亚洲免费av电影一区二区三区,日韩爱爱视频,51精品视频一区二区三区,91视频爱爱,日韩欧美在线播放视频,中文字幕少妇AV,亚洲电影中文字幕,久久久久亚洲av成人网址,久久综合视频网站,国产在线不卡免费播放

        ?

        Ki20227 aggravates apoptosis, inflammatory response, and oxidative stress after focal cerebral ischemia injury

        2022-06-13 08:30:10ChengJiangZeNingWangYuChenKangYiChenWeiXinLuHaiJunRenBoRuHou

        Cheng Jiang , Ze-Ning Wang , Yu-Chen Kang , Yi Chen Wei-Xin LuHai-Jun Ren Bo-Ru Hou

        Abstract The survival of microglia depends on the colony-stimulating factor-1 receptor (CSF1R) signaling pathway under physiological conditions.Ki20227 is a highly selective CSF1R inhibitor that has been shown to change the morphology of microglia. However, the effects of Ki20227 on the progression of ischemic stroke are unclear. In this study, male C57BL/6 mouse models of focal cerebral ischemic injury were established through the occlusion of the middle cerebral artery and then administered 3 mg/g Ki20227 for 3 successive days. The results revealed that the number of ionized calcium-binding adaptor molecule 1/bromodeoxyuridine double positive cells in the infarct tissue was reduced, the degree of edema was increased, neurological deficits were aggravated, infarct volume was increased, and the number of peri-infarct Nissl bodies was reduced. The number of terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells in the peri-infarct tissue was increased. The expression levels of Bax and Cleaved caspase-3 were up-regulated. Bcl-2 expression was downregulated. The expression levels of inflammatory factors and oxidative stress-associated factors were increased. These findings suggested that Ki20227 blocked microglial proliferation and aggravated the pathological progression of ischemia/reperfusion injury in a transient middle cerebral artery occlusion model. This study was approved by the Animal Ethics Committee of Lanzhou University Second Hospital (approval No. D2020-68) on March 6,2020.

        Key Words: apoptosis; colony-stimulating factor-1 receptor; inflammatory response; ischemia/reperfusion; Ki20227; microglia; oxidative stress; transient middle cerebral artery occlusion

        Introduction

        Ischemic stroke is a disease characterized by a high incidence of disability and mortality (Datta et al., 2020). Neurons,glial cells, and peripheral blood vessels are damaged after ischemic stroke due to the disruption of the blood supply,resulting in corresponding neurological impairment, such as hemiplegia, aphasia, and impaired cognitive abilities (Hillis et al., 2002; Jiang et al., 2018). The current treatment for stroke is mainly focused on improving the plasticity of the neural loop (Wang et al., 2019; Al-Kuraishy et al., 2020;Huang et al., 2020). Therapeutic research has focused on the complex relationship between the various cell types involved in tissue repair (Hayakawa et al., 2016; Xiong et al.,2016; Upadhya et al., 2020). Microglia are immunoreactive cells in the brain that react rapidly after stroke (Kanazawa et al., 2017). During ischemic injury, microglia undergo large changes in morphology, density, and function, including cell body enlargement, debranching, cell wall thickening,inflammatory protein production, and various states of proliferation, migration, and phagocytosis (Hou et al., 2016;Qin et al., 2019; Mo et al., 2020). The chronic inflammatory activation of microglia can last for years after ischemia/reperfusion (I/R) injury (Sekeljic et al., 2012; Cui et al., 2020;Radenovic et al., 2020). However, the function of microglia in stroke remains controversial (Ma et al., 2017; Qin et al.,2019). Activated microglia release pro-inflammatory factors that cause neuronal death and contribute to secondary tissue damage (Kanazawa et al., 2017). However, activated microglia also remove damaged neural cells, cellular debris, and dysfunctional synapses and produce anti-inflammatory factors to promote recovery after stroke (Wang et al., 2018). Microglia may, therefore, serve a dual role during the pathological process of ischemic stroke (Xiong et al., 2016; Ma et al., 2017),and the specific functions of microglia remain debatable.

