Glycine is a simple nonessential amino acid known to have neuroprotective properties. Treatment with glycine results in reduced infarct volume of the brain, neurologic function scores, and neuronal and microglial death in ischemic stroke injury. Neuroinflammation has been considered a major contributor to cerebral ischemia–induced brain damage. However, the role of glycine in neuroinflammation following ischemic stroke is unclear. The present study aimed to determine whether neuroinflammation is involved in the neuroprotective effects of glycine in cerebral ischemia injury. Ischemic stroke promotes M1 microglial polarization. Interestingly, we found that the injection of glycine in rats after injury can inhibit ischemia-induced inflammation and promote M2 microglial polarization in vivo (Sprague–Dawley rats) and in vitro (cortical microglia and BV-2 cells). We show that glycine suppresses Hif-1α by inhibiting the upregulation of NF-κB p65 after ischemia-reperfusion injury, resulting in the inhibition of proinflammatory activity. The activation of AKT mediates the inhibition of NF-κB p65/Hif-1α signaling by glycine. Moreover, we confirm that glycine-regulated AKT activation is mediated by the inhibition of PTEN in a PTEN depletion cell line, U251 cells. Glycine modulates microglial polarization after ischemic stroke, which indirectly inhibits ischemia-induced neuronal death and functional recovery. Taken together, our findings provide a new understanding of glycine in neuroprotection by inhibiting M1 microglial polarization and promoting anti-inflammation by suppressing NF-κB p65/Hif-1α signaling.

Ischemic stroke is a disease that results in high disability and high mortality worldwide. Because of its increasing prevalence, it has attracted widespread attention. Ischemic stroke is caused by arterial embolism, microangiopathy, or macroangiopathy, resulting in oxygen and glucose deprivation in the brain and leading to brain damage and neurologic deficits (1). Oxidative stress, neuroinflammation, and excitotoxicity are thought to be the main causes of the neuronal death and brain damage caused by ischemia (2). Neuroprotection has been a related problem due to the complex environment after ischemic stroke (3). Although we have extensively described the pathogenesis of ischemic stroke and physiological changes after onset, treatment options are limited. The immune system is closely involved in the various stages of brain damage and tissue repair processes induced by ischemic stroke (4, 5). Immune system–mediated neuroinflammation is heavily involved in determining the fate of the brain after ischemic stroke (6).

Microglia are innate immune cells of the CNS and are the major mediators of neuroinflammation (7, 8). Microglia are called resident macrophages in the brain, and their functions and characteristics are very similar to those of macrophages (9, 10). Microglia can be activated to be polarized after being stimulated, exerting proinflammatory or anti-inflammatory functions by secreting different substances (1113). Hif-1α is a major player in inflammation and controls the expression of genes related to promoting inflammation (14, 15). Hif-1α–regulated inflammation can be mediated through the nuclear translocation of PKM2 (16). However, NF-κB regulates the expression of Hif-1α, and the activation of NF-κB p65 is positively correlated with the expression of Hif-1α (17). AKT is upstream of p65, and the level of p65 can be negatively regulated by AKT activation (18). PTEN is a very important regulator of the PI3K/AKT signaling pathway, which negatively regulates AKT activation (19). Previous results have shown that the inhibition of PTEN not only results in the neuroprotection of neurons but also inhibits inflammation (2022).

Glycine is a simple nonessential amino acid that is an important component of many proteins. Glycine is also crucial for the synthesis of many biomolecules, such as porphyrin, creatine, and purine nucleotides (23). In the CNS, glycine is a major inhibitory neurotransmitter that binds to glycine receptors to inhibit postsynaptic neurons, and it is also a coagonist of excitatory NMDA receptors (a calcium-permeable ion channel) (24, 25). Previous studies have demonstrated that glycine has neuroprotective effects in many disease models, including ischemic stroke injury, hypoxia, anoxia, and intracerebral hemorrhage (26, 27). Clinical trials show that glycine treatment can improve the prognosis of patients with ischemic stroke. Although the neuroprotective effect on glycine has been studied, it remains largely unknown.

In this study, we demonstrated that glycine exhibited neuroprotective effects on neurons and microglia after ischemic stroke injury. Glycine plays a direct role in protecting neurons; furthermore, its beneficial effects are also indirectly mediated by microglia. In terms of mechanism, glycine inhibits NF-κB p65 and Hif-1α by activating AKT and downregulating PTEN, which suppresses ischemia-induced M1 microglia polarization and inhibiting inflammation, resulting in the inhibition of neuronal death followed by ischemic stroke.

Adult male Sprague–Dawley rats were housed with three rats per cage on a 12-h light/dark cycle in a temperature-controlled room (23–25°C) with free access to water and food. Animals were allowed at least 3 d for acclimation before experimentation. We used a total of 214 male rats in our in vivo experiments, and 46 male rats were discarded during the operational procedures. Fifteen adult pregnant female rats and 65 embryos were used in our cortical neuronal cultures experiments. All animal use and experimental protocols were approved and carried out in compliance with the Institutional Animal Care and Use Committee guidelines and the Animal Care and Ethics Committee of Wuhan University School of Medicine. Assignment to the experimental groups and the collection and processing of samples were performed randomly. The experiments were performed by investigators blinded to the assigned group of each animal.

Transient focal cerebral ischemia was induced using the suture occlusion technique (28). The entire process is the same as our laboratory previously described (29). Male Sprague–Dawley rats weighing 250–300 g were anesthetized with 4% isoflurane in 70% N2O and 30% O2 by using a mask. The rectal temperature was maintained at 37.0 ± 0.5°C using a homoeothermic blanket. A midline incision was made in the neck, the right external carotid artery (ECA) was carefully exposed and dissected, and a 3–0 monofilament nylon suture was inserted from the ECA into the right internal carotid artery to occlude the origin of the right middle cerebral artery (∼22 mm). After 90 min of occlusion, the suture was removed to allow reperfusion, the ECA was ligated, and the wound was closed. Sham-operated rats underwent identical surgery and/or intracerebroventricular (i.c.v.) injections except that the suture was inserted and withdrawn immediately. At 24 h after middle cerebral artery occlusion (MCAO), rats (n = 24) were reperfused with ice-cold 0.9% saline after anesthetization with 4% isoflurane in 70% N2O and 30% O2, and brains were quickly removed for Western blot analysis, RT-PCR analysis, behavioral testing, and 2,3,5-triphenyltetrazolium chloride (TTC) staining.

The brain was placed in a cooled matrix, and 2-mm coronal sections were cut. Individual sections were placed in 10-cm petri dishes and incubated for 30 min in a solution of 2% TTC in PBS at 37°C. The slices were fixed in 4% paraformaldehyde at 4°C for 24 h. All image collection, processing, and analysis were performed in a blinded manner and under controlled environmental lighting. The scanned images were analyzed using image analysis software (Image-Pro Plus version 6.0). The infarct volume was calculated to correct for edema. The normal volume of the contralateral hemisphere and the normal volume of the ipsilateral hemisphere were measured, and the infarct percentage was calculated as the percentage of contralateral structure to avoid mismeasurement secondary to edema (30).

Rats were anesthetized with a mixture of 4% isoflurane in 30% O2 and 70% N2O in a sealed perspective box. When the rats were deeply anesthetized, we used ear bars and an upper incisor bar to secure the rat’s head in a stereotaxic frame, and the rats were continuously under anesthesia with 4% isoflurane using a mask. Next, a small sagittal incision was made, and the bregma was located as the anatomical reference point. The cerebral ventricle (from the bregma: lateral, 1.5 mm; anteroposterior, −0.8 mm; depth, 3.5 mm) was performed using a 23-gauge needle attached via polyethylene tubing to a Hamilton microsyringe and drug infusion at a rate of 1.0 μl/min. Proper needle placement was verified by withdrawing a few microliters of clear cerebrospinal fluid into the Hamilton microsyringe.

Cortical neuron cultures were prepared from female Sprague–Dawley rats at 17 d of gestation (31). The pregnant rats were killed by cervical dislocation after anesthetization with 4% isoflurane in 70% N2O and 30% O2. The embryos were sprayed with 70% ethanol that was removed after the rats. The embryos were quickly decapitated, and the cortices were placed in ice-cold plating medium (neurobasal medium, 0.5% FBS, 2% B-27 supplement, 25 mM glutamic acid, and 0.5 mM L-GlutaMAX) following the removal of the meninges. The cortical neurons were plated on petri dishes coated with poly-d-lysine and suspended in plating medium. Half of the plating medium was removed and replaced with maintenance medium (neurobasal medium, 0.5 mM l-glutamine, and 2% B-27 supplement) in the same manner every 3 d. The cultured neurons were used in the experiments after 12 d.

