBI after TBI and enhanced neuroinflammation can aggravate EBI (10,47-49). Activation of the inflammatory process can induce the release of inflammatory cytokines, which include NF-κB, TNF-α IL-6 and IL-1β (39,50). Therefore, ELISA was employed to examine the hippocampal levels of NF-κB, TNF-α IL-6 and IL-1β. The findings revealed that the inflammatory cytokines were increased substantially after TBI, while they were significantly decreased after hydrogen-rich saline treatment (Fig. 3A-D). Thus, these results suggested that hydrogen-rich saline exhibited a potent anti-inflammatory activity against TBI-induced neuroinflammation.

Hydrogen-rich saline inhibits TBI-induced oxidative stress and decreases ROS levels

To clarify whether oxidative stress performs a crucial function in TBI and whether hydrogen-rich saline can regulate oxidative stress, the oxidative stress-related biomarker levels of GSH, MDA and SOD were detected in each cohort. The findings illustrated that MDA increased following TBI, but considerably decreased following hydrogen-rich saline treatment (Fig. 4A). Additionally, SOD and GSH decreased after TBI but increased considerably following hydrogen-rich saline treatment (Fig. 4B and C). ROS are considered to be a biomarker of oxidative stress activation initiating programmed cell and neuronal death (31,51). ROS levels were detected using the DCF-DA probe. The results demonstrated that ROS levels were increased after TBI in the hippocampus, while decreased after hydrogen-rich saline administration (Figs. 4D and S1).

Hydrogen-rich saline regulates necroptosis via the ROS/HO-1 signaling pathway after TBI

It was explored whether necroptosis inhibition occurred through the ROS/HO-1 signaling pathway after hydrogen-rich saline treatment. The mRNA expression levels of HO-1 and Nrf2 were detected via RT-qPCR. The findings illustrated that the expression levels of HO-1 and Nrf2 were considerably reduced in the TBI cohort, and were elevated following hydrogen-rich saline administration (Fig. 5A and B). The protein expression levels of HO-1 and Nrf2 were also detected via western blotting (Fig. 5C). Quantification of the protein levels of HO-1 and Nrf2 in the brain cortex relatively to the β-actin loading control revealed that hydrogen-rich saline increased HO-1 and Nrf2 expression after TBI in mice (Fig. 5D and E). Thus, the present findings collectively demonstrated that hydrogen-rich saline may inhibit TBI-induced necroptosis by regulating the ROS/HO-1 signaling pathway.

Discussion

The present study examined the therapeutic value of hydrogen-rich saline for ameliorating EBI in a TBI mouse model. The findings indicated that hydrogen-rich saline was a neuroprotective agent that can attenuate EBI following TBI. The results demonstrated that hydrogen-rich saline could achieve the following: i) Ameliorate neurological dysfunction following TBI; ii) relieve brain damage in a mouse TBI model; iii) relieve neuroinflammation after TBI and decrease brain inflammatory damage; and iv) prevent necroptosis after TBI and alleviate neuronal death. Furthermore, the anti-neuroinflammatory and anti-necroptotic roles of hydrogen-rich saline may be associated with the ROS/HO-1 pathway.

Hydrogen-rich saline or hydrogen gas can easily penetrate the BBB via gaseous diffusion, which is acknowledged to achieve protective effects in several CNS disorders, such as TBI, neurodegenerative diseases, intracranial hemorrhage and ischemic stroke (23-26). Hydrogen gas or hydrogen-rich saline serves an important antioxidant role with high tissue transferability, and it has been demonstrated that H2 is safe for patients and animals (37). The anti-oxidative stress and anti-inflammatory effects of hydrogen-rich saline or hydrogen gas are mediated by selective suppression of toxic ROS, which include peroxynitrite and hydroxyl radical (25). Liu et al (52) also reported that H2 can markedly improve cognitive dysfunction and survival rates, decrease inflammatory response and oxidative stress, and increase antioxidant enzyme activities in the serum and hippocampus in a mouse model of sepsis. In the intracerebral hemorrhage model, it has also been demonstrated that hydrogen performs a neuroprotective function against EBI following ICH, alleviating brain edema and neurologic deficits by regulating oxidative stress, neuroinflammation and apoptosis (53). In the hypoxic-ischemic brain injury neonatal rat model, H2 inhalation could alleviate brain damage and improve early neurological outcomes through anti-inflammatory, anti-apoptotic and antioxidant responses via the MAPK/HO-1/peroxisome proliferator-activated receptor γ coactivator 1a pathway (54). In the TBI model, molecular hydrogen water could also reverse the controlled cortical impact-induced brain edema through preserved or increased ATP levels (55). Tian et al (56) demonstrated that hydrogen-rich water could alleviate inflammation and decrease brain damage, exhibiting a neuroprotective function against TBI. Yuan et al (57) also discovered that hydrogen-rich water can inhibit oxidative stress and activate the Nrf2 pathway, thereby improving TBI-induced brain damage. Both aforementioned studies explored the neuroprotective effect of hydrogen-rich water associated with inflammation and oxidative stress without investigating further molecular mechanisms. However, the current study also revealed that hydrogen-rich saline considerably enhanced neurological function, alleviated brain edema, increased neuronal survival and downregulated the protein expression of RIP3 and RIP1, as well as the cytokines NF-κB, TNF-α, IL-1β and IL-6, following TBI.

