Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 762
Filter
1.
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
2.
Front Immunol ; 14: 1171150, 2023.
Article in English | MEDLINE | ID: mdl-37545495

ABSTRACT

Wear debris-induced osteolysis, especially titanium (Ti) particles-induced osteolysis, is the most common cause of arthroplasty failure with no effective therapy. Previous studies have suggested that inflammation and impaired osteogenesis are associated with Ti particles -induced osteolysis. Selenium (Se) is an essential trace element in the human body, which forms selenomethionine (Se-Met) in nature, and selenoproteins has strong anti-inflammatory and antioxidant stress effects. In this study, the effects of Se-Met on Ti particles-induced osteolysis were observed and the potential mechanism was explored. We found that exogenous Se-Met relieved osteolysis induced by Ti particles in two animal models and MC3T3-E1 cells. We found that the addition of Se-Met effectively inhibited Ti particle-induced inflammation by regulating reactive oxygen species-dependent (ROS-dependent) NOD-like receptor protein 3 (NLRP3) inflammasome activation. These therapeutic effects were abrogated in MC3T3-E1 cells that had received a ß-catenin antagonist, suggesting that Se-Met alleviates inflammatory osteolysis via the ß-catenin signaling pathway. Collectively, these findings indicated that Se-Met may serve as a potential therapeutic agent for treating Ti particle-induced osteolysis.


Subject(s)
Osteolysis , Selenomethionine , Titanium , Animals , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism , Inflammasomes , Inflammation/chemically induced , NLR Family, Pyrin Domain-Containing 3 Protein , Osteolysis/chemically induced , Osteolysis/metabolism , Reactive Oxygen Species , Selenomethionine/metabolism , Signal Transduction , Titanium/adverse effects , Mice , 3T3 Cells
3.
Oncol Rep ; 48(1)2022 Jul.
Article in English | MEDLINE | ID: mdl-35656884

ABSTRACT

Wnt/ß­catenin signaling is involved in endocrine resistance and stem cell­like properties of hormone receptor­positive breast cancer cells. Palbociclib is a well­known inhibitor of cyclin­dependent kinase 4 and 6 (CDK4/6 inhibitor) that downregulates the activation of retinoblastoma protein, thereby inhibiting the cell cycle in breast cancer cells. The inhibitory effects of a combination of palbociclib and ICG­001, a ß­catenin small­molecule inhibitor, were investigated in tamoxifen­resistant breast cancer cell lines. Tamoxifen­resistant MCF­7 (TamR) cells were established by continuously exposing MCF­7 cells to tamoxifen. The characteristics associated with the stem cell­like property of cancer were assessed using western blotting, cell cycle analysis, and the mammosphere assay. The effects of the combination of palbociclib and ICG­001 were evaluated in control MCF­7 and TamR cell lines. Compared with control cells, TamR cells exhibited elevated levels of Nanog, Sox2, ALDH1, and p­STAT3, indicating stem cell­like characteristics, and elevated ß­catenin activity. TamR cells also showed significantly higher mammosphere­forming efficiency. Several markers of stem cell­like nature of TamR cells showed reduced levels upon treatment of cells with the drug combination; there was a greater reduction in the levels of these markers when the cells were treated with the combination than in the case where cells were treated with one of the drugs individually (combination index value for 25 µM palbociclib and 50 µM ICG­001 was 1.1±0.02). TamR cells treated with the palbociclib and ICG­001 combination demonstrated significantly reduced cell proliferation and mammosphere­forming efficiency compared with the cells treated with one of these drugs. The combination of the drugs could additively inhibit proliferation and suppress stem cell­like characteristics. These results suggest that ß­catenin plays a role in endocrine­resistant breast cancer; the inhibition of ß­catenin and CDK4/6 together can overcome endocrine resistance in breast cancer cells.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , beta Catenin , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Catenins , Cell Proliferation , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Female , Humans , MCF-7 Cells , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , beta Catenin/antagonists & inhibitors
4.
Cell Rep ; 38(5): 110319, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108540

ABSTRACT

Wnt/ß-catenin signaling is a well-established driver of colon cancer; however, a targeted therapeutic agent has not reached clinics yet. In the present study, we report that the natural compound liquidambaric acid (LDA) inhibits oncogenic Wnt/ß-catenin signaling in vitro and in vivo through its direct target tumor necrosis factor receptor-associated factor 2 (TRAF2). Mechanistically, TRAF2 positively regulates Wnt signaling by interacting with the N-terminal of ß-catenin via its TRAF-C domain; this interaction is disrupted in presence of LDA. Particularly, a TRAF2/ß-catenin/TCF4/TNIK complex is present in colon cancer cells, where TRAF2 is indispensable for the complex formation, and TRAF2/ß-catenin and ß-catenin/TCF4 interactions are disrupted upon LDA treatment. Our findings not only highlight that TRAF2 is an oncogenic regulator of Wnt/ß-catenin signaling and colon cancer but also provide a lead compound targeting TRAF2 for cancer therapy.