Cortical microglia cultures were prepared from female Sprague–Dawley rats at 17 d of gestation. Pregnant rats died of cervical dislocation after anesthesia with 4% isoflurane with 70% N2O and 30% O2. Rats were sprayed with 70% ethanol and removed from the uterus of pregnant rats. The brain tissue of embryos was quickly separated. Pelleted cells were resuspended in warmed DMEM culture medium completed with 10% heat-inactivated FBS, 1% antibiotic-antimycotic, and 5 ng/ml carrier-free recombinant mouse GM-CS. A total of 1.3 × 106 cells were seeded into tissue culture–grade poly l-lysine–coated T75 cell culture–treated flasks and placed in a 37°C incubator with relative humidity. The culture supernatant was replaced twice weekly with 10 ml of fresh completed medium until confluency of cells was observed at approximately 3 wk.

For the oxygen-glucose deprivation (OGD) challenge, cells were transferred to a glucose-free extracellular solution (116 mM NaCl, 0.8 mM MgSO4, 5.4 mM KCl, 1.0 mM NaH2PO4, 26 mM NaHCO3, and 1.8 mM CaCl2) and deoxygenated environment, placed in a humidified chamber (Plas-Labs, Lansing, MI), and maintained at 37°C in 5% H2/85% CO2/10% N2 for 60 min. Next, the cells were replaced with fresh maintenance medium containing an appropriate concentration of reagents for 24 h during the recovery period in a 5% CO2/95% O2 incubator. The control cultures were first transferred to another extracellular solution (5.4 mM KCl, 116 mM NaCl, 0.8 mM MgSO4, 1.8 mM CaCl2, 1.0 mM NaH2PO4, 26 mM NaHCO3, and 33 mM glucose) and placed in the humidified chamber, which was maintained at 37°C in 95% O2/5% CO2 for 60 min (32). Finally, fresh maintenance medium was replaced for the whole period at 37°C in a 95% O2/5% CO2 incubator.

Western blotting was performed as previously described (33). Briefly, a polyvinylidene difluoride membrane from Millipore was used to incubate the samples with the primary Abs against NF-κB p65 (rabbit, 1:1000), Hif-1α (rabbit, 1:1000), AKT (mouse, 1:1000), phospho-AKT (Ser473) (rabbit, 1:2000), PTEN (rabbit, 1:1000), and actin (rabbit, 1:2000) from Cell Signaling Technology. The primary Abs were labeled with secondary Abs, and protein bands were imaged using SuperSignal West Femto Maximum Sensitivity Substrate (Pierce, Rockford, IL). The EC3 Imaging System (Uplant; UVP) was used to obtain blot images directly from the polyvinylidene difluoride membrane. The Western blot data were quantified using Image-Pro Plus version 6.0.

Rats were treated with an overdose of isoflurane and then intracardiac perfusion with 0.9% saline, placed in 4% paraformaldehyde at 4°C for 24 h, and transferred into 30% sucrose solution in 100 mol/ml phosphate buffer at 4°C for 72 h. Then, the brain tissue was kept in 4% paraformaldehyde solution at 4°C overnight. Brain tissues were cut into 16-μm coronal sections by a Leica VT1000 S Vibratome (Leica Microsystems AG, Nussloch, Germany). Fluoro Jade–C (FJC) labeling was performed by using the standard protocol (34). Brain sections were first immersed in 1% sodium hydroxide in 80% ethanol for 5 min, then rinsed in 70% ethanol for 2 min, then 2 min in distilled water, and then incubated in 0.06% potassium permanganate solution for 10 min. Following a 2-min distilled water rinse, the brain sections were transferred into 0.0001% solution of FJC (Sigma-Aldrich) 10 min, which was dissolved in 0.1% acetic acid. Brain sections were rinsed with water three times for 1 min, then air dried on a warmer at 50°C for at least 5 min, and finally immersed in xylene for at least 1 min. The brain sections were mounted with DPX Mountant media (Sigma-Aldrich). The brain sections were photographed by a blinded investigator using an Olympus fluorescence microscope (IX51; Olympus, Japan). A series of photomicrographs were taken from three regions of the ipsilateral cerebral cortex with a ×20 objective, and FJC-positive cells were counted by ImageJ software (ImageJ). The data were expressed as cells per mm2.

The immunostained brain sections were prepared as FJC staining, and the immunofluorescence staining steps were based on the description of the prior execution (35). The brain sections were treated with the primary Ab mouse anti-NeuN (neuronal-specific nuclear protein) from Chemicon. The secondary Ab was goat anti-mouse 488 from Molecular Probes (Eugene, OR). The DAPI probe was from Life Technologies. The sections were photographed by a blinded investigator using an Olympus fluorescence microscope (IX51; Olympus), and images were analyzed by ImageJ software (ImageJ).

Double-stranded oligonucleotides corresponding to the target sequences were cloned into the lenti-CRISPR-V2 vector and cotransfected packaging plasmids into HEK293 cells. Two days after transfection, the viruses were harvested, ultrafiltrated (0.22-mm filter; Millipore), and used to infect BV-2 cells in the presence of polybrene (8 mg/ml). The infected cells were selected with puromycin (1 mg/ml) for at least 5 d.

BV-2 cells and U251 cells were purchased from the Chinese Academy of Sciences Cell Bank. U251 cells were sown in a six-well plate (8 × 105 cells per well) in DMEM supplemented with 10% heat-inactivated FBS, penicillin G (100 U/ml), streptomycin (100 mg/ml), and l-glutamine (2.0 mM) and incubated at 37°C in a humidified atmosphere containing 5% CO2 and 95% air.

Short hairpin RNA (shRNA) targeting Hif-1α was inserted into the pGIPZ vector as previously reported, and the pGIPZ control was generated with the control oligonucleotide 5′-GCTTCTAACACCGGAGGTCTT-3′. PCR-amplified mouse Hif-1α was cloned into the pcDNA3.1/hypro (+) vector between BamH I and Not I. pCDNA3.1(+)-Hif-1α–WT (WT Hif-1α) was generated using the QuikChange Site-Directed Mutagenesis Kit (Stratagene, La Jolla, CA).

When the confluence of BV-2 cells reached 60–70% on the treatment day, the cells were transfected with Hif-1α shRNA or mouse Hif-1α plasmid WT Hif-1α for 8 h. The medium was then replaced with normal growth medium for 24 h. On the following day, the cells were then collected for Western blot analysis.

Total RNA was isolated from control brains and ischemic brains using the RNeasy Mini Kit (Qiagen) according to the manufacturer’s instructions; the first strand of cDNA was synthesized using 5-μg Superscript First-Strand Synthesis System for RT-PCR (Invitrogen). PCR was performed on the Opticon 2 Real-Time PCR Detection System (Bio-Rad) using the corresponding primers (Table I) and SYBR Green PCR Master Mix (Invitrogen). The cycle time value was normalized to the GAPDH level of the same sample. The mRNA expression level was then reported as the fold change compared with the control group.

Lactate dehydrogenase (LDH) release was analyzed using a colorimetric CytoTox 96 Cytotoxicity kit (Promega). Cell viability in the neuronal cultures was evaluated by the ability to take up thiazolyl blue tetrazolium bromide (mingest thiazolyl tetrazolium) (PowerWave X; BioTek, Winooski, VT). The two methods were performed following the manufacturer’s instructions.

The rats were subjected to a modified neurologic severity score test as reported previously. These tests are a battery of reflex, sensory, motor, and balance tests, which are similar to the contralateral neglect tests in humans. Neurological function was graded on a scale of 0 to 18 (normal score, 0; maximal deficit score, 18) (36).

The beam-walk test measures the complex neuromotor function of animals. The animal was timed as it walked across a (100 × 2-cm) beam. A box for the animal to feel safe was placed at one end of the beam. A loud noise was created to stimulate the animal to walk toward and into the box. Scoring was based on the time it took the rat to go into the box. The higher the score was, the more severe the neurologic deficit (37).

A modified sticky-tape test was performed to evaluate forelimb function. A sleeve was created using a 3.0 × 1.0-cm piece of yellow paper tape and was subsequently wrapped around the forepaw so that the tape attached to itself and allowed the digits to protrude slightly from the sleeve. The typical response is for the rat to vigorously attempt to remove the sleeve by either pulling at the tape with its mouth or brushing the tape with its contralateral paw. The rat was placed in its cage and observed for 30 s. Two timers were started; the first ran without interruption and the second was turned on only while the animal attempted to remove the tape sleeve. The ratio of the left (affected)/right (unaffected) forelimb performance was recorded. The contralateral and ipsilateral limbs were tested separately. The test was repeated three times per test day, and the best two scores of the day were averaged. The lower the ratio, the more severe the neurologic deficit (38).

