Neuritin Inhibits Astrogliosis To Ameliorate Diabetic Cognitive Dysfunction

1y ago
11 Views
3 Downloads
6.64 MB
14 Pages
Last View : 1m ago
Last Download : 3m ago
Upload by : Kaydence Vann
Transcription

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.66 :4Neuritin inhibits diabeticastrogliosis259–272RESEARCHNeuritin inhibits astrogliosis to amelioratediabetic cognitive dysfunctionZuo Zhang*, Hongli Zhou* and Jiyin ZhouNational Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, ChinaCorrespondence should be addressed to J Zhou: zhoujiyin@gmail.com*(Z Zhang and H Zhou contributed equally to this work)AbstractEarlier, it was shown that reversing the downregulation of neuritin expression in the brainimproves central neuropathy in diabetic rats. We investigated the protective mechanismof neuritin in diabetic cognitive dysfunction via astrocytes. Further, the impact of theoverexpression of neuritin in the cortex and the hippocampus on diabetic cognitivedysfunction and astrogliosis in type 2 diabetic (db/db) mice was assessed. Antagonistswere used to inhibit the JAK2/STAT3 signaling pathway in U-118MG, an astrocyte cell line.Immunofluorescence, Western blotting, and real-time PCR were performed. Neuritinoverexpression in the hippocampus of db/db mice significantly ameliorated cognitivedysfunction, hippocampal neuronal impairment, and synaptic plasticity deterioration, andinhibited astrogliosis and the JAK2/STAT3 signaling pathway in the hippocampus. Neuritinsuppressed the JAK2/STAT3 signaling pathway to inhibit lipopolysaccharide-inducedgliosis in U-118MG cells. It was observed that neuritin regulates the JAK2/STAT3 signalingpathway in astrocytes to inhibit astrogliosis and improve diabetic cognitive dysfunction.Key Wordsff neuritinff diabetic cognitivedysfunctionff gliosisff astrocyteff JAK2/STAT3 signalingpathwayJournal of MolecularEndocrinology(2021) 66, 259–272IntroductionThe incidence of cognitive dysfunction in patients withtype 2 diabetes is 1.5 times higher than that in nondiabeticpatients and 60 to 70% of diabetes patients have cognitivedysfunction. Currently, the management strategies fortype 2 diabetes cannot benefit cognitive dysfunction,which place a great burden on type 2 diabetes, theirfamilies, and society (Yuan & Wang 2017). Progressivecognitive dysfunction is a central characteristic of diabeticencephalopathy (Xu et al. 2017). Prediabetes is linked tostructural brain abnormalities, with further exacerbationof type 2 diabetes (van Agtmaal et al. 2018). Impairedcognition during type 2 diabetes is particularly evidentin the scope of memory and executive function (AreosaSastre et al. g/10.1530/JME-20-0321 2021 The authorsPublished by Bioscientifica Ltd.Printed in Great BritainHippocampal synaptic plasticity is the neurobiologicalbasis of learning and memory in cognitive functionand participates in the occurrence and development ofcognitive dysfunction in type 2 diabetes (Huang et al.2016). Hippocampal synaptic plasticity is regulated byseveral neurotrophic factors, including neuritin (Anet al. 2014). Astrocytes not only bridge the gap betweenmetabolic supplies by blood vessels and neurons but alsoallow the fine control of neurotransmission by providingappropriate signaling molecules and insulation throughtight enwrapping of synapses (Dallerac & Rouach 2016).Abnormalities in synaptic transmission lead to cognitivedysfunction (Koyama 2014). Astrocytes play an importantrole in cognitive functions, including learning andThis work is licensed under a Creative CommonsAttribution 4.0 International License.Downloaded from Bioscientifica.com at 06/20/2022 01:32:09AMvia free access