Subject(s)
Carcinogenesis/drug effects , Colonic Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 2/drug effects , TNF Receptor-Associated Factor 2/metabolism , Wnt Proteins/drug effects , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Zebrafish
5.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35156681

ABSTRACT

Axolotls are an important model organism for multiple types of regeneration, including functional spinal cord regeneration. Remarkably, axolotls can repair their spinal cord after a small lesion injury and can also regenerate their entire tail following amputation. Several classical signaling pathways that are used during development are reactivated during regeneration, but how this is regulated remains a mystery. We have previously identified miR-200a as a key factor that promotes successful spinal cord regeneration. Here, using RNA-seq analysis, we discovered that the inhibition of miR-200a results in an upregulation of the classical mesodermal marker brachyury in spinal cord cells after injury. However, these cells still express the neural stem cell marker sox2. In vivo cell tracking allowed us to determine that these cells can give rise to cells of both the neural and mesoderm lineage. Additionally, we found that miR-200a can directly regulate brachyury via a seed sequence in the 3'UTR of the gene. Our data indicate that miR-200a represses mesodermal cell fate after a small lesion injury in the spinal cord when only glial cells and neurons need to be replaced.


Subject(s)
MicroRNAs/metabolism , Spinal Cord Regeneration/genetics , Spinal Cord/metabolism , 3' Untranslated Regions , Ambystoma mexicanum/metabolism , Animals , Antagomirs/metabolism , Cell Differentiation , Fetal Proteins/genetics , Fetal Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Spinal Cord/cytology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Stem Cells/cytology , Stem Cells/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tail/physiology , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors , beta Catenin/chemistry , beta Catenin/metabolism
6.
Int J Mol Sci ; 23(4)2022 Feb 19.
Article in English | MEDLINE | ID: mdl-35216447

ABSTRACT

Oncolytic bovine herpesvirus type 1 (BoHV-1) infection induces DNA damage in human lung adenocarcinoma cell line A549. However, the underlying mechanisms are not fully understood. We found that BoHV-1 infection decreased the steady-state protein levels of p53-binding protein 1 (53BP1), which plays a central role in dictating DNA damage repair and maintaining genomic stability. Furthermore, BoHV-1 impaired the formation of 53BP1 foci, suggesting that BoHV-1 inhibits 53BP1-mediated DNA damage repair. Interestingly, BoHV-1 infection redistributed intracellular ß-catenin, and iCRT14 (5-[[2,5-Dimethyl-1-(3-pyridinyl)-1H-pyrrol-3-yl]methylene]-3-phenyl-2,4-thiazolidinedione), a ß-catenin-specific inhibitor, enhanced certain viral protein expression, such as the envelope glycoproteins gC and gD, and enhanced virus infection-induced DNA damage. Therefore, for the first time, we provide evidence showing that BoHV-1 infection disrupts 53BP1-mediated DNA damage repair and suggest ß-catenin as a potential host factor restricting both virus replication and DNA damage in A549 cells.


Subject(s)
Adenocarcinoma of Lung/genetics , DNA Damage/drug effects , Herpesviridae Infections/genetics , Lung Neoplasms/genetics , Pyridines/pharmacology , Pyrroles/pharmacology , Thiazolidinediones/pharmacology , Viral Proteins/genetics , beta Catenin/antagonists & inhibitors , A549 Cells , Cell Line, Tumor , DNA Damage/genetics , Herpesvirus 1, Bovine/pathogenicity , Humans , Virus Replication/drug effects
7.
PLoS Negl Trop Dis ; 16(1): e0010074, 2022 01.
Article in English | MEDLINE | ID: mdl-34986160