The experimental design and analysis of this study resulted in data and statistical analysis that comply with recommendations. All data are expressed as the mean ± SE. Student t test and variance analysis were used for differences among groups. p < 0.05 was considered statistically significant.

We generated an experimental ischemic stroke rat model induced by MCAO for 1.5 h followed by various periods of reperfusion (39) and showed the structure of coronal brain slices after MCAO injury in rats (Fig. 1A, Supplemental Fig. 1). TTC staining was performed to detect the infarct volume of the rat brain 24 h after ischemia-reperfusion (I/R), and the results showed that the ischemia-induced cerebral infarction accounts for 21% of the total brain volume (Fig. 1B, 1C). Additionally, the i.c.v. injection of glycine (100 μg/100 g) immediately after MCAO (40) significantly improved ischemic stroke–induced cerebral infarction volume (Fig. 1B, 1C). In the pathologic condition of ischemic stroke, the cortex of the peri-infarct area is mainly impacted. Therefore, we performed FJC staining to evaluate the degenerate neurons in the peri-infarct area 24 h after I/R. The results showed that the number of FJC-positive degenerating neurons was downregulated by treatment with glycine (Fig. 1D). In addition, we performed a sequence of neurobehavioral tests in rats and showed that glycine promoted functional recovery after ischemic stroke (Fig. 1E–G). Together, glycine treatment reduces brain damage and promotes functional recovery after cerebral ischemia injury.

FIGURE 1.

Glycine reduces ischemic stroke–induced brain damage and promotes functional recovery. (A) Schematic diagram of pero-infarcts and core anatomy of the rat brain after ischemic stroke. (B and C) TTC-stained brain slice images in rats 24 h following I/R (B) and quantification analysis of the infarct volume (C) (n = 6 in each group, *p < 0.05 versus the I/R + vehicle). Scale bar, 5 mm. (D) FJC staining (bottom) and quantification (top) analysis of FJC-positive neurons in rats 24 h following I/R (n = 6 at each group, *p < 0.05 versus the I/R + vehicle). Scale bar, 20 μm. (E) Rats treated with glycine had lower scores in the mNSS test on days 7 and 14 after I/R injury than rats in the I/R + vehicle group. (n = 6 in each group, *p < 0.05 versus the I/R + vehicle). (F) Rats treated with glycine had lower scores in the beam-walking test on days 7 and 14 after I/R injury than rats in the I/R + vehicle group. (n = 6 in each group, *p < 0.05 versus the I/R + vehicle). (G) Rats treated with glycine had a higher ratio in the MST test at days 7 and 14 after I/R injury than rats in the I/R + vehicle group (n = 6 in each group, *p < 0.05 versus the I/R + vehicle). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis. mNSS, modified neurologic severity score; MST, modified sticky-tape.

FIGURE 1.

Glycine reduces ischemic stroke–induced brain damage and promotes functional recovery. (A) Schematic diagram of pero-infarcts and core anatomy of the rat brain after ischemic stroke. (B and C) TTC-stained brain slice images in rats 24 h following I/R (B) and quantification analysis of the infarct volume (C) (n = 6 in each group, *p < 0.05 versus the I/R + vehicle). Scale bar, 5 mm. (D) FJC staining (bottom) and quantification (top) analysis of FJC-positive neurons in rats 24 h following I/R (n = 6 at each group, *p < 0.05 versus the I/R + vehicle). Scale bar, 20 μm. (E) Rats treated with glycine had lower scores in the mNSS test on days 7 and 14 after I/R injury than rats in the I/R + vehicle group. (n = 6 in each group, *p < 0.05 versus the I/R + vehicle). (F) Rats treated with glycine had lower scores in the beam-walking test on days 7 and 14 after I/R injury than rats in the I/R + vehicle group. (n = 6 in each group, *p < 0.05 versus the I/R + vehicle). (G) Rats treated with glycine had a higher ratio in the MST test at days 7 and 14 after I/R injury than rats in the I/R + vehicle group (n = 6 in each group, *p < 0.05 versus the I/R + vehicle). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis. mNSS, modified neurologic severity score; MST, modified sticky-tape.

Close modal

Neurons are structural and functional units of the CNS, whereas glial cells mainly play a supporting role; however, both cell types are very important (41). We cultured primary neurons and microglia from the rat cortex and challenged the neurons and microglia with OGD for 1.5 h to simulate ischemic stroke injury in vitro (42). The cell viability and LDH release results showed that treatment with glycine (100 μM) 1 h before OGD insult decreased OGD-induced neuronal death (Fig. 2A, 2B). Additionally, a similar result was observed in microglia (Fig. 2C, 2D). Because the function of glial cells is to support neurons in the CNS, we tested the indirect effects of microglial glycine treatment on neurons. Glycine treatment was administered to microglia 1 h before the OGD challenge. Then, we recovered the microglia culture medium (MCM) 22.5 h after OGD insult, mixed it with DMEM at a 1:1 ratio, and used this mixed medium to culture cortical neurons after OGD insult (Fig. 2E). We found that OGD-induced neuronal death was upregulated after treatment with MCM without glycine; however, glycine-treated MCM reduced OGD-induced neuronal death compared with the non-MCM group (Fig. 2F, 2G). In summary, treatment with glycine not only reduced OGD-induced neuronal death but also OGD-induced microglial death, and the treatment of microglia with glycine also has an indirect neuroprotective effect on neurons.

FIGURE 2.

Glycine downregulates OGD-induced neuronal and microglial death. (A and B) Cell viability (A) and LDH release (B) in cultured cortical neurons after OGD insult shows that glycine (100 μM) decreased OGD-induced neuronal death (n = 6 at each time point, *p < 0.05 versus vehicle). (C and D) Cell viability (C) and LDH release (D) in cultured cortical microglia after OGD insult shows that glycine (100 μM) decreased OGD-induced microglial death (n = 6 at each time point, *p < 0.05 versus vehicle). (E) Schematic diagram of neuron processing MCM. (F and G) Cell viability (F) and LDH release (G) in cultured cortical neurons after OGD insult shows that non-MCM increased the OGD-induced neuronal death compared with that of vehicle and that glycine-MCM decreased the OGD-induced neuronal death compared with that of non-MCM (n = 6 at each time point, *p < 0.05 versus vehicle, #p < 0.05 versus non-MCM). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

FIGURE 2.

Glycine downregulates OGD-induced neuronal and microglial death. (A and B) Cell viability (A) and LDH release (B) in cultured cortical neurons after OGD insult shows that glycine (100 μM) decreased OGD-induced neuronal death (n = 6 at each time point, *p < 0.05 versus vehicle). (C and D) Cell viability (C) and LDH release (D) in cultured cortical microglia after OGD insult shows that glycine (100 μM) decreased OGD-induced microglial death (n = 6 at each time point, *p < 0.05 versus vehicle). (E) Schematic diagram of neuron processing MCM. (F and G) Cell viability (F) and LDH release (G) in cultured cortical neurons after OGD insult shows that non-MCM increased the OGD-induced neuronal death compared with that of vehicle and that glycine-MCM decreased the OGD-induced neuronal death compared with that of non-MCM (n = 6 at each time point, *p < 0.05 versus vehicle, #p < 0.05 versus non-MCM). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

Close modal

Microglia are innate immune cells of the CNS and are characterized by resident macrophages in the brain, which are major mediators of neuroinflammation (43). Microglia can be activated after ischemic stroke injury. Activated microglia have two phenotypes: classically activated (M1 microglia) and alternatively activated (M2 microglia), as described (13, 44). The representative markers of M1 microglia are IL-1β, TNF-α, and CD32. Arg-1, CD206, and YM-1 are markers of M2 microglia. To further investigate the mechanism by which glycine regulates microglia and indirectly exerts neuroprotection of neurons, RT-PCR analysis of mRNA levels of M1 and M2 microglial markers in rats 24 h after I/R was performed. The results showed that ischemic stroke injury promoted M1 microglial polarization, whereas glycine treatment inhibited ischemia-induced inflammation and promoted M2 microglial polarization (Fig. 3A, 3B, Table I). Previous studies have demonstrated that NF-κB and Hif-1α play a crucial role in inflammation (15, 45). We showed that p65 and Hif-1α were increased after cerebral ischemia, which supports the function of p65 and Hif-1α in promoting inflammation (Fig. 3C, 3D); however, glycine treatment inhibited these processes (Fig. 3C, 3D). Similar results were observed in primary cultured microglia in vitro (Fig. 3E–G). Together, glycine-induced anti-inflammatory effects may be mediated by NF-κB and Hif-1α.

FIGURE 3.

