Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Clinics (Sao Paulo) ; 79: 100407, 2024.
Article in English | MEDLINE | ID: mdl-38889502

ABSTRACT

BACKGROUND: NSCLC is one of the most common causes of death. The hypoxia microenvironment contributes to cancer progression. The purpose was to explore the effects and mechanism of melittin on NSCLC cells in the hypoxic microenvironment. METHODS: NSCLC cell lines (A549 and H1299) were cultured in normoxia or hypoxia conditions with or without melittin treatment. The viability of the cells was detected via MTT assay and the proliferation ability was evaluated by EdU assay. QRT-PCR was performed to evaluate GLUT1, LDHA, HK2, VEGF and LATS2 mRNA levels. Glucose transport was assessed by the 2-NBDG uptake assay. The angiogenesis was determined by the tubule formation assay. The protein expressions of GLUT1, LDHA, HK2, VEGF, LATS2, YAP, p-YAP and HIF-1α were detected via western blotting assay. The tumor formation assay was conducted to examine the roles of melittin and LATS2 in vivo. RESULTS: Melittin inhibited hypoxia-induced cell viability, proliferation, glycolysis and angiogenesis as well as suppressed YAP binding to HIF-1α in NSCLC. Melittin inactivated the YAP/HIF-1α pathway via up-regulation of LATS2, ultimately inhibiting cancer progression of NSCLC. Moreover, melittin suppressed tumor growth via up-regulation of LATS2 in vivo. CONCLUSION: Melittin inactivated the YAP/HIF-1α pathway via up-regulation of LATS2 to contribute to the development of NSCLC. Therefore, melittin is expected to become a potential prognostic drug for the therapy of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Proliferation , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Lung Neoplasms , Melitten , Neovascularization, Pathologic , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins , Up-Regulation , YAP-Signaling Proteins , Humans , Protein Serine-Threonine Kinases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Up-Regulation/drug effects , Glycolysis/drug effects , Tumor Suppressor Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , YAP-Signaling Proteins/metabolism , Melitten/pharmacology , Melitten/therapeutic use , Cell Line, Tumor , Transcription Factors/metabolism , Animals , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction/drug effects , Cell Survival/drug effects , Phosphoproteins/metabolism , Angiogenesis
2.
FASEB J ; 38(9): e23633, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38690712

ABSTRACT

Recent reports suggest that the Hippo signaling pathway regulates testis development, though its exact roles in Sertoli cell differentiation remain unknown. Here, we examined the functions of the main Hippo pathway kinases, large tumor suppressor homolog kinases 1 and 2 (Lats1 and Lats2) in developing mouse Sertoli cells. Conditional inactivation of Lats1/2 in Sertoli cells resulted in the disorganization and overgrowth of the testis cords, the induction of a testicular inflammatory response and germ cell apoptosis. Stimulated by retinoic acid 8 (STRA8) expression in germ cells additionally suggested that germ cells may have been preparing to enter meiosis prior to their loss. Gene expression analyses of the developing testes of conditional knockout animals further suggested impaired Sertoli cell differentiation, epithelial-to-mesenchymal transition, and the induction of a specific set of genes associated with Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ)-mediated integrin signaling. Finally, the involvement of YAP/TAZ in Sertoli cell differentiation was confirmed by concomitantly inactivating Yap/Taz in Lats1/2 conditional knockout model, which resulted in a partial rescue of the testicular phenotypic changes. Taken together, these results identify Hippo signaling as a crucial pathway for Sertoli cell development and provide novel insight into Sertoli cell fate maintenance.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Protein Serine-Threonine Kinases , Sertoli Cells , Tumor Suppressor Proteins , YAP-Signaling Proteins , Animals , Sertoli Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Male , Mice , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Cell Differentiation/physiology , Mice, Knockout , Signal Transduction , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Testis/metabolism , Epithelial-Mesenchymal Transition/physiology , Transcription Factors/metabolism , Transcription Factors/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics
3.
Mutat Res ; 829: 111858, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38788314

