Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.159
Filter
1.
Mol Biol Rep ; 51(1): 842, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042261

ABSTRACT

BACKGROUND: Although Huaier granules can be used as prospective anti-cholangiocarcinoma drugs, the mechanism of action of Huaier granules in cholangiocarcinoma is not clear. The anti-cholangiocarcinoma effect of Huaier granules was validated in cell line research. In vitro experiments were conducted to investigate the signalling pathways affected by Huaier in CCA cells. METHODS AND RESULTS: Real-time quantitative PCR (RT‒qPCR) and Western blot analysis were performed to analyse gene expression in CCA cells. MTT assays, scratch tests, and Transwell assays were used to explore the effects on the proliferation and metastasis of CCA cells. Chromatin immunoprecipitation assays were performed to reveal the potential underlying mechanisms involved. Twist1 was upregulated in human CCA tissues. In addition, its expression levels were negatively related to FBP1 expression levels. Mechanistically, Twist1 can bind to the region of the FBP1 promoter to reduce its expression. Huaier plays an indispensable role in suppressing Twist1 expression to inhibit the Twist1/FBP1/Wnt/ß-catenin axis. Then, we verified the effect of Huaier in vitro. CONCLUSIONS: These findings suggested that Huaier granules were capable of inhibiting CCA development through regulating the Twist1/FBP1/Wnt/ß-catenin signalling axis and provided a novel orientation for the development of novel anti-CCA drugs.


Subject(s)
Bile Duct Neoplasms , Cell Proliferation , Cholangiocarcinoma , Gene Expression Regulation, Neoplastic , Nuclear Proteins , Twist-Related Protein 1 , Wnt Signaling Pathway , beta Catenin , Humans , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Cholangiocarcinoma/drug therapy , Wnt Signaling Pathway/drug effects , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cell Line, Tumor , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Cell Proliferation/drug effects , beta Catenin/metabolism , Cell Movement/drug effects , Cell Movement/genetics
2.
Oncol Rep ; 52(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38963044

ABSTRACT

Lysine methyltransferase 5A (KMT5A) is the sole mammalian enzyme known to catalyse the mono­methylation of histone H4 lysine 20 and non­histone proteins such as p53, which are involved in the occurrence and progression of numerous cancers. The present study aimed to determine the function of KMT5A in inducing docetaxel (DTX) resistance in patients with breast carcinoma by evaluating glucose metabolism and the underlying mechanism involved. The upregulation or downregulation of KMT5A­related proteins was examined after KMT5A knockdown in breast cancer (BRCA) cells by Tandem Mass Tag proteomics. Through differential protein expression and pathway enrichment analysis, the upregulated key gluconeogenic enzyme fructose­1,6­bisphosphatase 1 (FBP1) was discovered. Loss of FBP1 expression is closely related to the development and prognosis of cancers. A dual­luciferase reporter gene assay confirmed that KMT5A inhibited the expression of FBP1 and that overexpression of FBP1 could enhance the chemotherapeutic sensitivity to DTX through the suppression of KMT5A expression. The KMT5A inhibitor UNC0379 was used to verify that DTX resistance induced by KMT5A through the inhibition of FBP1 depended on the methylase activity of KMT5A. According to previous literature and interaction network structure, it was revealed that KMT5A acts on the transcription factor twist family BHLH transcription factor 1 (TWIST1). Then, it was verified that TWSIT1 promoted the expression of FBP1 by using a dual­luciferase reporter gene experiment. KMT5A induces chemotherapy resistance in BRCA cells by promoting cell proliferation and glycolysis. After the knockdown of the KMT5A gene, the FBP1 related to glucose metabolism in BRCA was upregulated. KMT5A knockdown expression and FBP1 overexpression synergistically inhibit cell proliferation and block cells in the G2/M phase. KMT5A inhibits the expression of FBP1 by methylating TWIST1 and weakening its promotion of FBP1 transcription. In conclusion, KMT5A was shown to affect chemotherapy resistance by regulating the cell cycle and positively regulate glycolysis­mediated chemotherapy resistance by inhibiting the transcription of FBP1 in collaboration with TWIST1. KMT5A may be a potential therapeutic target for chemotherapy resistance in BRCA.