        Colony-stimulating factor 1 receptor (CSF1R)—the specific receptor for colony-stimulating factor 1 (CSF1) and interleukin(IL)-34—regulates the proliferation, differentiation, and function of myeloid lineage cells, including microglia,macrophages, and osteoclasts (Hume et al., 2020). Microglia are the predominant CSF1R-expressing cell type in the central nervous system under physiological conditions (Hu et al.,2020). The selective inhibition of CSF1R was shown to change the physiological state of microgliain vivo(Hou et al., 2016).Although CSF1R has been detected in other cell types, such as neurons and astrocytes, the expression levels were minimal,and no significant changes were observed in these cells after CSF1R inhibition (Hou et al., 2016; Fu et al., 2020; Neal et al.,2020). The specific effect of CSF1R inhibition on microglia represents a new strategy for use in microglial research (Huang et al., 2018; Neal et al., 2020). The highly selective CSF1R tyrosine kinase inhibitor Ki20227 has been administered orally to manipulate myeloid cells in different animal models(Ohno et al., 2006; Hou et al., 2016; Xie et al., 2020). Although Ki20227 administration has been reported to result in morphological changes in microglia (Hou et al., 2016), the effect of Ki20227 on I/R injury remains unclear. In this study,we investigated the impacts of Ki20227 in a mouse model of transient middle cerebral artery occlusion (tMCAO).

        Materials and Methods

        Animals

        Male C57BL/6 mice (specific-pathogen-free level,n= 108,aged 10–12 weeks, weighing 23–25 g) were used for all experiments. All animals were purchased from the Animal Experimental Center of Lanzhou University Second Hospital[license No. SYXK (Gan) 2018-0003]. Mice had free access to water and food at all times and were maintained under the conditions of a 12-hour light/dark cycle at 22 ± 2°C. All animal procedures were performed strictly following the guidelines set by Lanzhou University Second Hospital, and the study was approved by the Animal Ethics Committee of Lanzhou University Second Hospital (approval No. D2020-68) on March 6, 2020. The mice were randomly divided into four groups:sham + vehicle (n= 16), sham + Ki20227 (n= 16), tMCAO +vehicle (n= 38), and tMCAO + Ki20227 (n= 38).

        tMCAO model

        The method used to generate the tMCAO model, in which a focal I/R injury was induced, was described in a previous study (Chiang et al., 2011). The mice were anesthetized with isoflurane (RWD, Shenzhen, China) inhalation and fixed in a supine position on a mouse plate. After iodophor disinfection,the median skin of the neck was cut open to expose the leftcommon carotid artery, internal carotid artery, and external carotid artery. The external carotid artery was cut after coagulation, and the proximal end was placed in a straight line with the internal carotid artery. A thread bolted with a silicone head (Doccol, Sharon, MA, USA) was inserted from the external carotid artery, through the internal carotid artery, to the entrance of the middle cerebral artery. The embolization process lasted for 60 minutes, and then the threaded bolt was removed. To ensure that ischemia was effectively induced in mice, reduced blood flow was confirmed by laser Doppler flowmetry (RWD). The mice were placed on a constant temperature test pad to ensure that the anal temperature was maintained at approximately 37°C. In the sham groups,the left common carotid artery, internal carotid artery, and external carotid artery were exposed for 60 minutes without thread insertion.