ABSTRACT

BACKGROUND: Ovarian cancer (OC) is a frequent malignancy of the female reproductive system. Recently, the aberrant expression of numerous lncRNAs has been confirmed as a key factor for cancer development. The regulatory role of PAX8-AS1 in some cancers has been investigated, but its role in OC progression remains unclear. This study focuses on the role and molecular mechanism of PAX8-AS1 in the malignant progression of OC. METHODS: Bioinformatics means were adopted to analyze the expression of PAX8-AS1, microRNA-25-3p, and LATS2 in OC tissues and the binding sites between the three. qRT-PCR was employed to determine the expression of these genes in OC cells. CCK-8, colony formation, scratch healing, and Transwell assays were used to see cell viability, proliferation, migration, and invasion, respectively. Fluorescence in situ Hybridization was performed to probe the subcellular localization of PAX8-AS1. Western blot was applied to evaluate the expression and phosphorylation levels of YAP and TAZ, and an immunofluorescence assay was used to detect the translocation of them. Dual luciferase assay was applied to validate the binding relationship between PAX8-AS1 and microRNA-25-3p, as well as between microRNA-25-3p and LATS2. RESULTS: PAX8-AS1 and LATS2 were lowly expressed. MicroRNA-25-3p was highly expressed in OC. PAX8-AS1 was expressed in cytoplasm and regulated LATS2 expression by sponging microRNA-25-3p. Overexpressing PAX8-AS1 can suppress the malignant behaviors of OC cells, whereas treatment with microRNA-mimic can reverse these results. In addition, the phosphorylation levels of YAP and TAZ increased upon oe-LATS2 treatment, and oe-LATS2 could promote YAP and TAZ translocate from the nucleus to cytoplasm. Rescue experiments demonstrated that sh-PAX8-AS1 fostered malignant progression of OC, which was reversed by simultaneous oe-LATS2. CONCLUSION: In summary, PAX8-AS1/microRNA-25-3p/LATS2 regulated the malignant progression of OC through Hippo signaling, which suggested that PAX8-AS1/microRNA-25-3p/LATS2 axis may be a novel target for OC treatment.

4.
J Cell Mol Med ; 28(8): e18216, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38652219

ABSTRACT

We tried to elucidate the possible roles of maternal embryonic leucine pull chain kinase (MELK) in lung adenocarcinoma (LUAD) growth and metastasis. Differentially expressed genes in LUAD samples were analysed by the GEPIA database. Clinical tissue samples and cells were collected for MELK, EZH2 and LATS2 expression determination. Co-IP assay was used to verify the interaction between EZH2 and MELK; CHX tracking assay and ubiquitination assay detected the degradation of MELK on EZH2 ubiquitination. ChIP assay detected the enrichment of EZH2 and H3K27me3 on the LATS2 promoter region. LUAD cells were selected for in vitro validation, and the tumorigenic ability of LUAD cells was also observed in a transplantation tumour model of LUAD nude mice. MELK and EZH2 were highly expressed in LUAD samples, while LATS2 was lowly expressed. MELK interacted with EZH2 to inhibit its ubiquitination degradation; EZH2 elevated H3K27me3 modification in the LATS2 promoter to lower LATS2 expression. Silencing MELK or EZH2 or overexpressing LATS2 restrained LUAD cell proliferation and invasion, and facilitated their apoptosis. Silencing MELK or EZH2 or overexpressing LATS2 suppressed tumour formation in nude mice. This study demonstrated that MELK aggravated LUAD by upregulating EZH2 and downregulating LATS2.


Subject(s)
Adenocarcinoma of Lung , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , Histones , Lung Neoplasms , Mice, Nude , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins , Ubiquitination , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Animals , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Histones/metabolism , Mice , Cell Proliferation/genetics , Methylation , Cell Line, Tumor , Promoter Regions, Genetic/genetics , Apoptosis/genetics , Female , Male
5.
World J Gastrointest Surg ; 16(2): 518-528, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38463354