Glycine inhibits ischemia-induced M1 microglial polarization and promotes anti-inflammatory activity. (A and B) RT-PCR analysis of M1 microglial markers (A) and M2 microglial markers (B) in rats 24 h after I/R (n = 6 in each group, *p < 0.05 versus sham, #p < 0.05 versus I/R + vehicle). (C and D) Western blotting analysis of p65 (C) and Hif-1α (D) in rats 24 h after I/R (n = 6 in each group, *p < 0.05 versus sham, #p < 0.05 versus I/R + vehicle). (E) Western blotting analysis of Hif-1α and p65 in cultured cortical microglia 24 h after OGD insult (n = 6 in each group, *p < 0.05 versus control, #p < 0.05 versus OGD + vehicle). (F and G) RT-PCR analysis of M1 microglial markers (F) and M2 microglial markers (G) in microglia 24 h after OGD insult (n = 6 in each group, *p < 0.05 versus control, #p < 0.05 versus OGD + vehicle). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

FIGURE 3.

Glycine inhibits ischemia-induced M1 microglial polarization and promotes anti-inflammatory activity. (A and B) RT-PCR analysis of M1 microglial markers (A) and M2 microglial markers (B) in rats 24 h after I/R (n = 6 in each group, *p < 0.05 versus sham, #p < 0.05 versus I/R + vehicle). (C and D) Western blotting analysis of p65 (C) and Hif-1α (D) in rats 24 h after I/R (n = 6 in each group, *p < 0.05 versus sham, #p < 0.05 versus I/R + vehicle). (E) Western blotting analysis of Hif-1α and p65 in cultured cortical microglia 24 h after OGD insult (n = 6 in each group, *p < 0.05 versus control, #p < 0.05 versus OGD + vehicle). (F and G) RT-PCR analysis of M1 microglial markers (F) and M2 microglial markers (G) in microglia 24 h after OGD insult (n = 6 in each group, *p < 0.05 versus control, #p < 0.05 versus OGD + vehicle). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

Close modal

To further investigate whether glycine-regulated microglial polarization is mediated by p65 and Hif-1α, we first determined the relationship between p65 and Hif-1α. Western blotting analysis of Hif-1α in rats 1 h after treatment with the NF-κB p65 inhibitor helenalin (200 μM, 2 μl) by i.c.v injection showed that Hif-1α was downregulated after the inhibition of p65 (Supplemental Fig. 2A). Similar results were observed in vitro (Supplemental Fig. 2B). Meanwhile, we treated cultured cortical microglia with helenalin (2 μM) 1 h after OGD insult, and the results showed that the level of Hif-1α was downregulated by the inhibition of p65 (Supplemental Fig. 2C). In addition, the transfection of p65 shRNA or the expression of WT p65 in BV-2 cells showed that the level of Hif-1α was downregulated or upregulated, respectively (Fig. 4A, 4B). To further confirm the relationship between glycine, NF-κB p65, and Hif-1α, we established a p65 knockout BV-2 cell line and showed that the regulation of Hif-1α by glycine after the knockout of p65 was no longer observed (Fig. 4C). These results suggest that glycine-regulated Hif-1α is mediated by p65. Furthermore, the transfection of Hif-1α shRNA or expression of WT Hif-1α in BV-2 cells did not result in a significant difference in p65 levels (Fig. 4D, 4E). NF-κB p65 is upstream of Hif-1α. Moreover, we pretreated rats with a p65 inhibitor 1 h before MCAO. Western blotting analysis of p65 and Hif-1α showed that glycine-regulated p65 and Hif-1α expression disappeared after I/R (Fig. 4F). RT-PCR analysis of M1 and M2 microglial markers in rats after I/R 24 h revealed that glycine-mediated microglial polarization was significantly inhibited (Figs. 4G, 4H, 3A, 3B). Similar results were also observed in cultured microglia in vitro (Supplemental Fig. 2D, 2E). Together, these results indicate that NF-κB p65/Hif-1α mediates glycine-regulated microglial polarization after ischemic stroke injury.

FIGURE 4.

Glycine promotes M2 microglial polarization by inhibiting NF-κB p65 and Hif-1α. (A and B) Western blotting analysis of p65 and Hif-1α in BV-2 cells after transfection of p65 shRNA or WT p65 (n = 6 in each group, *p < 0.05 versus EV). (C) Western blotting analysis of p65 and Hif-1α in p65 knockout BV-2 cells after glycine treatment (n = 6 in each group). (D and E) Western blotting analysis of p65 and Hif-1α in BV-2 cells after transfection of Hif-1α shRNA or WT Hif-1α (n = 6 in each group, *p < 0.05 versus EV). (F) Western blotting analysis of Hif-1α and p65 in rats after glycine treatment with I/R + helenalin (n = 6 in each group). (G and H) RT-PCR analysis of M1 microglial markers (G) and M2 microglial markers (H) in rats after glycine treatment with I/R + helenalin (n = 6 in each group). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

FIGURE 4.

Glycine promotes M2 microglial polarization by inhibiting NF-κB p65 and Hif-1α. (A and B) Western blotting analysis of p65 and Hif-1α in BV-2 cells after transfection of p65 shRNA or WT p65 (n = 6 in each group, *p < 0.05 versus EV). (C) Western blotting analysis of p65 and Hif-1α in p65 knockout BV-2 cells after glycine treatment (n = 6 in each group). (D and E) Western blotting analysis of p65 and Hif-1α in BV-2 cells after transfection of Hif-1α shRNA or WT Hif-1α (n = 6 in each group, *p < 0.05 versus EV). (F) Western blotting analysis of Hif-1α and p65 in rats after glycine treatment with I/R + helenalin (n = 6 in each group). (G and H) RT-PCR analysis of M1 microglial markers (G) and M2 microglial markers (H) in rats after glycine treatment with I/R + helenalin (n = 6 in each group). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

Close modal

Previous studies have demonstrated that AKT activation inhibits M1 macrophage polarization and that AKT is upstream of p65 (18). In this study, we confirmed that glycine-regulated microglial polarization after ischemic stroke is dependent on p65 and Hif-1α. We hypothesized that AKT activation mediates glycine-regulated microglial polarization. Western blotting analysis of S473 p-AKT showed that the level of p-AKT was downregulated after I/R in vivo and in vitro. Meanwhile, treatment with glycine increased the level of p-AKT (Fig. 5A, Supplemental Fig. 3A). Next, Western blotting analysis of p65 and Hif-1α in rats 1 h after treatment with the AKT inhibitor IV (IV) (100 μM, 2 μl) by i.c.v injection showed that p65 and Hif-1α were upregulated after the inhibition of AKT (Fig. 5B, 5C), and similar results were observed in vitro (Supplemental Fig. 3B, 3C). Furthermore, Western blotting analysis of p65 and Hif-1α after pretreatment with IV 1 h before I/R showed that treatment with glycine did not result in significant changes in the levels of p65 and Hif-1α after the inhibition of AKT in vivo and in vitro (Fig. 5D, Supplemental Fig. 3D). In addition, RT-PCR analysis of M1 and M2 microglial markers revealed that the inhibition of AKT significantly suppressed glycine-induced M2 microglial polarization (Fig. 5E, 5F, Supplemental Fig. 3E, 3F). Together, glycine regulates the AKT activation that reduces the levels of NF-κB p65 and Hif-1α, subsequently promoting anti-inflammatory activity following ischemic stroke in vitro.

FIGURE 5.

Glycine regulates NF-κB p65 and Hif-1α by modulating AKT activation and regulating ischemia-induced microglial polarization. (A) Western blotting analysis of p-AKT in rats after glycine treatment 24 h following I/R (n = 6 in each group, *p < 0.05 versus sham, #p < 0.05 versus I/R + vehicle). (B) Western blotting analysis of p-AKT in rats after treatment with IV (n = 6 in each group, *p < 0.05 versus vehicle). (C) Western blotting analysis of p65 and Hif-1α in rats after treatment of IV (n = 6 in each group, *p < 0.05 versus vehicle). (D) Western blotting analysis of Hif-1α and p65 in rats after glycine treatment with I/R + IV (n = 6 in each group). (E and F) RT-PCR analysis of M1 microglial markers (E) and M2 microglial markers (F) in rats after pretreatment with IV (n = 6 in each group). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

FIGURE 5.