ABSTRACT

The protozoan parasite, Trypanosoma cruzi, causes severe morbidity and mortality in afflicted individuals. Approximately 30% of T. cruzi infected individuals present with cardiac pathology. The invasive forms of the parasite are carried in the vascular system to infect other cells of the body. During transportation, the molecular mechanisms by which the parasite signals and interact with host endothelial cells (EC) especially heart endothelium is currently unknown. The parasite increases host thrombospondin-1 (TSP1) expression and activates the Wnt/ß-catenin and hippo signaling pathways during the early phase of infection. The links between TSP1 and activation of the signaling pathways and their impact on parasite infectivity during the early phase of infection remain unknown. To elucidate the significance of TSP1 function in YAP/ß-catenin colocalization and how they impact parasite infectivity during the early phase of infection, we challenged mouse heart endothelial cells (MHEC) from wild type (WT) and TSP1 knockout mice with T. cruzi and evaluated Wnt signaling, YAP/ß-catenin crosstalk, and how they affect parasite infection. We found that in the absence of TSP1, the parasite induced the expression of Wnt-5a to a maximum at 2 h (1.73±0.13), P< 0.001 and enhanced the level of phosphorylated glycogen synthase kinase 3ß at the same time point (2.99±0.24), P<0.001. In WT MHEC, the levels of Wnt-5a were toned down and the level of p-GSK-3ß was lowest at 2 h (0.47±0.06), P< 0.01 compared to uninfected control. This was accompanied by a continuous significant increase in the nuclear colocalization of ß-catenin/YAP in TSP1 KO MHEC with a maximum Pearson correlation coefficient of (0.67±0.02), P< 0.05 at 6 h. In WT MHEC, the nuclear colocalization of ß-catenin/YAP remained steady and showed a reduction at 6 h (0.29±0.007), P< 0.05. These results indicate that TSP1 plays an important role in regulating ß-catenin/YAP colocalization during the early phase of T. cruzi infection. Importantly, dysregulation of this crosstalk by pre-incubation of WT MHEC with a ß-catenin inhibitor, endo-IWR 1, dramatically reduced the level of infection of WT MHEC. Parasite infectivity of inhibitor treated WT MHEC was similar to the level of infection of TSP1 KO MHEC. These results indicate that the ß-catenin pathway induced by the parasite and regulated by TSP1 during the early phase of T. cruzi infection is an important potential therapeutic target, which can be explored for the prophylactic prevention of T. cruzi infection.


Subject(s)
Chagas Disease/pathology , Hippo Signaling Pathway/physiology , Thrombospondin 1/metabolism , Wnt Signaling Pathway/physiology , YAP-Signaling Proteins/metabolism , beta Catenin/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Endothelial Cells/parasitology , Endothelium/cytology , Endothelium/parasitology , Glycogen Synthase Kinase 3 beta/metabolism , Heart/parasitology , Mice , Mice, Knockout , Rats , Thrombospondin 1/genetics , Trypanosoma cruzi/metabolism , Wnt-5a Protein/metabolism , beta Catenin/antagonists & inhibitors
8.
Int J Cancer ; 150(5): 727-740, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34536299

ABSTRACT

Wnt signaling plays an essential role in the initiation and progression of various types of cancer. Besides, the Wnt pathway components have been established as reliable biomarkers and potential targets for cancer therapy. Wnt signaling is categorized into canonical and noncanonical pathways. The canonical pathway is involved in cell survival, proliferation, differentiation and migration, while the noncanonical pathway regulates cell polarity and migration. Apart from its biological role in development and homeostasis, the Wnt pathway has been implicated in several pathological disorders, including cancer. As a result, inhibiting this pathway has been a focus of cancer research with multiple targetable candidates in development. In this review, our focus will be to summarize information about ongoing and completed clinical trials targeting various Wnt pathway components, along with describing current and emerging Wnt targeted therapies. In addition, we will discuss potential opportunities and associated challenges of inhibiting Wnt signaling for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Acyltransferases/antagonists & inhibitors , Animals , Humans , Membrane Proteins/antagonists & inhibitors , Tankyrases/antagonists & inhibitors , Wnt Signaling Pathway/physiology , beta Catenin/antagonists & inhibitors
9.
Gastroenterology ; 162(1): 253-268.e13, 2022 01.
Article in English | MEDLINE | ID: mdl-34534538