ABSTRACT

BACKGROUND: Gastric cancer is a leading cause of cancer-related deaths worldwide. Prognostic assessments are typically based on the tumor-node-metastasis (TNM) staging system, which does not account for the molecular heterogeneity of this disease. LATS2, a tumor suppressor gene involved in the Hippo signaling pathway, has been identified as a potential prognostic biomarker in gastric cancer. AIM: To construct and validate a nomogram model that includes LATS2 expression to predict the survival prognosis of advanced gastric cancer patients following radical surgery, and compare its predictive performance with traditional TNM staging. METHODS: A retrospective analysis of 245 advanced gastric cancer patients from the Fourth Hospital of Hebei Medical University was conducted. The patients were divided into a training group (171 patients) and a validation group (74 patients) to develop and test our prognostic model. The performance of the model was determined using C-indices, receiver operating characteristic curves, calibration plots, and decision curves. RESULTS: The model demonstrated a high predictive accuracy with C-indices of 0.829 in the training set and 0.862 in the validation set. Area under the curve values for three-year and five-year survival prediction were significantly robust, suggesting an excellent discrimination ability. Calibration plots confirmed the high concordance between the predictions and actual survival outcomes. CONCLUSION: We developed a nomogram model incorporating LATS2 expression, which significantly outperformed conventional TNM staging in predicting the prognosis of advanced gastric cancer patients postsurgery. This model may serve as a valuable tool for individualized patient management, allowing for more accurate stratification and improved clinical outcomes. Further validation in larger patient cohorts will be necessary to establish its generalizability and clinical utility.

6.
Biochim Biophys Acta Gen Subj ; 1868(5): 130592, 2024 May.
Article in English | MEDLINE | ID: mdl-38395204

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) cell-intrinsic programmed death 1 (PD-1) promotes tumor progression. However, the mechanisms that regulate its expression are unclear. This study investigated the impact of alpha-fetoprotein (AFP) on HCC cell-intrinsic PD-1 expression. METHODS: The expression of PD-1 and AFP at the gene and protein levels was detected using real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) and western blotting (WB). Proteins interacting with AFP were examined by co-immunoprecipitation (CO-IP). Chromatin immunoprecipitation (ChIP) and dual luciferase reporter assays were used to identify transcription-enhanced association domain 1 (TEAD1) binding to the promoter of PD-1. RESULTS: The expression of HCC cell-intrinsic PD-1 was positively correlated with AFP. Mechanistically, AFP inhibited the phosphorylation of large tumor suppressor 2 (LATS2) and yes-associated protein (YAP). As a result, YAP is transferred to the nucleus and forms a transcriptional complex with TEAD1, promoting PD-1 transcription by binding to its promoter. CONCLUSION: AFP is an upstream regulator of the HCC cell-intrinsic PD-1 and increases PD-1 expression via the LATS2/YAP/TEAD1 axis. GENERAL: Our findings provide insight into the mechanisms of HCC development and offer new ideas for further in-depth studies of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , alpha-Fetoproteins/metabolism , Liver Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , TEA Domain Transcription Factors
7.
J Mol Histol ; 54(6): 593-607, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37861952

ABSTRACT

Circular RNAs (circRNAs) play critical roles in the initiation and progression of various cancers. However, the potential functional roles of circSNX14 in hepatocellular carcinoma (HCC) remain largely unknown. CircSNX14 expression pattern was analyzed in HCC tissues and cell lines via qRT-PCR. The effects of circSNX14 on cell proliferation, invasion, angiogenesis, and Epithelial-mesenchymal transition (EMT) were investigated by overexpression experiments. The role of circSNX14 in the tumorigenesis of HCC cells was examined using in vivo xenograft mouse model. The interaction between circSNX14, miR-562, and Large Tumor Suppressor Kinase 2 (LATS2) mRNA was confirmed by Luciferase reporter assay and RNA immunoprecipitation (RIP) analysis. CircSNX14 was significantly down-regulated in HCC tissues and cell lines, and its down-regulation was correlated with a poor prognosis in HCC patients. In the following functional experiments, circSNX14 overexpression remarkably suppressed the proliferation and invasion of HCC cells, and attenuated the mesenchymall status. circSNX14 overexpression also suppressed the tumorigenesis of HCC cells in the mouse model. We further revealed the interaction of circSNX14 and miR-562, and miR-562 could suppress the expression of LATS2 by interacting with its mRNA. The negative correlation of circSNX14 and miR-562, negative correlation of miR-562 and LATS2, and positive correlation of circSNX14 and LATS2 have been confirmed by Pearson correlation in the HCC samples. Collectively, these results reveal a novel role of circSNX14/miR-562/LATS2 axis in regulating the malignant progression of HCC cancer progression, indicating the tumor suppressor role of circSNX14 and its potential as a prognostic biomarker.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Humans , Animals , Mice , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , RNA, Messenger , Carcinogenesis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Int J Mol Sci ; 24(20)2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37894939