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.memory (Santello et al. 2019). Reactive gliosis is a processin which astrocytes maintain the stability of the neuronalmicroenvironment and play a protective role in the earlystage of the injury, but persistent reactive gliosis forms aglial scar at the injury site to repair the missing site andfurther block the nerve. Diabetic rats display astrogliosis inthe cortex and hippocampus (Tomassoni et al. 2013); whiletype 2 diabetic mice also show synaptic dysfunction andastrogliosis with memory impairment (Duarte et al. 2012).Scar formation in astrocytes after spinal injury isregulated by the STAT3 signaling pathway (Wanner et al.2013). The JAK2/STAT3 signaling pathway is involvedin several diabetic complications, such as diabeticneuropathy (Li et al. 2019), diabetic cardiomyopathy (Gaoet al. 2019), and diabetic nephropathy (Wang et al. 2012).The JAK2/STAT3 signaling pathway in the hippocampus ofdiabetic rats induced by streptozotocin is one of the mostsignificant signaling pathways that regulates the processduring such complications (Gurzov et al. 2016). TheJAK2/STAT3 signaling pathway is involved tion (Chen et al. 2018) and is necessary and sufficientto induce and maintain astrocyte reactivity (Ceyzeriatet al. 2018). Over the years, the JAK2/STAT3 signalingpathway has emerged as a central regulator of astrocytereactivity and plays a critical role in animal models thatregulate synaptic plasticity, reactive gliosis, and cognitivedysfunction (Ceyzeriat et al. 2016).Neuritin (also named cpg15) is an red axonal proteinthat is mainly expressed in the brain (Zhou & Zhou2014). Neuritin ameliorates neurite outgrowth recoveryof hippocampal neurons after mouse cerebral ischemia(Zhao et al. 2017) and improves depression and cognitivefunction during schizophrenia (Son et al. 2012). Nervegrowth factor treatment restores neuritin levels in thedorsal root ganglia and sciatic nerves of diabetic rats(Karamoysoyli et al. 2008). In a previous study, we showedthat berberine benefits diabetic neuropathy by improvingmicropathology and increasing neuritin expression viathe mitogen-activated protein kinase signaling pathway(Zhou et al. 2016). Administration of exogenous neuritinimproves the viability and function of Schwann cells indiabetic neuropathy rats (Xi et al. 2020).In the present study, we employed the overexpressionof neuritin in the cortex and hippocampus of type 2diabetic (db/db) mice and lipopolysaccharide inductionof U-118MG astrocyte cell line to investigate the effects ofneuritin on diabetic cognitive dysfunction and astrogliosisthrough the JAK2/STAT3 signaling pathway.https://jme.bioscientifica.com https://doi.org/10.1530/JME-20-0321 2021 The authorsPublished by Bioscientifica Ltd.Printed in Great Britain66 :4Neuritin inhibits diabeticastrogliosis260Material and methodsdb/db/neuritin/Emx1-Cre miceA transgenic mouse line harboring the CMV-LoxP-STOPLoxP-tagged human neuritin transgene was establishedusing C57BL/6J mice generated by Cyagen Biosciences Inc.((Guangzhou, China) Certificate No. TGBS141013BA1).A transgenic mouse model with high cortical andhippocampal tissue-specific overexpression of neuritinwas established by crossing neuritin transgenic mice withEmx1-Cre mice (B6.129S2Emx1tm1(cre)Krj/J, https://www.jax.org/strain/005628). C57BL/6J-Leprdb/ heterozygouslittermate (db/m) mice were purchased from the JacksonLaboratory (Stock Number: 000699). Nondiabetic db/mmice were used to crossbreed C57BL/6J-Leprdb/dbdiabetic (db/db) mice. The db/m mice were crossed withthe neuritin-Cre transgenic mice to yield db/db/neuritin/Exm1-Cre (neuritin overexpression db/db) mice, whichare triple transgenic diabetic mice overexpressing neuritinin the cortex and hippocampus. All mice were backcrossedonto the C57BL/6J background for ten generations.Cre-mediated excision of neuritin was assessed by PCRusing genomic DNA derived from the tail. In all animalstudies, male mice were used, and littermates served ascontrols. All mice were bred in a specific vironmentwith a 12 h light: 12 h darkness cycle and allowed freeaccess to food and water. The animal experiments wereapproved by the Army Medical University according tothe guidelines of the Institutional Animal Care and UseCommittee.Drug treatmentBoth standard and high-fat diets containing 45% fatwere purchased from Mediscience Ltd., Nanjing, China.The high-fat diet contained 24.0% protein, 41.0%carbohydrate, and 24.0% fat. Six-week-old male mice wereseparated into four groups, with six animals per group.One group of mice was fed a standard diet, while the othergroups were fed a high-fat diet instead of a standard diet for8 weeks. (i) db/m mice were fed a standard diet, (ii) db/dbmice fed a high-fat diet, (iii) neuritin-overexpressingdb/db mice fed a high-fat diet, (iv) db/db mice fed ahigh-fat diet JAK2 inhibitor (AG490, 15 mg/kg, Abcam,Catalog #ab120950). During the 8 weeks of treatment, themice were intraperitoneally injected with JAK2 inhibitoror its dilution vehicle (PBS containing 5% dimethylsulfoxide) (Ignarro et al. 2013).This work is licensed under a Creative CommonsAttribution 4.0 International License.Downloaded from Bioscientifica.com at 06/20/2022 01:32:09AMvia free access