        Ki20227 treatment

        Mice were treated intragastrically with Ki20227 (Med Chem Express, Deer Park, NJ, USA) at a dose of 2 mg/g daily for 3 days starting immediately after tMCAO to inhibit CSF1Rin vivo(Figure 1A). The sham + vehicle and tMCAO + vehicle groups received the same volume (0.04 mL/g) of vehicle [0.5%methylcellulose (Sigma, St. Louis, MO, USA), which promotes the dispersion and dissolution of Ki20227, in distilled water].Bromodeoxyuridine (BrdU; 50 mg/kg, Sigma) was also administered intragastrically to mice twice a day, starting immediately following the surgery. All mice were sacrificed on the 3rdday after I/R, except for those used for neurological functional assessment (n= 8 in each of the tMCAO + vehicle and tMCAO + Ki20227 groups).

        Peri-infarct microglial proliferation assessment

        The proliferation of peri-infarct microglia was analyzed by immunofluorescence staining. Mice were perfused via the leftheart ventricle with phosphate-buffered saline (PBS), followed by 4% paraformaldehyde. After brain tissues were fixed overnight in 4% paraformaldehyde at 4°C, the brain tissues were dehydrated in 30% sucrose/0.01 M PBS for several days, and then the brain tissues were sectioned (30 μm) on a microtome (Leica, Wetzlar, Hessen, Germany). To label microglia, three or four brain slices were selected from each mouse in each treatment group and stained in a 24-well plate.First, the sections were rinsed three times with 0.1 mL PBS for 5 minutes and treated with 0.5% PBS-Triton X-100 for 10 minutes to increase cell membrane permeability. The sections were blocked in 1% bovine serum albumin solution for 60 minutes at 37°C and incubated overnight at 4°C with rabbit anti-ionized calcium-binding adapter protein 1 (Iba1) antibody(1:100, Abcam, Cambridge, MA, USA, Cat# EPR16589).The brain sections were incubated with the corresponding secondary antibody (fluorescein isothiocyanate, goat antirabbit, 1:200, ZSGB-BIO, Beijing, China, Cat# ZF-0312) for 1 hour at 37°C. For BrdU immunofluorescence, the brain sections were incubated with 2 M HCl for 30 minutes at 37°C,neutralized by three incubations with 0.1 M borate buffer (pH 8.5) for 15 minutes each, and incubated overnight with rat anti-BrdU monoclonal antibody (1:500; Abcam, Cat# ab6326).The brain sections were incubated with the corresponding secondary antibody (1:200, tetramethylrhodamine, goat anti-rat, ZSGB-BIO, Cat# ZF-0318) for 1 hour at 37°C. The sections were incubated with Hoechst 33258 (1:10,000; Sigma) for 15 minutes to label the nucleus. The brain slices stained with immunofluorescent labels were scanned sequentially using a laser-scanning confocal microscope (Olympus, Tokyo, Japan).Cells co-expressing Iba1 and BrdU were considered newly proliferated microglia.

        Infarct volume assessment

        The infarct volume was measured by 2,3,5-triphenyl tetrazolium chloride (TTC) staining. Briefly, the mouse brain was cut into seven 2-mm-thick coronal slices, incubated with 2% TTC (Sigma) at 37°C for 30 minutes, and fixed in 10%formalin to stop the staining. The infarct area was measured using ImageJ software (National Institutes of Health, Bethesda,MD, USA). The percentage of infarct volume = total infarct volume/volume of the contralateral hemisphere × 100%.The analysis was conducted by researchers blinded to the treatment strategy.

        Nissl staining

        Sectioned brain tissues (30 μm) were obtained using a vibrating microtome (Leica) after cardiac perfusion, as described above. The brain slices were incubated in 1%methyl violet for 15 minutes, treated with Nissl differentiation solution for 4–8 seconds, and then immersed in an ethanol gradient and xylene. Finally, the sections were observed with an optical microscope (Olympus).

        Neurological functional assessment

        The modified neurological severity score (Bieber et al., 2019)was determined based on the performance on a series of behavioral tests, including sensory, motor, reflex, and balance disturbance tests, to assess the neurological function of mice 1,3, 7, and 14 days after tMCAO. The total score was the sum of the test scores, ranging from 0 to 18. A higher score indicates more serious neurological impairment. The assessment was conducted by researchers blinded to the treatment strategy.

