Objective To study the p-mammalian target of rapamyein(p-roTOR)expression and its prognostic significance in non-small cell lung cancer(NSCLC).Methods Immunohistochemical staining of EnVision was applied to investigate the expression of p-roTOR in lung specimens from 59 cases with NSCLC and 10 cases with benign pulmonary diseases(3 pulmonary tuberculosises and 7 inflammatory pseudotumors 1.Results The positive rate of p-mTOR was 40.7% in NSCLC which was significantly higher than that in the benign pulmonary diseases(x =6.237,P=0.013).The expression of p-mTOR was closely correlated with age,sex and pTNM stage.Kaplan-Meire survival analysis revealed that the expression of p-mTOR was not correlated significantly with survival days(Log rank test P =0.055).Conclusion P-mTOR might be a biomark for differential diagnosis of malignant lung disease,but has poor prognostic value.
Objective To review the possible mechanisms of the mammal ian target of rapamycin (mTOR) in theneuronal restoration process after nervous system injury. Methods The related l iterature on mTOR in the restoration ofnervous system injury was extensively reviewed and comprehensively analyzed. Results mTOR can integrate signals fromextracellular stress and then plays a critical role in the regulation of various cell biological processes, thus contributes to therestoration of nervous system injury. Conclusion Regulating the activity of mTOR signaling pathway in different aspects cancontribute to the restoration of nervous system injury via different mechanisms, especially in the stress-induced brain injury.mTOR may be a potential target for neuronal restoration mechanism after nervous system injury.
Objective To investigate the mechanism of adenosine-tri phosphate (ATP) activated mammal ian target of rapamycin (mTOR)/signal transducer and activator of transcription 3 (STAT3) signal pathway in the physiology and pathology of spinal cord injury (SCI). Methods Ninety-six adult healthy female Sprague-Dawley rats were randomly divided into 4 groups (groups A, B, C and D, n=24). In groups A, B and C, the rats were made the SCI models at T8-10 levels by using a modified Allen’ s stall, and in group D, rats were given laminectomy without SCI. The rats were subjected to the administration of ATP (40 mg/kg) for 7 days in group A, to the administration of physiological sal ine (equal-volume) for 7 days in group B, to the administration of ATP (40 mg/kg) and rapamycin (3 mg/kg) for 7 days in group C, and to the administration of physiological sal ine (equal-volume) for 7 days in group D. Locomotor activity was evaluated using the Basso-Beattie-Bresnahan rating scale at the postoperative 1st, 2nd, 3rd, and 4th weeks. Then, the expressions of spinal cord cell marker [Nestin, neuron-specific enolase (NSE), gl ial fibrillary acidic protein (GFAP)] and the mTOR/STAT3 pathway factors (mTOR, STAT3) were detected at the postoperative 1st, 2nd, 3rd, and 4th weeks by immunohistochemistry analysis, Western blot assay, and real-time fluorescence PCR analysis. Results The BBB scores in group A showed a steady increase in the postoperative 1st-4th weeks and were significantly higher than those in groups B and C (P lt; 0.01), but were lower than that in group D (P lt; 0.01). Real-time fluorescence PCR results showed that the mRNA expressions of mTOR, STAT3, NSE of group A steadily increased, however, the Nestin mRNA expression gradually decreased in the postoperative 1st-4th weeks, which were all significantly higher than those of groups B, C, and D (P lt; 0.01). The mRNA expression of GFAP showed a steady increase in group A and was significantly less than those of groups B and C, but was higher than that of group D (P lt; 0.01). There were significant differences (Plt; 0.01) in all markers between groups B, C, and group D; there were significant differences in mTOR, P-mTOR, STAT3, and P-STAT3 mRNA between groups B and C at 1st-4th weeks (P lt; 0.05). The similar changes were found by Western blot assay. Conclusion ATP can activate the mTOR/STAT3 pathway to induce endogenic NSCs to prol iferate and differentiate into neurons in rats, it enhances the heal ing of SCI.