ABSTRACT

The Hippo pathway mediates renal maladaptive repair after acute kidney injury (AKI), which has been considered a driving force in the progression to chronic kidney disease (CKD). LATS2, a core kinase of the Hippo pathway, exerts non-Hippo-dependent functions in the regulation of the cell cycle and cell fate, providing new insights into AKI and further repair. However, its role remains unknown. Here, we utilized a proximal tubular Lats2 conditional knockout mouse strain (Lats2-CKO) to evaluate the effect of LATS2 deficiency on ischemia/reperfusion-induced AKI-to-CKD transition. Lats2-CKO mice presented with more severe tubular maladaptive repair, inflammatory infiltration, interstitial fibrosis, and apoptosis following AKI. Importantly, we discovered that Lats2 ablation caused the activation of p53, with increased levels of cellular apoptotic molecules (p21, Bax, and cleaved caspase-3), and decreased levels of anti-apoptotic molecules (Bcl-2 and Bcl-xL). Pifithirin-α (p53 inhibitor) effectively attenuated renal fibrosis, inflammation, and apoptosis in Lats2-CKO mice after AKI. Consistently, in vitro Lats2 overexpression decreased p53, p21, Bax and cleaved caspase 3 expression after hypoxia/reoxygenation (H/R) treatment. Of note, the phosphorylation of MDM2, which promotes the ubiquitination degradation of p53, at site Ser186 was decreased in Lats2-CKO kidneys, but increased by Lats2 overexpression in vitro. Therefore, LATS2 deficiency aggravated ischemia/reperfusion injury (IRI)-induced maladaptive repair via regulating the tubular MDM2-p53 axis in AKI-to-CKD transition.


Subject(s)
Acute Kidney Injury , Renal Insufficiency, Chronic , Reperfusion Injury , Animals , Mice , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Apoptosis , bcl-2-Associated X Protein/metabolism , Kidney/metabolism , Mice, Inbred C57BL , Renal Insufficiency, Chronic/metabolism , Reperfusion Injury/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
9.
Diagnostics (Basel) ; 13(18)2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37761387

ABSTRACT

Cryptorchidism (CO) is a risk factor for the development of testicular germ-cell tumors (TGCT). This is supported by reports showing the persistence of gonocytes in CO patients. These cells are proposed to be related to the development of germ-cell neoplasia in situ (GCNIS), which is considered the precursor stage/lesion of TGCT. Therefore, it is proposed that some patients with CO could express some molecular markers related to TGCT. In this study, we analyzed testicular tissue samples from CO, TGCT, and controls. We determined the expression of POU5F1, PLAP, and KIT by immunohistochemistry and that of the hsa-miR-371-373 cluster, hsa-miR-367, and LATS2, PTEN, and IGFR1 genes by RT-qPCR. We then carried out a bioinformatic analysis to identify other possible candidate genes as tumor biomarkers. We found that 16.7% (2/12) of the CO patients presented increased expression of POU5F1, KIT, PLAP, hsa-miR-371-373, and hsa-miR-367 and decreased expression of LATS2 and IGF1R. Finally, the genes ARID4B, GALNT3, and KPNA6 were identified as other possible candidate tumor biomarkers. This is the first report describing the expression of the hsa-miR-371-373 cluster, hsa-miR-367, LATS2, and IGF1R in the testicular tissues of two CO patients with cells immune-positive to POU5F1, PLAP, and KIT, which is similar to what is observed in TGCT.

