Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.226
Filter
1.
Biol Direct ; 19(1): 35, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715121

ABSTRACT

BACKGROUND: Ubiquitin-conjugating enzyme E2 N (UBE2N) is recognized in the progression of some cancers; however, little research has been conducted to describe its role in prostate cancer. The purpose of this paper is to explore the function and mechanism of UBE2N in prostate cancer cells. METHODS: UBE2N expression was detected in Cancer Genome Atlas Prostate Adenocarcinoma (TCGA-PRAD) data, prostate cancer tissue microarrays, and prostate cancer cell lines, respectively. UBE2N knockdown or overexpression was used to analyze its role in cell viability and glycolysis of prostate cancer cells and tumor growth. XAV939 or Axin1 overexpression was co-treated with UBE2N overexpression to detect the involvement of the Wnt/ß-catenin signaling and Axin1 in the UBE2N function. UBE2N interacting with Axin1 was analyzed by co-immunoprecipitation assay. RESULTS: UBE2N was upregulated in prostate cancer and the UBE2N-high expression correlated with the poor prognosis of prostate cancer. UBE2N knockdown inhibited cell viability and glycolysis in prostate cancer cells and restricted tumor formation in tumor-bearing mice. Wnt/ß-catenin inhibition and Axin1 overexpression reversed the promoting viability and glycolysis function of UBE2N. UBE2N promoted Axin1 ubiquitination and decreased Axin1 protein level.


Subject(s)
Axin Protein , Cell Survival , Glycolysis , Prostatic Neoplasms , Ubiquitin-Conjugating Enzymes , Ubiquitination , Animals , Humans , Male , Mice , Axin Protein/metabolism , Axin Protein/genetics , Cell Line, Tumor , Mice, Nude , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Wnt Signaling Pathway
2.
Med Oncol ; 41(6): 147, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38733492

ABSTRACT

Wnt/ß-catenin signaling plays important role in cancers. Compound 759 is one of the compounds previously screened to identify inhibitors of the Wnt/ß-catenin pathway in A549 cells [Lee et al. in Bioorg Med Chem Lett 20:5900-5904, 2010]. However, the mechanism by which Compound 759 induces the inhibition of the Wnt/ß-catenin pathway remains unknown. In our study, we employed various assays to comprehensively evaluate the effects of Compound 759 on lung cancer cells. Our results demonstrated that Compound 759 significantly suppressed cell proliferation and Wnt3a-induced Topflash activity and arrested the cell cycle at the G1 stage. Changes in Wnt/ß-catenin signaling-related protein expression, gene activity, and protein stability including Axin, and p21, were achieved through western blot and qRT-PCR analysis. Compound 759 treatment upregulated the mRNA level of p21 and increased Axin protein levels without altering the mRNA expression in A549 cells. Co-treatment of Wnt3a and varying doses of Compound 759 dose-dependently increased the amounts of Axin1 in the cytosol and inhibited ß-catenin translocation into the nucleus. Moreover, Compound 759 reduced tumor size and weight in the A549 cell-induced tumor growth in the in vivo tumor xenograft mouse model. Our findings indicate that Compound 759 exhibits potential anti-cancer activity by inhibiting the Wnt/ß-catenin signaling pathway through the increase of Axin1 protein stability.


Subject(s)
Axin Protein , Cell Proliferation , Lung Neoplasms , Mice, Nude , Wnt Signaling Pathway , Humans , Axin Protein/metabolism , Wnt Signaling Pathway/drug effects , Animals , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Protein Stability/drug effects , Xenograft Model Antitumor Assays , A549 Cells , beta Catenin/metabolism , beta Catenin/antagonists & inhibitors , Wnt3A Protein/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C
3.
Mol Biol Cell ; 35(7): ar93, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38656801