Necroptosis is a cell death type of modulated necrosis, requiring the proteins MLKL and RIP3, and is stimulated by death receptors (12). Increasing evidence suggests that necroptosis performs an instrumental function in CNS diseases, such as TBI (13-15). Our previous studies have also demonstrated that necroptosis has an integral function in the pathophysiology of neuronal death following in vitro traumatic neuronal injury, and that the potential regulatory mechanisms may be associated with the Akt/GSK-3β and the metabotropic glutamate receptor 1 signaling pathways (13,14). Therefore, necroptosis is a vital mechanism in TBI, and necroptosis inhibition via drugs may decrease neuronal death. Jia et al (58) reported that hydrogen can decrease the expression levels of necroptosis-related proteins in the hippocampus, including MLKL, phosphorylated MLKL and RIP3, thereby partly preventing neuronal and astrocyte necroptosis in the lithium-pilocarpine model of status epilepticus. Dong et al (59) also reported that high concentrations of hydrogen inhalation can alleviate skin ischemia/reperfusion injury by regulating necroptosis and the RIP-MLKL-serine/threonine-protein phosphatase PGAM5/dynamin-related protein 1 necrotic pathway. In the present study, the results confirmed that the mRNA and protein expression levels of RIP3 and RIP1 were elevated 72 h after TBI, while hydrogen-rich saline treatment could reduce the RIP3 and RIP1 expression levels.

The molecular mechanism of necroptosis and neuroinflammation is complex, and the specific mechanisms of the neuroprotective benefits of hydrogen-rich saline treatment are yet to be elucidated. Wang et al (60) reported that rehmapicrogenin can improve adriamycin-induced nephropathy in vitro and in vivo by reducing ROS accumulation, thereby regulating the expression levels of Nrf2. Nrf2 is an important transcriptional regulation factor that can regulate the expression of >250 genes and is characterized by its binding site ‘antioxidant response element’; the majority of regulated genes by Nrf2 are involved in oxidative stress and cell apoptosis, necroptosis, autophagy and ferroptosis (31). Yu et al (61) discovered that the inhalation of 2% molecular hydrogen gas may enhance the survival rates, reduce the lung edema and the lung injury score, and ameliorate the injuries induced by inflammation and oxidative stress in the septic mouse model, while Nrf2 knockout could reverse or weaken the protection of H2 gas on lung damage, which was also dependent on HO-1 and high mobility group protein B1. Additionally, Chen et al (62) demonstrated that H2 decreased endothelial inflammation and injury and increased the expression and activity of HO-1 in vivo and in vitro. Nrf2 knockout or HO-1 inhibition reversed the protection of H2, indicating that this was dependent on the activity of the Nrf2/HO-1 signaling pathway. However, the exact mechanism requires further elucidation.

In summary, the present findings demonstrated that necroptosis, which is mediated by the RIP1 and RIP3 proteins, is a key cellular regulatory mechanism that may lead to EBI following TBI. The present study, for the first time to the best of our knowledge, revealed that hydrogen-rich saline induced modulation of necroptosis and neuroinflammation via the ROS/HO-1 pathway, and also provided a novel insight into evaluating the biological impacts as well as the mechanisms that underly neuroprotection and inhibition of inflammation and necroptosis by hydrogen-rich saline.

Supplementary Material

Reactive oxygen species levels as detected via flow cytometry. TBI, traumatic brain injury; HS, hydrogen-rich saline.
Neurological behavior scores.