10.
Int J Biol Sci ; 19(11): 3428-3440, 2023.
Article in English | MEDLINE | ID: mdl-37497006

ABSTRACT

Large tumor suppressor kinase 2 (Lats2) is a member of the Hippo pathway, a critical regulator of organ size. Since Lats2 activity may trigger mitochondrial dysfunction, a key pathogenic factor in acute myocardial infarction (AMI), this study sought to investigate whether Lats2 deletion confers cardioprotection in AMI. AMI was induced in cardiomyocyte-specific Lats2 knockout (Lats2Cko) and control (Lats2flox) mice. Twenty-eight days after AMI surgery, myocardial performance and mitochondrial homeostasis were impaired in Lats2floxmice. In contrast, Lats2Cko mice exhibited markedly preserved cardiac structure and contraction/relaxation activity, decreased fibrosis, reduced circulating cardiac injury biomarker levels, and enhanced cardiomyocyte viability. Consistent with these findings, siRNA-mediated Lats2 silencing sustained mitochondrial respiration and inhibited apoptosis in hypoxia-treated HL-1 cardiomyocytes. Notably, Lats2 deficiency inhibited AMI/hypoxia-related mitochondrial fission and inactivated STING/p65 signaling by preventing hypoxia-induced release of mtDNA into the cytosol. Accordingly, pharmacological reactivation of STING signaling abolished the cardioprotective effects of Lats2 ablation. Those data suggest that AMI-induced Lats2 upregulation is associated with impaired cardiomyocyte viability and function resulting from enhanced mitochondrial fission, mtDNA release, and STING/p65 pathway activation.


Subject(s)
Mitochondrial Dynamics , Myocardial Infarction , Animals , Mice , Apoptosis/genetics , DNA, Mitochondrial/metabolism , Hypoxia/metabolism , Inflammation/metabolism , Mitochondrial Dynamics/genetics , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism
11.
Genes (Basel) ; 14(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-37239394

ABSTRACT

Progressive corneal opacification can result from multiple etiologies, including corneal dystrophies or systemic and genetic diseases. We describe a novel syndrome featuring progressive epithelial and anterior stromal opacification in a brother and sister and their mildly affected father, with all three family members having sensorineural hearing loss and two also with tracheomalacia/laryngomalacia. All carried a 1.2 Mb deletion at chromosome 13q12.11, with no other noteworthy co-segregating variants identified on clinical exome or chromosomal microarray. RNAseq analysis from an affected corneal epithelial sample from the proband's brother revealed downregulation of XPO4, IFT88, ZDHHC20, LATS2, SAP18, and EEF1AKMT1 within the microdeletion interval, with no notable effect on the expression of nearby genes. Pathway analysis showed upregulation of collagen metabolism and extracellular matrix (ECM) formation/maintenance, with no significantly down-regulated pathways. Analysis of overlapping deletions/variants demonstrated that deleterious variants in XPO4 were found in patients with laryngomalacia and sensorineural hearing loss, with the latter phenotype also being a feature of variants in the partially overlapping DFNB1 locus, yet none of these had reported corneal phenotypes. Together, these data define a novel microdeletion-associated syndromic progressive corneal opacification and suggest that a combination of genes within the microdeletion may contribute to ECM dysregulation leading to pathogenesis.


Subject(s)
Hearing Loss, Sensorineural , Laryngomalacia , Male , Female , Humans , Hearing Loss, Sensorineural/genetics , Syndrome , Siblings , Microarray Analysis , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins
12.
Arch Biochem Biophys ; 742: 109640, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37207933

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the most lethal solid tumors in China, with the 5-year overall survival rate less than 20%. Although the carcinogenic process of ESCC is still not clear, recent studies using whole genomic profiling revealed that dysregulation of Hippo signaling pathway might play important roles in ESCC progression. The ubiquitin-like with PHD and RING finger domain 1 (RNF106) was a modifier of DNA methylation and histone ubiquitination. In this study, we evaluate the oncogenic function of RNF106 in ESCC both in vitro and in vivo. Wound healing and transwell data showed that RNF106 was required for ESCC cell migration and invasion. RNF106 depletion dramatically restrained Hippo signaling targeted gene expression. The bioinformatics analysis displayed that RNF106 was increased in ESCC tumor tissues and related with poor survival in ESCC patients. Mechanistic studies demonstrated that RNF106 was associated with LATS2 and facilitate LATS2 K48-linked ubiquitination and degradation, which subsequently inhibited YAP phosphorylation and promoted YAP oncogenic function in ESCC. Taken together, our study revealed a novel link between RNF106 and Hippo signaling in ESCC, suggesting that RNF106 could be a promising target for ESCC therapy.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
Neurosci Lett ; 801: 137126, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36796622