ABSTRACT

Wnt signaling is a crucial developmental pathway involved in early development as well as stem-cell maintenance in adults and its misregulation leads to numerous diseases. Thus, understanding the regulation of this pathway becomes vitally important. Axin2 and Nkd1 are widely utilized negative feedback regulators in Wnt signaling where Axin2 functions to destabilize cytoplasmic ß-catenin, and Nkd1 functions to inhibit the nuclear localization of ß-catenin. Here, we set out to further understand how Axin2 and Nkd1 regulate Wnt signaling by creating axin2gh1/gh1, nkd1gh2/gh2 single mutants and axin2gh1/gh1;nkd1gh2/gh2 double mutant zebrafish using sgRNA/Cas9. All three Wnt regulator mutants were viable and had impaired heart looping, neuromast migration defects, and behavior abnormalities in common, but there were no signs of synergy in the axin2gh1/gh1;nkd1gh2/gh2 double mutants. Further, Wnt target gene expression by qRT-PCR and RNA-seq, and protein expression by mass spectrometry demonstrated that the double axin2gh1/gh1;nkd1gh2/gh2 mutant resembled the nkd1gh2/gh2 phenotype demonstrating that Nkd1 functions downstream of Axin2. In support of this, the data further demonstrates that Axin2 uniquely alters the properties of ß-catenin-dependent transcription having novel readouts of Wnt activity compared with nkd1gh2/gh2 or the axin2gh1/gh1;nkd1gh2/gh2 double mutant. We also investigated the sensitivity of the Wnt regulator mutants to exacerbated Wnt signaling, where the single mutants displayed characteristic heightened Wnt sensitivity, resulting in an eyeless phenotype. Surprisingly, this phenotype was rescued in the double mutant, where we speculate that cross-talk between Wnt/ß-catenin and Wnt/Planar Cell Polarity pathways could lead to altered Wnt signaling in some scenarios. Collectively, the data emphasizes both the commonality and the complexity in the feedback regulation of Wnt signaling.


Subject(s)
Axin Protein , Wnt Signaling Pathway , Zebrafish Proteins , Zebrafish , beta Catenin , Animals , Axin Protein/metabolism , Axin Protein/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , beta Catenin/metabolism , Mutation/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins
4.
Cancer Res ; 84(9): 1443-1459, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38359148

ABSTRACT

AXIN1 is a major component of the ß-catenin destruction complex and is frequently mutated in various cancer types, particularly liver cancers. Truncating AXIN1 mutations are recognized to encode a defective protein that leads to ß-catenin stabilization, but the functional consequences of missense mutations are not well characterized. Here, we first identified the GSK3ß, ß-catenin, and RGS/APC interaction domains of AXIN1 that are the most critical for proper ß-catenin regulation. Analysis of 80 tumor-associated variants in these domains identified 18 that significantly affected ß-catenin signaling. Coimmunoprecipitation experiments revealed that most of them lost binding to the binding partner corresponding to the mutated domain. A comprehensive protein structure analysis predicted the consequences of these mutations, which largely overlapped with the observed effects on ß-catenin signaling in functional experiments. The structure analysis also predicted that loss-of-function mutations within the RGS/APC interaction domain either directly affected the interface for APC binding or were located within the hydrophobic core and destabilized the entire structure. In addition, truncated AXIN1 length inversely correlated with the ß-catenin regulatory function, with longer proteins retaining more functionality. These analyses suggest that all AXIN1-truncating mutations at least partially affect ß-catenin regulation, whereas this is only the case for a subset of missense mutations. Consistently, most colorectal and liver cancers carrying missense variants acquire mutations in other ß-catenin regulatory genes such as APC and CTNNB1. These results will aid the functional annotation of AXIN1 mutations identified in large-scale sequencing efforts or in individual patients. SIGNIFICANCE: Characterization of 80 tumor-associated missense variants of AXIN1 reveals a subset of 18 mutations that disrupt its ß-catenin regulatory function, whereas the majority are passenger mutations.


Subject(s)
Axin Protein , Mutation, Missense , beta Catenin , Axin Protein/genetics , Axin Protein/metabolism , Humans , beta Catenin/genetics , beta Catenin/metabolism , Signal Transduction/genetics , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , HEK293 Cells , Cell Line, Tumor , Protein Binding
5.
Cell Commun Signal ; 22(1): 77, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38291457

ABSTRACT

AXIN1, has been initially identified as a prominent antagonist within the WNT/ß-catenin signaling pathway, and subsequently unveiled its integral involvement across a diverse spectrum of signaling cascades. These encompass the WNT/ß-catenin, Hippo, TGFß, AMPK, mTOR, MAPK, and antioxidant signaling pathways. The versatile engagement of AXIN1 underscores its pivotal role in the modulation of developmental biological signaling, maintenance of metabolic homeostasis, and coordination of cellular stress responses. The multifaceted functionalities of AXIN1 render it as a compelling candidate for targeted intervention in the realms of degenerative pathologies, systemic metabolic disorders, cancer therapeutics, and anti-aging strategies. This review provides an intricate exploration of the mechanisms governing mammalian AXIN1 gene expression and protein turnover since its initial discovery, while also elucidating its significance in the regulation of signaling pathways, tissue development, and carcinogenesis. Furthermore, we have introduced the innovative concept of the AXIN1-Associated Phosphokinase Complex (AAPC), where the scaffold protein AXIN1 assumes a pivotal role in orchestrating site-specific phosphorylation modifications through interactions with various phosphokinases and their respective substrates.