Glycine regulates NF-κB p65 and Hif-1α by modulating AKT activation and regulating ischemia-induced microglial polarization. (A) Western blotting analysis of p-AKT in rats after glycine treatment 24 h following I/R (n = 6 in each group, *p < 0.05 versus sham, #p < 0.05 versus I/R + vehicle). (B) Western blotting analysis of p-AKT in rats after treatment with IV (n = 6 in each group, *p < 0.05 versus vehicle). (C) Western blotting analysis of p65 and Hif-1α in rats after treatment of IV (n = 6 in each group, *p < 0.05 versus vehicle). (D) Western blotting analysis of Hif-1α and p65 in rats after glycine treatment with I/R + IV (n = 6 in each group). (E and F) RT-PCR analysis of M1 microglial markers (E) and M2 microglial markers (F) in rats after pretreatment with IV (n = 6 in each group). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

Close modal

PTEN is a tumor suppressor, and the inhibition of PTEN activates AKT through the PI3K signaling pathway (46, 47). The inhibition of PTEN reduces ischemia-induced neuronal death (48). Western blotting analysis of PTEN in rats 1 h after glycine treatment demonstrated that PTEN was downregulated by glycine (Fig. 6A). To confirm whether glycine-induced AKT activation is mediated by PTEN, we used a PTEN-deficient U251 cell line (Fig. 6B). Western blotting analysis of p-AKT in U251 cells 1 h after treatment with glycine showed that the level of p-AKT did not significantly change (Fig. 6C). These results indicate that glycine-mediated AKT activation is dependent on PTEN. Together, glycine inhibits PTEN regulation of the AKT/NF-κB/Hif-1α signaling pathway and subsequently suppresses M1 microglia polarization.

FIGURE 6.

Glycine regulates OGD-induced microglia polarization by inhibiting PTEN regulation of the AKT/NF-κB/Hif-1α signaling pathway. (A) Western blotting analysis of PTEN in rats after treatment with glycine (n = 6 in each group, *p < 0.05 versus vehicle). (B) Western blotting analysis of PTEN shows no PTEN expression in U251 cells (n = 6). (C) Western blotting analysis of p-AKT in U251 cells shows that the level of p-AKT was not significantly different after treatment with glycine (n = 6 in each group). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

FIGURE 6.

Glycine regulates OGD-induced microglia polarization by inhibiting PTEN regulation of the AKT/NF-κB/Hif-1α signaling pathway. (A) Western blotting analysis of PTEN in rats after treatment with glycine (n = 6 in each group, *p < 0.05 versus vehicle). (B) Western blotting analysis of PTEN shows no PTEN expression in U251 cells (n = 6). (C) Western blotting analysis of p-AKT in U251 cells shows that the level of p-AKT was not significantly different after treatment with glycine (n = 6 in each group). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

Close modal

In Fig. 2F, 2G, we demonstrated that glycine-treated microglia can indirectly reduce OGD-induced neuronal death. We also confirmed that glycine-mediated microglial polarization is dependent on the PTEN/AKT/NF-κB/Hif-1α signaling pathway. In this study, we pretreated cultured cortical microglia with the NF-κB inhibitor helenalin (2 μM) or IV (1 μM) 1 h before glycine treatment and then used the MCM to culture neurons (Fig. 7A). Cell viability and LDH release showed that the inhibition of AKT or NF-κB inhibited the indirect neuroprotection of neurons by glycine (Fig. 7B, 7C). Meanwhile, we pretreated cultured cortical neurons with the NF-κB inhibitor helenalin or IV and demonstrated that the OGD-induced neuronal death was directly decreased or increased, respectively (Fig. 7D, 7E). Furthermore, TTC staining, FJC staining, and neurologic function scores showed that the inhibition of AKT or NF-κB blocked the glycine-induced decrease in infarct volume and reduction in FJC-positive degenerative neurons and improved functional recovery (Fig. 8A–F).

FIGURE 7.

Glycine indirectly reduces OGD-induced neuronal death by modulating microglial polarization through the regulation of the PTEN/AKT/NF-κB/Hif-1α signaling pathway (A) A diagram showing the time of OGD insult and I/R injury and the helenalin, IV, and glycine treatment procedure. (B and C) Cell viability (B) and LDH release (C) in cultured cortical neurons 22.5 h after OGD insult with MCM treatment (n = 6 in each group, *p < 0.05 versus non-MCM, #p < 0.05 versus helenalin + MCM, §p < 0.05 versus IV + MCM). (D and E) Cell viability (D) and LDH release (E) in cultured cortical neurons 22.5 h after OGD insult with helenalin or IV treatment (n = 6 in each group, *p < 0.05 versus vehicle). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

FIGURE 7.

Glycine indirectly reduces OGD-induced neuronal death by modulating microglial polarization through the regulation of the PTEN/AKT/NF-κB/Hif-1α signaling pathway (A) A diagram showing the time of OGD insult and I/R injury and the helenalin, IV, and glycine treatment procedure. (B and C) Cell viability (B) and LDH release (C) in cultured cortical neurons 22.5 h after OGD insult with MCM treatment (n = 6 in each group, *p < 0.05 versus non-MCM, #p < 0.05 versus helenalin + MCM, §p < 0.05 versus IV + MCM). (D and E) Cell viability (D) and LDH release (E) in cultured cortical neurons 22.5 h after OGD insult with helenalin or IV treatment (n = 6 in each group, *p < 0.05 versus vehicle). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis.

Close modal
FIGURE 8.

Glycine reduces ischemia-induced neuronal death and brain damage through regulation of the PTEN/AKT/NF-κB/Hif-1α signaling pathway. (A and B) TTC-stained rat brain slice images 24 h following I/R (A) and quantification of the infarct volume (B) after I/R with glycine ± helenalin ± IV treatment (n = 6 in each group, *p < 0.05 versus I/R + vehicle, #p < 0.05 versus I/R + helenalin, §p < 0.05 versus I/R + IV). Scale bar, 5 mm. (C) FJC staining analysis of FJC-positive neurons with glycine ± helenalin ± IV treatment (n = 6 in each group, *p < 0.05 versus I/R + vehicle, #p < 0.05 versus I/R + helenalin, §p < 0.05 versus I/R + IV). Scale bar, 20 μm. (DF) mNSS test, beam-walking test, and MST test results show that glycine does not promote functional recovery after ischemic stroke with helenalin or IV pretreatment (n = 6 in each group, *p < 0.05 versus I/R + vehicle, #p < 0.05 versus I/R + helenalin, §p < 0.05 versus I/R + IV). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis. mNSS, modified neurologic severity score; MST, modified sticky-tape.

FIGURE 8.

Glycine reduces ischemia-induced neuronal death and brain damage through regulation of the PTEN/AKT/NF-κB/Hif-1α signaling pathway. (A and B) TTC-stained rat brain slice images 24 h following I/R (A) and quantification of the infarct volume (B) after I/R with glycine ± helenalin ± IV treatment (n = 6 in each group, *p < 0.05 versus I/R + vehicle, #p < 0.05 versus I/R + helenalin, §p < 0.05 versus I/R + IV). Scale bar, 5 mm. (C) FJC staining analysis of FJC-positive neurons with glycine ± helenalin ± IV treatment (n = 6 in each group, *p < 0.05 versus I/R + vehicle, #p < 0.05 versus I/R + helenalin, §p < 0.05 versus I/R + IV). Scale bar, 20 μm. (DF) mNSS test, beam-walking test, and MST test results show that glycine does not promote functional recovery after ischemic stroke with helenalin or IV pretreatment (n = 6 in each group, *p < 0.05 versus I/R + vehicle, #p < 0.05 versus I/R + helenalin, §p < 0.05 versus I/R + IV). Data are expressed as the mean ± SE. Statistical differences were determined by Student t test and variance analysis. mNSS, modified neurologic severity score; MST, modified sticky-tape.

Close modal
Table I.
Primers for RT-PCR
GenePrimer
M1  
 IL-1β SENS: 5′-GAGGACATGAGCACCTTCTTT-3′ 
REVS: 5′-GCCTGTAGTGCAGTTGTCTAA-3′ 
 TNF-α SENS: 5′-ACCACGCTCTTCTGTCTACT-3′ 
REVS: 5′-GTTTGTGAGTGTGAGGGTCTG-3′ 
 CD32 SENS: 5′-AATCCTGCCGTTCCTACTGATC-3′ 
REVS: 5′-GTGTCACCGTGTCTTCCTTGAG-3′ 
M2  
 Arg-1 SENS: 5′-TCACCTGAGCTTTGATGTCG-3′ 
REVS: 5′-CTGAAAGGAGCCCTGTCTTG-3′ 
 CD206 SENS: 5′-CAAGGAAGGTTGGCATTTGT-3′ 
REVS: 5′-CCTTTCAGTCCTTTGCAAGC-3′ 
 YM-1 SENS: 5′-CAGGGTAATGAGTGGGTTGG-3′ 
REVS: 5′-CACGGCACCTCCTAAATTGT-3′ 
GenePrimer
M1  
 IL-1β SENS: 5′-GAGGACATGAGCACCTTCTTT-3′ 
REVS: 5′-GCCTGTAGTGCAGTTGTCTAA-3′ 
 TNF-α SENS: 5′-ACCACGCTCTTCTGTCTACT-3′ 
REVS: 5′-GTTTGTGAGTGTGAGGGTCTG-3′ 
 CD32 SENS: 5′-AATCCTGCCGTTCCTACTGATC-3′ 
REVS: 5′-GTGTCACCGTGTCTTCCTTGAG-3′ 
M2  
 Arg-1 SENS: 5′-TCACCTGAGCTTTGATGTCG-3′ 
REVS: 5′-CTGAAAGGAGCCCTGTCTTG-3′ 
 CD206 SENS: 5′-CAAGGAAGGTTGGCATTTGT-3′ 
REVS: 5′-CCTTTCAGTCCTTTGCAAGC-3′ 
 YM-1 SENS: 5′-CAGGGTAATGAGTGGGTTGG-3′ 
REVS: 5′-CACGGCACCTCCTAAATTGT-3′ 

