Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119741, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697304

ABSTRACT

Prostate cancer (PCa) is the second leading cause of death in males. It has been reported that δ-catenin expression is upregulated during the late stage of prostate cancer. Palmitoylation promotes protein transport to the cytomembrane and regulates protein localization and function. However, the effect of δ-catenin palmitoylation on the regulation of cancer remains unknown. In this study, we utilized prostate cancer cells overexpressing mutant δ-catenin (J6A cells) to induce a depalmitoylation phenotype and investigate its effect on prostate cancer. Our results indicated that depalmitoylation of δ-catenin not only reduced its membrane expression but also promoted its degradation in the cytoplasm, resulting in a decrease in the effect of EGFR and E-cadherin signaling. Consequently, depalmitoylation of δ-catenin reduced the proliferation and metastasis of prostate cancer cells. Our findings provide novel insights into potential therapeutic strategies for controlling the progression of prostate cancer through palmitoylation-based targeting of δ-catenin.


Subject(s)
Cadherins , Catenins , Cell Proliferation , Delta Catenin , Disease Progression , Lipoylation , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Catenins/metabolism , Catenins/genetics , Cell Line, Tumor , Cadherins/metabolism , Cadherins/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Signal Transduction , Animals , Cell Movement , Gene Expression Regulation, Neoplastic
2.
Clin Genet ; 106(2): 180-186, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38604781

ABSTRACT

CTNND2 encodes δ-catenin, a component of an adherens junction complex, and plays an important role in neuronal structure and function. To date, only heterozygous loss-of-function CTNND2 variants have been associated with mild neurodevelopmental delay and behavioral anomalies, a condition, which we named Rauch-Azzarello syndrome. Here, we report three siblings of a consanguineous family of Syrian descent with a homozygous deletion encompassing the last 19 exons of CTNND2 predicted to disrupt the transcript. All presented with severe neurodevelopmental delay with absent speech, profound motor delay, stereotypic behavior, microcephaly, short stature, muscular hypotonia with lower limb hypertonia, and variable eye anomalies. The parents and the fourth sibling were heterozygous carriers of the deletion and exhibited mild neurodevelopmental impairment resembling that of the previously described heterozygous individuals. The present study unveils a severe manifestation of CTNND2-associated Rauch-Azzarello syndrome attributed to biallelic loss-of-function aberrations, clinically distinct from the already described mild presentation of heterozygous individuals. Furthermore, we demonstrate novel clinical features in homozygous individuals that have not been reported in heterozygous cases to date.


Subject(s)
Delta Catenin , Neurodevelopmental Disorders , Child , Child, Preschool , Female , Humans , Infant , Male , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Alleles , Catenins/genetics , Consanguinity , Homozygote , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Pedigree , Phenotype , Sequence Deletion/genetics
3.
Neuropharmacology ; 253: 109963, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38657945

ABSTRACT

Valproic acid (VPA) is an effective and commonly prescribed drug for epilepsy and bipolar disorder. However, children born from mothers treated with VPA during pregnancy exhibit an increased incidence of autism spectrum disorder (ASD). Although VPA may impair brain development at the cellular level, the mechanism of VPA-induced ASD has not been completely addressed. A previous study has found that VPA treatment strongly reduces δ-catenin mRNA levels in cultured human neurons. δ-catenin is important for the control of glutamatergic synapses and is strongly associated with ASD. VPA inhibits dendritic morphogenesis in developing neurons, an effect that is also found in neurons lacking δ-catenin expression. We thus hypothesize that prenatal exposure to VPA significantly reduces δ-catenin levels in the brain, which impairs glutamatergic synapses to cause ASD. Here, we found that prenatal exposure to VPA markedly reduced δ-catenin levels in the brain of mouse pups. VPA treatment also impaired dendritic branching in developing mouse cortical neurons, which was partially reversed by elevating δ-catenin expression. Prenatal VPA exposure significantly reduced synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor levels and postsynaptic density 95 (PSD95) in the brain of mouse pups, indicating dysfunctions in glutamatergic synaptic transmission. VPA exposure also significantly altered ultrasonic vocalization (USV) in newly born pups when they were isolated from their nest. Moreover, VPA-exposed pups show impaired hypothalamic response to isolation, which is required to produce animals' USVs following isolation from the nest. Therefore, these results suggest that VPA-induced ASD pathology can be mediated by the loss of δ-catenin functions.