Subject(s)
Wnt Signaling Pathway , beta Catenin , Animals , Gene Ontology , Axin Protein/genetics , Axin Protein/metabolism , Wnt Signaling Pathway/genetics , Phosphorylation , Proteolysis , beta Catenin/metabolism , Mammals/metabolism
6.
Tohoku J Exp Med ; 262(4): 269-276, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38233113

ABSTRACT

Osimertinib, a promising and approved third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), is a standard strategy for EGFR-mutant non-small cell lung cancer (NSCLC) patients. However, developed resistance is unavoidable, which reduces its long-term effectiveness. In this study, RNA sequencing was performed to analyze differentially expressed genes (DEGs). The PrognoScan database and Gene Expression Profiling Interactive Analysis (GEPIA) were used to identify the key genes for clinical prognosis and gene correlation respectively. Protein expression was determined by western blot analysis. Cell viability assay and Ki67 staining were used to evaluate the effect of osimertinib on tumor cells. Finally, we screened out two hub genes, myelocytomatosis oncogene (Myc) and axis inhibition protein 1 (Axin1), upregulated in three osimertinib-resistant cell lines through RNA sequencing and bioinformatics analysis. Next, cell experiment confirmed that expression of C-MYC and AXIN1 were elevated in different EGFR mutant NSCLC cell lines with acquired resistance to osimertinib, compared with their corresponding parental cell lines. Furthermore, we demonstrated that AXIN1 upregulated the expression of C-MYC and mediated the acquired resistance of EGFR mutant NSCLC cells to osimertinib in vitro. In conclusion, AXIN1 affected the sensitivity of EGFR mutant NSCLC to osimertinib via regulating C-MYC expression in vitro. Targeting AXIN1/MYC signaling may be a potential new strategy for overcoming acquired resistance to osimertinib.


Subject(s)
Acrylamides , Aniline Compounds , Axin Protein , Carcinoma, Non-Small-Cell Lung , Drug Resistance, Neoplasm , ErbB Receptors , Gene Expression Regulation, Neoplastic , Indoles , Lung Neoplasms , Mutation , Proto-Oncogene Proteins c-myc , Pyrimidines , Humans , Acrylamides/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Aniline Compounds/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Drug Resistance, Neoplasm/genetics , Axin Protein/genetics , Axin Protein/metabolism , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mutation/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Int J Mol Sci ; 25(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38203748

ABSTRACT

Colorectal cancer (CRC) is one of the most common neoplasms worldwide. Among the risk factors of CRC, inflammatory bowel disease (IBD) is one of the most important ones leading to the development of colitis-associated CRC (CAC). G-protein coupled receptors (GPCR) are transmembrane receptors that orchestrate a multitude of signaling cascades in response to external stimuli. Because of their functionality, they are promising targets in research on new strategies for CRC diagnostics and treatment. Recently, regulators of G-proteins (RGS) have been attracting attention in the field of oncology. Typically, they serve as negative regulators of GPCR responses to both physiological stimuli and medications. RGS activity can lead to both beneficial and harmful effects depending on the nature of the stimulus. However, the atypical RGS-AXIN uses its RGS domain to antagonize key signaling pathways in CRC development through the stabilization of the ß-catenin destruction complex. Since AXIN does not limit the efficiency of medications, it seems to be an even more promising pharmacological target in CRC treatment. In this review, we discuss the current state of knowledge on RGS significance in sporadic CRC and CAC with particular emphasis on the regulation of GPCR involved in IBD-related inflammation comprising opioid, cannabinoid and serotonin receptors.


Subject(s)
Colitis-Associated Neoplasms , Inflammatory Bowel Diseases , Humans , Axin Protein , GTP-Binding Proteins , Signal Transduction , Inflammatory Bowel Diseases/complications
8.
Cell Commun Signal ; 22(1): 14, 2024 01 05.
Article in English | MEDLINE | ID: mdl-38183076

ABSTRACT

R-spondins (RSPOs) are secreted signaling molecules that potentiate the Wnt/ß-catenin pathway by cooperating with Wnt ligands. RSPO1 is crucial in tissue development and tissue homeostasis. However, the molecular mechanism by which RSPOs activate Wnt/ß-catenin signaling remains elusive. In this study, we found that RSPOs could mediate the degradation of Axin through the ubiquitin-proteasome pathway. The results of Co-IP showed that the recombinant RSPO1 protein promoted the interaction between Axin1 and CK1ε. Either knockout of the CK1ε gene or treatment with the CK1δ/CK1ε inhibitor SR3029 caused an increase in Axin1 protein levels and attenuated RSPO1-induced degradation of the Axin1 protein. Moreover, we observed an increase in the number of associations of LRP6 with CK1ε and Axin1 following RSPO1 stimulation. Overexpression of LRP6 further potentiated Axin1 degradation mediated by RSPO1 or CK1ε. In addition, recombinant RSPO1 and Wnt3A proteins synergistically downregulated the protein expression of Axin1 and enhanced the transcriptional activity of the SuperTOPFlash reporter. Taken together, these results uncover the novel mechanism by which RSPOs activate Wnt/ß-catenin signaling through LRP6/CK1ε-mediated degradation of Axin.