ObjectiveTo investigate the effect of dexamethasone on mammalian target of rapamycin (mTOR) expression of astrocytes in hippocampus of rats with sepsis associated encephalopathy (SAE). MethodsTotally, 90 cases of 30-day-old male Wistar rats were randomly divided into sham-operation group (n=10) and cecal ligation and puncture (CLP) group (n=80). Models of rats with sepsis were established by CLP. At 12 hours after CLP, if rats appeared lower neurobehavioral scores, abnormal electroencephalogram (EEG) and somatosensory evoked potential (SEP), they were diagnosed with SAE. And then, they were randomly divided into non-treated group and dexamethasone group. Rats in the dexamethasone group were injected with dexamethasone (1 mg/kg) via tail vein every other day for a total of 3 times. The same dose of saline was used in the non-treated group. The neurobehavioral score was measured, SEP and EEG were examined in the age of 40 days, and then the rats were killed and the hippocampus was taken. Expressions of mTOR protein were measured by Western blot. The glial fibrillary acidic protein (GFAP) and mTOR were detected by immunofluorescence assay, and the number of positive cells was calculated by image analysis system software. ResultsSix of 80 CLP rats died in 12 hours after operation, and 28 of 74 rats were diagnosed as SAE because they appeared lower neurobehavioral scores, abnormal EEG and SEP at 12 hours after CLP. The incidence of SAE was 37.84% (28/74). In the age of 40 days, compared with non-treated group, neurobehavioral score of rats in the dexamethasone group was low, the amount of alpha waves in EEG reduced, delta waves increased, the amplitude of P1 waves in SEP was decreased, and the latencies of P1 and N1 waves were prolonged (P<0.05). GFAP immunofluorescence staining showed astrocytic body and processes were small in the sham operation group. However, astrocytes in the non-treated group had large body and hypertrophic processes, and compared with the sham operation group, the number of these cells increased significantly (P<0.05). Astrocytic body and processes were small in the dexamethasone group compared with the non-treated group, and the number of cells also decreased (P<0.05). The mTOR positive astrocytes in the non-treated group were more than those in the sham operation group (P<0.05). But mTOR positive astrocytes in the dexamethasone group were fewer than those in the non-treated group (P<0.05). ConclusionsAstrocytes are activated in the hippocampus of rats with SAE. They show features of reactive hyperplasia, and the expression of mTOR is up-regulated, while dexamethasone can inhibit effects on these.
ObjectiveTo explore the involvement of miR-126 and the role of mammalian target of rapamycin (mTOR)/hypoxia-induced factor 1 α (HIF-1 α) pathway in regulating human umbilical cord mesenchymal stem cells (hUCMSCs) exosomes (Exo) on vascular endothelial growth factor (VEGF)-A levels in high glucose-induced human retinal vascular endothelial cells (HRECs). MethodsThe hREC was cultured in EGM-2-MV endothelial cell culture medium with 30 mmol/L glucose and placed in hypoxic cell incubator by 1% oxygen concentration. The cell model of high glucose and low oxygen was established. After modeling, divided HRECs into Exo group, phosphate buffer saline (PBS) group, PBS+anti-miR126 group, Exo+anti-miR126 group, PBS+anti-mTOR group, and PBS+anti-HIF-1 α group. High-glucose and hypoxia-induced hREC in the PBS and Exo groups were respectively co-cultured with PBS and 100 μg/ml hUCMSC Exo. PBS+anti-mTOR group, PBS+anti-HIF-1 α group: 500 nmol/L mTOR inhibitor ADZ2014, 25 μmol/L HIF-1 α inhibitor YC-1 pretreatment for hREC 12 h, and then co-culture with PBS after High-glucose and hypoxia-induced. PBS+anti-miR126 group, Exo+anti-miR126 group: miR-126 LNA power inhibitor probe was transfected with high glucose, and co-cultured with PBS and hUCMSC Exo 6 h after transfection. Real-time polymerase chain reaction (qPCR) measured miRNA-126 expression levels in PBS, and Exo groups for 0, 8, 16 and 24 h. After 24 hof co-culture, the levels of mTOR and HIF-1 α in the cells of PBS and Exo groups were detected by immunofluorescence, Western blot and qPCR, respectively. Western blot, qPCR detection of VEGF-A expression levels in cells of the PBS+anti-mTOR and PBS+anti-HIF-1 α groups. The expression of VE GF-A, mTOR, and HIF-1 α mRNA was measured in cells of PBS+anti-miR126 group and Exo+anti-miR126 group by qPCR. Comparison between two groups was performed by t-test; one-way ANOVA was used for comparison between multiple groups. ResultsAt 0, 8, 16 and 24 h, the relative mRNA expression of miR-126 gradually increased in the Exo group (F=95.900, P<0.05). Compared with the PBS group, The mTOR, HIF-1 α protein (t=3.466, 6.804), mRNA in HRECs in the Exo group, VEGF-A mRNA expression (t=8.642, 7.897, 6.099) were all downregulated, the difference was statistically significant (P<0.05). The relative expression level of VEGF-Aprotein (t=3.337, 7.380) and mRNA (t=8.515, 10.400) was decreased in HRECs of the anti-mTOR+PBS group and anti-HIF-1 α+PBS group, differences were statistically significant (P<0.05). The relative expression of VEGF-A, mTOR, and HIF-1 α mRNA was significantly increased in the cells of the Exo+anti-miR126 group, the differences were all statistically significant (t=4.664, 6.136, 6.247; P<0.05). ConclusionsmiR-126 plays a role in regulating the effect of hUCMSCs exosomes on VEGF-A levels in high glucose-induced HRECs via mTOR-HIF-1 α pathway.