Subject(s)
Breast Neoplasms , Docetaxel , Drug Resistance, Neoplasm , Fructose-Bisphosphatase , Gene Expression Regulation, Neoplastic , Nuclear Proteins , Twist-Related Protein 1 , Humans , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Female , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Fructose-Bisphosphatase/genetics , Fructose-Bisphosphatase/metabolism , Docetaxel/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Cell Proliferation/drug effects , DNA Methylation
3.
Dis Model Mech ; 17(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38967226

ABSTRACT

Robinow syndrome is a rare disease caused by variants of seven WNT pathway genes. Craniofacial features include widening of the nasal bridge and jaw hypoplasia. We used the chicken embryo to test whether two missense human FZD2 variants (1301G>T, p.Gly434Val; 425C>T, p.Pro142Lys) were sufficient to change frontonasal mass development. In vivo, the overexpression of retroviruses with wild-type or variant human FZD2 inhibited upper beak ossification. In primary cultures, wild-type and variant human FZD2 significantly inhibited chondrogenesis, with the 425C>T variant significantly decreasing activity of a SOX9 luciferase reporter compared to that for the wild type or 1301G>T. Both variants also increased nuclear shuttling of ß-catenin (CTNNB1) and increased the expression of TWIST1, which are inhibitory to chondrogenesis. In canonical WNT luciferase assays using frontonasal mass cells, the variants had dominant-negative effects on wild-type FZD2. In non-canonical assays, the 425C>T variant failed to activate the reporter above control levels and was unresponsive to exogenous WNT5A. This is the first single amino acid change to selectively alter ligand binding in a FZD receptor. Therefore, FZD2 missense variants are pathogenic and could lead to the altered craniofacial morphogenesis seen in Robinow syndrome.


Subject(s)
Chondrogenesis , Craniofacial Abnormalities , Frizzled Receptors , Animals , Chick Embryo , Humans , Beak , beta Catenin/metabolism , Cell Nucleus/metabolism , Chondrogenesis/genetics , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Dwarfism , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Limb Deformities, Congenital , Skull/pathology , Skull/embryology , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Urogenital Abnormalities , Wnt Signaling Pathway
4.
J Clin Invest ; 134(16)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042469

ABSTRACT

Crohn's disease (CD) is marked by recurring intestinal inflammation and tissue injury, often resulting in fibrostenosis and bowel obstruction, necessitating surgical intervention with high recurrence rates. To elucidate the mechanisms underlying fibrostenosis in CD, we analyzed the transcriptome of cells isolated from the transmural ileum of patients with CD, including a trio of lesions from each patient: non-affected, inflamed, and stenotic ileum samples, and compared them with samples from patients without CD. Our computational analysis revealed that profibrotic signals from a subset of monocyte-derived cells expressing CD150 induced a disease-specific fibroblast population, resulting in chronic inflammation and tissue fibrosis. The transcription factor TWIST1 was identified as a key modulator of fibroblast activation and extracellular matrix (ECM) deposition. Genetic and pharmacological inhibition of TWIST1 prevents fibroblast activation, reducing ECM production and collagen deposition. Our findings suggest that the myeloid-stromal axis may offer a promising therapeutic target to prevent fibrostenosis in CD.