ABSTRACT

Glioma is the most prevalent brain tumor with a poor prognosis. Circular RNA (circ) (PKD2) has been identified as a potential tumor suppressor. However, the effect of circPKD2 on glioma has been unknown. circPKD2 expression in glioma and its potential targets were analyzed by bioinformatics methods, qRT-PCR, dual luciferase reporter, RNA-pull down and RNA immunoprecipitation assays. Overall survival was analyzed by Kaplan-Meier method. The correlation of circPKD2 expression with patient's clinical characteristics was assessed by Chi-square test. Glioma cell invasion was detected by Transwell invasion assay, and cell proliferation was determined by CCK8 and EdU assays. ATP level, Lactate production, and glucose consumption were measured by commercial assay kits, and glycolysis-related protein (Ki-67, VEGF, HK2, LDHA) levels were evaluated by western blot. circPKD2 expression was downregulated in glioma, but circPKD2 overexpression inhibited the cell proliferation, invasion, and glycolytic metabolism. Besides, patients with low circPKD2 expression had a worse prognosis. circPKD2 level was correlated with distant metastasis, WHO grade, and Karnofsky, KPS score. circPKD2 acted as a sponge of miR-1278, and LATS2 was a target gene of miR-1278. Moreover, circPKD2 could target miR-1278 to up-regulate LATS2 expression to suppress the cell proliferation, invasion, and glycolytic metabolism. These findings display that circPKD2 can function as a tumor suppressor in glioma by controlling the miR-1278/LATS2 axis and provide the potential biomarkers for glioma treatment.


Subject(s)
Glioma , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Glioma/metabolism , Cell Proliferation/genetics , Glycolysis , Cell Movement , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
14.
Dig Dis Sci ; 68(5): 1913-1922, 2023 05.
Article in English | MEDLINE | ID: mdl-36515852

ABSTRACT

BACKGROUND: Dysregulation of ubiquitin ligases plays a crucial role in the development and progression of various human tumors. F-box only protein 22 (FBXO22), an F-box E3 ubiquitin ligase, has been reported to participate in diverse aspects of cancer progression. However, the clinical significance and biological function of FBXO22 in pancreatic cancer remain poorly understood. AIMS: This study aimed to investigate the role of FBXO22 in promoting pancreatic cancer growth. METHODS: FBXO22 expression was detected in pancreatic cancer and adjacent normal tissues using qRT-PCR, western blotting, and immunohistochemistry. Ectopic expression and knockdown of FBXO22 were performed to measure the impact on pancreatic cancer cells growth by CCK-8, colony formation, and tumorigenicity assay. Bioinformatics analysis uncovered the potential correlation between FBXO22 and various signaling pathways. Western blotting and immunoprecipitation were performed to identify FBXO22-interacting proteins. RESULTS: We observed that FBXO22 was upregulated in samples obtained from patients with pancreatic cancer compared with its levels in the adjacent normal tissues, and an elevated FBXO22 level was obviously associated with poor prognosis among patients with pancreatic cancer. FBXO22 knockdown impaired pancreatic cancer cell growth both in vitro and in vivo, whereas FBXO22 overexpression accelerated pancreatic cancer cell growth. Furthermore, we found that FBXO22 contributed to pancreatic cancer cell growth by deactivating the Hippo pathway. Mechanistically, FBXO22 directly interacts with and destabilizes the large tumor suppressor 2 (LATS2), which is a critical regulator of the Hippo pathway. Blocking LATS2 leads to the loss of FBXO22-mediated oncogenic effect in pancreatic cancer. CONCLUSIONS: These findings provide new insights into the upstream regulation of the Hippo pathway inactivation in pancreatic cancer growth and identify FBXO22 as a potential therapeutic target for this lethal malignant tumor.


Subject(s)
F-Box Proteins , Pancreatic Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , F-Box Proteins/genetics , F-Box Proteins/metabolism , Hippo Signaling Pathway , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
Anticancer Res ; 43(1): 45-51, 2023 01.
Article in English | MEDLINE | ID: mdl-36585159