Subject(s)
Axin Protein , Thrombospondins , Wnt Signaling Pathway , beta Catenin , Biological Transport , Wnt3A Protein , Humans , Thrombospondins/metabolism
9.
Cell Mol Life Sci ; 81(1): 57, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38279052

ABSTRACT

The Wnt/ß-catenin pathway is critical to maintaining cell fate decisions. Recent study showed that liquid-liquid-phase separation (LLPS) of Axin organized the ß-catenin destruction complex condensates in a normal cellular state. Mutations inactivating the APC gene are found in approximately 80% of all human colorectal cancer (CRC). However, the molecular mechanism of the formation of ß-catenin destruction complex condensates organized by Axin phase separation and how APC mutations impact the condensates are still unclear. Here, we report that the ß-catenin destruction complex, which is constructed by Axin, was assembled condensates via a phase separation process in CRC cells. The key role of wild-type APC is to stabilize destruction complex condensates. Surprisingly, truncated APC did not affect the formation of condensates, and GSK 3ß and CK1α were unsuccessfully recruited, preventing ß-catenin phosphorylation and resulting in accumulation in the cytoplasm of CRCs. Besides, we propose that the phase separation ability of Axin participates in the nucleus translocation of ß-catenin and be incorporated and concentrated into transcriptional condensates, affecting the transcriptional activity of Wnt signaling pathway.


Subject(s)
Axin Signaling Complex , beta Catenin , Humans , Axin Signaling Complex/genetics , Axin Protein/genetics , Axin Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Phase Separation , Mutation/genetics , Wnt Signaling Pathway/genetics , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism
10.
Arch Dermatol Res ; 316(2): 64, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38170256

ABSTRACT

BACKGROUND: A myriad of therapeutic modalities for alopecia areata are available; however, none is of high level of evidence, creating an immense need for the evaluation of other treatment modalities, of which topical sodium valproate is of potential role via proposed decrease in beta-catenin breakdown, despite its well-known side effect of hair fall as an oral therapy. OBJECTIVE: Evaluating the efficacy and the safety of sodium valproate (SV)-loaded nanospanlastics, in comparison to topical corticosteroids, this is the currently available gold standard topical treatment for patchy AA. METHODOLOGY: A total of 66 patients with patchy AA were randomly assigned to receive either topical mometasone furoate lotion or topical SV applied twice daily to all patches except a control patch, which was left untreated. Clinical, trichoscopic and biochemical assessments of beta-catenin tissue levels and Axin-2 gene expression were carried out at baseline and after 3 months. RESULTS: Both therapeutic modalities were comparable. Potential efficacy was highlighted by significant improvement in the representative patch, the largest treated patch, to the control patch, the smallest untreated patch in both steroid and valproate groups (p = 0.027, 0.003 respectively). Both beta-catenin levels and Axin-2 gene expression were reduced after treatment, pointing to the inhibitory effect of dominating uncontrolled inflammatory milieu. Baseline beta-catenin was found to significantly negatively correlate with improvement in the representative patch in patients with baseline level above 0.42 ng/ml (p = - 0.042). CONCLUSION: Both topical SV and steroids are of comparable modest efficacy. Thus, further evaluation of SV is due in combination with intralesional steroids and other anti-inflammatory treatment modalities, together with developing individualized approaches based on baseline beta-catenin level. GOV IDENTIFIER: NCT05017454, https://clinicaltrials.gov/ct2/show/NCT05017454 .