Acknowledgements

Not applicable.

Funding

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

YH wrote the manuscript. YH, XF, YW, LS and JC performed the experiments and prepared the figures. YH and JC confirm the authenticity of all the raw data. YH and JC designed the study and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study protocol was approved by the Research Ethics Committee of Wuxi Clinical College of Anhui Medical University (approval no. YXLL-2020-012; Wuxi, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jiang JY, Gao GY, Feng JF, Mao Q, Chen LG, Yang XF, Liu JF, Wang YH, Qiu BH and Huang XJ: Traumatic brain injury in China. Lancet Neurol. 18:286–295. 2019.PubMed/NCBI View Article : Google Scholar

2 

Chen J, Li M, Chen L, Chen W, Zhang C, Feng Y, Wang Y and Chen Q: The effect of controlled decompression for severe traumatic brain injury: A randomized, controlled trial. Front Neurol. 11(107)2020.PubMed/NCBI View Article : Google Scholar

3 

Chen JH, Li PP, Yang LK, Chen L, Zhu J, Hu X and Wang YH: Value of ventricular intracranial pressure monitoring for traumatic bifrontal contusions. World Neurosurg. 113:e690–e701. 2018.PubMed/NCBI View Article : Google Scholar

4 

Nichol A, French C, Little L, Haddad S, Presneill J, Arabi Y, Bailey M, Cooper DJ, Duranteau J, Huet O, et al: Erythropoietin in traumatic brain injury (EPO-TBI): A double-blind randomised controlled trial. Lancet. 386:2499–2506. 2015.PubMed/NCBI View Article : Google Scholar

5 

Hutchinson PJ, Kolias AG, Timofeev IS, Corteen EA, Czosnyka M, Timothy J, Anderson I, Bulters DO, Belli A, Eynon CA, et al: Trial of decompressive craniectomy for traumatic intracranial hypertension. N Engl J Med. 375:1119–1130. 2016.PubMed/NCBI View Article : Google Scholar

6 

Cooper DJ, Nichol AD, Bailey M, Bernard S, Cameron PA, Pili-Floury S, Forbes A, Gantner D, Higgins AM, Huet O, et al: Effect of early sustained prophylactic hypothermia on neurologic outcomes among patients with severe traumatic brain injury: The POLAR randomized clinical trial. JAMA. 320:2211–2220. 2018.PubMed/NCBI View Article : Google Scholar

7 

Wright DW, Yeatts SD, Silbergleit R, Palesch YY, Hertzberg VS, Frankel M, Goldstein FC, Caveney AF, Howlett-Smith H, Bengelink EM, et al: Very early administration of progesterone for acute traumatic brain injury. N Engl J Med. 371:2457–2466. 2014.PubMed/NCBI View Article : Google Scholar

8 

Robertson CS, Hannay HJ, Yamal JM, Gopinath S, Goodman JC and Tilley BC: Epo Severe TBI Trial Investigators. Baldwin A, Rivera Lara L, Saucedo-Crespo H, et al: Effect of erythropoietin and transfusion threshold on neurological recovery after traumatic brain injury: A randomized clinical trial. JAMA. 312:36–47. 2014.PubMed/NCBI View Article : Google Scholar

9 

Wang Y, Wang L, Hu T, Wang F, Han Z, Yin Z, Ge X, Xie K and Lei P: Hydrogen improves cell viability partly through inhibition of autophagy and activation of PI3K/Akt/GSK3β signal pathway in a microvascular endothelial cell model of traumatic brain injury. Neurol Res. 42:487–496. 2020.PubMed/NCBI View Article : Google Scholar

10 

Li H, Lu C, Yao W, Xu L, Zhou J and Zheng B: Dexmedetomidine inhibits inflammatory response and autophagy through the circLrp1b/miR-27a-3p/Dram2 pathway in a rat model of traumatic brain injury. Aging (Albany NY). 12:21687–21705. 2020.PubMed/NCBI View Article : Google Scholar

11 

Wang Y, Zhao M, Shang L, Zhang Y, Huang C, He Z, Luo M, Wu B, Song P, Wang M and Duan F: Homer1a protects against neuronal injury via PI3K/AKT/mTOR signaling pathway. Int J Neurosci. 130:621–630. 2020.PubMed/NCBI View Article : Google Scholar

12 

Vandenabeele P, Galluzzi L, Vanden Berghe T and Kroemer G: Molecular mechanisms of necroptosis: An ordered cellular explosion. Nat Rev Mol Cell Biol. 11:700–714. 2010.PubMed/NCBI View Article : Google Scholar