        Cerebral edema assessment

        On the third day after the operation, seven mice in each group were killed to determine brain edema. The cerebral hemispheres on the infarct side were separated and weighed to obtain the wet weight and measured again after 24 hours of air drying to obtain the dry weight. The formula for calculating edema is (wet weight – dry weight)/wet weight × 100%.

        TdT-mediated dUTP nick-end labeling apoptosis assessment

        TdT-mediated dUTP nick-end labeling (TUNEL) assay was used to determine peri-infarct apoptosis 72 hours after tMCAO. The process was conducted in accordance with the manufacturer’s instructions. The 30-μm-thick coronal brain slices were washed in PBS, incubated in 0.3% PBS-Tween 20,at room temperature for 5 minutes, and then incubated with the TUNEL reaction mixture (Roche, Basel, Switzerland) in the dark at 37°C for 1 hour. To label the nucleus, the sections were incubated with Hoechst 33258 (1:10,000; Sigma) for 15 minutes. The brain slices were scanned sequentially by a laserscanning confocal microscope (Olympus). Quantification of TUNEL assay was described as the positive cell density in the peri-infarct region, as measured by ImageJ software.

        Western blot analysis

        The total proteins of ischemic tissues were extracted with radioimmunoprecipitation assay buffer, evenly separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis,and transferred to a polyvinylidene difluoride membrane.After transfer, the membrane was incubated in 5% skim milk at 37°C for 1 hour and then incubated with gentle shaking with primary antibodies at 4°C overnight. Primary antibodies against Cleaved caspase-3 (rabbit, 1:1000, Cat# 9664, Cell Signaling Technology, Danvers, MA, USA), Bcl-2 (rabbit, 1:1000,Cat# 12789-1-AP, Proteintech Group, Wuhan, China), Bax(rabbit, 1:1000, Cat# 50599-2-lg, Proteintech Group) and β-actin (mouse, 1:3000, Cat# TA-09, ZSGB-BIO) were used in our study. After incubation with the primary antibody,the membrane was exposed to goat anti-rabbit secondary antibody (1:5000, ZSGB-BIO, Cat# ZB-2301) or goat antimouse secondary antibody (1:5000, Cat# ZB-2305, ZSGB-BIO)at 37°C for 1 hour. Enhanced chemiluminescence was used for visualization, and ImageJ software was used to measure the gray value of each protein. Target proteins were normalized against β-actin expression.

        Quantitative real-time-polymerase chain reaction

        Total RNA from ischemic tissues was extracted by RNA prep pure Tissue Kit (TIANGEN, Beijing, China) and reverse transcribed to complementary DNA using a reverse-transcription kit with gDNA Eraser (TIANGEN). Quantitative polymerase chain reaction was performed using a SYBR Green kit (TIANGEN) in accordance with the manufacturer’s instructions. All mRNA expression levels were normalized against the expression of glyceraldehyde 3-phosphate dehydrogenase as the endogenous control.The fold-change rate was calculated using the comparative threshold cycle (Ct) method. The primer sequences are shown inAdditional Table 1.

        Enzyme-linked immunosorbent assay

        Supernatants of homogenates extracted from ischemic tissues were assessed using a commercial enzyme-linked immunosorbent assay (ELISA) kit (Mlbio, Shanghai, China). The expression levels of superoxide dismutase (SOD), glutathione(GSH), and malondialdehyde (MDA) were quantified following the ELISA kit’s protocols.

        Statistical analysis

        GraphPad Prism 8.3 (GraphPad Software, San Diego, CA, USA)was used for statistical analysis. Two-tailed unpairedt-test were used to analyze changes in the quantification of Iba1+/BrdU+cells, infarct volume, brain water content, neurological severity score, TUNEL-positive cells, and apoptotic protein expression. One-way analysis of variance followed by Tukey’spost hoctest was used to analyze changes in Nissl bodies,inflammatory factors, and oxidative stress-associated factors.Data are expressed as the mean ± standard deviation (SD).P<0.05 indicated a significant difference.