Subject(s)
Animals, Newborn , Prenatal Exposure Delayed Effects , Valproic Acid , Vocalization, Animal , Animals , Valproic Acid/pharmacology , Valproic Acid/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Female , Vocalization, Animal/drug effects , Vocalization, Animal/physiology , Mice , Synapses/drug effects , Synapses/metabolism , Catenins/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Receptors, AMPA/metabolism , Brain/drug effects , Brain/metabolism , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/metabolism
4.
Am J Cancer Res ; 12(10): 4853-4864, 2022.
Article in English | MEDLINE | ID: mdl-36381334

ABSTRACT

δ-Catenin is expressed abundantly in various human cancers, including prostate, brain, breast, and lung carcinomas, and is recognized as an oncogene that promotes cancer cell growth and tumorigenesis. Although several transcriptional and post-translational pathways for δ-catenin regulation have been identified in cancer cells, the potential effects of microRNA-mediated regulation remain elusive. Here, we used a δ-catenin 3'-UTR luciferase reporter assay to identify regulatory microRNAs. Subsequent bioinformatics analyses and molecular studies revealed that overexpression of miR-122 downregulated δ-catenin expression significantly via targeted binding to a seed sequence in the 3'-UTR region of δ-catenin, and suppressed the invasion, migration, and proliferation of prostate cancer cells in vitro. In a TRAMP-C2 mouse syngeneic prostate tumor model, stable expression of miR-122 decreased both δ-catenin expression and tumor growth. Mechanistically, overexpression of miR-122 inhibited the expression of δ-catenin-mediated downstream factors significantly in prostate cancer cells, including c-myc and cyclin D1. In cells overexpressing miR-122, there was no additive or synergistic effect of siRNA-mediated knockdown of δ-catenin on cell invasiveness, and overexpression of miR-122 alone had a more pronounced suppressive effect on cell invasion than knockdown of δ-catenin alone. These results suggest that miR-122 acts as tumor suppressor in prostate cancer, mainly by downregulating δ-catenin expression, but also by targeting other factors. Indeed, subsequent experiments showed that overexpression of miR-122 reduced the levels of the mRNAs encoding myc, snail, and VEGF in prostate cancer cells. Overall, our findings demonstrate that targeting of δ-catenin by miR-122 represses the motility and tumorigenesis of prostate cancer cells, indicating a tumor suppressive effect of this miRNA in prostate cancer.

5.
Mol Cancer ; 21(1): 141, 2022 07 02.
Article in English | MEDLINE | ID: mdl-35780119

ABSTRACT

BACKGROUND: Increasing studies suggest that circular RNAs (circRNAs) are critical regulators of cancer development and progression. However, the biological roles and mechanisms of circRNAs in gastric cancer (GC) remain largely unknown. METHODS: We identified the differentially expressed circRNAs in GC by analyzing Gene Expression Omnibus (GEO) datasets. We explored the biological roles of circRNAs in GC by in vitro functional assays and in vivo animal studies. We performed tagged RNA affinity purification (TRAP), RNA immunoprecipitation (RIP), mass spectrometry (MS), RNA sequencing, luciferase reporter assays, and rescue experiments to investigate the mechanism of circRNAs in GC. RESULTS: Downregulated expression of circular RNA EIF4G3 (circEIF4G3; hsa_circ_0007991) was found in GC and was associated with poor clinical outcomes. Overexpression of circEIF4G3 suppressed GC growth and metastasis through the inhibition of ß-catenin signaling, whereas knockdown of circEIF4G3 showed the opposite effects. Mechanistic studies revealed that circEIF4G3 bound to δ-catenin protein to promote its TRIM25-mediated ubiquitin degradation and interacted with miR-4449 to upregulate SIK1 expression. CONCLUSION: Our findings uncovered a tumor suppressor function of circEIF4G3 in GC through the regulation of δ-catenin protein stability and miR-4449/SIK1 axis. CircEIF4G3 may act as a promising prognostic biomarker and therapeutic target for GC.