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.Morris water maze testAfter 7 weeks of drug administration, the animals weretested in a spatial version of the Morris water maze testas previously described (Si et al. 2016). The Morris watermaze consisted of a circular water tank (120 cm diameter,50 cm height) that was partially filled with water (25 C).Milk powder was used to render the water opaque. Thetraining started by acclimating the mouse to the taskenvironment with 2 days of free-swimming in the poolwith no platform. Each session lasted for 2 min. The poolwas virtually divided into four equal quadrants, labeledas N-S-E-W. A platform (10 cm diameter) was placed inone of the four maze quadrants (the target quadrant)and submerged 0.5 cm below the water surface. Theplatform remained in the same quadrant throughout theexperiment. The mice were required to find the platformusing only the distal spatial cues available in the testingroom. The cues were maintained throughout the time ofthe test. The mice received four consecutive daily trainingtrials in the following 5 days, with each trial having aceiling time of 60 s and a trial interval of approximately30 s. The mouse had to swim until it climbed onto theplatform and then submerged beneath the water. Afterclimbing onto the platform, the animal remained therefor 30 s before the commencement of the next trial. Theescape platform was kept at the same position relative tothe distal cues. If the mouse failed to reach the escapeplatform within the maximum allowed time of 60 s, itwas gently placed on the platform and allowed to remainthere for the same amount of time. The time taken toreach the platform (latency in seconds) was measured.A probe trial was performed to assess the extent ofmemory consolidation. The time spent in the targetquadrant indicates the degree of memory consolidationthat occurs after learning. In the probe trial, the mousewas placed into the pool as in the training trial, exceptthat the hidden platform was removed from the pool. Thetime of crossing the former platform quadrant and thetotal time of crossing all quadrants were recorded for 60 s.Tissue preparationAfter the Morris water maze test, mice were allowedto recover for a day, then fasted overnight, and wereanesthetized with chloral hydrate (ip, 400 mg/kg). Bloodof mice from each group was collected from the heart,transferred immediately into microcentrifuge tubes,and allowed to clot to obtain the serum. It was thenperfused with 0.9% sodium chloride solution containinghttps://jme.bioscientifica.com https://doi.org/10.1530/JME-20-0321 2021 The authorsPublished by Bioscientifica Ltd.Printed in Great Britain66 :4Neuritin inhibits diabeticastrogliosis2610.1% diethylpyrocarbonate at 25 C followed by 4%paraformaldehyde in 0.1 mol/L PBS. After removing fromthe skull, the brains were fixed in 4% paraformaldehydeovernight, dehydrated in a 30% sucrose solution for3–5 days at 4 C. Serial coronal sections (25 μm thick)of the whole hippocampus were cut using a slidingmicrotome and stored at 20 C until used for Nissl andimmunofluorescence staining. The same sequence numbersection of serial sagittal sections of the brain containingthe hippocampus was used for each experiment.Nissl stainingThe frozen sections were fixed with 70% ethanol for 30 sand rinsed in DEPC-treated water for 30 s. The sectionswere then stained with 1% toluidine blue dye for 10 minat room temperature. After washing in distilled waterfor 1 min, the sections were dehydrated in a gradientalcohol and mounted with neutral resins. Nissl substancewas observed under a light microscope (Olympus) withlive neurons being highlighted by blue staining (Su et al.2017). ImageJ 1.50 (National Institutes of Health) wasused to analyze the average gray value of images.In vitro U-118MG cells experimentU-118MG cells were maintained in a humidified incubatorwith 5% CO2 and maintained at 37 C in Dulbecco’smodified Eagle’s medium (DMEM), supplemented with10% fetal bovine serum. Recombinant human neuritin(Sigma Co. Ltd.), JAK2 inhibitor (AG490), and STAT3inhibitor (Stattic, Abcam Catalog #ab120952) wereadministered 30 min before stimulation with 1 μg/mLlipopolysaccharide. After 48 h of treatment, cells werecollected and lysed, and cell extracts were analyzed byreal-time PCR and Western blotting.Immunofluorescence stainingEnzymatic retrieval was performed by incubating thesections in proteinase K for 10 min at 25 C. The sectionswere rinsed with PBS, permeabilized with 0.3% TritonX-100 in PBS for 30 min, blocked using blocking buffer(PBS containing 5% normal serum and 0.3% TritonX-100) for 1 h, and incubated with primary antibodies(4 C, 12 h) and secondary antibodies (37 C, 2 h) in PBScontaining 0.05% Tween 20. The primary antibodies usedwere as follows: GFAP (1:100, Abcam, Catalog #ab7260),JAK2 (1:100, Abcam, Catalog #ab108596), p-JAK2 (1:100,Abcam, Catalog #ab32101), STAT3 (1:100, Abcam,This work is licensed under a Creative CommonsAttribution 4.0 International License.Downloaded from Bioscientifica.com at 06/20/2022 01:32:09AMvia free access

