12 research outputs found
DataSheet_1_CSTF2 Promotes Hepatocarcinogenesis and Hepatocellular Carcinoma Progression via Aerobic Glycolysis.docx
BackgroundThe shortening of 3’ untranslated regions (3’UTRs) of messenger RNAs(mRNAs) by alternative polyadenylation (APA) is an important mechanism for oncogene activation. Cleavage stimulation factor 2 (CSTF2), an important regulator of APA, has been reported to have a tumorigenic function in urothelial carcinoma of the bladder and lung cancers. However, the tumor-promoting role of CSTF2 in hepatocellular carcinoma (HCC) and its underlying molecular mechanism remains unclear.MethodsMultiple databases were used to analyze the expression level and prognostic value of CSTF2 in HCC. Function enrichment analysis was used to investigate the molecular mechanism of CSTF2 for the occurrence and development of HCC. The biological function in HCC cell lines in vitro was determined by CCK8, colony formation, Transwell migration, and invasion assay. Moreover, the tumorigenic function of CSTF2 in vivo was measured by a subcutaneous tumor formation or injecting four plasmids into a mouse tail vein within 5–7 s in an immunocompetent HCC mouse model. In addition, aerobic glycolysis in HCC cells was determined by measuring the extracellular acid rate (ECAR) and extracellular glucose and lactate levels.ResultsBioinformatics analysis revealed that CSTF2 was overexpressed in HCC tissues. The high expression of CSTF2 was correlated with a poor prognosis and high histological grades. CSTF2 knockout inhibited the proliferation, migration, and invasion of HCC cells. In addition, CSTF2 knockout HCC cells failed to form tumors by a subcutaneous graft experiment. Furthermore, endogenous CSTF2 knockout attenuated hepatocarcinogenesis in an immunocompetent HCC mouse model. Function enrichment analysis suggested that the high expression of CSTF2 was associated with enhanced glycolysis. Moreover, we found that CSTF2 knockout reduced the level of the short 3’ UTR isoform of hexokinase 2 and increased its level of long 3’UTR. Furthermore, CSTF2 knockout inhibited ECAR levels, glucose uptake, and lactate production.ConclusionOur results indicated that CSTF2 is highly expressed in HCC and is correlated with a poor prognosis and high histological grade. The knockout of CSTF2 inhibits the tumorigenesis and procession of HCC both in vitro and in vivo. Moreover, CSTF2 is associated with enhanced glycolysis. Therefore, this study suggests that CSTF2 might be a new prognostic biomarker and therapeutic target for HCC.</p
Data_Sheet_1_hnRNPA2B1 Promotes Colon Cancer Progression via the MAPK Pathway.docx
HNRNPA2B1, an RNA-binding protein, plays a key role in primary microRNA processing, alternative splicing, mRNA metabolism and transport. Interestingly, hnRNPA2B1 also works as an N6-methyladenosine (m6A) reader and is critical during tumorigenesis of various tissue types. However, its role in colon cancer is still unclear. In this study, we aimed to elucidate the biological functions of hnRNPA2B1 and to explore its underlying mechanisms in colon cancer. We examined the expression of hnRNPA2B1 in Oncomine and TCGA databases. Then verified the findings in colon cancer cells and clinical samples with western blotting and immunohistochemistry (IHC). We used CRISPR/Cas9 directed gene editing to knockout hnRNPA2B1 expression in human colon cancer cell line SW480 and HCT-116 and carried out both in vivo and in vitro experiments. The results were further confirmed by RNA-seq analyses. We found that hnRNPA2B1 significantly promoted colon cancer cell proliferation both in vitro and in vivo, while knockout of hnRNPA2B1 induced apoptosis and cell cycle arrest in SW480. RNA-seq analyses revealed that the ERK/MAPK pathway was activated by hnRNPA2B1 upregulation. In addition, both hnRNPA2B1 and MAPK pathway were activated in clinical colon cancer specimens and positively correlated. Mechanistically, hnRNPA2B1 appeared to be an upstream regulator of the ERK/MAPK pathway and inhibition of MAPK signaling blocked the effects of hnRNPA2B1. Taken together, our data demonstrated that the RNA-binding protein hnRNPA2B1 promotes cell proliferation and regulates cell cycle and apoptosis of human colon cancer by activating the ERK/MAPK signaling, which may provide a new insight into the development of hnRNPA2B1 as a potential therapeutic target for treatment of colon cancer.</p
Data_Sheet_1_Prognosis and Immunotherapy Significances of a Cancer-Associated Fibroblasts-Related Gene Signature in Gliomas.docx