Subject(s)
MicroRNAs , Stomach Neoplasms , Animals , Catenins , Cell Line, Tumor , Cell Proliferation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , Stomach Neoplasms/pathology , Ubiquitin , beta Catenin/genetics , Delta Catenin
6.
Int J Stem Cells ; 15(3): 324-333, 2022 Aug 30.
Article in English | MEDLINE | ID: mdl-35769053

ABSTRACT

Background and Objectives: This study was to investigate the role of microRNA-29a-3p (miR-29a-3p) in human bone marrow mesenchymal stem cells (hBMSCs), and its relationship with steroid-associated osteonecrosis. Methods and Results: The online tool GEO2R was used to screen out the differentially expressed genes (DEGs) in GSE123568 dataset. Quantitative real time-polymerase chain reaction (qRT-PCR) was performed to detect the expression of miR-29a-3p, forkhead box O3 (FOXO3), alkaline phosphatase (ALP), bone gamma-carboxyglutamate protein (OCN) and RUNX family transcription factor 2 (Runx2) in the hBMSCs isolated from the patients with steroid- associated osteonecrosis. CCK-8 assay was executed to measure cell viability; western blot assay was utilized to detect FOXO3, ALP, Runx2, OCN and ß-catenin expression. Cell apoptosis and cell cycle were detected by flow cytometry. Immunofluorescence assay was used to detect the sub-cellular localization of ß-catenin. Bioinformatics analysis and luciferase reporter gene assay were performed to confirm whether miR-29a-3p can combine with FOXO3 3'UTR. MiR-29a-3p was markedly up-regulated in the hBMSCs of patients with steroid-associated osteonecrosis, while FOXO3 mRNA was significantly down-regulated. Transfection of miR-29a-3p mimics significantly inhibited the hBMSCs' proliferation, osteogenic differentiation markers' expressions, including ALP, Runx2, OCN, and repressed the ALP activity, as well as promoted cell apoptosis and cell-cycle arrest. FOXO3 was identified as a target gene of miR-29a-3p, and miR-29a-3p can inhibit the expression of FOXO3 and ß-catenin, and inhibition of miR-29a-3p promoted translocation of ß-catenin to the nucleus. Conclusions: MiR-29a-3p can modulate FOXO3 expression and Wnt/ß-catenin signaling to inhibit viability and osteogenic differentiation of hBMSCs, thereby promoting the development of steroid-associated osteonecrosis.

7.
Am J Cancer Res ; 12(1): 108-122, 2022.
Article in English | MEDLINE | ID: mdl-35141007

ABSTRACT

As a member of the catenin family, δ-catenin is overexpressed in many cancers, including prostate cancer, and the role of δ-catenin in prostate tumor growth has been reported. However, the involvement of δ-catenin in the migration and invasion of prostate cancer has rarely been studied. In this study, we innovatively proposed that δ-catenin would enhance the migration and invasion ability of prostate cancer cells. It is worth noting that the molecular mechanism underlying the effect involved the downregulation of autophagy. We demonstrated that δ-catenin could suppress autophagy by Bcl-2-regulated disruption of the Beclin1-Vps34 autophagosome complex. Furthermore, the effect of δ-catenin on promoting cell migration and invasion was dependent upon ß-catenin-mediated Bcl-2 transcription. Finally, using rapamycin and bafilomycin, we largely confirmed that the degradation of Snails by autolysosomes may be related to δ-catenin regulated migration and invasion. Overall, our results indicated that δ-catenin promoted cell migration and invasion of prostate cancer cells via Bcl-2-regulated autophagy suppression.

8.
Am J Cancer Res ; 11(8): 3877-3892, 2021.
Article in English | MEDLINE | ID: mdl-34522455

ABSTRACT

Recently, we have shown that δ-catenin strengthened the epidermal growth factor receptor (EGFR)/Erk1/2 signaling pathway through the association between EGFR and δ-catenin. Now, we further analyzed the correlation between basic fibroblast growth factor (bFGF)/fibroblast growth factor receptor 1 (FGFR1) and δ-catenin in prostate cancer and investigated the molecular mechanism underlying the role of bFGF/FGFR1 modulation in CWR22Rv-1 (Rv-1) cells. Here, we demonstrated that bFGF phosphorylated the tyrosine residues of δ-catenin in Rv-1 cells and further proved that the bFGF mediated FGFR1/δ-catenin tyrosine phosphorylation was time dependent. Furthermore, we demonstrated that bFGF stabilized the expression of δ-catenin through weakening its association with GSK3ß and enhancing its stability to induce ß-catenin into the nuclear by strengthening the processing of E-cadherin. In a word, these results indicated that bFGF/FGFR1 signaling pathway could enhance the tumor progression of prostate cancer via δ-catenin.