        Results

        Ki20227 inhibits the proliferation of microglia in the peri-infarct region after tMCAO

        Iba1 and BrdU immunofluorescence was performed to evaluate the effects of Ki20227 on the proliferation of microglia. We quantified the Iba1+/BrdU+cells in the periinfarct region through immunofluorescence staining and found that a large number of microglia were activated after tMCAO. Furthermore, stroke-induced microglial proliferation was significantly inhibited by Ki20227 treatment. Fewer Iba1+/BrdU+cells were observed in mice treated with Ki20227 than in mice treated with vehicle after tMCAO (P< 0.05,Figure 2AandB).

        Ki20227 aggravates tMCAO-induced I/R injury

        Assessments of infarct volume, Nissl body density, brain water content, and neurologic deficits tests were performed to evaluate the pathological progression of I/R injury. We applied TTC staining to measure the infarct volume 3 days after tMCAO and found that Ki20227 treatment significantly increased the infarct volume compared with that in the untreated tMCAO group (P< 0.05;Figure 3AandB). No infarct area was observed in the sham groups. Fewer peri-infarct Nissl bodies were observed in Ki20227-treated mice after tMCAO compared with those in the tMCAO + vehicle group(P< 0.05;Figure 3CandD). No influence of Ki20227 on Nissl bodies was observed in the sham group. Worse neurological behavioral performance was detected in the Ki20227-treated stroke model mice than in the vehicle-treated tMCAO mice on days 3, 5, 7, and 14 after reperfusion (Figure 3E). Brain edema increased in mice treated with Ki20227 compared with that in mice in the tMCAO + vehicle group (P< 0.05;Figure 3F).

        Figure 1|Treatment steps before sample harvest.

        Figure 2|Ki20227 blocks microglial proliferation in the peri-infarct region after tMCAO.

        Figure 3|Ki20227 aggravates tMCAO-induced infarct, apoptosis, and edema in mice with ischemia/reperfusion injury.

        Ki20227 promotes peri-infarct apoptosis after tMCAO

        TUNEL staining was performed to evaluate the effects of Ki20227 on peri-infarct apoptosis after tMCAO. We found that the density of TUNEL-positive cells in the tMCAO + Ki20227 group was significantly higher than that in the tMCAO +vehicle group (P< 0.05,Figure 4AandB). Furthermore, the results of western blot analysis showed that the expression levels of pro-apoptotic proteins Bax and Cleaved caspase-3 increased in the tMCAO + vehicle group compared with those in the sham + vehicle group (Figure 4D–F). The expression level of the anti-apoptotic factor Bcl-2 was downregulated in the tMCAO groups compared with that in the sham groups.Comparing between the tMCAO + vehicle and tMCAO +Ki20227 groups, the mice treated with Ki20227 showed a lower expression level of Bcl-2 (P< 0.05,Figure 4D) and higher expression levels of Bax (P< 0.05,Figure 4E) and Cleaved caspase-3 (P< 0.05,Figure 4F). No significant change was observed after Ki20227 administration in the sham group.

        Ki20227 exacerbates the inflammatory response and oxidative stress in the peri-infarct region after tMCAO