Ischemic stroke is a global disease that has drawn increasing attention because of its high disability and mortality rates; however, the therapeutic options for it are limited. Glycine confers protection against neuronal death under in vitro and in vivo experimental conditions (49, 50). Clinical trials show that glycine treatment can improve the prognosis of patients with ischemic stroke (51). However, the mechanism of the neuroprotective role of glycine remains unclear. In this study, we demonstrate that glycine not only inhibits neuronal death directly after I/R but indirectly through microglia. Glycine inhibits ischemia-induced M1 microglial polarization and promotes anti-inflammatory effects by inhibiting PTEN and activating AKT and then inhibiting NF-κB p65 and Hif-1α, resulting in the indirect inhibition of ischemia-induced neuronal death.

Glycine is a simple nonessential amino acid that is an important part of many proteins and is also a major inhibitory neurotransmitter that binds to glycine receptors to inhibit postsynaptic neurons (52). Treatment with glycine reduced the ischemic stroke–induced brain damage and neuronal death. We found that glycine can directly reduce OGD-induced neuronal death. However, glycine can inhibit OGD-induced microglial death and indirectly reduce neuronal death. Previous studies have demonstrated that the activation of GluN2A-containing NMDA receptors, but not GluN2B-containing NMDA receptors, contributes to glycine-induced neuroprotection in ischemic stroke injury (40). These studies show that the neuroprotection of neurons by glycine can also be achieved by microglia. Microglia are innate immune cells of the CNS and major mediators of neuroinflammation (7). Microglia are characterized as the resident macrophages in the brain and can be activated during pathological conditions (10). Activated microglia have two phenotypes: M1 microglia and M2 microglia (11). M1 microglia are proinflammatory and secrete oxidative metabolites and cytokines such as IL-1β, TNF-α, CD32, and iNOS, which promote brain damage under pathological conditions (53). In contrast, M2 microglia express anti-inflammatory mediators, such as Arg-1, CD206, and YM-1, which prevent inflammation and contribute to recovery after brain injury (54). Microglia are activated to polarize into the M1 phenotype after 24 h of ischemic stroke. Treatment with glycine resulted in the suppression of ischemia-mediated M1 microglia polarization and promotion of M2 microglia polarization. M2 microglia polarization promotes anti-inflammatory effects that indirectly reduce OGD-induced neuronal death.

Previous studies have shown that NF-κB and Hif-1α play a crucial role in inflammation (15, 45). The activation of the NF-κB pathway leads to the expression of various proinflammation-associated genes, including cytokines, chemokines, and adhesion molecules (55). In macrophages, the upregulation of Hif-1α promotes M1 macrophage polarization and inflammation (56). Several studies have shown that controlling the HIF-1α gene by NF-κB provides important additional and parallel regulatory levels for the HIF-1α pathway. The levels of NF-κB p65 and Hif-1α increased after ischemic stroke, and treatment with glycine suppressed the upregulation of NF-κB p65 and the subsequent inhibition of Hif-1α. We confirmed that NF-κB p65/Hif-1α mediates glycine-regulated microglial polarization. AKT is upstream of NF-κB. The activation of AKT can negatively regulate NF-κB p65. We further indicated that glycine-regulated NF-κB p65 and Hif-1α are mediated by AKT activation. Moreover, PTEN mediates glycine-regulated AKT/NF-κB/Hif-1α signaling pathways. PTEN is downregulated by glycine in microglia. PTEN is a tumor suppressor, and the inhibition of PTEN activates AKT through the PI3K signaling pathway (46, 47). The downregulation of PTEN has a neuroprotective effect on ischemic stroke injury. We demonstrated that the inhibition of PTEN lipid phosphatase activity activates AKT and that the inhibition of PTEN protein phosphatase activity inhibits synaptic GluN2B-expressing NMDA receptors, which leads to protection against ischemic neuronal death (22). We have also shown that the inhibition of PTEN by enhancing GABAA receptor expression and function prevents neuronal death in OGD, in vitro OGD, and ischemic stroke models in vivo (57). These studies show that glycine modulates microglial polarization through regulation of the PTEN/AKT/NF-κB/Hif-1α signaling pathway and subsequently indirectly reduces ischemia-induced neuronal death, brain damage, and functional recovery.

The results of the current study reveal that neuroinflammation after ischemic stroke can be mediated by glycine. In the pathologic condition of ischemic stroke, glycine inhibits PTEN, which activates AKT and subsequently inhibits NF-κB p65 and Hif-1α, thereby inhibiting ischemia-induced M1 microglia polarization and promoting anti-inflammatory effects, which indirectly result in reduced ischemia-induced neuronal death. These findings provide a new understanding of the function of glycine in ischemic stroke and provide a basis for the extending the use of glycine in clinical trials.

We thank American Journal Experts for linguistic assistance.

This work was supported by the China Key Project of Basic Research (“973” Project; 2014CB541606), the Natural Science Foundation of China (81470599), The Fund of Collaborative Innovation Center for Brain Science to Q.W., and the QingHai Science and Technology Department Applied Basic Research Project (2015-ZJ-749).

The online version of this article contains supplemental material.

Abbreviations used in this article:

ECA

external carotid artery

FJC

Fluoro Jade–C

i.c.v.

intracerebroventricular

I/R

ischemia-reperfusion

IV

AKT inhibitor IV

LDH

lactate dehydrogenase

MCAO

middle cerebral artery occlusion

MCM

microglia culture medium

OGD

oxygen-glucose deprivation

shRNA

short hairpin RNA

TTC

2,3,5-triphenyltetrazolium chloride

WT Hif-1α

pCDNA3.1(+)-Hif-1α–WT.