9.
Int J Stem Cells ; 14(4): 434-446, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34456188

ABSTRACT

BACKGROUND AND OBJECTIVES: MiR-148a-3p has been reported to regulate the differentiation of marrow stromal cell osteoblast. In this study, whether miR-148a-3p regulated the odontoblastic differentiation of human dental pulp stem cells (hDPSCs) or not was explored. METHODS AND RESULTS: The hDPSCs were isolated and identified via flow cytometry. Targets of miR-148a-3p were identified via bioinformatics and dual-luciferase reporter assay. After the cell was cultured in the odontogenic differentiation medium or infected, cell viability, invasion, and odontoblastic differentiation were detected via MTT, transwell, and Alizarin Red S staining, respectively. The miR-148a-3p, Wnt1, ß-catenin, DSPP, DMP-1, RUNX2, OCN, and Smad4 expressions were determined by RT-qPCR and Western blot. The hDPSCs odontoblastic differentiation downregulated the miR-148a-3p expression and upregulated Wnt1 expression. Wnt1 was determined as the target for miR-148a-3p. MiR-148a-3p mimic and siWnt1 suppressed the cell viability, invasion, and odontoblastic differentiation of hDPSCs and inhibited the Wnt1, ß-catenin, DSPP, DMP-1, RUNX2, OCN, and Smad4 expressions. In contrast, miR-148a-3p inhibitor and overexpressed Wnt1 promoted the cell viability, invasion, and odontoblastic differentiation of hDPSCs, and upregulated the Wnt1, ß-catenin, DSPP, DMP-1, RUNX2, OCN, and Smad4 expressions. Also, miR-148a-3p mimic and inhibitor reversed the effects of Wnt1 overexpression and siWnt1. CONCLUSIONS: MiR-148a-3p modulated the invasion and odontoblastic differentiation of hDPSCs through the Wnt1/ß-catenin pathway.

10.
Int J Mol Sci ; 22(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34069970

ABSTRACT

Prostate cancer (PCa) is the second most leading cause of death in males. Our previous studies have demonstrated that δ-catenin plays an important role in prostate cancer progression. However, the molecular mechanism underlying the regulation of δ-catenin has not been fully explored yet. In the present study, we found that δ-catenin could induce phosphorylation of p21Waf and stabilize p21 in the cytoplasm, thus blocking its nuclear accumulation for the first time. We also found that δ-catenin could regulate the interaction between AKT and p21, leading to phosphorylation of p21 at Thr-145 residue. Finally, EGF was found to be a key factor upstream of AKT/δ-catenin/p21 for promoting proliferation and metastasis in prostate cancer. Our findings provide new insights into molecular controls of EGF and the development of potential therapeutics targeting δ-catenin to control prostate cancer progression.


Subject(s)
Catenins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epidermal Growth Factor/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Active Transport, Cell Nucleus , Binding Sites/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p21/chemistry , Cyclin-Dependent Kinase Inhibitor p21/genetics , Humans , Ligands , Male , Models, Biological , Mutagenesis, Site-Directed , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , PC-3 Cells , Phosphorylation , Prostatic Neoplasms/genetics , Protein Interaction Domains and Motifs , Protein Stability , Proto-Oncogene Proteins c-akt/chemistry , Signal Transduction , Threonine/chemistry , Delta Catenin
11.
J Biol Chem ; 295(32): 10988-11001, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32554807

ABSTRACT

The development of the dendritic arbor in pyramidal neurons is critical for neural circuit function. Here, we uncovered a pathway in which δ-catenin, a component of the cadherin-catenin cell adhesion complex, promotes coordination of growth among individual dendrites and engages the autophagy mechanism to sculpt the developing dendritic arbor. Using a rat primary neuron model, time-lapse imaging, immunohistochemistry, and confocal microscopy, we found that apical and basolateral dendrites are coordinately sculpted during development. Loss or knockdown of δ-catenin uncoupled this coordination, leading to retraction of the apical dendrite without altering basolateral dendrite dynamics. Autophagy is a key cellular pathway that allows degradation of cellular components. We observed that the impairment of the dendritic arbor resulting from δ-catenin knockdown could be reversed by knockdown of autophagy-related 7 (ATG7), a component of the autophagy machinery. We propose that δ-catenin regulates the dendritic arbor by coordinating the dynamics of individual dendrites and that the autophagy mechanism may be leveraged by δ-catenin and other effectors to sculpt the developing dendritic arbor. Our findings have implications for the management of neurological disorders, such as autism and intellectual disability, that are characterized by dendritic aberrations.