        We detected the mRNA expression of CSF1R using quantitative real-time polymerase chain reaction. CSF1R mRNA was expressed at a higher level after tMCAO and decreased significantly after Ki20227 treatment (P< 0.05,Figure 5A).To explore the role played by the CSF1R signaling pathway in the tMCAO-induced inflammatory response and oxidative stress, we used quantitative real-time polymerase chain reaction to measure the levels of inflammatory factors and ELISA to quantify the expression of oxidative stress-associated proteins. A significant change in inflammatory gene expression levels was observed after tMCAO, showing the activation of the inflammatory response cause by tMCAO. The mRNA expression levels of pro-inflammatory factors [interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, inducible nitric oxide synthase (iNOS)] were higher in Ki20227-treated mice than in the vehicle-treated group after tMCAO (allP< 0.05,Figure 5B–E). In contrast, transforming growth factor (TGF)-β, which is an anti-inflammatory factor (Ma et al., 2017), showed lower expression in the tMCAO + Ki20227 group than in the tMCAO+ vehicle group (P< 0.05,Figure 5F). The increased expression level of MDA [a pro-oxidant factor (Del Rio et al., 2005)] and the reduced expression of antioxidant factors (including SOD and GSH) were detected in the Ki20227-treated group after tMCAO compared with the vehicle-treated group (allP< 0.05,Figure 5G–I).

        Figure 4|Ki20227 promotes peri-infarct apoptosis after tMCAO.

        Figure 5|Ki20227 exacerbates the inflammatory response and oxidative stress in the peri-infarct region after tMCAO.

        Discussion

        Estrogen has been shown to exert a protective effect against stroke (Koellhoffer and McCullough, 2013; Ritzel et al., 2013;Nematipour et al., 2020). Following the same ischemic intervention treatment, the infarct degree in female mice was shown to be smaller than that of male mice (Manwani et al., 2015). To avoid the potentially confounding effects of estrogen on the experimental results, female mice were not used in the course of this experiment. Age is another factor that affects the severity of stroke (Dinapoli et al., 2010; Zhao et al., 2017); therefore, we used mice at the same age. Young adult mice (10–12 weeks old) display strong vitality, which can reduce the death rate associated with the modeling process.Furthermore, mice at this age range had blood vessels that were the optimal size for the 0.21-mm diameter threat plugs used to generate the model. Some studies have shown the beneficial effects of CSF1R signaling on a variety of nervous system diseases (Fu et al., 2020; Hu et al., 2020), whereas other studies have reported the opposite results (Du et al.,2020; Kerkhofs et al., 2020; Neal et al., 2020). To investigate the role played by CSF1R in I/R injury, we used Ki20227 to inhibit the signaling pathway and observed its effects in tMCAO model mice.

        The infarct volume is the most direct reflection of injury degree after cerebral I/R injury (Kalogeris et al., 2016). Nissl bodies are large basophilic masses and granules found in neuronal cell bodies or dendrites. When the neuron is damaged, the number of Nissl bodies decrease, whereas Nissl bodies increase during the recovery process; Nissl body can, therefore, be used as a sign of the functional recovery state of neurons (Niu et al., 2015). After I/R, large numbers of necrotic brain cells can result in an increase in vascular wall permeability, resulting in brain edema (Shadman et al., 2019).Neurobehavioral outcomes are macroscopic reflections of the neurological state of mice (Bieber et al., 2019). In our study,we found that the intragastric injection of Ki20227 into I/R mice could increase the infarct volume, reduce the number of Nissl bodies, increase the degree of hemispheric edema,and worsen behavioral outcomes, indicating that the use of Ki20227 increased the degree of I/R injury in mice.

        Iba1 is a specific cytoskeleton protein expressed in microglia(Gheorghe et al., 2020). BrdU is a thymidine analog that binds to the DNA of mitotic cells during the S phase of the cell cycle. Therefore, Iba1 has become a widely accepted microglial marker, and BrdU has been used to monitor cell proliferation (Taupin, 2007). In this study, we use the doubleimmunofluorescence analysis of Iba1 and BrdU to evaluate microglial proliferation in different treatment groups. This analysis was performed 3 days after reperfusion when microglial proliferation and activation peaked (Inamasu et al.,2000). We found that mice treated with Ki20227 had fewer Iba1+/BrdU+cells in the peri-infarct zone after tMCAO than mice in the vehicle group, suggesting that the inhibition of CSF1R with Ki20227 blocked the proliferation of microglia.