Subject(s)
Crohn Disease , Fibroblasts , Fibrosis , Monocytes , Twist-Related Protein 1 , Crohn Disease/metabolism , Crohn Disease/pathology , Crohn Disease/immunology , Humans , Fibroblasts/metabolism , Fibroblasts/pathology , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Monocytes/metabolism , Monocytes/pathology , Monocytes/immunology , Male , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Female , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Ileum/pathology , Ileum/metabolism , Ileum/immunology , Cell Communication , Adult , Endopeptidases/metabolism , Endopeptidases/genetics , Animals , Mice
5.
Exp Mol Pathol ; 138: 104909, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38876079

ABSTRACT

Epithelial-mesenchymal transition (EMT) plays a pivotal role in the development and progression of many cancers. Partial EMT (pEMT) could represent a critical step in tumor migration and dissemination. Sarcomatoid renal cell carcinoma (sRCC) is an aggressive form of renal cell carcinoma (RCC) composed of a carcinomatous (sRCC-Ca) and sarcomatous (sRCC-Sa) component. The role of (p)EMT in the progression of RCC to sRCC remains unclear. The aim of this study was to investigate the involvement of (p)EMT in RCC and sRCC. Tissue samples from 10 patients with clear cell RCC (ccRCC) and 10 patients with sRCC were selected. The expression of main EMT markers (miR-200 family, miR-205, SNAI1/2, TWIST1/2, ZEB1/2, CDH1/2, VIM) was analyzed by qPCR in ccRCC, sRCC-Ca, and sRCC-Sa and compared to non-neoplastic tissue and between both groups. Expression of E-cadherin, N-cadherin, vimentin and ZEB2 was analyzed using immunohistochemistry. miR-200c was downregulated in sRCC-Ca compared to ccRCC, while miR-200a was downregulated in sRCC-Sa compared to ccRCC. CDH1 was downregulated in sRCC-Sa when compared to any other group. ZEB2 was downregulated in ccRCC and sRCC compared to corresponding non-neoplastic kidney. A positive correlation was observed between CDH1 expression and miR-200a/b/c. Our results suggest that full EMT is not present in sRCC. Instead, discreet molecular differences exist between ccRCC, sRCC-Ca, and sRCC-Sa, possibly representing distinct intermediary states undergoing pEMT.


Subject(s)
Biomarkers, Tumor , Carcinoma, Renal Cell , Epithelial-Mesenchymal Transition , Kidney Neoplasms , MicroRNAs , Vimentin , Zinc Finger E-box Binding Homeobox 2 , Humans , Epithelial-Mesenchymal Transition/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , MicroRNAs/genetics , Male , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Vimentin/metabolism , Vimentin/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism , Aged , Cadherins/genetics , Cadherins/metabolism , Gene Expression Regulation, Neoplastic , Antigens, CD/genetics , Antigens, CD/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/metabolism , Adult , Nuclear Proteins
6.
Arch Oral Biol ; 165: 106017, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38852529

ABSTRACT

OBJECTIVE: To evaluate and compare the expression of E-cadherin, Snail1 and Twist1 in pleomorphic adenomas (PAs), adenoid cystic carcinomas (AdCCa) and carcinoma ex-pleomorphic adenomas (CaexPA) of salivary glands, as well as investigate possible associations with clinicopathological parameters. STUDY DESIGN: E-cadherin, Snail1 and Twist1 antibody immunostaining were analyzed semiquantitatively in 20 PAs, 20 AdCCas and 10 CaexPAs. Cases were classified as low and high expression for analysis of the association with clinicopathological parameters. RESULTS: Compared to PAs, AdCCas and CaexPAs exhibited higher nuclear expression of Snail1 (p = 0.021 and p = 0.028, respectively) and Twist1 (p = 0.009 and p = 0.001). Membranous and cytoplasmic expression of E-cadherin were positively correlated in PAs, AdCCas and CaexPAs (r = 0.645, p = 0.002; r = 0.824, p < 0.001; r = 0.677, p = 0.031). In PAs, positive correlation was found between nuclear expression of Snail1 and membrane expression of E-cadherin (r = 0.634; p = 0.003), as well as between nuclear expression of Snail1 and Twist1 (r = 0.580; p = 0.007). Negative correlations were detected between membrane expression of E-cadherin and cytoplasmic expression of Snail1 in AdCCas (r = - 0.489; p = 0.029). CONCLUSIONS: E-cadherin, Twist1, and Snail1 may participate in modulating events related to cell differentiation and adhesion in PAs and to biological behavior in AdCCas and CaexPAs, which indicates the involvement of EMT in these processes. Furthermore, the expression of these proteins in these carcinomas may reflect the plasticity feature of EMT.