ABSTRACT

BACKGROUND & AIMS: A major clinical challenge for patients with pancreatic cancer (PC) is metabolic adaptation. Neoplastic cells harboring molecular perturbations suffice for their increased anabolic demand and nucleotide biosynthesis to acquire chemoresistance. The mucin 5AC expressed de novo in malignant pancreas promotes cancer cell stemness and is significantly associated with poor patient survival. Identification of MUC5AC-associated drivers of chemoresistance through metabolic alterations may facilitate the sculpting of a new combinatorial regimen. METHODS: The contributions of MUC5AC to glutaminolysis and gemcitabine resistance were examined by The Cancer Genome Atlas data analysis, RNA sequencing, and immunohistochemistry analysis on pancreatic tissues of KrasG12D;Pdx1-Cre (KC) and KrasG12D;Pdx1-Cre;Muc5ac-/- mice. These were followed by metabolite flux assays as well as biochemical and xenograft studies on MUC5AC-depleted human and murine PC cells. Murine and human pancreatic 3-dimensional tumoroids were used to evaluate the efficacy of gemcitabine in combination with ß-catenin and glutaminolysis inhibitors. RESULTS: Transcriptional analysis showed that high MUC5AC-expressing human and autochthonous murine PC tumors exhibit higher resistance to gemcitabine because of enhanced glutamine use and nucleotide biosynthesis. Gemcitabine treatment led to MUC5AC overexpression, resulting in disruption of E-cadherin/ß-catenin junctions and the nuclear translocation of ß-catenin, which increased c-Myc expression, with a concomitant rise in glutamine uptake and glutamate release. MUC5AC depletion and glutamine deprivation sensitized human PC cells to gemcitabine, which was obviated by glutamine replenishment in MUC5AC-expressing cells. Coadministration of ß-catenin and glutaminolysis inhibitors with gemcitabine abrogated the MUC5AC-mediated resistance in murine and human tumoroids. CONCLUSIONS: The MUC5AC/ß-catenin/c-Myc axis increases the uptake and use of glutamine in PC cells, and cotargeting this axis along with gemcitabine may improve therapeutic efficacy in PC.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Energy Metabolism/drug effects , Glutamine/metabolism , Mucin 5AC/metabolism , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Databases, Genetic , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic , Glutaminase/antagonists & inhibitors , Glutaminase/metabolism , Humans , Male , Mice, Knockout , Mice, Nude , Mucin 5AC/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , Gemcitabine
10.
J Enzyme Inhib Med Chem ; 37(1): 118-124, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34894966

ABSTRACT

Hepatic stellate cells activation (HSCs) plays a crucial role in the pathogenesis of liver fibrosis. Specific microRNAs have been suggested to affect the activation of HSCs via various signalling pathways including TGF-ß/smads and Wnt/ß-catenin pathways. Dasatinib is a multitarget inhibitor of many tyrosine kinases has recently studied for its anti-fibrotic effects in a variety of fibrous diseases. This study investigated the role of modulation of miRNA-378 and miRNA-17 in the pathogenesis of liver fibrosis through altering Wnt/ß-catenin and TGF-ß/smads pathways and evaluated the beneficial effect of the tyrosine kinase inhibitor, dasatinib, in thioacetamide-induced liver fibrosis model in mice. Treatment with dasatinib down-regulated miRNA-17 expression, leading to the restoration of WiF-1 and smad-7 which cause the inhibition of both Wnt/ß-catenin and TGF-ß/smads signalling. In addition, it upregulated miRNA-378 leading to the decrease of Wnt-10 which contributes to the suppression of activated HSCs.


Subject(s)
Dasatinib/pharmacology , Liver Cirrhosis/drug therapy , MicroRNAs/antagonists & inhibitors , Smad7 Protein/metabolism , Thioacetamide/antagonists & inhibitors , Animals , Dasatinib/chemistry , Dose-Response Relationship, Drug , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mice , MicroRNAs/metabolism , Molecular Structure , Structure-Activity Relationship , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
11.
Comput Math Methods Med ; 2021: 6753926, 2021.
Article in English | MEDLINE | ID: mdl-34966442