Subject(s)
Alopecia Areata , Humans , Alopecia Areata/drug therapy , Valproic Acid/therapeutic use , beta Catenin , Axin Protein , Treatment Outcome
11.
Int Immunopharmacol ; 127: 111319, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38064812

ABSTRACT

Mannose is a unique natural sugar that can be found in a variety of fruits and vegetables. During the past decades, mannose has been reported to be effective in promoting immune tolerance and suppressing inflammatory diseases. Metabolic dysfunction and altered inflammation have clear implications for the development and progression of inflammatory diseases. Herein, we intended to reveal the molecular mechanism of mannose in protecting against intestinal epithelial damage in experimental colitis. We showed that mannose treatment significantly attenuated dextran sodium sulfate (DSS)-induced intestinal barrier damage. The AMPK pathway was responsible for the mannose-mediated protective effect in DSS-induced intestinal epithelial damage. Mechanistically, mannose promoted the axis inhibition protein (AXIN)-based AMPK activation, thereby preventing mitochondrial dysfunction and tight junction disruption in response to the DSS challenge. Cumulatively, the results indicate the use of mannose as a novel approach to treat IBD and other diseases involving tight junction dysfunction. The therapeutic effect of mannose is related to its regulatory function in AMPK pathway activation.


Subject(s)
Colitis , Mannose , Animals , Mice , Mannose/therapeutic use , AMP-Activated Protein Kinases/metabolism , Axin Protein/metabolism , Axin Protein/pharmacology , Tight Junctions , Intestinal Mucosa , Dextran Sulfate/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Mice, Inbred C57BL , Disease Models, Animal
12.
Inflamm Res ; 73(3): 407-414, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38158447

ABSTRACT

OBJECTIVE AND DESIGN: This study aimed to investigate Axin2 effects on myocardial infarction (MI) using a macrophage Axin2 conditional knockout (cKO) mouse model, RAW264.7 cell line, and human subepicardial tissues from patients with coronary artery bypass graft (CABG). MATERIAL OR SUBJECTS: Axin2 cKO mice showed decreased cardiac function, reduced edema, increased lymphangiogenesis, and improved repair in MI Few studies border zones. Hypoxic macrophages with Axin2 depletion exhibited decreased senescence, elevated IL6 expression, and increased LYVE1 transcription. Senescent macrophages decreased in patients with CABG and low Axin2 expression. TREATMENT: Treatment options included in this study were MI induction in Axin2 cKO mice, in vitro experiments with RAW264.7 cells, and analysis of human subepicardial tissues. METHODS: Assays included MI induction, in vitro experiments, and tissue analysis with statistical tests applied. RESULTS: Axin2 cKO improved cardiac function, reduced edema, enhanced lymphangiogenesis, and decreased senescence. Hypoxic macrophages with Axin2 depletion showed reduced senescence, increased IL6 expression, and elevated LYVE1 transcription. Senescent macrophages decreased in patients with CABG and low Axin2 expression. CONCLUSION: Targeting Axin2 emerges as a novel therapeutic strategy for regulating cardiac lymphatics and mitigating cell senescence post-MI, evidenced by improved outcomes in Axin2-deficient conditions.


Subject(s)
Interleukin-6 , Myocardial Infarction , Humans , Mice , Animals , Interleukin-6/metabolism , Myocardial Infarction/genetics , Macrophages , Immunity , Edema/metabolism , Mice, Inbred C57BL , Myocardium , Axin Protein/genetics , Axin Protein/metabolism
13.
Int J Biol Macromol ; 258(Pt 1): 128570, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38096938

ABSTRACT

The ß-catenin has two intrinsically disordered regions in both C- and N-terminal domains that trigger the formation of phase-separated condensates. Variants in its C-terminus are associated with familial exudative vitreoretinopathy (FEVR), yet the pathogenesis and the role of these variants in inducing abnormal condensates, are unclear. In this study, we identified a novel heterozygous frameshift variant, c.2104-2105insCC (p.Gln703ProfsTer33), in CTNNB1 from a FEVR-affected family. This variant encodes an unstable truncated protein that was unable to activate Wnt signal transduction, which could be rescued by the inhibition of proteasome or phosphorylation. Further functional experiments revealed the propensity of the Gln703ProfsTer33 variant to form cytoplasmic condensates, exhibiting a lower turnover rate after fluorescent bleaching due to enhanced interaction with AXIN1. LiCl, which specifically blocks GSK3ß-mediated phosphorylation, restored signal transduction, cell proliferation, and junctional integrity in primary human retinal microvascular endothelial cells over-expressed with Gln703ProfsTer33. Finally, experiments on two reported FEVR-associated mutations in the C-terminal domain of ß-catenin exhibited several functional defects similar to the Gln703ProfsTer33. Together, our findings unravel that the C-terminal region of ß-catenin is pivotal for the regulation of AXIN1/ß-catenin interaction, acting as a switch to mediate nucleic and cytosolic condensates formation that is implicated in the pathogenesis of FEVR.