Objective To explore the influence and mechanism of mechanistic target of rapamycin kinase (mTOR)/ receptor of advanced glycation end products (RAGE) pathway mediated-ferritinophagy on high glucose consumption promoting invasion and migration of colorectal cancer (CRC). Methods① Patients and tissue samples. Clinical data and tissues were collected from CRC patients underwent surgery and completed the dietary questionnaire in the Second Affiliated Hospital of Harbin Medical University between October 2022 and October 2023. Real-time quantitative reverse transcription PCR (qRT-PCR) was used to analyzed the expression of nuclear receptor coactivator 4 (NCOA4) and ferritin in CRC and para-carcinoma tissues.② Cell culture and treatment. The HT29 and HCT116 cells were treated by RPMI1640 medium containing 0, 35, 70, 105, 140 mmol/L glucose, and cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) activity analysis were performed to confirm 105 mmol/L glucose was the optimal concentration in the current study. Then the HT29 and HCT116 cells were randomly divided into: control group, glucose group; control group, glucose group, si-RAGE group, and glucose+si-RAGE group; control group, glucose group, rapamycin group, and glucose+rapamycin group. Untreated HT29 and HCT116 cells were considered as control group. The cells in glucose group were treated with 105 mmol/L glucose for 48 h. The CRC cells in si-RAGE group were transfected with si-RAGE for 6 h. The CRC cells in rapamycin group were treated with 10 nmol/L rapamycin for 48 h. The CRC cells in glucose+si-RAGE group were treated with 105 mmol/L glucose for 48 h combination transfected with si-RAGE for 6 h. The CRC cells in glucose+rapamycin group were treated with 105 mmol/L glucose for 48 h combination treated with 10 nmol/L rapamycin for 48 h. Then electron microscopy and western blot, wound healing assay and transwell assay were exhibited, respectively.③ Azoxymethane (AOM)-induced CRC rat model. The effects of glucose consumption on malignant behavior and ferritinophagy mediated by mTOR/RAGE pathway were evaluated in AOM-induced CRC rat models. A total of 16 rats were randomly divided into control group and glucose group, the colorectal tumor number was record and HE staining of colorectal tumor tissues was further performed. The expression of RAGE, mTOR, NCOA4, and ferritin in colorectal tissues of rats from each group was detected by qRT-PCR. Results① More lymphatic node metastasis and TNM Ⅲ/Ⅳ stages was observed in CRC patients from high glucose consumption group (P=0.004, P=0.004). Moreover, we confirmed that NCOA4 expression was significantly decreased (P<0.001) while ferritin was significantly increased (P<0.001) in CRC tissues especially in the CRC tissues from patients with positive lymph nodes metastasis. Additionally, high glucose consumption of CRC patients was negatively correlated with ferritinophagy flux. ② High glucose treatment significantly decreased autophagosomes in HT29 and HCT116 cells while si-RAGE transfection increased autophagic vacuoles compared to the control group. When compared with the glucose group, autophagosomes were increased in the glucose+si-RAGE group. Moreover, compared to the control group, the expression of RAGE, p-mTOR, and ferritin was increased (P<0.001) while the expression of NCOA4 was decreased (P<0.001) in glucose group, but the expression of RAGE, p-mTOR and ferritin was decreased (P<0.001) while the expression of NCOA4 was increased (P<0.001) in si-RAGE group; when compared with the glucose group, the expression of RAGE, p-mTOR and ferritin was downregulated (P<0.001) while the expression of NCOA4 was upregulated (P<0.001) in HT29 and HCT116 cells from the glucose+siRAGE group. Compared to the control group, the HT29 and HCT116 cells in the glucose group performed enhanced wound scratch healing and migration, invasion viability (P<0.05); but the HT29 and HCT116 cells in the si-RAGE group performed impaired wound scratch healing and migration, invasion viability (P<0.05). When compared with the glucose group, the HT29 and HCT116 cells in the glucose+si-RAGE group performed impaired wound scratch healing and migration, invasion viability (P<0.05).③ Rapamycin treatment significantly inhibited the expression of RAGE, p-mTOR and ferritin (P<0.05) but induced the expression of NCOA4 (P<0.05) compared to the control group. When compared with the glucose group, the expression of RAGE, p-mTOR and ferritin was downregulated (P<0.05) while the expression of NCOA4 was upregulated (P<0.05) in HT29 and HCT116 cells from the glucose+rapamycin group. Additionally, compared to the control group, rapamycin treatment performed inhibited effect on wound scratch healing and migration, invasion viability in the HT29 and HCT116 cells (P<0.05); while the HT29 and HCT116 cells in the glucose+rapamycin group performed impaired wound scratch healing and migration, invasion viability (P<0.05) when compared with the glucose group.④ In the AOM induced CRC rat model, we found the more colorectal tumors, aggravated cellular pleomorphism and upregulate expression of RAGE, p-mTOR, ferritin (P<0.05) while downregulated expression of NCOA4 (P<0.05) in the control group than those of the glucose group. ConclusionHigh glucose consumption promote invasion and migration in CRC through suppressing ferritinophagy via activating the mTOR/RAGE pathway.