13 

Chen T, Yang LK, Zhu J, Hang CH and Wang YH: The AMPAR antagonist perampanel regulates neuronal necroptosis via Akt/GSK3β signaling after acute traumatic injury in cortical neurons. CNS Neurol Disord Drug Targets. 20:266–272. 2021.PubMed/NCBI View Article : Google Scholar

14 

Chen T, Zhu J, Wang YH and Hang CH: Arc silence aggravates traumatic neuronal injury via mGluR1-mediated ER stress and necroptosis. Cell Death Dis. 11(4)2020.PubMed/NCBI View Article : Google Scholar

15 

Bao Z, Fan L, Zhao L, Xu X, Liu Y, Chao H, Liu N, You Y, Liu Y, Wang X and Ji J: Silencing of A20 aggravates neuronal death and inflammation after traumatic brain injury: A potential trigger of necroptosis. Front Mol Neurosci. 12(222)2019.PubMed/NCBI View Article : Google Scholar

16 

Laird MD, Wakade C, Alleyne CH Jr and Dhandapani KM: Hemin-induced necroptosis involves glutathione depletion in mouse astrocytes. Free Radic Biol Med. 45:1103–1114. 2008.PubMed/NCBI View Article : Google Scholar

17 

Shen H, Liu C, Zhang D, Yao X, Zhang K, Li H and Chen G: Role for RIP1 in mediating necroptosis in experimental intracerebral hemorrhage model both in vivo and in vitro. Cell Death Dis. 8(e2641)2017.PubMed/NCBI View Article : Google Scholar

18 

Zhang Y, Li M, Li X, Zhang H, Wang L, Wu X, Zhang H and Luo Y: Catalytically inactive RIP1 and RIP3 deficiency protect against acute ischemic stroke by inhibiting necroptosis and neuroinflammation. Cell Death Dis. 11(565)2020.PubMed/NCBI View Article : Google Scholar

19 

Yuan J, Amin P and Ofengeim D: Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat Rev Neurosci. 20:19–33. 2019.PubMed/NCBI View Article : Google Scholar

20 

Liu C, Chen Y, Cui W, Cao Y, Zhao L, Wang H, Liu X, Fan S, Huang K, Tong A and Zhou L: Inhibition of neuronal necroptosis mediated by RIP1/RIP3/MLKL provides neuroprotective effects on kaolin-induced hydrocephalus in mice. Cell Prolif. 54(e13108)2021.PubMed/NCBI View Article : Google Scholar

21 

Wu Y, Zheng Z, Cao X, Yang Q, Norton V, Adini A, Maiti AK, Adini I and Wu H: RIP1/RIP3/MLKL mediates myocardial function through necroptosis in experimental autoimmune myocarditis. Front Cardiovasc Med. 8(696362)2021.PubMed/NCBI View Article : Google Scholar

22 

Linkermann A and Green DR: Necroptosis. N Engl J Med. 370:455–465. 2014.PubMed/NCBI View Article : Google Scholar

23 

Zou R, Wang MH, Chen Y, Fan X, Yang B, Du J, Wang XB, Liu KX and Zhou J: Hydrogen-rich saline attenuates acute lung injury induced by limb ischemia/reperfusion via down-regulating chemerin and NLRP3 in rats. Shock. 52:134–141. 2019.PubMed/NCBI View Article : Google Scholar

24 

Ning K, Liu WW, Huang JL, Lu HT and Sun XJ: Effects of hydrogen on polarization of macrophages and microglia in a stroke model. Med Gas Res. 8:154–159. 2019.PubMed/NCBI View Article : Google Scholar

25 

Kumagai K, Toyooka T, Takeuchi S, Otani N, Wada K, Tomiyama A and Mori K: Hydrogen gas inhalation improves delayed brain injury by alleviating early brain injury after experimental subarachnoid hemorrhage. Sci Rep. 10(12319)2020.PubMed/NCBI View Article : Google Scholar

26 

Ohno K and Ito M, Ichihara M and Ito M: Molecular hydrogen as an emerging therapeutic medical gas for neurodegenerative and other diseases. Oxid Med Cell Longev. 2012(353152)2012.PubMed/NCBI View Article : Google Scholar