ABSTRACT

MicroRNA (miRNA) dysfunction has been confirmed as a key event of ischemic stroke appearance. This study is aimed at revealing the role of miR-429 in the angiogenesis of HBMECs. The HBMECs were treated with oxygen and glucose deprivation (OGD) to establish the ischemic cell model. The qRT-PCR was used to measure the expression levels of the miR-429 in the serums of the patients or cells, and CCK-8, wound healing assay, and tube formation assay were used to observe the effects of miR-429 on the phenotype of HBMECs. Moreover, the Targetscan, dual-luciferase reporter assay, and Western blot were used to reveal the downstream target and regulation mechanism of miR-429 in OGD-induced HBMECs. The results showed that miR-429 was significantly upregulated in the serums of the patients, and overexpressed miR-429 could extremely inhibit the viability, migration, and tube formation of OGD-induced HBMECs. Furthermore, it was found that SNAI2 was a downstream factor of miR-429, and SNAI2 could rescue the effects of miR-429 on OGD-induced HBMECs. Besides, the Western blot showed that miR-429 could affect the activity of GSK-3ß/ß-catenin pathway via inhibiting the expression of SNAI2. In conclusion, this study suggests that miR-429 inhibits the angiogenesis of HBMECs through SNAI2-mediated GSK-3ß/ß-catenin pathway.


Subject(s)
Brain/blood supply , Glycogen Synthase Kinase 3 beta/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Snail Family Transcription Factors/genetics , beta Catenin/genetics , 3' Untranslated Regions , Brain/metabolism , Brain/pathology , Cells, Cultured , Computational Biology , Disease Progression , Endothelial Cells/metabolism , Endothelial Cells/pathology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Ischemic Stroke/blood , Ischemic Stroke/genetics , MicroRNAs/metabolism , Models, Cardiovascular , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Signal Transduction/genetics , Snail Family Transcription Factors/metabolism , Up-Regulation , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
12.
Exp Cell Res ; 409(2): 112912, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34762897

ABSTRACT

Rapid proliferation, high stemness potential, high invasiveness and apoptotic evasion are the distinctive hallmarks of glioma malignancy. The dysregulation of the Wnt/ß-catenin pathway is the key factor regulating glioma malignancy. Wnt antagonist, secreted frizzled-related protein 4 (sFRP4), which has a prominent pro-apoptotic role in glioma stem cells, has two functional domains, the netrin-like domain (NLD), and cysteine-rich domain (CRD) both of which contribute to apoptotic properties of the whole protein. However, there are no reports elucidating the specific effects of individual domains of sFRP4 in inhibiting the invasive properties of glioma. This study explores the efficacy of the domains of sFRP4 in inhibiting the key hallmarks of glioblastoma such as invasion, metastasis, and stemness. We overexpressed sFRP4 and its domains in the glioblastoma cell line, U87MG cells and observed that both CRD and NLD domains played prominent roles in attenuating cancer stem cell properties. Significantly, we could demonstrate for the first time that both NLD and CRD domains negatively impacted the key driver of metastasis and migration, the matrix metalloproteinase-2 (MMP-2). Mechanistically, compared to CRD, NLD domain suppressed MMP-2 mediated invasion more effectively in glioma cells as observed in matrigel invasion assay and a function-blocking antibody assay. Fluorescent matrix degradation assay further revealed that NLD reduces matrix degradation. NLD also significantly disrupted fibronectin assembly and decreased cell adhesion in another glioma cell line LN229. In conclusion, the NLD peptide of sFRP4 could be a potent short peptide therapeutic candidate for targeting MMP-2-mediated invasion in the highly malignant glioblastoma multiforme.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Matrix Metalloproteinase 2/chemistry , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Apoptosis , Cell Cycle , Cell Movement , Cell Proliferation , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , beta Catenin/genetics , beta Catenin/metabolism
13.
Bioorg Chem ; 117: 105449, 2021 12.
Article in English | MEDLINE | ID: mdl-34736136

ABSTRACT

Lung cancer is one of the most malignant tumors with the highest mortality and morbidity. The tubers of Bletilla striata are known as "an excellent medicine for lung diseases" in traditional Chinese medicine. This study performed a targeted study to explore compounds with anti-lung cancer activity and the molecular mechanisms using A549 cells. Eighteen bibenzyl derivatives, including four new compounds (13, 14, 16, and 18), were isolated from the tubers of B. striata. Analysis of the structure-activity relationship indicated that the cytotoxicity of the bibenzyls against A549 cells increased gradually as the number of the benzyl groups in the structures increased. Bletillain (18), an unusual benzyl polymer, was found to be the most active compound. Further flow cytometric analysis, dual-luciferase assays, real-time PCR assays, and western blot assays revealed that bletillain induced autophagy in A549 cells by regulating the Akt/GSK-3ß/ß-catenin signaling pathway. Beclin 1, LC3, and p62 are downstream autophagy factors of Akt, and Beclin 1 was the key autophagy factor. These results suggested that bibenzyls of B. striata play important roles in the treatment of lung cancer and provided scientific evidence illustrating why the tubers of B. striata are a suitable medicine for the treatment of lung cancer in traditional Chinese medicine.