ABSTRACT

BACKGROUND/AIM: Kidney and brain expressed protein (KIBRA), a member of the WW domain-containing protein family, has an important role in tumour growth and progression in various cancers. However, the pathological significance of KIBRA expression in clear cell renal cell carcinoma (ccRCC) tissues is not fully understood. The aim of this study was to clarify the pathological significance and prognostic roles of KIBRA expression in patients with ccRCC. MATERIALS AND METHODS: KIBRA immunoreactivity, proliferation index (PI; with anti-Ki-67 antibody), apoptotic index (AI; using anti-cleaved caspase-3), and large tumour suppressor kinases (LATS-2) were evaluated in 157 ccRCC specimens by immunohistochemistry. Fifty normal kidney tissues were also evaluated as controls. The relationships between KIBRA expression and these cancer-related variables as well as clinicopathological features and survival were analysed. RESULTS: Moderate to strong immunoreactivity of KIBRA was identified in all normal kidney tissues; however, ccRCC cells with strong KIBRA expression was rare. The immunoreactivity score (IRS) of KIBRA was negatively associated with grade, T stage, tumour diameter, and metastasis. Kaplan-Meier survival curves showed that high KIBRA expression was a favourite predictor for overall survival. KIBRA IRS was negatively associated with PI and positively associated with the IRS of LATS-2 by univariate analysis. In addition, multivariate analysis showed that KIBRA was significantly associated with PI. CONCLUSION: KIBRA demonstrated important roles as a tumour suppressor in ccRCC. In addition, its expression was significantly associated with survival in these patients. Several such KIBRA-related functions were speculated to be modulated by cancer cell proliferation and LATS-2.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Kidney/pathology , Prognosis , Kaplan-Meier Estimate , Brain/pathology , Biomarkers, Tumor/metabolism
16.
Res Vet Sci ; 152: 417-426, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36126508

ABSTRACT

As an important downstream effector gene in the hippo signaling pathway, large tumor suppressor gene 2 (LATS2) is involved in cell proliferation and differentiation, organ size and tissue regeneration, and plays an important role in regulating the growth and development of animal muscles. The purpose of this study is to explore the temporal expression of bovine LATS2 gene, and determine the key transcription factors for regulating bovine LATS2 gene. The result showed that bovine LATS2 gene was highly expressed in liver and longissimus dorsi, and was up-regulated in infancy muscle. In addition, it was highly expressed on the 2th day during the differentiation stage of myoblast. The upstream 1.7 Kb sequence of the 5 'translation region of bovine LATS2 gene was cloned, and 7 different deletion fragments were amplified by the upstream primers. These fragments were constructed into double luciferase reporter vectors and transfected into myoblasts and myotubes cells, respectively to detect the core promoter regions. In addition, the key transcription factors of the core promoter sequence of the bovine LATS2 gene were analyzed and predicted by online software. Combining with site-directed mutations, siRNA interference and chromatin immunoprecipitation technology, it was identified that MEF2A and MyoG combined in core promoter region (-248/-56) to regulate the transcription activity of bovine LATS2 gene. The results have laid a theoretical foundation for exploring the molecular regulation mechanism of LATS2 gene in the process of muscle growth.


Subject(s)
Gene Expression Regulation , Transcription Factors , Cattle , Animals , Transcription Factors/metabolism , Cell Proliferation , Promoter Regions, Genetic , Cell Differentiation
17.
Front Genet ; 13: 952528, 2022.
Article in English | MEDLINE | ID: mdl-36118851

ABSTRACT

According to the TIMER database, large tumor suppressor 2 (LATS2) is differentially expressed in various tumors. However, the correlation between LATS2 and esophageal squamous cell carcinoma (ESCC) and the association between LATS2 and immune infiltration in ESCC remain unclear. Our synthetic research on LATS2 in ESCC revealed that the expression was low in esophageal squamous epithelium tissues, revealing the pernicious and adverse prognosis of ESCC. The Kaplan-Meier survival investigation pointed out that low LATS2 expression would result in an adverse prognosis. Biological investigation indicated that LATS2 was engaged in cell migration, adhesion, and junction. To further explore the relationship between LATS2 and tumor immunity, we utilized CIBERSORT to assess immune infiltration. The findings revealed that specimens with lower LATS2 expression showed higher immune infiltration, including T-cell follicular helper cells, M0 macrophages, M1 macrophages, and myeloid dendritic cell resting. An association investigation indicated that LATS2 was negatively relevant to immune checkpoints that restrain operative antitumor immune reactions. We also conducted immunohistochemical staining to explore the link between LATS2 expression and immunophenotype. The indicated association between low LATS2 expression and an immunophenotype is conducive to our understanding of ESCC mini-environments and might offer new indications for enhancing new therapeutic targets.