Subject(s)
Autophagy , Catenins/metabolism , Dendritic Cells/metabolism , Animals , Autophagy-Related Protein 7/genetics , Catenins/genetics , Cells, Cultured , Gene Knockdown Techniques , Hippocampus/cytology , Hippocampus/metabolism , Mice , Pyramidal Cells/metabolism , Rats , Delta Catenin
12.
Onco Targets Ther ; 13: 2771-2781, 2020.
Article in English | MEDLINE | ID: mdl-32308412

ABSTRACT

INTRODUCTION: It has been found that mannose exerts antitumoural properties in vitro and in animal models. Whether mannose has potential anti-proliferative and anti-metastatic properties against non-small-cell lung cancer (NSCLC) is still unclear. METHODS: Here, we performed ex vivo experiments and established a nude mouse model to evaluate the anticancer effects of mannose on NSCLC cells and its effects on the ERK/GSK-3ß/ß-catenin/SNAIL axis. A CCK-8 assay was conducted to evaluate the effects of mannose on lung cancer cells (A549 and HCC827) and normal lung cells (HPAEpiC). Transwells were used to examine the motility of cancer cells. qRT-PCR was used to evaluate the effects of mannose on the mRNA expression of ß-catenin. Western blotting was conducted to explore the effects of mannose on the ERK/GSK-3ß/ß-catenin/SNAIL axis and nuclear accumulation of ß-catenin. An animal model was established to evaluate the antitumoural effect of mannose on hepatic metastasis in vivo. RESULTS: In this study, we found that mannose inhibited the proliferation of A549 and HCC827 cells in vitro both time- and dose-dependently. However, it exerted only a slight influence on the viability of normal lung cells in vitro. Moreover, mannose also inhibited the migrating and invading capacity of NSCLC cells in vitro. Using Western blotting, we observed that mannose reduced SNAIL and ß-catenin expression and ERK activation and promoted phospho-GSK-3ß expression. The ERK agonist LM22B-10 promoted the metastatic ability of NSCLC cells and increased SNAIL and ß-catenin expression in cancer cells, which could be reversed by mannose. Furthermore, ERK-mediated phosphorylation of the ß-catenin-Tyr654 residue might participate in the nuclear accumulation of ß-catenin and its transcriptional function. The results from animal experiments showed that mannose effectively reduced hepatic metastasis of A549 cells in vivo. Furthermore, mannose inhibited ERK/GSK-3ß/ß-catenin/SNAIL in tumour tissues obtained from nude mice. DISCUSSION: Collectively, these findings suggest that mannose exerts anti-metastatic activity against NSCLC by inhibiting the activation of the ERK/GSK-3ß/ß-catenin/SNAIL axis, which indicates the potential anticancer effects of mannose.

13.
Int J Biol Sci ; 16(4): 671-681, 2020.
Article in English | MEDLINE | ID: mdl-32025214

ABSTRACT

Background: Activation of macrophages and infiltration are key events in acute liver injury (ALI). Kv1.3 plays an important role in regulating immunologic functions of macrophages and is extensively recognized as a potential ion channel for immunological diseases. Objective: We hypothesized that blockage of Kv1.3 may influence ALI by inhibiting macrophages infiltration in damaged liver tissues. Methods: Margatoxin was administered into the peritoneal cavity of ALI mice. The impact of this treatment on ALI and macrophage migration in vivo and in vitro was determined using immunohistochemistry, transwell migration, and wound healing assays. Results: MgTX treatment alleviated ALI in mice, as evidenced by reduced macrophage infiltration in liver tissues and lower serum levels of liver ALT and AST. RNA-seq profiling analysis showed that the most obvious change by MgTX treatment was downregulation of δ-catenin, a protein known to be associated with macrophage migration. The effect of MgTX on macrophage migration and involvement of δ-catenin was confirmed by transwell and wound healing assays. Overexpression of δ-catenin in RAW264.7 cells promoted migration, an event that was suppressed upon silencing of δ-catenin. Mechanistically, the expression of RhoA was regulated by the overexpression or knockdown of δ-catenin. Conclusion: These findings suggest a role for blockage of Kv1.3 channel in macrophage migration and reveal a new target in the treatment of ALI.