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.Catalog #ab68153), p-STAT3 (1:100, Abcam, Catalog#ab76315), and neuritin (1:100, Abcam, Catalog #64186).The secondary antibodies Alexa 488-labeled goat antirabbit IgG (1:500, Catalog#A0423) and Alexa 647-labeledgoat anti-rabbit IgG (1:500, Catalog#A0468) werepurchased from Beyotime (Shanghai, China). Nuclei werestained with DAPI (Beyotime). Finally, slides were washedfive times in PBS and coverslips were mounted in 90%glycerol for microscopic analysis.Western blotThe cortex and hippocampus of the mice were dissectedon ice. The proteins in the cortex, hippocampus, andU-118MG cells were extracted using RIPA lysis buffer(Beyotime, Catalog #P0013B), and total proteins in thesupernatant were determined using a BCA protein assaykit (Beyotime, Catalog #P0012). Then, 40 μg of proteinwas mixed in a buffer (25% glycerol, 2% SDS, 0.01%bromophenol blue, Tris–HCl, pH 6.8) and heated at100 C for 5 min. The samples were subjected to 10%SDS-PAGE, followed by transfer onto a PVDF membrane(Roche) using the GelDoc XR system (Bio-Rad) (Tang et al.2017). The membrane was washed with Tris-buffered154 mmol/L NaCl solution with 0.1% Tween 20, andincubated with anti-rabbit neuritin (Abcam, Catalog#64186), GFAP (Abcam, Catalog #ab7260), JAK2 (Abcam,Catalog # ab108596), p-JAK2 (Abcam, Catalog #ab32101),STAT3 (Abcam, Catalog #ab68153), p-STAT3 (Abcam,Catalog #ab76315), or β-actin polyclonal antibody (100in dilution, Sigma, Catalog #A2103) for 1 h at 25 C, andincubated with peroxidase-conjugated anti-rabbit IgG(1:1000) for 1 h at 25 C. After the reaction, proteins werevisualized with an ECL kit and images were obtained usingImageQuant LAS4010 (GE Healthcare). Samples were runin duplicate for each experiment. Densitometry analysisof the images was performed using ImageJ 1.50.Real-time PCRTotal RNA from the cortex, hippocampus, and U-118MGcells was extracted using RNAiso Plus (TAKARA,Catalog#9108/9109) (Tang et al. 2017). cDNA wassynthesized using a Reverse-Transcription Reagent Kit(TAKARA, Catalog#RR047A) (Tang et al. 2017). Real-timePCR measurements of individual cDNAs were performedusing SYBR Premix Ex Taq II (TAKARA, Catalog#RR820A)to measure the duplex DNA formation with the ABI Prism7500 Sequence Detection System (Applied Biosystems)(Tang et al. 2017), normalized to the amount of β-actin RNAhttps://jme.bioscientifica.com https://doi.org/10.1530/JME-20-0321 2021 The authorsPublished by Bioscientifica Ltd.Printed in Great Britain66 :4Neuritin inhibits diabeticastrogliosis262and analyzed by the 2 CT method (Livak & Schmittgen2001). The following primers were used: β-actin sense5-CTCTAGACTTCGAGCAGGAGAT-3; β-actin antisense5-CAGGATTCCATACCCAAGAAGG-3; neuritin sense5-GCGGTGCAAATAGCTTACCTG-3, neuritin antisense5-CGGTCTTGATGTTCGTCTTGTC-3′.Statistical analysisAll data are presented as means s.d. All grouped data wereanalyzed using SPSS 13.0. Comparisons between groupswere made by one-way ANOVA followed by Tukey’s testto analyze the differences. Statistical significance was setat P 0.05.ResultOverexpression of neuritin in hippocampus of miceReal-time PCR analysis showed significantly increasedmRNA expression of neuritin in the hippocampus ofneuritin-overexpressing transgenic C57BL/6J micecompared to the WT mice but not in the cortex (Fig. 1A).Immunochemical staining (Fig. 1B) and Western blotting(Fig. 1C) analysis also confirmed increased neuritinexpression. However, neuritin expression was not affectedin the other tissues (data not shown). Since there wasno difference in the expression of neuritin in the cortex,the subsequent experiments were only focused on thehippocampus.Effect of neuritin on memoryCognitive function was assessed using the Morris watermaze test. The mean escape latency for the trained micedecreased from 70 to 17 s over the course of the 20learning trials. The mean escape latency did not differbetween any of the groups on the first and the seconddays of testing in the Morris water maze. However, fromthe third day onwards, there was a significant differencein the transfer latency between db/db and db/m mice.db/db mice showed a lower ability to find the platform andlearned its location on the fifth day of training. Neuritinoverexpression significantly decreased the mean transferlatency in db/db mice (Fig. 2A). This poorer performancewas also improved upon treatment with the JAK2inhibitor, as evident from the animal’s decreased latencyto find the platform from the third day of training. Figure2B displays the representative swimming paths of mice inthe four groups on the fourth day of training.This work is licensed under a Creative CommonsAttribution 4.0 International License.Downloaded from Bioscientifica.com at 06/20/2022 01:32:09AMvia free access