18.
Development ; 149(18)2022 09 15.
Article in English | MEDLINE | ID: mdl-36125128

ABSTRACT

Hippo signaling, an evolutionarily conserved kinase cascade involved in organ size control, plays key roles in various tissue developmental processes, but its role in craniofacial development remains poorly understood. Using the transgenic Wnt1-Cre2 driver, we inactivated the Hippo signaling components Lats1 and Lats2 in the cranial neuroepithelium of mouse embryos and found that the double conditional knockout (DCKO) of Lats1/2 resulted in neural tube and craniofacial defects. Lats1/2 DCKO mutant embryos had microcephaly with delayed and defective neural tube closure. Furthermore, neuroepithelial cell shape and architecture were disrupted within the cranial neural tube in Lats1/2 DCKO mutants. RNA sequencing of embryonic neural tubes revealed increased TGFB signaling in Lats1/2 DCKO mutants. Moreover, markers of epithelial-to-mesenchymal transition (EMT) were upregulated in the cranial neural tube. Inactivation of Hippo signaling downstream effectors, Yap and Taz, suppressed neuroepithelial defects, aberrant EMT and TGFB upregulation in Lats1/2 DCKO embryos, indicating that LATS1/2 function via YAP and TAZ. Our findings reveal important roles for Hippo signaling in modulating TGFB signaling during neural crest EMT.


Subject(s)
Cell Cycle Proteins , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Mice , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Skull , Transforming Growth Factor beta/metabolism
19.
Mol Ther ; 30(12): 3714-3728, 2022 12 07.
Article in English | MEDLINE | ID: mdl-35923112

ABSTRACT

Activation of hepatic stellate cells (HSCs) is a central driver of liver fibrosis. Previous investigations have identified various altered epigenetic landscapes during the cellular progression of HSC activation. N6-methyladenosine (m6A) is the most abundant internal RNA modification in eukaryotic cells and is dynamically regulated under various physiological and pathophysiological conditions. However, the functional role of Mettl3-mediated m6A in liver fibrosis remains elusive. Here, we found that the HSC-specific knockout of m6A methyltransferase Mettl3 suppressed HSC activation and significantly alleviated liver fibrosis. Multi-omics analysis of HSCs showed that Mettl3 depletion reduced m6A deposition on mRNA transcripts of Lats2 (a central player of the Hippo/YAP signaling pathway) and slowed down their degradation. Elevated Lats2 increased phosphorylation of the downstream transcription factor YAP, suppressed YAP nuclear translocation, and decreased pro-fibrotic gene expression. Overexpressing YAP mutant resistant to phosphorylation by Lats2 partially rescued the activation and pro-fibrotic gene expression of Mettl3-deficient HSCs. Our study revealed that disruption of Mettl3 in HSCs mitigated liver fibrosis by controlling the Hippo/YAP signaling pathway, providing potential therapeutic strategies to alleviate liver fibrosis by targeting epitranscriptomic machinery.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , Methyltransferases , Liver Cirrhosis/genetics , Methyltransferases/deficiency , Methyltransferases/genetics , Multiomics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins , Animals , Mice
20.
Med Oncol ; 39(5): 97, 2022 May 23.
Article in English | MEDLINE | ID: mdl-35599283

ABSTRACT

Myeloproliferative neoplasms (MPN) are hematological disorders characterized by increased proliferation of precursor and mature myeloid cells. MPN patients may present driver mutations in JAK2, MPL, and CALR genes, which are essential to describe the molecular mechanisms of MPN pathogenesis. Despite all the new knowledge on MPN pathogenesis, many questions remain to be answered to develop effective therapies to cure MPN or impair its progression to acute myeloid leukemia. The present study examined the expression levels of the Hippo signaling pathway members in patients with polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), as well as the role that they play in disease pathogenesis. The Hippo pathway is a tumor suppressor pathway that participates in the regulation of cell proliferation, differentiation, and death. Our main finding was that the expression of tumor suppressor genes from Hippo pathway were downregulated and seemed to be associated with cell resistance to apoptosis and increased proliferation rate. Therefore, the decreased expression of Hippo pathway-related genes may contribute to the malignant phenotype, apoptosis resistance, and cell proliferation in MPN pathogenesis.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Primary Myelofibrosis , Calreticulin/genetics , Hippo Signaling Pathway , Humans , Janus Kinase 2/genetics , Mutation , Myeloproliferative Disorders/genetics , Polycythemia Vera/genetics , Receptors, Thrombopoietin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...