As a cold tumor, malignant glioma has strong immunosuppression and immune escape characteristics. The tumor microenvironment (TME) provides the “soil” for the survival of malignant tumors, and cancer-associated fibroblasts (CAFs) are the architects of matrix remodeling in TME. Therefore, CAFs have potent regulatory effects on the recruitment and functional differentiation of immune cells, whereby they synthesize and secrete numerous collagens, cytokines, chemokines, and other soluble factors whose interaction with tumor cells creates an immunosuppressive TME. This consequently facilitates the immune escape of tumor cells. Targeting CAFs would improve the TME and enhance the efficacy of immunotherapy. Thus, regulation of CAFs and CAFs-related genes holds promise as effective immunotherapies for gliomas. Here, by analyzing the Chinese Glioma Genome Atlas and the Cancer Genome Atlas database, the proportion of CAFs in the tumor was revealed to be associated with clinical and immune characteristics of gliomas. Moreover, a risk model based on the expression of CAFs-related six-gene for the assessment of glioma patients was constructed using the least absolute shrinkage and selection operator and the results showed that a high-risk group had a higher expression of the CAFs-related six-genes and lower overall survival rates compared with those in the low-risk group. Additionally, patients in the high-risk group exhibited older age, high tumor grade, isocitrate dehydrogenase wildtype, 1p/19q non-codeletion, O-6-methylguanine-DNA methyltransferase promoter unmethylation and poor prognosis. The high-risk subtype had a high proportion CAFs in the TME of glioma, and a high expression of immune checkpoint genes. Analysis of the Submap algorithm indicated that the high-risk patients could show potent response to anti-PD-1 therapy. The established risk prediction model based on the expression of six CAFs-related genes has application prospects as an independent prognostic indicator and a predictor of the response of patients to immunotherapy.</p
Additional file 7 of Topical administration of the secretome derived from human amniotic epithelial cells ameliorates psoriasis-like skin lesions in mice
Additional file 7. Table S3: A table of normalized fluorescence signal intensity of anti-inflammatory related mediators in the HEF-SC, UMSC-SC, AEC-SC
Additional file 4 of Topical administration of the secretome derived from human amniotic epithelial cells ameliorates psoriasis-like skin lesions in mice
Additional file 4. Fig. S3: Representative images of H&E staining of the back skin of mice in different groups. Scale bar: 50 μm. At least 4 mice were shown in each group
Additional file 8 of Topical administration of the secretome derived from human amniotic epithelial cells ameliorates psoriasis-like skin lesions in mice
Additional file 8. Fig. S5: Immunostaining of lesion skin tissue of mice with human IL-1ra antibody in different groups and the representative images are shown. Scale bar: 50 μm. White arrows represent IL-1ra signals in the dermis in the skin
Additional file 2 of Topical administration of the secretome derived from human amniotic epithelial cells ameliorates psoriasis-like skin lesions in mice
Additional file 2. Fig. S1: Characterization of hAECs and hUMSCs by flow cytometry with antibody against Epcam, CD49f, CD90, CD105, CD73, CD29, CD44, CD31, CD45 and HLA-DR. a hAECs. b hUMSCs
Additional file 3 of Topical administration of the secretome derived from human amniotic epithelial cells ameliorates psoriasis-like skin lesions in mice
Additional file 3. Fig. S2: Phenotypic images of mouse back skin after 6-day IMQ administration and different medium treatment in various groups. CON represents normal control mice. At lease 4 mice were shown in each group
Additional file 1 of Uncovering impaired mitochondrial and lysosomal function in adipose-derived stem cells from obese individuals with altered biological activity
Additional file 1:Figure S1. Lysosomal morphology of L-ADSCs and O-ADSCs from more donors and the representative images are shown. a Immunofluorescence staining of L-ADSCs and O-ADSCs with antibodies against LC3 and LAMP1. Scale bar, 50 μm. b Phagocytosis of L-ADSC and O-ADSCs was measured by phagocytic FITC-Aβ after being treated with FITC-Aβ for 4 hours. Scale bar, 50 μm
Additional file 5 of Topical administration of the secretome derived from human amniotic epithelial cells ameliorates psoriasis-like skin lesions in mice
Additional file 5. Fig. S4: Flow cytometry analysis of lesion skin tissues in IMQ-induced mice with mouse IL 17A and δγ TCR antibodies