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.Neuritin inhibits diabeticastrogliosis66 :4263Figure 1Expression profile of neuritin in the cortex and hippocampus of mice overexpressing neuritin. Neuritin mRNA expression in cortex and hippocampus wasmeasured by real-time PCR in WT and neuritin-overexpressing mice (A). Neuritin expression in the cortex and hippocampus was observed byimmunofluorescence (B), the quantification of fluorescence-integrated intensity (C), and by Western blot and its quantification (D) in WT and neuritinoverexpressing mice. Mean standard deviation (s.d.), n 6. *P 0.01, compared with WT mice. Overexpression, neuritin overexpression. A full colorversion of this figure is available at https://doi.org/10.1530/JOE-20-0321.Animals showed a significant difference in the probetrial of the Morris water maze study, which measuredhow well the animals had learned and consolidated theplatform location during the 5 days of training (Fig. 2C).db/db mice spent less time in the target quadrant thancontrol mice. The time spent in the target quadrant wassignificantly higher in mice with neuritin overexpressionand JAK2 inhibitor-treated db/db mice than in db/m mice.Figure 2Effects of neuritin on cognitive dysfunction ofdb/db mice. The alteration of transfer latency (A),pathway maps of searching for the hiddenplatform at the fourth day of training (B), and thealteration of time spent in the target quadrant (C)during the Morris water maze test. Mean s.d.,n 6. *P 0.01, compared with db/m mice;#P 0.01, compared with db/db mice. db/dbneuritin, neuritin overexpression db/db; db/dbinhibitor, db/db JAK2 inhibitor. A full color versionof this figure is available at oscientifica.com https://doi.org/10.1530/JME-20-0321 2021 The authorsPublished by Bioscientifica Ltd.Printed in Great BritainThis work is licensed under a Creative CommonsAttribution 4.0 International License.Downloaded from Bioscientifica.com at 06/20/2022 01:32:09AMvia free access