        Unlike the ischemic core, where cells die primarily due to necrosis, apoptosis plays a dominant role in the penumbra.The penumbra represents the potentially salvageable transition zone in the peri-infarct region that expands after stroke (Uzdensky, 2019). Bcl-2 is an anti-apoptotic protein that inhibits the release of cytochrome c from the mitochondria by preventing mitochondrial membrane permeabilization in cells.Bax is a protein in the Bcl-2 family that facilitates apoptosis (Cui and Placzek, 2018). Cytochrome c further activates various proteases and nucleases, stimulating the caspase cascade(Siddiqui et al., 2015). More TUNEL-positive apoptotic cells were observed in the peri-infarcted regions in mice treated with Ki20227 than in mice treated with vehicle after I/R injury.Additionally, a reduced level of Bcl-2 and higher levels of proapoptotic proteins (Bax and Cleaved caspase-3) were detected in Ki20227-treated ischemic mice compared with those in the vehicle-treated group. Our results indicated that treatment with the CSF1R kinase inhibitor Ki20227 further aggravated apoptosis in the peri-infarcted region.

        In this study, we observed the increased release of inflammatory cytokines in response to ischemic injury,suggesting that these molecules play crucial roles in the pathological progression of ischemia. Pro-inflammatory cytokines, such as IL-6 and TNF-α, were further released in mice treated with Ki20227, whereas anti-inflammatory cytokines, such as TGF-β, were suppressed, suggesting the inflammatory effects of CSF1R signaling. Microglia play a controversial role in the inflammatory process after stroke and have been divided into M1 (pro-inflammatory) and M2(anti-inflammatory) phenotypes according to their effects(Kanazawa et al., 2017; Qin et al., 2019). Our study found changes in inflammatory cytokine expression after Ki20227 treatment; however, whether Ki20227 functions to affect the microglial polarization phenotypes requires additional studies.In addition to changes in inflammatory cytokine expression,our results showed that CSF1R was expressed at a significantly higher level after stroke, which was concurrent with changes in microglial proliferation. The expression of CSF1R decreased significantly after Ki20227 treatment. The mechanism of this phenomenon may be associated with the inhibitory effect of Ki20227, which reduces the further expression of CSF1R,inhibiting the proliferation of microglia (Neal et al., 2020).Additionally, the decrease in the microglial population may also contribute to reduced CSF1R expression.

        Oxidative stress damages the integrity of the genome and leads to cell death (Li et al., 2018). In this study, the expression levels of MDA, SOD, and GSH were determined to analyze the degree of oxidative stress. Free radicals act on lipid peroxidation, and the final product of oxidation is MDA, which can cause the cross-linking polymerization of macromolecules.Therefore, the MDA concentration has become an important indicator that reflects the degree of tissue peroxidation (Del Rio et al., 2005). SOD is an important antioxidant enzyme within the biological system. GSH can protect sulfhydryl groups in proteins from oxidation and has been widely accepted as a physiological antioxidant against free radicals(Dinapoli et al., 2010). The production and metabolism of reactive oxygen species are maintained in a dynamic balance under physiological conditions. Our study found that MDA increased sharply, whereas the antioxidant factors (SOD and GSH) decreased significantly, indicating that this balance was disrupted after tMCAO. More importantly, we found that mice treated with Ki20227 expressed higher levels of MDA and lower levels of antioxidant factors, such as SOD and GSH, after tMCAO, indicating that CSF1R inhibition exacerbated oxidative stress.

        Our study had several limitations. First, although CSF1R is present primarily in microglia in the central nervous system, CSF1R is also expressed at low levels on neurons and astrocytes. The impacts of Ki20227 on other cell types cannot be excluded, and the experimental results cannot be attributed solely to microglial CSF1R signaling. Second, we analyzed peri-infarct apoptosis in a general manner without identifying the types of cells that are affected, such as neurons, astrocytes, and microglia. All cell types will undergo apoptosis under I/R conditions, but our study did not address the specific outcomes of any specific cell types.