Subject(s)
Adenoma, Pleomorphic , Cadherins , Carcinoma, Adenoid Cystic , Epithelial-Mesenchymal Transition , Nuclear Proteins , Salivary Gland Neoplasms , Snail Family Transcription Factors , Twist-Related Protein 1 , Humans , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/metabolism , Snail Family Transcription Factors/metabolism , Cadherins/metabolism , Female , Male , Twist-Related Protein 1/metabolism , Middle Aged , Carcinoma, Adenoid Cystic/pathology , Carcinoma, Adenoid Cystic/metabolism , Nuclear Proteins/metabolism , Adult , Adenoma, Pleomorphic/metabolism , Adenoma, Pleomorphic/pathology , Aged , Twist Transcription Factors/metabolism , Immunohistochemistry , Transcription Factors/metabolism , Biomarkers, Tumor/metabolism
7.
Carbohydr Polym ; 341: 122298, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38876710

ABSTRACT

Cutaneous melanoma is a lethal skin cancer variant with pronounced aggressiveness and metastatic potential. However, few targeted medications inhibit the progression of melanoma. Ganoderma lucidum, which is a type of mushroom, is widely used as a non-toxic alternative adjunct therapy for cancer patients. This study determines the effect of WSG, which is a water-soluble glucan that is derived from G. lucidum, on melanoma cells. The results show that WSG inhibits cell viability and the mobility of melanoma cells. WSG induces changes in the expression of epithelial-to-mesenchymal transition (EMT)-related markers. WSG also downregulates EMT-related transcription factors, Snail and Twist. Signal transduction assays show that WSG reduces the protein levels in transforming growth factor ß receptors (TGFßRs) and consequently inhibits the phosphorylation of intracellular signaling molecules, such as FAK, ERK1/2 and Smad2. An In vivo study shows that WSG suppresses melanoma growth in B16F10-bearing mice. To enhance transdermal drug delivery and prevent oxidation, two highly biocompatible compounds, polyvinyl alcohol (PVA) and polyvinylpyrrolidone (PVP), are used to synthesize a dissolvable microneedle patch that is loaded with WSG (MN-WSG). A functional assay shows that MN-WSG has an effect that is comparable to that of WSG alone. These results show that WSG has significant potential as a therapeutic agent for melanoma treatment. MN-WSG may allow groundbreaking therapeutic approaches and offers a novel method for delivering this potent compound effectively.


Subject(s)
Reishi , Snail Family Transcription Factors , Animals , Mice , Reishi/chemistry , Snail Family Transcription Factors/metabolism , Humans , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Cell Line, Tumor , Twist-Related Protein 1/metabolism , Epithelial-Mesenchymal Transition/drug effects , Cell Survival/drug effects , Mice, Inbred C57BL , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Cell Movement/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Polyvinyl Alcohol/chemistry , Polysaccharides/pharmacology , Polysaccharides/chemistry , Signal Transduction/drug effects
8.
PeerJ ; 12: e17360, 2024.
Article in English | MEDLINE | ID: mdl-38737746

ABSTRACT

Breast cancer is the most common invasive neoplasm and the leading cause of cancer death in women worldwide. The main cause of mortality in cancer patients is invasion and metastasis, where the epithelial-mesenchymal transition (EMT) is a crucial player in these processes. Pharmacological therapy has plants as its primary source, including isoflavonoids. Brazilin is an isoflavonoid isolated from Haematoxilum brasiletto that has shown antiproliferative activity in several cancer cell lines. In this study, we evaluated the effect of Brazilin on canonical markers of EMT such as E-cadherin, vimentin, Twist, and matrix metalloproteases (MMPs). By Western blot, we evaluated E-cadherin, vimentin, and Twist expression and the subcellular localization by immunofluorescence. Using gelatin zymography, we determined the levels of secretion of MMPs. We used Transwell chambers coated with matrigel to determine the in vitro invasion of breast cancer cells treated with Brazilin. Interestingly, our results show that Brazilin increases 50% in E-cadherin expression and decreases 50% in vimentin and Twist expression, MMPs, and cell invasion in triple-negative breast cancer (TNBC) MDA-MB-231 and to a lesser extend in MCF7 ER+ breast cancer cells. Together, these findings position Brazilin as a new molecule with great potential for use as complementary or alternative treatment in breast cancer therapy in the future.