Subject(s)
Autophagy/drug effects , Drug Discovery , A549 Cells , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Molecular Structure , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Tumor Cells, Cultured , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
14.
Cell Mol Life Sci ; 78(23): 7831-7849, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34724098

ABSTRACT

Randall's plaques (RP) are well established as precursor lesions of idiopathic calcium oxalate (CaOx) stones, and the process of biomineralization driven by osteogenic-like cells has been highlighted in RP formation, but the mechanism is poorly understood. Given the inhibitory role of α-Klotho (KL), an aging suppressor protein with high expression in kidneys, in ectopic calcification and the close association between KL gene polymorphisms and urolithiasis susceptibility, we determined the potential role of KL in RP formation. This study found that both soluble KL (s-KL) and transmembrane KL (m-KL) were downregulated, and that s-KL but not m-KL was inversely correlated with upregulation of osteogenic markers in RP tissues. Additionally, s-KL expression was markedly suppressed in human renal interstitial fibroblasts (hRIFs) and slightly suppressed in HK-2 cells after osteogenic induction, intriguingly, which was echoed to the greater osteogenic capability of hRIFs than HK-2 cells. Further investigations showed the inhibitory effect of s-KL on hRIF osteogenic differentiation in vitro and in vivo. Moreover, coculture with recombinant human KL (r-KL) or HK-2 cells suppressed osteogenic differentiation of hRIFs, and this effect was abolished by coculture with KL-silenced HK-2 cells or the ß-catenin agonist SKL2001. Mechanistically, s-KL inactivated the Wnt-ß-catenin pathway by directly binding to Wnt2 and upregulating SFRP1. Further investigations identified activation of the Wnt-ß-catenin pathway and downregulation of SFRP1 and DKK1 in RP tissues. In summary, this study identified s-KL deficiency as a pathological feature of RP and revealed that s-KL released from HK-2 cells inhibited osteogenic differentiation of hRIFs by inactivating the Wnt-ß-catenin pathway, not only providing in-depth insight into the role of s-KL in renal interstitial biomineralization but also shedding new light on the interaction of renal tubular epithelial cells with interstitial cells to clarify RP formation.


Subject(s)
Cell Differentiation , Fibroblasts/pathology , Kidney Calculi/pathology , Klotho Proteins/metabolism , Osteogenesis , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Calculi/genetics , Kidney Calculi/metabolism , Kidney Medulla/metabolism , Kidney Medulla/pathology , Klotho Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
15.
Life Sci ; 286: 120027, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34627778

ABSTRACT

Pulmonary fibrosis (PF) caused by paraquat remains a critical issue, and the molecular mechanisms are still unclear. Epithelial-mesenchymal transition (EMT) is regarded as a hallmark of PF, conferring alveolar epithelial cells partial mesenchymal characteristics, facilitating migration, expressing excessive extracellular matrix components, and participating in lung parenchyma remodeling and stiffening. Aberration of Wnt signaling has been identified in EMT and PF, and Wnt protein family consists of 19 ligands. The relationship of the specific Wnt ligands and fibrogenesis induced by PQ was not well defined. In current study, PQ-induced lung fibrosis rat model and EMT cell model were utilized to investigate the underlying molecular mechanisms both in vivo and in vitro. The results demonstrated that canonical Wnt/ß-catenin signaling was highly activated and Wnt10b was the most affected. Additionally, suppression of Wnt10b by RNA interference could reverse EMT in vitro and detain the process of PF in vivo. These data establish Wnt10b as the key regulator of EMT and lung fibrogenesis, and suggest the potential of targeted interference against Wnt10b as a promising therapeutic strategy for lung fibrosis.