        In conclusion, this study found that the inhibition of CSF1R with Ki20227 blocked microglial proliferation and aggravated the pathological progression of ischemia. Enhanced apoptosis,an aggravated inflammatory response, and intense oxidative stress may be associated with the damage and deterioration observed after I/R. Therefore, the CSF1R signaling pathway may play a beneficial role in the pathological process and can potentially be considered as a therapeutic target for ischemic stroke.

        Author contributions:Study design: BRH, HJR, CJ, ZNW; experiment performance: CJ, ZNW, YCK, WXL; data analysis: WXL, YC; manuscript preparation and revision: CJ, ZNW, YCK. All authors approved the final version of the manuscript.

        Conflicts of interest:The authors declare no competing interests.

        Financial support:This study was supported by the Natural Science Foundation of Gansu Province, China, Nos. 20JR5RA337 (to BRH),20JR5RA336 (to HJR); Cuiying Graduate Supervisor Applicant Training Program of Lanzhou University Second Hospital, China, No.CYDSPY201902 (to BRH); Cuiying Students Research Ability Training Program of Lanzhou University Second Hospital, China, No. CYXZ2020-14(to BRH); and Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital, China, No. CY2018-MS08 (to BRH).The funding sources had no role in study conception and design, data analysis or interpretation, paper writing or deciding to submit this paper for publication.

        Institutional review board statement:The study was approved by Animal Ethics Committee of Lanzhou University Second Hospital (approval No. D2020-68) on March 6, 2020.

        Copyright license agreement:The Copyright License Agreement has been signed by all authors before publication.

        Data sharing statement:Datasets analyzed during the current study are available from the corresponding author on reasonable request.

        Plagiarism check:Checked twice by iThenticate.

        Peer review:Externally peer reviewed.

        Open access statement:This is an open access journal, and articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix,tweak, and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.

        Open peer reviewers:Ryszard Pluta, Polish Academy of Sciences, Poland;María José Pérez-Alvarez, Universidad Autonoma de Madrid, Spain.

        Additional files:

        Additional Table 1: Primer sequences for quantitative polymerase chain reaction.

        Additional file 1: Open peer review reports 1 and 2.

        Additional file 2: Original data of the experiment.

        91青草久久久久久清纯| 我爱我色成人网| 色欲aⅴ亚洲情无码av蜜桃| 日韩av一区二区三区四区av| 国产一区二区美女主播| 精品国产粉嫩内射白浆内射双马尾| 国产成人无码a区在线观看视频| 亚洲天堂中文| 白白色福利视频在线观看| 亚洲av无码国产精品色| 国产精品无圣光一区二区| 天天狠天天透天干天天| 国产丝袜在线福利观看| 337p日本欧洲亚洲大胆色噜噜 | 精品人妻伦九区久久aaa片69| 四虎成人精品国产一区a| 亚洲中文字幕第一页免费| 国产99视频精品免视看7 | а天堂中文在线官网在线| 国产99久久久久久免费看| 国产精品白浆视频免费观看| 亚洲国产一区二区视频| 午夜三级a三级三点在线观看| 亚洲 欧美 激情 小说 另类| 中文字幕人妻丝袜成熟乱| 麻豆人妻性色av专区0000| 久久9精品区-无套内射无码| 亚洲综合欧美在线| 国产自拍伦理在线观看| 成年av动漫网站18禁| 亚洲一区精品无码色成人| 国产高清一级毛片在线看| 91精品人妻一区二区三区水蜜桃| 尤物在线精品视频| 久久久精品3d动漫一区二区三区 | 久久久国产打桩机| 国产伦精品一区二区三区视| 中文字幕人妻乱码在线| 无码乱肉视频免费大全合集| 国产精品久久久久久人妻精品| 国产三级国产精品三级在专区|