Subject(s)
Benzopyrans , Breast Neoplasms , Cadherins , Epithelial-Mesenchymal Transition , Female , Humans , Benzopyrans/pharmacology , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Cadherins/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/genetics , MCF-7 Cells , Neoplasm Invasiveness/genetics , Nuclear Proteins , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/genetics , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Vimentin/metabolism , Vimentin/genetics
9.
Clin Transl Med ; 14(5): e1690, 2024 May.
Article in English | MEDLINE | ID: mdl-38760896

ABSTRACT

INTRODUCTION: Radiation-induced pulmonary fibrosis (RIPF) is a chronic, progressive, irreversible lung interstitial disease that develops after radiotherapy. Although several previous studies have focused on the mechanism of epithelial-mesenchymal transition (EMT) in lung epithelial cells, the essential factors involved in this process remain poorly understood. The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) exhibits strong repair capacity when cells undergo radiation-induced damage; whether DNA-PKcs regulates EMT during RIPF remains unclear. OBJECTIVES: To investigate the role and molecular mechanism of DNA-PKcs in RIPF and provide an important theoretical basis for utilising DNA-PKcs-targeted drugs for preventing RIPF. METHODS: DNA-PKcs knockout (DPK-/-) mice were generated via the Cas9/sgRNA technique and subjected to whole chest ionizing radiation (IR) at a 20 Gy dose. Before whole chest IR, the mice were intragastrically administered the DNA-PKcs-targeted drug VND3207. Lung tissues were collected at 1 and 5 months after IR. RESULTS: The expression of DNA-PKcs is low in pulmonary fibrosis (PF) patients. DNA-PKcs deficiency significantly exacerbated RIPF by promoting EMT in lung epithelial cells. Mechanistically, DNA-PKcs deletion by shRNA or inhibitor NU7441 maintained the protein stability of Twist1. Furthermore, AKT1 mediated the interaction between DNA-PKcs and Twist1. High Twist1 expression and EMT-associated changes caused by DNA-PKcs deletion were blocked by insulin-like growth factor-1 (IGF-1), an AKT1 agonist. The radioprotective drug VND3207 prevented IR-induced EMT and alleviated RIPF in mice by stimulating the kinase activity of DNA-PKcs. CONCLUSION: Our study clarified the critical role and mechanism of DNA-PKcs in RIPF and showed that it could be a potential target for preventing RIPF.


Subject(s)
DNA-Activated Protein Kinase , Epithelial-Mesenchymal Transition , Nuclear Proteins , Proto-Oncogene Proteins c-akt , Pulmonary Fibrosis , Twist-Related Protein 1 , Epithelial-Mesenchymal Transition/drug effects , Animals , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Mice , Proto-Oncogene Proteins c-akt/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/etiology , Ubiquitination , Humans , Mice, Knockout , DNA-Binding Proteins
10.
Kaohsiung J Med Sci ; 40(7): 650-659, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38757734