Subject(s)
Proteasome Endopeptidase Complex , Ubiquitin , Humans , Familial Exudative Vitreoretinopathies/genetics , Proteasome Endopeptidase Complex/genetics , Ubiquitin/genetics , beta Catenin/metabolism , Endothelial Cells/metabolism , Mutation , DNA Mutational Analysis , Axin Protein/genetics
14.
Sci Rep ; 13(1): 20099, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37973999

ABSTRACT

Mounting evidence has proposed the importance of the Wnt/ß-catenin pathway and tripartite motif 31 (TRIM31) in certain malignancies. Our research aimed to clarify the correlation between aberrant TRIM31 expression and the Wnt/ß-catenin pathway during gastric cancer (GC) oncogenesis and development. TRIM31 was drastically elevated in GC tissues and was closely associated with aggressive clinical outcomes and poor prognosis. Moreover, TRIM31 downregulation attenuated GC cell proliferation and invasion in vitro. Mechanistically, TRIM31 could bind and ubiquitinate Axin1 protein, thereby facilitating the activation of the Wnt/ß-catenin pathway. Additionally, Axin1 knockdown partially abrogated the inhibitory effects on the proliferative, invasive and migratory abilities of GC cells induced by TRIM31 silencing. Furthermore, TRIM31 was negatively correlated with Axin1 protein expression in GC tissues. In summary, we revealed a new TRIM31-Axin1-Wnt/ß-catenin axis that contributed greatly to the progression of GC, and targeting this regulatory axis may represent an effective treatment for GC patients.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , beta Catenin/metabolism , Cell Line, Tumor , Wnt Signaling Pathway , Cell Proliferation , Protein Stability , Gene Expression Regulation, Neoplastic , Axin Protein/genetics , Axin Protein/metabolism , Cell Movement , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
15.
Gut Microbes ; 15(2): 2286674, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38010886

ABSTRACT

Classically, Axin1 is considered a regulator of Wnt/ß-catenin signaling. However, Axin1's roles in host-microbial interactions have been unknown. Our recent study has demonstrated that deletion of intestinal epithelial Axin1 in epithelial cells and Paneth cells protects the host against colitis by enhancing Akkermansia muciniphila. Loss of intestinal epithelial or Paneth cell Axin1 results in increased Wnt/ß-catenin signaling, proliferation, and cell migration. This is associated with morphologically altered goblet and Paneth cells, including increased Muc2 and decreased lysozyme. Axin1 deletion specifically enriched Akkermansia muciniphila. Akkermansia muciniphila in Axin1 knockout mice is the driver of protection against DSS-induced inflammation. Here, we feature several significant conceptual changes, such as differences between Axin1 and Axin2, Axin1 in innate immunity and microbial homeostasis, and Axin1 reduction of Akkermansia muciniphila. We discuss an important trend in the field related to Paneth cells and tissue-specific Axin1 manipulation of microbiome in health and inflammation.


Subject(s)
Axin Protein , Colitis , Gastrointestinal Microbiome , Microbiota , Animals , Mice , Axin Protein/genetics , beta Catenin , Colitis/chemically induced , Colitis/genetics , Inflammation , Paneth Cells
16.
Front Immunol ; 14: 1271603, 2023.
Article in English | MEDLINE | ID: mdl-38035113

ABSTRACT

Given the increasing incidence of pancreatic cancer and the low survival rate, the exploration of the complex tumor microenvironment and the development of novel treatment options is urgent. NK cells, known for their cytotoxic abilities and modulation of other immune cells, are vital in recognizing and killing cancer cells. However, hypoxic conditions in the tumor microenvironment have been found to impair NK cell functionality and contribute to tumor immune escape. Therefore, we aimed to uncover the mechanism through which hypoxia mediates the immune escape of pancreatic cancer cells, focusing on the influence of miR-1275/AXIN2 on NK cells. Using a combination of GEO dataset screening, Tumor Immune Estimation Resource 2.0 immunoscore screening, and the Cancer Genome Atlas data, we identified a correlation between miR-1275 and NK cells. The down-regulation of miR-1275 was associated with decreased NK cell activity and survival in patients with pancreatic cancer. Pathway analysis further linked miR-1275 expression with the hypoxic HIF1A pathway. In vitro experiments were conducted using the NK-92 cell, revealing that hypoxia significantly reduced miR-1275 expression and correspondingly decreased the cell-killing ability of NK cells. Upregulation of miR-1275 increased perforin, IFN-γ and TNF-α expression levels and enhanced NK cell cytotoxicity. Additionally, miR-1275 was found to bind to and inhibit AXIN2 expression, which when overexpressed, partially alleviated the promotive effect of upregulated miR-1275 on NK-92 cell killing ability. In conclusion, this research underscores the critical role of the miR-1275/AXIN2 axis in hypoxia-mediated immune escape in pancreatic cancer, thus opening new potential avenues for treatment strategies.