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.Effects of neuritin on body weight and brain weightAs shown in Fig. 3A, db/db mice showed significantlyhigher body weight than db/m animals that were fed astandard diet. Neuritin overexpression slightly decreasedthe body weight of db/db mice However, JAK2 inhibitoradministration for 8 weeks did not change the bodyweight of db/db mice.db/db mice showed a significant decrease in brainweight compared to db/m mice (Fig. 3B). Neuritinoverexpression slightly ameliorated the brain weight ofdb/db mice However, JAK2 inhibitor administration for8 weeks did not affect the brain weight in db/db mice.Neuritin inhibits diabeticastrogliosis66 :4264Neuritin improved neuronal impairment inthe hippocampusNissl staining revealed a significantly lower number ofneurons in db/db mice than in db/m mice (Fig. 4A). DAPIstaining showed a significantly lower number of all cells,including neurons, in db/db mice than in db/m mice(Fig. 4B). Overexpression of neuritin and JAK2 inhibitortreatment ameliorated these changes in the hippocampusof diabetic mice.Neuritin ameliorated astrogliosis and synapticplasticity in hippocampusIn Fig. 5A, we demonstrated the astrocyte markerGFAP in the hippocampus of each group of mice byimmunohistochemistry. There was a significant increasein the expression of GFAP in the hippocampus of db/dbmice compared to that of standard diet-fed db/m mice.GFAP expression was downregulated by both neuritinoverexpression and JAK2 inhibitor treatment in db/db mice.db/db mice expressed lower levels of synaptophysinin the hippocampus than db/m mice (Fig. 5B).Overexpression of neuritin and JAK2 inhibitor treatmentupregulated the expression of synaptophysin in thehippocampus of db/db mice.Neuritin regulated JAK2/STAT3 signaling pathwayin hippocampusTo further explore the potential mechanism by whichneuritin attenuates hippocampal astrogliosis, wedetermined the effect of neuritin on the JAK2/STAT3signaling pathway. Protein expression of p-JAK2 andp-STAT3 was significantly upregulated in db/db micecompared to that in db/m mice, thereby indicating theactivation of the JAK2/STAT3 signaling pathway (Fig.6A and B). This activation was markedly reversed by theoverexpression of neuritin and JAK2 inhibitor treatment,as shown by the significantly decreased expressionof p-JAK2 and p-STAT3 in db/db mice. This providedcompelling evidence that the neuritin interfered with theJAK2/STAT3 signaling pathway in the hippocampus.Figure 3Effects of neuritin on body weight and brain weight of db/db mice. Thechanges in body weight (A) and brain weight (B). Mean s.d., n 6.*P 0.01, compared with db/m mice; #P 0.05, compared with db/dbmice. db/db neuritin, neuritin overexpression db/db; db/db inhibitor,db/db JAK2 inhibitor. A full color version of this figure is available bioscientifica.com https://doi.org/10.1530/JME-20-0321 2021 The authorsPublished by Bioscientifica Ltd.Printed in Great BritainNeuritin inhibited lipopolysaccharide-inducedgliosis in U-118MG cellsLipopolysaccharide significantly upregulated GFAPexpression in U-118G cells compared to that in the controlgroup (Fig. 7). However, when pretreated with recombinantThis work is licensed under a Creative CommonsAttribution 4.0 International License.Downloaded from Bioscientifica.com at 06/20/2022 01:32:09AMvia free access