ABSTRACT

Colorectal cancer (CRC) is notable for its high mortality and high metastatic characteristics. The shear force generated by bloodstream provides mechanical signals regulating multiple responses of cells, including metastatic cancer cells, dispersing in blood vessels. We, therefore, studied the effect of shear flow on circulating CRC cells in the present study. The CRC cell line SW620 was subjected to shear flow of 12.5 dynes/cm2 for 1 and 2 h separately. Resulting elevated caspase-9 and -3 indicated that shear flow initiated the apoptosis of SW620. Enlarged cell size associated with a higher level of cyclin D1 was coincident with the flow cytometric results indicating that the cell cycle was arrested at the G1 phase. An elevated phosphor-eNOSS1177 increased the production of nitric oxide and led to reactive oxygen species-mediated oxidative stress. Shear flow also regulated epithelial-mesenchymal transition (EMT) by increasing E-cadherin and ZO-1 while decreasing Snail and Twist1. The migration and invasion of sheared SW620 were also substantially decreased. Further investigations showed that mitochondrial membrane potential was significantly decreased, whereas mitochondrial mass and ATP production were not changed. In addition to the shear flow of 12.5 dynes/cm2, the expressions of EMT were compared at lower (6.25 dynes/cm2) and at higher (25 dynes/cm2) shear flow. The results showed that lower shear flow increased mesenchymal characteristics and higher shear flow increased epithelial characteristics. Shear flow reduces the malignancy of CRC in their metastatic dispersal that opens up new ways to improve cancer therapies by applying a mechanical shear flow device.


Subject(s)
Apoptosis , Cell Movement , Colorectal Neoplasms , Epithelial-Mesenchymal Transition , Reactive Oxygen Species , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Stress, Mechanical , Membrane Potential, Mitochondrial , Cyclin D1/metabolism , Oxidative Stress , Cadherins/metabolism , Nitric Oxide/metabolism , Caspase 9/metabolism , Caspase 3/metabolism , Zonula Occludens-1 Protein/metabolism , Twist-Related Protein 1/metabolism
11.
Integr Cancer Ther ; 23: 15347354241247223, 2024.
Article in English | MEDLINE | ID: mdl-38646808

ABSTRACT

BACKGROUND: Luteolin, a common dietary flavonoid found in plants, has been shown to have anti-cancer properties. However, its exact mechanisms of action in non-small cell lung cancer (NSCLC) are still not fully understood, particularly its role in regulating broader genomic networks and specific gene targets. In this study, we aimed to elucidate the role of microRNAs (miRNAs) in NSCLC treated with luteolin, using A549 cells as a model system. MATERIALS AND METHODS: miRNA profiling was conducted on luteolin-treated A549 cells using Exiqon microarrays, with validation of selected miRNAs by qRT-PCR. Bioinformatic analysis identified the regulatory roles of miRNAs in biological processes and pathways following luteolin treatment. Computational algorithms were employed to identify potential target genes. A549 cells were transfected with miR-106a-5p mimic and inhibitor or their corresponding controls. The expression levels of 2 genes, twist basic helix-loop-helix transcription factor 1 (TWIST1) and matrix metallopeptidase 2 (MMP2), and cell migration were assessed. RESULTS: miRNA profiling identified 341 miRNAs, with 18 exhibiting significantly altered expression (P < 0.05). Subsequent qRT-PCR analysis confirmed altered expression of 6 selected miRNAs. KEGG and GO analyses revealed significant alterations in pathways and biological processes crucial for tumor biology. TWIST1 and MMP2, which both contain conserved miR-106a-5p binding sites, exhibited an inverse correlation with the expression levels of miR-106a-5p. Dual-luciferase reporter assays confirmed TWIST1 and MMP2 as direct targets of miR-106a-5p. Luteolin treatment led to a reduction in A549 cell migration, and this reduction was further amplified by the overexpression of miR-106a-5p. CONCLUSION: Luteolin inhibits A549 cell migration by modulating the miRNA landscape, shedding light on its mechanisms and laying the foundation for miRNA-based therapeutic approaches for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Movement , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Luteolin , Matrix Metalloproteinase 2 , MicroRNAs , Nuclear Proteins , Twist-Related Protein 1 , Up-Regulation , Humans , Luteolin/pharmacology , MicroRNAs/genetics , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Cell Movement/drug effects , Cell Movement/genetics , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , A549 Cells , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Up-Regulation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics
12.
Cell Tissue Res ; 397(1): 37-50, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38602543