Subject(s)
Catenins/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Kv1.3 Potassium Channel/metabolism , Macrophages/cytology , Macrophages/metabolism , rhoA GTP-Binding Protein/metabolism , Alanine Transaminase/genetics , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/genetics , Aspartate Aminotransferases/metabolism , Blotting, Western , Catenins/genetics , Cell Movement/genetics , Cell Movement/physiology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Kv1.3 Potassium Channel/genetics , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , rhoA GTP-Binding Protein/genetics , Delta Catenin
14.
Cancer Med ; 9(6): 2201-2212, 2020 03.
Article in English | MEDLINE | ID: mdl-31991069

ABSTRACT

δ-Catenin is a unique member of the catenin family and is proved to be overexpressed in diverse human cancer types. However, the clinical significance and underling mechanism of δ-catenin expression in renal cell carcinoma (RCC) remain elusive. Herein, we detected the protein expression of δ-catenin in 28 clinical specimens of paired renal cancer tissues and normal renal tissues by Western blot analysis. δ-Catenin expression in 58 cases of renal cell carcinoma was also examined by immunohistochemistry, and its association with clinicopathological factors was analyzed by statistical analysis. In vitro and in vivo assays were employed to further explore the biological role of δ-catenin in RCC. The results showed that δ-catenin was highly expressed in both clinical samples and cell lines of RCC. RCC patients with higher δ-catenin expression had a more advanced pTNM stage and tumor stage as well as lymph nodes metastasis than those with lower expression. By regulating the nuclear translocation of ß-catenin and ß-catenin-mediated oncogenic signals, δ-catenin promoted proliferation and inhibited apoptosis in RCC. In vivo assay indicated δ-catenin facilitated tumor growth in ACHN cell xenograft mouse model. Taken together, our study suggests that δ-catenin might be considered as a novel prognostic indicator and actionable target for gene therapy in renal cell carcinoma.


Subject(s)
Carcinoma, Renal Cell/genetics , Catenins/metabolism , Kidney Neoplasms/genetics , beta Catenin/metabolism , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Carcinoma, Renal Cell/pathology , Catenins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Kidney/cytology , Kidney/pathology , Kidney Neoplasms/pathology , Male , Mice , Middle Aged , Xenograft Model Antitumor Assays , Delta Catenin
15.
Clin Exp Allergy ; 50(1): 29-40, 2020 01.
Article in English | MEDLINE | ID: mdl-31520422

ABSTRACT

BACKGROUND: microRNA (miR)-218-5p is involved in cigarette smoke-induced airway inflammation. In our earlier asthma epithelial miRNA profiling data, miR-218-5p was the top 2 down-regulated miRNA. We hypothesize that miR-218-5p plays a role in asthma airway inflammation. OBJECTIVE: To unveil the role of miR-218-5p and its target gene in asthma airway inflammation. METHODS: We measured miR-218-5p expression in bronchial brushings of asthma patients (n = 50) and healthy controls (n = 15), and analysed the correlations between miR-218-5p expression and airway eosinophilia. We examined whether CTNND2 was a target of miR-218-5p, and the expression of 12 catenin family members in bronchial brushings, in cultured human bronchial epithelial (HBE) cells and BEAS-2B cells. We explored the role of miR-218-5p-CTNND2 pathway using a murine model of allergic airway inflammation. RESULTS: Epithelial miR-218-5p expression was significantly decreased and negatively correlated with eosinophils in induced sputum and bronchial biopsies, and other type 2 biomarkers in asthma patients. We verified that CTNND2 (encoding δ-catenin) was a target of miR-218-5p. Remarkably, CTNND2 was the most significantly up-regulated catenin compared with the other 11 catenin family members in bronchial brushings of asthma patients, IL-13-stimulated HBE and BEAS-2B cells. Moreover, epithelial CTNND2 expression positively correlated with airway eosinophilia in asthma. Airway mmu-miR-218-5p expression was also decreased, and Ctnnd2 expression was increased in a murine model of allergic airway inflammation. Intriguingly, mmu-miR-218-5p overexpression suppressed airway hyperresponsiveness, eosinophilic airway inflammation and Ctnnd2 up-regulation in the mouse model. Finally, perturbation of miR-218-5p or CTNND2 expression significantly altered chemokine CCL26 expression in the cell cultures and the mouse model. CONCLUSIONS AND CLINICAL RELEVANCE: Epithelial miR-218-5p plays a protective role in eosinophilic airway inflammation via targeting CTNND2, a novel catenin in asthma, and suppressing chemokine CCL26 expression.