Subject(s)
MicroRNAs , Pancreatic Neoplasms , Humans , Killer Cells, Natural , Hypoxia/genetics , Hypoxia/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Tumor Microenvironment/genetics , Axin Protein/metabolism
17.
J Neuropathol Exp Neurol ; 82(12): 995-1009, 2023 11 20.
Article in English | MEDLINE | ID: mdl-37944015

ABSTRACT

Parkinson disease (PD) is a major neurodegenerative disease that greatly undermines people's health and for which effective therapeutic strategies are currently limited. This study dissected the effects of expression changes of AXIN2, a modulator of the Wnt/beta-catenin signaling pathway, the transcription factor CREB1, and of the microRNA miR-15b-5p on apoptosis and the inflammatory response in a PD mouse model in vivo and in a cellular PD model in vitro. The analyses demonstrated low CREB1 and miR-15b-5p expression and high AXIN2 expression in both models. miR-15b-5p overexpression or AXIN2 knockdown alleviated the inflammatory response indicated by decreased levels of TNF-α, IL-6, and IL-1ß and apoptosis indicated by decreased levels of cleaved caspase-3 and Bax and elevated Bcl-2. Protection by miR-15b-5p upregulation was counteracted by the simultaneous overexpression of AXIN2. miR-15b-5p targeted AXIN2. CREB1 promoted miR-15b-5p expression, which activated the Wnt/ß-catenin pathway by inhibiting AXIN2. Collectively, the data indicate that transcriptional expression of miR-15b-5p can be promoted by CREB1 to inhibit AXIN2 and activate Wnt/ß-catenin, thereby reducing the inflammatory response and apoptosis in these PD models. These data suggest the CREB1/miR-15b-5p/AXIN2 axis is a potential therapeutic target in PD patients.


Subject(s)
MicroRNAs , Neurodegenerative Diseases , Parkinson Disease , Animals , Mice , Humans , beta Catenin , Parkinson Disease/genetics , Apoptosis , Inflammation , MicroRNAs/genetics , Cyclic AMP Response Element-Binding Protein , Axin Protein/genetics
18.
J Dent Res ; 102(13): 1478-1487, 2023 12.
Article in English | MEDLINE | ID: mdl-37837227

ABSTRACT

Vital pulp therapy and root canal therapy (RCT) are the dominant treatment for irreversible pulpitis. While the success rate of these procedures is favorable, they have some limitations. For instance, RCT leads to removing significant dentin in the coronal third of the tooth that increases root-fracture risk, which forces tooth removal. The ideal therapeutic goal is dental pulp regeneration, which is not achievable with RCT. Specialized proresolving mediators (SPMs) are well known for inflammatory resolution. The resolution of inflammation and tissue restoration or regeneration is a dynamic and continuous process. SPMs not only have potent immune-modulating functions but also effectively promote tissue homeostasis and regeneration. Resolvins have been shown to promote dental pulp regeneration. The purpose of this study was to explore further the cellular target of Resolvin E1 (RvE1) therapy in dental pulp regeneration and the impact of RvE1 in infected pulps. We investigated the actions of RvE1 on experimentally exposed pulps with or without microbial infection in an Axin2Cre-Dox;Ai14 genetically defined mouse model. Our results showed RvE1 promoted Axin2-tdTomato+ cell expansion and odontoblastic differentiation after direct pulp capping in the mouse, which we used to mimic reversible pulpitis cases in the clinic. In cultured mouse dental pulp stem cells (mDPSCs), RvE1 facilitated Axin2-tdTomato+ cell proliferation and odontoblastic differentiation and also rescued impaired functions after lipopolysaccharide stimulation. In infected pulps exposed to the oral environment for 24 h, RvE1 suppressed inflammatory infiltration, reduced bacterial invasion in root canals, and prevented the development of apical periodontitis, while its proregenerative impact was limited. Collectively, topical treatment with RvE1 facilitated dental pulp regenerative properties by promoting Axin2-expressing cell proliferation and differentiation. It also modulated the resolution of inflammation, reduced infection severity, and prevented apical periodontitis, presenting RvE1 as a novel therapeutic for treating endodontic diseases.