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.Neuritin inhibits diabeticastrogliosis66 :4265Figure 4Neuritin ameliorated neuronal impairment in hippocampus. The Nissl staining (A) and its quantification analysis (B) and DAPI staining (C) and itsquantification analysis (D) in the hippocampus. Mean s.d., n 6. *P 0.01, compared with db/m mice; #P 0.01, compared with db/db mice. Scalebar 100 μm in A, scale bar 50 μm in B. db/db neuritin, neuritin overexpression db/db; db/db inhibitor, db/db JAK2 inhibitor. A full color version of thisfigure is available at https://doi.org/10.1530/JOE-20-0321.neuritin (100 ng/mL), JAK2 inhibitor, or STAT3 inhibitorfor 30 min, there was a suppressed GFAP expression.Neuritin suppressed lipopolysaccharide-stimulatedJAK2/STAT3 pathway activation in U-118MG osphorylation of JAK2 and STAT3 in U-118MG cellswithout affecting the total levels of the proteins. However,30 min pretreatment with recombinant neuritin(100 ng/mL) decreased the phosphorylation of JAK2 andSTAT3, while the phosphorylation of JAK2 and STAT3showed a similar trend when the cells were incubatedwith JAK2 inhibitor. However, the STAT3 inhibitor onlydownregulated the expression of p-STAT3 (Fig. 8).DiscussionOur study found that neuritin overexpression in thehippocampus of db/db mice significantly amelioratedhttps://jme.bioscientifica.com https://doi.org/10.1530/JME-20-0321 2021 The authorsPublished by Bioscientifica Ltd.Printed in Great Britaincognitive dysfunction, neuronal impairment, and synapticplasticity, and inhibited astrogliosis and the JAK2/STAT3signaling pathway in the hippocampus. Neuritin alsosuppressed the JAK2/STAT3 signaling pathway to inhibitlipopolysaccharide-induced gliosis in U-118MG cells.Obesity is the single best predictor of whether aperson would develop type 2 diabetes. In our study,db/db mice were fed a high-fat diet to induce diabeticneuropathy (Islam 2013). Our results show that there issignificant downregulation in the expression of neuritinand increased body weight in db/db mice compared tocontrol mice. The downregulated expression of neuritinmight, thus, be body weight dependent, which is inaccordance with the results of previous investigationsin streptozotocin-induced diabetic rats (Karamoysoyliet al. 2008, Xi et al. 2020). There are no reports on theside effects of acute and chronic exogenous neuritinadministration in mice or rats (An et al. 2014, Gao et al.2016, Xi et al. 2020).The diabetic brain has structural and functionalabnormalities, including atrophy of the whole brain,This work is licensed under a Creative CommonsAttribution 4.0 International License.Downloaded from Bioscientifica.com at 06/20/2022 01:32:09AMvia free access

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.Neuritin inhibits diabeticastrogliosis66 :4266Figure 5Neuritin ameliorated astrogliosis and synaptic plasticity in the hippocampus. GFAP protein was measured using immunofluorescence in thehippocampus (A) and its quantification of fluorescence integrated intensity (B). Synaptophysin protein was measured using immunofluorescence (C) andits quantification of fluorescence integrated intensity (D). Mean s.d., n 6. *P 0.01, compared with db/m mice; #P 0.01, compared with db/db mice.db/db neuritin, neuritin overexpression db/db; db/db inhibitor, db/db JAK2 inhibitor. A full color version of this figure is available at https://doi.org/10.1530/JOE-20-0321.gray matter, hippocampus, and amygdala. Progressivebrain atrophy (Zhou et al. 2020), axon loss, and neuronaldegeneration in the cortex and hippocampus have beenobserved in diabetic animals and humans (Klein &Waxman 2003). Consistent with previous reports in db/dbmice showing that the brains of db/db mice are smallerand lighter than those of control mice (Sheppard et al.1985, Makar et al. 1995, Vannucci et al. 1997, Ahima et al.1999), our results showed that the brain weight of db/dbmice was remarkably lower than that of db/m mice.According to the Jackson Laboratory, recombination(overexpression of neuritin) occurs in approximately 88%neurons of the neocortex and hippocampus, and in theglia of the cerebral cortex. Cortical excitatory neurons andhttps://jme.bioscientifica.com https://doi.org/10.1530/JME-20-0321 2021 The authorsPublished by Bioscientifica Ltd.Printed in Great Britainglia (radial glia, astrocytes, and oligodendrocytes), but notGABAergic neurons, are produced in the Emx1-expressinglineage (Gorski et al. 2002). Overexpression of neuritinin the cortex and hippocampus increases the lower brainweight of db/db mice, but JAK2 inhibitor has no effecton the brain weight of db/db mice. Consistent withprevious reports following Nissl and DAPI staining (Yanet al. 2009, Zhang et al. 2018), db/db mice showed a lowernumber of neurons and all cells in the hippocampus thanthat in db/m mice. Overexpression of neuritin preventsthe loss of neurons in db/db mice, but JAK2 inhibitorscould not restore the lost neurons in db/db mice. Globalknockout of neuritin in mice accelerates retinal ganglioncell loss and retinal degeneration following optic nerveThis work is licensed under a Creative CommonsAttribution 4.0 International License.Downloaded from Bioscientifica.com at 06/20/2022 01:32:09AMvia free access