Subject(s)
Asthma/genetics , Catenins/genetics , Chemokine CCL26/metabolism , Eosinophilia/genetics , MicroRNAs/genetics , Animals , Asthma/metabolism , Bronchi/metabolism , Case-Control Studies , Cell Line , Cells, Cultured , Chemokine CCL11/metabolism , Chemokine CCL24/metabolism , Eosinophilia/metabolism , Gene Expression , Humans , Mice , Delta Catenin
16.
Folia Neuropathol ; 58(4): 324-333, 2020.
Article in English | MEDLINE | ID: mdl-33480237

ABSTRACT

INTRODUCTION: Triptolide, extracted from Chinese medicinal materials Tripterygium wilfordii Hook F (TwHF), has immunosuppressive, anti-inflammatory and anti-tumour effects. The purpose of this study was to examine whether triptolide has the neuroprotective effect on cerebral ischemia-reperfusion (I/R) injury and to explore its possible mechanism. MATERIAL AND METHODS: The rat model of focal cerebral I/R was established by the suture-occluded method. The SD rats were randomly divided into five groups: sham operation group (Sham group), ischemia-reperfusion model group (I/R group), low concentration of triptolide group (12.5 mg/kg, TL-L group), medium concentration of triptolide group (25 mg/kg, TL-M group) and high concentration of triptolide group (50 mg/kg, TL-H group). The neurological function of the rats was scored, the degree of brain oedema was detected by the dry-wet method, and the cerebral infarction area was determined by TTC staining. Nissl staining was used to detect neuronal damage. The contents of reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD) were also detected. Meanwhile, the expression level of proteins related to Wnt/-catenin signalling pathway was measured by Western blot. RESULTS: Compared with the I/R group, cerebral oedema, infarct volume, neurological impairment, the contents of MDA and ROS were reduced, while the SOD level was increased in the TL-L, TL-M, and TL-H groups. The results of Nissl staining showed that triptolide could reduce the nerve cell injury caused by cerebral I/R. In addition, the results of Western blot confirmed that the expression of Wnt1, -catenin, c-Myc, and Cyclin-D1 were down-regulated after triptolide intervention, that is, inhibited the activation of Wnt/-catenin signalling pathway. CONCLUSIONS: Triptolide mediates Wnt/-catenin signalling pathway to alleviate cerebral I/R injury in rats. This study provides ideas and experimental basis for the treatment of ischemic stroke patients.


Subject(s)
Brain Ischemia/pathology , Diterpenes/pharmacology , Immunosuppressive Agents/pharmacology , Phenanthrenes/pharmacology , Reperfusion Injury/pathology , Wnt Signaling Pathway/drug effects , Animals , Brain/drug effects , Brain/pathology , Epoxy Compounds/pharmacology , Male , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley
17.
Exp Neurol ; 318: 22-31, 2019 08.
Article in English | MEDLINE | ID: mdl-30981806

ABSTRACT

δ-Catenin is abundant in the brain and affects its synaptic plasticity. Furthermore, loss of δ-catenin is related to the deficits of learning and memory, mental retardation (cri-du-chat syndrome), and autism. A few studies about δ-catenin deficiency mice were performed. However, the effect of δ-catenin overexpression in the brain has not been investigated as yet. Therefore we generated a δ-catenin overexpressing mouse model. To generate a transgenic mouse model overexpressing δ-catenin in the brain, δ-catenin plasmid having a Thy-1 promotor was microinjected in C57BL/6 mice. Our results showed δ-catenin transgenic mice expressed higher levels of N-cadherin, ß-catenin, and p120-catenin than did wild type mice. Furthermore, δ-catenin transgenic mice exhibited better object recognition, better sociability, and lower anxiety than wild type mice. However, both mice groups showed a similar pattern in locomotion tests. Although δ-catenin transgenic mice show similar locomotion, they show improved sociability and reduced anxiety. These characteristics are opposite to the symptoms of autism or mental retardation, which are caused when δ-catenin is deficient. These results suggest that δ-catenin may alleviate symptoms of autism, Alzheimer's disease and mental retardation.