Subject(s)
Herbicides/toxicity , Paraquat/toxicity , Proto-Oncogene Proteins/antagonists & inhibitors , Pulmonary Fibrosis/prevention & control , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Cell Line , Epithelial-Mesenchymal Transition , Humans , Male , Proto-Oncogene Proteins/metabolism , Pulmonary Fibrosis/chemically induced , Rats , Rats, Sprague-Dawley , Wnt Proteins/metabolism , beta Catenin/metabolism
16.
Oncogene ; 40(50): 6720-6735, 2021 12.
Article in English | MEDLINE | ID: mdl-34657130

ABSTRACT

Epithelial-mesenchymal transition (EMT) refers to the acquisition of mesenchymal properties in cells participating in tumor progression. One hallmark of EMT is the increased level of active ß-catenin, which can trigger the transcription of Wnt-specific genes responsible for the control of cell fate. We investigated how Monocyte Chemotactic Protein-1-Induced Protein-1 (MCPIP1), a negative regulator of inflammatory processes, affects EMT in a clear cell renal cell carcinoma (ccRCC) cell line, patient tumor tissues and a xenotransplant model. We showed that MCPIP1 degrades miRNAs via its RNase activity and thus protects the mRNA transcripts of negative regulators of the Wnt/ß-catenin pathway from degradation, which in turn prevents EMT. Mechanistically, the loss of MCPIP1 RNase activity led to the upregulation of miRNA-519a-3p, miRNA-519b-3p, and miRNA-520c-3p, which inhibited the expression of Wnt pathway inhibitors (SFRP4, KREMEN1, CXXC4, CSNK1A1 and ZNFR3). Thus, the level of active nuclear ß-catenin was increased, leading to increased levels of EMT inducers (SNAI1, SNAI2, ZEB1 and TWIST) and, consequently, decreased expression of E-cadherin, increased expression of mesenchymal markers, and acquisition of the mesenchymal phenotype. This study revealed that MCPIP1 may act as a tumor suppressor that prevents EMT by stabilizing Wnt inhibitors and decreasing the levels of active ß-catenin and EMT inducers.


Subject(s)
Carcinoma, Renal Cell/pathology , Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/physiology , MicroRNAs/antagonists & inhibitors , Ribonucleases/metabolism , Transcription Factors/metabolism , Wnt1 Protein/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Ribonucleases/genetics , Transcription Factors/genetics , Tumor Cells, Cultured , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
17.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34614511

ABSTRACT

The Wnt/ß-catenin pathway is upregulated in uterine leiomyomas, the most common benign tumors in the female reproductive tract. Simvastatin is an antihyperlipidemic drug, and previous in vitro and in vivo reports showed that it may have therapeutic effects in treating leiomyomas. The objective of this study was to examine the effects of simvastatin on the Wnt/ß-catenin signaling pathway in leiomyoma. We treated primary and immortalized human leiomyoma cells with simvastatin and examined its effects using quantitative real-time polymerase chain reaction, Western blotting, and immunocytochemistry. We also examined the effects using human leiomyoma tissues from an ongoing randomized controlled trial in which women with symptomatic leiomyoma received simvastatin (40 mg) or placebo for 3 months prior to their surgery. The results of this study revealed that simvastatin significantly reduced the expression of Wnt4 and its co-receptor LRP5. After simvastatin treatment, levels of total ß-catenin and its active form, nonphosphorylated ß-catenin, were reduced in both cell types. Additionally, simvastatin reduced the expression of Wnt4 and total ß-catenin, as well as nonphosphorylated ß-catenin protein expression in response to estrogen and progesterone. Simvastatin also inhibited the expression of c-Myc, a downstream target of the Wnt/ß-catenin pathway. The effect of simvastatin on nonphosphorylated-ß-catenin, the key regulator of the Wnt/ß-catenin pathway, was recapitulated in human leiomyoma tissue. These results suggest that simvastatin may have a beneficial effect on uterine leiomyoma through suppressing the overactive Wnt/ß-catenin pathway.