Subject(s)
Periapical Periodontitis , Pulpitis , Mice , Animals , Dental Pulp/physiology , Periapical Periodontitis/therapy , Inflammation , Bacteria , Regeneration/physiology , Axin Protein
19.
Nan Fang Yi Ke Da Xue Xue Bao ; 43(8): 1322-1332, 2023 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-37712268

ABSTRACT

OBJECTIVE: To investigate the role of Rho GTPase-activating protein 21 (ARHGAP21) in regulating the migration and metastasis of non-small cell lung cancer (NSCLC) cells. METHODS: TCGA, CPTAC database were used to analyze the correlation of ARHGAP21 expression level in NSCLC and the patients' prognosis. The expression of ARHGAP21 in clinical specimens of NSCLC tissues was examined using Western blotting and immunohistochemistry. The effect of ARHGAP21 knockdown on migration ability of lung cancer cell lines was examined using Transwell assay and wound healing assay. A nude mouse model with injection of lung cancer H1299 cells via the tail vein was used to examine the effect of ARHGAP21 knockdown on the metastatic ability of the tumor cells. The possible mechanism of ARHGAP21 was predicted by bioinformatics analysis and verified using Western blotting. RESULTS: A low ARHGAP21 expression was associated with poor prognosis of patients with NSCLC (P < 0.05). ARHGAP21 expression was significantly downregulated in lung cancer tissues as compared with the adjacent tissues (P < 0.001). In cultured lung cancer cells, ARHGAP21 knockdown obviously promoted the migration ability of the cells (P < 0.001). In the nude mouse models, injection of H1299 cells with ARHGAP21 knockdown, as compared with the negative control cells, resulted in a greater number of metastatic lung cancer nodules (P < 0.05), which expressed higher levels of N-cadherin and vimentin. Bioinformatic analysis showed a close correlation of ARHGAP21 with APC, GSK3ß, and Axin (P < 0.001). Western blotting showed that ARHGAP21 knockdown significantly decreased ubiquitination of ß-catenin, upregulated N-cadherin and activated the WNT signaling pathway in the lung cancer cells. CONCLUSION: ARHGAP21 downregulation can significantly promote the migration and metastatic ability of NSCLC possibly as a result of WNT signaling pathway activation, which reduces the ubiquitination of ß-catenin by affecting the expressions of APC, GSK3ß, and Axin.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Mice , Axin Protein , beta Catenin , Cadherins , Epithelial-Mesenchymal Transition , Glycogen Synthase Kinase 3 beta , Mice, Nude , Wnt Signaling Pathway
20.
Mol Biol Rep ; 50(11): 8937-8947, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37710072

ABSTRACT

Increased hyaluronan deposition (HA) in various cancer tissues, including sarcomas, correlates with disease progression. The receptor for hyaluronic acid-mediated motility (RHAMM) expression is elevated in most human cancers. ß-catenin is a critical downstream mediator of the Wnt signaling pathways, facilitating carcinogenic events characterized by deregulated cell proliferation. We previously showed that low molecular weight (LMW) HA/RHAMM/ß-catenin signaling axis increases HT1080 fibrosarcoma cell growth. Here, focusing on mechanistic aspects and utilizing immunofluorescence and immunoprecipitation, we demonstrate that LMW HA treatment enhanced RHAMM intracellular localization (p ≤ 0.001) and RHAMM/ß-catenin colocalization in HT1080 fibrosarcoma cells (p ≤ 0.05). Downregulating endogenous HA attenuated the association of RHAMM/ß-catenin in HT1080 fibrosarcoma cells (p ≤ 0.0.01). Notably, Axin-2, the key ß-catenin degradation complex component, and RHAMM were demonstrated to form a complex primarily to cell membranes, enhanced by LMW HA (p ≤ 0.01). In contrast, LMW HA attenuated the association of ß-catenin and Axin-2 (p ≤ 0.05). The utilization of FH535, a Wnt signaling inhibitor, showed that LMW HA partially rescued the Wnt-dependent growth of HT1080 cells and restored the expression of Wnt/ß-catenin mediators, cyclin-D1 and c-myc (p ≤ 0.05). B6FS fibrosarcoma cells with different HA metabolism do not respond to the LMW HA growth stimulus (p = NS). The present study identifies a novel LMW HA/RHAMM mechanism in a fibrosarcoma model. LMW HA regulates intracellular RHAMM expression, which acts as a scaffold protein binding ß-catenin and Axin-2 at different cellular compartments to increase ß-catenin expression, transcriptional activity, and fibrosarcoma growth.


Subject(s)
Fibrosarcoma , Hyaluronic Acid , Humans , Hyaluronic Acid/pharmacology , Axin Protein/genetics , Axin Protein/metabolism , beta Catenin/metabolism , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Cell Proliferation , Fibrosarcoma/metabolism , Cell Movement , Carrier Proteins , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...