1
Han
,
Z.
,
L.
Li
,
L.
Wang
,
V.
Degos
,
M.
Maze
,
H.
Su
.
2014
.
Alpha-7 nicotinic acetylcholine receptor agonist treatment reduces neuroinflammation, oxidative stress, and brain injury in mice with ischemic stroke and bone fracture.
J. Neurochem.
131
:
498
508
.
2
Wu
,
K. J.
,
M. T.
Hsieh
,
C. R.
Wu
,
W. G.
Wood
,
Y. F.
Chen
.
2012
.
Green tea extract ameliorates learning and memory deficits in ischemic rats via its active component polyphenol epigallocatechin-3-gallate by modulation of oxidative stress and neuroinflammation.
Evid. Based Complement. Alternat. Med.
2012
:
163106
.
3
Blasi
,
F.
,
F.
Herisson
,
S.
Wang
,
J.
Mao
,
C.
Ayata
.
2015
.
Late-onset thermal hypersensitivity after focal ischemic thalamic infarcts as a model for central post-stroke pain in rats.
J. Cereb. Blood Flow Metab.
35
:
1100
1103
.
4
Zhu
,
Z.
,
Y.
Fu
,
D.
Tian
,
N.
Sun
,
W.
Han
,
G.
Chang
,
Y.
Dong
,
X.
Xu
,
Q.
Liu
,
D.
Huang
,
F. D.
Shi
.
2015
.
Combination of the immune modulator fingolimod with alteplase in acute ischemic stroke: a pilot trial.
Circulation
132
:
1104
1112
.
5
Ruhnau
,
J.
,
K.
Schulze
,
B.
Gaida
,
S.
Langner
,
C.
Kessler
,
B.
Bröker
,
A.
Dressel
,
A.
Vogelgesang
.
2014
.
Stroke alters respiratory burst in neutrophils and monocytes. [Published erratum appears in 2014 Stroke 45: e68.]
Stroke
45
:
794
800
.
6
Hao
,
M.
,
X.
Li
,
J.
Feng
,
N.
Pan
.
2015
.
Triptolide protects against ischemic stroke in rats.
Inflammation
38
:
1617
1623
.
7
Skaper
,
S. D.
,
L.
Facci
,
P.
Giusti
.
2014
.
Neuroinflammation, microglia and mast cells in the pathophysiology of neurocognitive disorders: a review.
CNS Neurol. Disord. Drug Targets
13
:
1654
1666
.
8
Amor
,
S.
,
M. N.
Woodroofe
.
2014
.
Innate and adaptive immune responses in neurodegeneration and repair.
Immunology
141
:
287
291
.
9
Perry
,
V. H.
,
J. A.
Nicoll
,
C.
Holmes
.
2010
.
Microglia in neurodegenerative disease.
Nat. Rev. Neurol.
6
:
193
201
.
10
AlShakweer
,
W.
,
Y.
Alwelaie
,
A. M.
Mankung
,
M. B.
Graeber
.
2011
.
Bone marrow-derived microglia in pilocytic astrocytoma.
Front. Biosci. (Elite Ed.)
3
:
371
379
.
11
Xia
,
C. Y.
,
S.
Zhang
,
Y.
Gao
,
Z. Z.
Wang
,
N. H.
Chen
.
2015
.
Selective modulation of microglia polarization to M2 phenotype for stroke treatment.
Int. Immunopharmacol.
25
:
377
382
.
12
Yu
,
Z.
,
D.
Sun
,
J.
Feng
,
W.
Tan
,
X.
Fang
,
M.
Zhao
,
X.
Zhao
,
Y.
Pu
,
A.
Huang
,
Z.
Xiang
, et al
.
2015
.
MSX3 switches microglia polarization and protects from inflammation-induced demyelination.
J. Neurosci.
35
:
6350
6365
.
13
Herder
,
V.
,
C. D.
Iskandar
,
K.
Kegler
,
F.
Hansmann
,
S. A.
Elmarabet
,
M. A.
Khan
,
A.
Kalkuhl
,
U.
Deschl
,
W.
Baumgärtner
,
R.
Ulrich
,
A.
Beineke
.
2015
.
Dynamic changes of microglia/macrophage M1 and M2 polarization in Theiler’s murine encephalomyelitis.
Brain Pathol.
25
:
712
723
.
14
Liu
,
Z.
,
R.
Xi
,
Z.
Zhang
,
W.
Li
,
Y.
Liu
,
F.
Jin
,
X.
Wang
.
2014
.
4-hydroxyphenylacetic acid attenuated inflammation and edema via suppressing HIF-1α in seawater aspiration-induced lung injury in rats.
Int. J. Mol. Sci.
15
:
12861
12884
.
15
Palazon
,
A.
,
A. W.
Goldrath
,
V.
Nizet
,
R. S.
Johnson
.
2014
.
HIF transcription factors, inflammation, and immunity.
Immunity
41
:
518
528
.
16
Palsson-McDermott
,
E. M.
,
A. M.
Curtis
,
G.
Goel
,
M. A.
Lauterbach
,
F. J.
Sheedy
,
L. E.
Gleeson
,
M. W.
van den Bosch
,
S. R.
Quinn
,
R.
Domingo-Fernandez
,
D. G.
Johnston
, et al
.
2015
.
Pyruvate kinase M2 regulates Hif-1α activity and IL-1β induction and is a critical determinant of the warburg effect in LPS-activated macrophages. [Published erratum appears in 2015 Cell Metab. 21: 347.]
Cell Metab.
21
:
65
80
.
17
Azoitei
,
N.
,
A.
Becher
,
K.
Steinestel
,
A.
Rouhi
,
K.
Diepold
,
F.
Genze
,
T.
Simmet
,
T.
Seufferlein
.
2016
.
PKM2 promotes tumor angiogenesis by regulating HIF-1α through NF-κB activation.
Mol. Cancer
15
:
3
.
18
Chen
,
Y.
,
Q.
Tang
,
J.
Wu
,
F.
Zheng
,
L.
Yang
,
S. S.
Hann
.
2015
.
Inactivation of PI3-K/Akt and reduction of SP1 and p65 expression increase the effect of solamargine on suppressing EP4 expression in human lung cancer cells.
J. Exp. Clin. Cancer Res.
34
:
154
.
19
Makker
,
A.
,
M. M.
Goel
,
A. A.
Mahdi
.
2014
.
PI3K/PTEN/Akt and TSC/mTOR signaling pathways, ovarian dysfunction, and infertility: an update.
J. Mol. Endocrinol.
53
:
R103
R118
.
20
Wang
,
G.
,
Y.
Shi
,
X.
Jiang
,
R. K.
Leak
,
X.
Hu
,
Y.
Wu
,
H.
Pu
,
W. W.
Li
,
B.
Tang
,
Y.
Wang
, et al
.
2015
.
HDAC inhibition prevents white matter injury by modulating microglia/macrophage polarization through the GSK3β/PTEN/Akt axis.
Proc. Natl. Acad. Sci. USA
112
:
2853
2858
.
21
Chen
,
J.
,
Y.
Zhuang
,
Z. F.
Zhang
,
S.
Wang
,
P.
Jin
,
C.
He
,
P. C.
Hu
,
Z. F.
Wang
,
Z. Q.
Li
,
G. M.
Xia
, et al
.
2016
.
Glycine confers neuroprotection through microRNA-301a/PTEN signaling.
Mol. Brain
9
:
59
.
22
Ning
,
K.
,
L.
Pei
,
M.
Liao
,
B.
Liu
,
Y.
Zhang
,
W.
Jiang
,
J. G.
Mielke
,
L.
Li
,
Y.
Chen
,
Y. H.
El-Hayek
, et al
.
2004
.
Dual neuroprotective signaling mediated by downregulating two distinct phosphatase activities of PTEN.
J. Neurosci.
24
:
4052
4060
.
23
Tibbetts
,
A. S.
,
D. R.
Appling
.
2010
.
Compartmentalization of Mammalian folate-mediated one-carbon metabolism.
Annu. Rev. Nutr.
30
:
57
81
.
24
Betz
,
H.
,
B.
Laube
.
2006
.
Glycine receptors: recent insights into their structural organization and functional diversity.
J. Neurochem.
97
:
1600
1610
.
25
Lynch
,
J. W.
2009
.
Native glycine receptor subtypes and their physiological roles.
Neuropharmacology
56
:
303
309
.
26
Wheeler
,
M. D.
,
R. G.
Thurman
.
1999
.
Production of superoxide and TNF-alpha from alveolar macrophages is blunted by glycine.
Am. J. Physiol.
277
:
L952
L959
.
27
Zhao
,
D.
,
J.
Chen
,
Y.
Zhang
,
H.-B.
Liao
,
Z.-F.
Zhang
,
Y.
Zhuang
,
M.-X.
Pan
,
J.-C.
Tang
,
R.
Liu
,
Y.
Lei
, et al
.
2018
.
Glycine confers neuroprotection through PTEN/AKT signal pathway in experimental intracerebral hemorrhage.
Biochem. Biophys. Res. Commun.
501
:
85
91
.
28
Wang
,
L.
,
Y.
Lu
,
H.
Guan
,
D.
Jiang
,
Y.
Guan
,
X.
Zhang
,
H.
Nakano
,
Y.
Zhou
,
Y.
Zhang
,
L.
Yang
,
H.
Li
.
2013
.
Tumor necrosis factor receptor-associated factor 5 is an essential mediator of ischemic brain infarction.
J. Neurochem.
126
:
400
414
.
29
Zhang
,
Z.-F.
,
J.
Chen
,
X.
Han
,
Y.
Zhang
,
H.-B.
Liao
,
R.-X.
Lei
,
Y.
Zhuang
,
Z.-F.
Wang
,
Z.
Li
,
J.-C.
Chen
, et al
.
2017
.
Bisperoxovandium (pyridin-2-squaramide) targets both PTEN and ERK1/2 to confer neuroprotection.
Br. J. Pharmacol.
174
:
641
656
.
30
Liu
,
F.
,
D. P.
Schafer
,
L. D.
McCullough
.
2009
.
TTC, fluoro-Jade B and NeuN staining confirm evolving phases of infarction induced by middle cerebral artery occlusion.
J. Neurosci. Methods
179
:
1
8
.
31
Shan
,
Y.
,
B.
Liu
,
L.
Li
,
N.
Chang
,
L.
Li
,
H.
Wang
,
D.
Wang
,
H.
Feng
,
C.
Cheung
,
M.
Liao
, et al
.
2009
.
Regulation of PINK1 by NR2B-containing NMDA receptors in ischemic neuronal injury.
J. Neurochem.
111
:
1149
1160
.
32
Liu
,
B.
,
M.
Liao
,
J. G.
Mielke
,
K.
Ning
,
Y.
Chen
,
L.
Li
,
Y. H.
El-Hayek
,
E.
Gomez
,
R. S.
Zukin
,
M. G.
Fehlings
,
Q.
Wan
.
2006
.
Ischemic insults direct glutamate receptor subunit 2-lacking AMPA receptors to synaptic sites.
J. Neurosci.
26
:
5309
5319
.
33
Chen
,
J.
,
R.
Hu
,
H.
Liao
,
Y.
Zhang
,
R.
Lei
,
Z.
Zhang
,
Y.
Zhuang
,
Y.
Wan
,
P.
Jin
,
H.
Feng
,
Q.
Wan
.
2017
.
A non-ionotropic activity of NMDA receptors contributes to glycine-induced neuroprotection in cerebral ischemia-reperfusion injury.
Sci. Rep.
7
:
3575
.
34
Schmued
,
L. C.
,
C. C.
Stowers
,
A. C.
Scallet
,
L.
Xu
.
2005
.
Fluoro-Jade C results in ultra high resolution and contrast labeling of degenerating neurons.
Brain Res.
1035
:
24
31
.
35
Mason
,
D. Y.
,
K.
Micklem
,
M.
Jones
.
2000
.
Double immunofluorescence labelling of routinely processed paraffin sections.
J. Pathol.
191
:
452
461
.
36
Chen
,
J.
,
P. R.
Sanberg
,
Y.
Li
,
L.
Wang
,
M.
Lu
,
A. E.
Willing
,
J.
Sanchez-Ramos
,
M.
Chopp
.
2001
.
Intravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in rats.
Stroke
32
:
2682
2688
.
37
Clifton
,
G. L.
,
J. Y.
Jiang
,
B. G.
Lyeth
,
L. W.
Jenkins
,
R. J.
Hamm
,
R. L.
Hayes
.
1991
.
Marked protection by moderate hypothermia after experimental traumatic brain injury.
J. Cereb. Blood Flow Metab.
11
:
114
121
.
38
Sughrue
,
M. E.
,
J.
Mocco
,
R. J.
Komotar
,
A.
Mehra
,
A. L.
D’Ambrosio
,
B. T.
Grobelny
,
D. L.
Penn
,
E. S.
Connolly
Jr
.
2006
.
An improved test of neurological dysfunction following transient focal cerebral ischemia in rats.
J. Neurosci. Methods
151
:
83
89
.
39
Fluri
,
F.
,
M. K.
Schuhmann
,
C.
Kleinschnitz
.
2015
.
Animal models of ischemic stroke and their application in clinical research.
Drug Des. Devel. Ther.
9
:
3445
3454
.
40
Chen
,
Z.
,
C.
Zhang
,
H.
Ma
,
T.
Zhou
,
B.
Jiang
,
M.
Chen
,
X.
Chen
.
2015
.
A non-aggregation spectrometric determination for mercury ions based on gold nanoparticles and thiocyanuric acid.
Talanta
134
:
603
606
.
41
Dambach
,
H.
,
D.
Hinkerohe
,
N.
Prochnow
,
M. N.
Stienen
,
Z.
Moinfar
,
C. G.
Haase
,
A.
Hufnagel
,
P. M.
Faustmann
.
2014
.
Glia and epilepsy: experimental investigation of antiepileptic drugs in an astroglia/microglia co-culture model of inflammation.
Epilepsia
55
:
184
192
.
42
Fann
,
D. Y.
,
S. Y.
Lee
,
S.
Manzanero
,
S. C.
Tang
,
M.
Gelderblom
,
P.
Chunduri
,
C.
Bernreuther
,
M.
Glatzel
,
Y. L.
Cheng
,
J.
Thundyil
, et al
.
2013
.
Intravenous immunoglobulin suppresses NLRP1 and NLRP3 inflammasome-mediated neuronal death in ischemic stroke.
Cell Death Dis.
4
:
e790
.
43
Cunningham
,
C.
2013
.
Microglia and neurodegeneration: the role of systemic inflammation.
Glia
61
:
71
90
.
44
Tam
,
W. Y.
,
C. H.
Ma
.
2014
.
Bipolar/rod-shaped microglia are proliferating microglia with distinct M1/M2 phenotypes.
Sci. Rep.
4
:
7279
.
45
Chen
,
Y. H.
,
Z.
Yu
,
L.
Fu
,
H.
Wang
,
X.
Chen
,
C.
Zhang
,
Z. M.
Lv
,
D. X.
Xu
.
2015
.
Vitamin D3 inhibits lipopolysaccharide-induced placental inflammation through reinforcing interaction between vitamin D receptor and nuclear factor kappa B p65 subunit.
Sci. Rep.
5
:
10871
.
46
Roa
,
I.
,
G.
de Toro
,
F.
Fernández
,
A.
Game
,
S.
Muñoz
,
X.
de Aretxabala
,
M.
Javle
.
2015
.
Inactivation of tumor suppressor gene pten in early and advanced gallbladder cancer.
Diagn. Pathol.
10
:
148
.
47
Lim
,
H. J.
,
P.
Crowe
,
J. L.
Yang
.
2015
.
Current clinical regulation of PI3K/PTEN/Akt/mTOR signalling in treatment of human cancer.
J. Cancer Res. Clin. Oncol.
141
:
671
689
.
48
Chen
,
Y.
,
C.
Luo
,
M.
Zhao
,
Q.
Li
,
R.
Hu
,
J. H.
Zhang
,
Z.
Liu
,
H.
Feng
.
2015
.
Administration of a PTEN inhibitor BPV(pic) attenuates early brain injury via modulating AMPA receptor subunits after subarachnoid hemorrhage in rats.
Neurosci. Lett.
588
:
131
136
.
49
Zhong
,
Z.
,
M. D.
Wheeler
,
X.
Li
,
M.
Froh
,
P.
Schemmer
,
M.
Yin
,
H.
Bunzendaul
,
B.
Bradford
,
J. J.
Lemasters
.
2003
.
L-Glycine: a novel antiinflammatory, immunomodulatory, and cytoprotective agent.
Curr. Opin. Clin. Nutr. Metab. Care
6
:
229
240
.
50
Chen
,
Z.
,
B.
Hu
,
F.
Wang
,
L.
Du
,
B.
Huang
,
L.
Li
,
J.
Qi
,
X.
Wang
.
2015
.
Glycine bidirectionally regulates ischemic tolerance via different mechanisms including NR2A-dependent CREB phosphorylation.
J. Neurochem.
133
:
397
408
.
51
Gusev
,
E. I.
,
V. I.
Skvortsova
,
S. A.
Dambinova
,
K. S.
Raevskiy
,
A. A.
Alekseev
,
V. G.
Bashkatova
,
A. V.
Kovalenko
,
V. S.
Kudrin
,
E. V.
Yakovleva
.
2000
.
Neuroprotective effects of glycine for therapy of acute ischaemic stroke.
Cerebrovasc. Dis.
10
:
49
60
.
52
Stephan
,
J.
,
E.
Friauf
.
2014
.
Functional analysis of the inhibitory neurotransmitter transporters GlyT1, GAT-1, and GAT-3 in astrocytes of the lateral superior olive.
Glia
62
:
1992
2003
.
53
Zhu
,
T.
,
A. G.
Miller
,
D.
Deliyanti
,
D. R.
Berka
,
A.
Agrotis
,
D. J.
Campbell
,
J. L.
Wilkinson-Berka
.
2015
.
Prorenin stimulates a pro-angiogenic and pro-inflammatory response in retinal endothelial cells and an M1 phenotype in retinal microglia.
Clin. Exp. Pharmacol. Physiol.
42
:
537
548
.
54
Xu
,
Y.
,
Y.
Xu
,
Y.
Wang
,
Y.
Wang
,
L.
He
,
Z.
Jiang
,
Z.
Huang
,
H.
Liao
,
J.
Li
,
J. M.
Saavedra
, et al
.
2015
.
Telmisartan prevention of LPS-induced microglia activation involves M2 microglia polarization via CaMKKβ-dependent AMPK activation.
Brain Behav. Immun.
50
:
298
313
.
55
Wang
,
Z.
,
Y.
Lee
,
J. S.
Eun
,
E. J.
Bae
.
2014
.
Inhibition of adipocyte inflammation and macrophage chemotaxis by butein.
Eur. J. Pharmacol.
738
:
40
48
.
56
Hood
,
J. L.
2016
.
Melanoma exosome induction of endothelial cell GM-CSF in pre-metastatic lymph nodes may result in different M1 and M2 macrophage mediated angiogenic processes.
Med. Hypotheses
94
:
118
122
.
57
Liu
,
B.
,
L.
Li
,
Q.
Zhang
,
N.
Chang
,
D.
Wang
,
Y.
Shan
,
L.
Li
,
H.
Wang
,
H.
Feng
,
L.
Zhang
, et al
.
2010
.
Preservation of GABAA receptor function by PTEN inhibition protects against neuronal death in ischemic stroke.
Stroke
41
:
1018
1026
.

The authors have no financial conflicts of interest.

Supplementary data