Subject(s)
Anxiety/metabolism , Catenins/metabolism , Memory/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Delta Catenin
18.
Mol Cells ; 42(1): 36-44, 2019 Jan 31.
Article in English | MEDLINE | ID: mdl-30622228

ABSTRACT

Alzheimer's disease (AD) is the most frequent age-related human neurological disorder. The characteristics of AD include senile plaques, neurofibrillary tangles, and loss of synapses and neurons in the brain. ß-Amyloid (Aß) peptide is the predominant proteinaceous component of senile plaques. The amyloid hypothesis states that Aß initiates the cascade of events that result in AD. Amyloid precursor protein (APP) processing plays an important role in Aß production, which initiates synaptic and neuronal damage. δ-Catenin is known to be bound to presenilin-1 (PS-1), which is the main component of the γ-secretase complex that regulates APP cleavage. Because PS-1 interacts with both APP and δ-catenin, it is worth studying their interactive mechanism and/or effects on each other. Our immunoprecipitation data showed that there was no physical association between δ-catenin and APP. However, we observed that δ-catenin could reduce the binding between PS-1 and APP, thus decreasing the PS-1 mediated APP processing activity. Furthermore, δ-catenin reduced PS-1-mediated stabilization of APP. The results suggest that δ-catenin can influence the APP processing and its level by interacting with PS-1, which may eventually play a protective role in the degeneration of an Alzheimer's disease patient.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Catenins/metabolism , Presenilin-1/metabolism , Animals , Cell Line , Cell Line, Tumor , Humans , Mice , Protein Binding , Protein Processing, Post-Translational , Protein Stability , Delta Catenin
19.
Exp Cell Res ; 374(1): 94-103, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30458179

ABSTRACT

Hypoxia frequently occurs in solid tumors, hepatocellular carcinoma included. Hypoxia-inducible factors (HIFs) upregulated in hypoxia can induce various downstream target genes to resist hypoxia stress, resulting in tumor growth, angiogenesis and metastasis in vivo. Therefore, hypoxia associated genes are usually cancer progression associated genes and can be potential therapy targets for cancer therapy. In our present work, we find that the hypoxia-inducible transcriptional factor, HIF1α, can directly upregulate the expression of the gene Ctnnd2, which codes the protein δ-Catenin. Then, δ-Catenin can stabilize ß-Catenin by disrupting the destruction complex, which leads to the activation of Wnt signaling. As a result, δ-Catenin can promote the proliferation and migration of HCC cells in vitro, further enhance mice HCC tumorigenesis in vivo. In summary, our work reveals that δ-Catenin is a direct downstream target gene of HIF1α. It can activate Wnt signaling via ß-Catenin stabilization. δ-Catenin can enhance HCC progression.


Subject(s)
Catenins/metabolism , Disease Progression , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Tumor Hypoxia , Wnt Signaling Pathway , Animals , Base Sequence , Carcinogenesis/pathology , Catenins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/genetics , Mice, Inbred C57BL , Mice, Inbred ICR , Promoter Regions, Genetic/genetics , Protein Binding , Protein Stability , Proteolysis , Tumor Hypoxia/genetics , Ubiquitination , Up-Regulation/genetics , beta Catenin/metabolism , Delta Catenin
20.
Oncoimmunology ; 7(8): e1450713, 2018.
Article in English | MEDLINE | ID: mdl-30221043

ABSTRACT

As classical therapy method of advanced hepatocellular carcinoma (HCC) is not effective enough, HCC immunotherapy is a hot spot for research in recent years. Although in recent years, immune checkpoint inhibitors are focused in cancer therapy, vaccines and adoptive cell therapy (ACT), as traditional immunotherapy methods for HCC are still promising. We found that δ-Catenin might be a new tumor-associated antigen for HCC, for it could be upregulated as a stress associated protein under hypoxia and irradiation treatment. δ-Catenin peptide vaccines could inhibit the growth of subcutaneous hepatocellular tumors in vivo. According to our work, δ-Catenin peptide vaccines could stimulate the activation of cytotoxic T lymphocytes (CTLs) and enhance the infiltration of CD8+ T cells into tumors. Moreover, δ-Catenin peptide vaccines could enhance the secretion of IFN-γ and the killing of tumor cells by T cells. Mechanistically, δ-Catenin peptide vaccines, presented by antigen-presenting cells to T cells, could enhance the activation of T cells via MAPK/ERK signaling and the transcriptional factors Eomes and T-bet. Our research results indicate new potential peptide vaccines, which can be applied in clinical HCC therapy.

SELECTION OF CITATIONS
SEARCH DETAIL