Subject(s)
Leiomyoma/pathology , Simvastatin/pharmacology , Uterine Neoplasms/pathology , Wnt Signaling Pathway/drug effects , Adolescent , Adult , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Clinical Trials, Phase II as Topic , Double-Blind Method , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Humans , Leiomyoma/genetics , Leiomyoma/metabolism , Middle Aged , Primary Cell Culture , Randomized Controlled Trials as Topic , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Young Adult , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
18.
Expert Opin Ther Targets ; 25(9): 701-719, 2021 09.
Article in English | MEDLINE | ID: mdl-34633266

ABSTRACT

AREAS COVERED: This perspective discusses the challenges of targeting the Wnt signaling cascade, the safety, efficacy, and therapeutic potential of specific CBP/ß-catenin antagonists and a rationale for the pleiotropic effects of CBP/ß-catenin antagonists beyond Wnt signaling. EXPERT OPINION: CBP/ß-catenin antagonists can correct lineage infidelity, enhance wound healing, both normal and aberrant (e.g. fibrosis) and force the differentiation and lineage commitment of stem cells and cancer stem cells by regulating enhancer and super-enhancer coactivator occupancy. Small molecule CBP/ß-catenin antagonists rebalance the equilibrium between CBP/ß-catenin versus p300/ß-catenin dependent transcription and may be able to treat or prevent many diseases of aging, via maintenance of our somatic stem cell pool, and regulating mitochondrial function and metabolism involved in differentiation and immune cell function.


Subject(s)
CREB-Binding Protein/antagonists & inhibitors , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors , Cell Differentiation/drug effects , Humans , Neoplastic Stem Cells/drug effects , Stem Cells/drug effects
19.
DNA Cell Biol ; 40(10): 1308-1316, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34520253

ABSTRACT

Diabetic nephropathy (DN) is a complication of diabetes, which leads to most end-stage kidney diseases and threatens health of patients. Mucin 1 (MUC1) is a heterodimeric oncoprotein, which is abnormally expressed in tumors and hematologic diseases. The aim of this study is to clarify the mechanism and role of MUC1 in DN. The mesangial cells (MCs) suffered from high glucose (HG) treatment to mimic DN in vitro. The cell proliferation was detected by Cell Counting Kit-8 assay and 5-ethynyl-2-deoxyuridine (EdU) staining assay. The expression of MUC1 and fibrosis markers: fibronectin, collagen I, and collagen IV were assessed by western blot. In this study, we demonstrated that HG treatment induced MUC1 expression in MCs. With knockdown of MUC1 or overexpressed MUC1 in MCs, the results indicated that knockdown of MUC1 inhibited MCs proliferation and reduced kidney fibrosis markers expression, including fibronectin, collagen I, and collagen IV, whereas overexpression of MUC1 led to opposite results. Mechanically, MUC1 activated signal transducers and activators of transcription (STAT) and ß-catenin signal pathway. After added AG490 (STAT inhibitor) or FH535 (ß-catenin inhibitor), blocking STAT3 and ß-catenin signal pathway attenuated MUC1-induced cell proliferation and fibronectin production in MCs. Finally, knockdown of MUC1 attenuated DN-induced kidney fibrosis in db/db mice. Therapeutic target for DN. In conclusion, MUC1 promotes MCs proliferation and kidney fibrosis in DN through activating STAT and ß-catenin signal pathway, which can help to provide a novel therapeutic target for DN.


Subject(s)
Cell Proliferation , Diabetic Nephropathies/metabolism , Mesangial Cells/metabolism , Mucin-1/metabolism , Signal Transduction , Animals , Cells, Cultured , Fibronectins/metabolism , Fibrosis , Kidney/metabolism , Kidney/pathology , Male , Mesangial Cells/physiology , Mice , Mice, Inbred C57BL , Mucin-1/genetics , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , Sulfonamides/pharmacology , Tyrphostins/pharmacology , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
20.
Cells ; 10(9)2021 09 20.
Article in English | MEDLINE | ID: mdl-34572136

ABSTRACT

Retrospective observational studies have reported that statins improve clinical outcomes in patients previously treated with programmed cell death protein 1 (PD-1)-targeting monoclonal antibodies for malignant pleural mesothelioma (MPM) and advanced non-small cell lung cancer (NSCLC). In multiple mouse cancer models, de novo synthesis of mevalonate and cholesterol inhibitors was found to synergize with anti-PD-1 antibody therapy. In the present study, we investigated whether statins affect programmed death-ligand 1 (PD-L1) expression in cancer cells. Four statins, namely simvastatin, atorvastatin, lovastatin, and fluvastatin, decreased PD-L1 expression in melanoma and lung cancer cells. In addition, we found that AKT and ß-catenin signaling involved PD-L1 suppression by statins. Our cellular and molecular studies provide inspiring evidence for extending the clinical evaluation of statins for use in combination with immune checkpoint inhibitor-based cancer therapy.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Apoptosis , Cell Proliferation , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...