Journal of MolecularEndocrinologyZ Zhang, H Zhou et al.Neuritin inhibits diabeticastrogliosis66 :4267Figure 6Neuritin suppressed JAK2/STAT3 signaling pathway in the hippocampus. Phosphorylation level of JAK2 was measured using immunofluorescence inhippocampus (A) and its quantification of fluorescence integrated intensity (B). Phosphorylation level

The high-fat diet contained 24.0% protein, 41.0% carbohydrate, and 24.0% fat. Six-week-old male mice were separated into four groups, with six animals per group. One group of mice was fed a standard diet, while the other groups were fed a high-fat diet instead of a standard diet for 8 weeks. (i) db/m mice were fed a standard diet, (ii) db/db

Related Documents:

the bonds are usually excitatory in nature. But not always. For example, Estes (1972) proposed a chain-association theory where the bonds are inhibitory rather than excitatory. The first unit inhibits the remaining units, the second inhibits all but the first, the third inhibits all but the first two, and so on. For example, in

work/products (Beading, Candles, Carving, Food Products, Soap, Weaving, etc.) ⃝I understand that if my work contains Indigenous visual representation that it is a reflection of the Indigenous culture of my native region. ⃝To the best of my knowledge, my work/products fall within Craft Council standards and expectations with respect to

Zyflamend, a polyherbal mixture, inhibits lipogenesis and mTORC1 signalling via activation of AMPK Yi Zhao a,1, Dallas Donohoe a, E-Chu Huang 2, Jay Whelan,b * a Department of Nutrition, University of Tennessee, 1215 West Cumberland Avenue, 229 Jessie Harris Building, Knoxville,TN 37996, USA b Tennessee Agricultural Experiment Station, University of Tennessee, Knoxville,TN 37996, USA

Sulfometuron - DuPont Oust XP Sulfonylurea family Inhibits branched amino acid synthesis ALS (or AHAS) – Acetolactase synthetase Isoleucine, leucine, valine Herbicides for Pine Silviculture Metsulfuron - DuPont Escort XP Sulfonylurea family Inhibits branched ami

Polyglutamine-expanded ataxin-7 inhibits STAGA histone acetyltransferase activity to produce retinal degeneration Vikas B. Palhan*†, Shiming Chen†‡§, Guang-Hua Peng‡, Agneta Tjernberg¶, Armin M. Gamper*, Yuxin Fan**, Brian T. Chait¶, Albert R. La Spada**††‡‡§§¶¶, and Robert G. Roeder*¶¶ Laboratories of *Biochemistry and Molecular Biology and ¶Mass Spectrometry and .

Robo4 is a vascular-specific receptor that inhibits endothelial migration Kye Won Park,a,b Clayton M. Morrison,a Lise K. Sorensen,a Christopher A. Jones,a,b Yi Rao,c Chi-Bin Chien,d Jane Y. Wu,e Lisa D. Urness,f and Dean Y. Lia,b,f,* a Program in Human Molecular Biology and Genetics, School of Medicine, Univers

meiosis II produces spermatids in the seminiferous tubules; 5.FSH and testosterone stimulates the Sertoli cells 7.release spermatozoa into the lumen 6.which stimulate sperm maturation 8.increase in testosterone inhibits GnRH 9.testosterone inhibits ICSH 10. inhibin suppresses FSH In exams, the

Syllabus for ANALYTICAL CHEMISTRY II: CHEM:3120 Spring 2017 Lecture: Monday, Wednesday, Friday, 10:30-11:20 am in W128 CB Discussion: CHEM:3120:0002 (Monday, 9:30-10:20 AM in C129 PC); CHEM:3120:0003 (Tuesday, 2:00-2:50 PM in C129 PC); or CHEM:3120:0004 (Wednesday, 11:30-12:20 PM in C139 PC) INSTRUCTORS Primary Instructor: Prof. Amanda J. Haes (amanda-haes@uiowa.edu; (319) 384 – 3695) Office .