27 

Takeuchi S, Mori K, Arimoto H, Fujii K, Nagatani K, Tomura S, Otani N, Osada H and Wada K: Effects of intravenous infusion of hydrogen-rich fluid combined with intra-cisternal infusion of magnesium sulfate in severe aneurysmal subarachnoid hemorrhage: Study protocol for a randomized controlled trial. BMC Neurol. 14(176)2014.PubMed/NCBI View Article : Google Scholar

28 

Schallner N, Pandit R, LeBlanc R III, Thomas AJ, Ogilvy CS, Zuckerbraun BS, Gallo D, Otterbein LE and Hanafy KA: Microglia regulate blood clearance in subarachnoid hemorrhage by heme oxygenase-1. J Clin Invest. 125:2609–2625. 2015.PubMed/NCBI View Article : Google Scholar

29 

Kaiser S, Frase S, Selzner L, Lieberum JL, Wollborn J, Niesen WD, Foit NA, Heiland DH and Schallner N: Neuroprotection after hemorrhagic stroke depends on cerebral heme oxygenase-1. Antioxidants (Basel). 8(496)2019.PubMed/NCBI View Article : Google Scholar

30 

Afonso MB, Rodrigues PM, Simão AL, Ofengeim D, Carvalho T, Amaral JD, Gaspar MM, Cortez-Pinto H, Castro RE, Yuan J and Rodrigues CM: Activation of necroptosis in human and experimental cholestasis. Cell Death Dis. 7(e2390)2016.PubMed/NCBI View Article : Google Scholar

31 

Chen J, Wang Y, Wu J, Yang J, Li M and Chen Q: The potential value of targeting ferroptosis in early brain injury after acute CNS disease. Front Mol Neurosci. 13(110)2020.PubMed/NCBI View Article : Google Scholar

32 

National Research Council (US). Committee for the Update of the Guide for the Care and Use of Laboratory Animals: The National Academies Collection: Reports funded by National Institutes of Health. In: Guide for the Care and Use of Laboratory Animals. 8th edition. National Academies Press, Washington, DC, 2011.

33 

Flierl MA, Stahel PF, Beauchamp KM, Morgan SJ, Smith WR and Shohami E: Mouse closed head injury model induced by a weight-drop device. Nat Protoc. 4:1328–1337. 2009.PubMed/NCBI View Article : Google Scholar

34 

Tian J, Yang L, Wang P, Yang L and Fan Z: Exogenous CGRP regulates apoptosis and autophagy to alleviate traumatic brain injury through Akt/mTOR signalling pathway. Neurochem Res. 45:2926–2938. 2020.PubMed/NCBI View Article : Google Scholar

35 

Zhuang Z, Zhou ML, You WC, Zhu L, Ma CY, Sun XJ and Shi JX: Hydrogen-rich saline alleviates early brain injury via reducing oxidative stress and brain edema following experimental subarachnoid hemorrhage in rabbits. BMC Neurosci. 13(47)2012.PubMed/NCBI View Article : Google Scholar

36 

Feng Y, Wang R, Xu J, Sun J, Xu T, Gu Q and Wu X: Hydrogen-rich saline prevents early neurovascular dysfunction resulting from inhibition of oxidative stress in STZ-diabetic rats. Curr Eye Res. 38:396–404. 2013.PubMed/NCBI View Article : Google Scholar

37 

Ohsawa I, Ishikawa M, Takahashi K, Watanabe M, Nishimaki K, Yamagata K, Katsura K, Katayama Y, Asoh S and Ohta S: Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals. Nat Med. 13:688–694. 2007.PubMed/NCBI View Article : Google Scholar

38 

Tang C, Shan Y, Hu Y, Fang Z, Tong Y, Chen M, Wei X, Fu X and Xu X: FGF2 attenuates neural cell death via suppressing autophagy after rat mild traumatic brain injury. Stem Cells Int. 2017(2923182)2017.PubMed/NCBI View Article : Google Scholar

39 

Chen J, Zhang C, Yan T, Yang L, Wang Y, Shi Z, Li M and Chen Q: Atorvastatin ameliorates early brain injury after subarachnoid hemorrhage via inhibition of pyroptosis and neuroinflammation. J Cell Physiol. 236:6920–6931. 2021.PubMed/NCBI View Article : Google Scholar

40 

Chen JH, Wu T, Xia WY, Shi ZH, Zhang CL, Chen L, Chen QX and Wang YH: An early neuroprotective effect of atorvastatin against subarachnoid hemorrhage. Neural Regen Res. 15:1947–1954. 2020.PubMed/NCBI View Article : Google Scholar

41 

Chen J, Xuan Y, Chen Y, Wu T, Chen L, Guan H, Yang S, He J, Shi D and Wang Y: Netrin-1 alleviates subarachnoid haemorrhage-induced brain injury via the PPARγ/NF-KB signalling pathway. J Cell Mol Med. 23:2256–2262. 2019.PubMed/NCBI View Article : Google Scholar

42 

Hollingshead JR and Phillips RK: Haemorrhoids: Modern diagnosis and treatment. Postgrad Med J. 92:4–8. 2016.PubMed/NCBI View Article : Google Scholar

43 

Das S, Chattopadhyay D, Chatterjee SK, Mondal SA, Majumdar SS, Mukhopadhyay S, Saha N, Velayutham R, Bhattacharya S and Mukherjee S: Increase in PPARγ inhibitory phosphorylation by Fetuin-A through the activation of Ras-MEK-ERK pathway causes insulin resistance. Biochim Biophys Acta Mol Basis Dis. 1867(166050)2021.PubMed/NCBI View Article : Google Scholar

44 

Li Y, Liu Y, Wu P, Tian Y, Liu B, Wang J, Bihl J and Shi H: Inhibition of ferroptosis alleviates early brain injury after subarachnoid hemorrhage in vitro and in vivo via reduction of lipid peroxidation. Cell Mol Neurobiol. 41:263–278. 2021.PubMed/NCBI View Article : Google Scholar

45 

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.PubMed/NCBI View Article : Google Scholar

46 

Wehn AC, Khalin I, Duering M, Hellal F, Culmsee C, Vandenabeele P, Plesnila N and Terpolilli NA: RIPK1 or RIPK3 deletion prevents progressive neuronal cell death and improves memory function after traumatic brain injury. Acta Neuropathol Commun. 9(138)2021.PubMed/NCBI View Article : Google Scholar

47 

Huang GR and Hao FG: Dexmedetomidine inhibits inflammation to alleviate early neuronal injury via TLR4/NF-κB pathway in rats with traumatic brain injury. Crit Rev Eukaryot Gene Expr. 31:41–47. 2021.PubMed/NCBI View Article : Google Scholar

48 

Li F, Wang X, Zhang Z, Zhang X and Gao P: Dexmedetomidine attenuates neuroinflammatory-induced apoptosis after traumatic brain injury via Nrf2 signaling pathway. Ann Clin Transl Neurol. 6:1825–1835. 2019.PubMed/NCBI View Article : Google Scholar

49 

Yang T, Feng X, Zhao Y, Zhang H, Cui H, Wei M, Yang H and Fan H: Dexmedetomidine enhances autophagy via α2-AR/AMPK/mTOR pathway to inhibit the activation of NLRP3 inflammasome and subsequently alleviates lipopolysaccharide-induced acute kidney injury. Front Pharmacol. 11(790)2020.PubMed/NCBI View Article : Google Scholar

50 

Fei W, Jiao W, Feng X, Chen X and Wang Y: Intermittent hypoxia mimicking obstructive sleep apnea aggravates early brain injury following ICH via neuroinflammation and apoptosis. Mol Med Rep. 24(824)2021.PubMed/NCBI View Article : Google Scholar

51 

Feng X, Ma W, Zhu J, Jiao W and Wang Y: Dexmedetomidine alleviates early brain injury following traumatic brain injury by inhibiting autophagy and neuroinflammation through the ROS/Nrf2 signaling pathway. Mol Med Rep. 24(661)2021.PubMed/NCBI View Article : Google Scholar

52 

Liu L, Xie K, Chen H, Dong X, Li Y and Yu Y, Wang G and Yu Y: Inhalation of hydrogen gas attenuates brain injury in mice with cecal ligation and puncture via inhibiting neuroinflammation, oxidative stress and neuronal apoptosis. Brain Res. 1589:78–92. 2014.PubMed/NCBI View Article : Google Scholar

53 

Choi KS, Kim HJ, Do SH, Hwang SJ and Yi HJ: Neuroprotective effects of hydrogen inhalation in an experimental rat intracerebral hemorrhage model. Brain Res Bull. 142:122–128. 2018.PubMed/NCBI View Article : Google Scholar

54 

Wang P, Zhao M, Chen Z, Wu G, Fujino M, Zhang C, Zhou W, Zhao M, Hirano SI, Li XK and Zhao L: Hydrogen gas attenuates hypoxic-ischemic brain injury via regulation of the MAPK/HO-1/PGC-1a pathway in neonatal rats. Oxid Med Cell Longev. 2020(6978784)2020.PubMed/NCBI View Article : Google Scholar

55 

Dohi K, Kraemer BC, Erickson MA, McMillan PJ, Kovac A, Flachbartova Z, Hansen KM, Shah GN, Sheibani N, Salameh T and Banks WA: Molecular hydrogen in drinking water protects against neurodegenerative changes induced by traumatic brain injury. PLoS One. 9(e108034)2014.PubMed/NCBI View Article : Google Scholar

56 

Tian R, Hou Z, Hao S, Wu W, Mao X, Tao X, Lu T and Liu B: Hydrogen-rich water attenuates brain damage and inflammation after traumatic brain injury in rats. Brain Res. 1637:1–13. 2016.PubMed/NCBI View Article : Google Scholar

57 

Yuan J, Wang D, Liu Y, Chen X, Zhang H, Shen F, Liu X and Fu J: Hydrogen-rich water attenuates oxidative stress in rats with traumatic brain injury via Nrf2 pathway. J Surg Res. 228:238–246. 2018.PubMed/NCBI View Article : Google Scholar

58 

Jia R, Jia N, Yang F, Liu Z, Li R, Jiang Y, Zhao J, Wang L, Zhang S, Zhang Z, et al: Hydrogen alleviates necroptosis and cognitive deficits in lithium-pilocarpine model of status epilepticus. Cell Mol Neurobiol. 39:857–869. 2019.PubMed/NCBI View Article : Google Scholar

59 

Dong XH, Liu H, Zhang MZ, Zhao PX, Liu S, Hao Y and Wang YB: Postconditioning with inhaled hydrogen attenuates skin ischemia/reperfusion injury through the RIP-MLKL-PGAM5/Drp1 necrotic pathway. Am J Transl Res. 11:499–508. 2019.PubMed/NCBI

60 

Wang M, Ke Y, Li Y, Shan Z, Mi W, Cao Y, Feng W and Zheng X: The nephroprotective effects and mechanisms of rehmapicrogenin include ROS inhibition via an oestrogen-like pathway both in vivo and in vitro. Biomed Pharmacother. 138(111305)2021.PubMed/NCBI View Article : Google Scholar

61 

Yu Y, Yang Y, Yang M, Wang C, Xie K and Yu Y: Hydrogen gas reduces HMGB1 release in lung tissues of septic mice in an Nrf2/HO-1-dependent pathway. Int Immunopharmacol. 69:11–18. 2019.PubMed/NCBI View Article : Google Scholar

62 

Chen H, Xie K, Han H, Li Y, Liu L, Yang T and Yu Y: Molecular hydrogen protects mice against polymicrobial sepsis by ameliorating endothelial dysfunction via an Nrf2/HO-1 signaling pathway. Int Immunopharmacol. 28:643–654. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

February-2022
Volume 23 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hu Y, Feng X, Chen J, Wu Y and Shen L: Hydrogen‑rich saline alleviates early brain injury through inhibition of necroptosis and neuroinflammation via the ROS/HO‑1 signaling pathway after traumatic brain injury. Exp Ther Med 23: 126, 2022
APA
Hu, Y., Feng, X., Chen, J., Wu, Y., & Shen, L. (2022). Hydrogen‑rich saline alleviates early brain injury through inhibition of necroptosis and neuroinflammation via the ROS/HO‑1 signaling pathway after traumatic brain injury. Experimental and Therapeutic Medicine, 23, 126. https://doi.org/10.3892/etm.2021.11049
MLA
Hu, Y., Feng, X., Chen, J., Wu, Y., Shen, L."Hydrogen‑rich saline alleviates early brain injury through inhibition of necroptosis and neuroinflammation via the ROS/HO‑1 signaling pathway after traumatic brain injury". Experimental and Therapeutic Medicine 23.2 (2022): 126.
Chicago
Hu, Y., Feng, X., Chen, J., Wu, Y., Shen, L."Hydrogen‑rich saline alleviates early brain injury through inhibition of necroptosis and neuroinflammation via the ROS/HO‑1 signaling pathway after traumatic brain injury". Experimental and Therapeutic Medicine 23, no. 2 (2022): 126. https://doi.org/10.3892/etm.2021.11049