ABSTRACT

Synovial chondromatosis (SC) is a disorder of the synovium characterized by the formation of osteochondral nodules within the synovium. This study aimed to identify the abnormally differentiated progenitor cells and possible pathogenic signaling pathways. Loose bodies and synovium were obtained from patients with SC during knee arthroplasty. Single-cell RNA sequencing was used to identify cell subsets and their gene signatures in SC synovium. Cells derived from osteoarthritis (OA) synovium were used as controls. Multi-differentiation and colony-forming assays were used to identify progenitor cells. The roles of transcription factors and signaling pathways were investigated through computational analysis and experimental verification. We identified an increased proportion of CD34+ sublining fibroblasts in SC synovium. CD34+CD31- cells and CD34-CD31- cells were sorted from SC synovium. Compared with CD34- cells, CD34+ cells had larger alkaline phosphatase (ALP)-stained area and calcified area after osteogenic induction. In addition, CD34+ cells exhibited a stronger tube formation ability than CD34- cells. Our bioinformatic analysis suggested the expression of TWIST1, a negative regulator of osteogenesis, in CD34- sublining fibroblasts and was regulated by the TGF-ß signaling pathway. The experiment showed that CD34+ cells acquired the TWIST1 expression during culture and the combination of TGF-ß1 and harmine, an inhibitor of Twist1, could further stimulate the osteogenesis of CD34+ cells. Overall, CD34+ synovial fibroblasts in SC synovium have multiple differentiation potentials, especially osteogenic differentiation potential, and might be responsible for the pathogenesis of SC.


Subject(s)
Antigens, CD34 , Chondromatosis, Synovial , Fibroblasts , Osteogenesis , Synovial Membrane , Humans , Antigens, CD34/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Chondromatosis, Synovial/pathology , Chondromatosis, Synovial/metabolism , Synovial Membrane/pathology , Synovial Membrane/metabolism , Female , Male , Middle Aged , Cell Differentiation , Aged , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Nuclear Proteins
13.
Cell Signal ; 119: 111182, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38640983

ABSTRACT

Cancer-associated Fibroblasts (CAFs) exert a tumor-promoting effect in various cancers, including breast cancer. CAFs secrete exosomes containing miRNA and proteins, influencing the tumor microenvironment. In this study, we identified CAF-derived exosomes that transport functional miR-92a from CAFs to tumor cells, thereby intensifying the aggressiveness of breast cancer. CAFs downregulate the expression of G3BP2 in breast cancer cells, and a significant elevation in miR-92a levels in CAF-derived exosomes was observed. Both in vitro and in vivo experiments demonstrate that miR-92a enhances breast cancer cell migration and invasion by directly targeting G3BP2, functioning as a tumor-promoting miRNA. We validated that the RNA-binding proteins SNRPA facilitate the transfer of CAF-derived exosomal miR-92a to breast cancer cells. The reduction of G3BP2 protein by CAF-derived exosomes releases TWIST1 into the nucleus, promoting epithelial-mesenchymal transition (EMT) and further exacerbating breast cancer progression. Moreover, CAF-derived exosomal miR-92a induces tumor invasion and metastasis in mice. Overall, our study reveals that CAF-derived exosomal miR-92a serves as a promoter in the migration and invasion of breast cancer cells by reducing G3BP2 and may represent a potential novel tumor marker for breast cancer.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Cell Movement , Epithelial-Mesenchymal Transition , Exosomes , Gene Expression Regulation, Neoplastic , MicroRNAs , Neoplasm Invasiveness , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carrier Proteins/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Exosomes/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Neoplasm Metastasis , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics
14.
Sci Rep ; 14(1): 8246, 2024 04 08.
Article in English | MEDLINE | ID: mdl-38589525

ABSTRACT

MicroRNAs are small RNA molecules that have a significant role in translational repression and gene silencing through binding to downstream target mRNAs. MiR-762 can stimulate the proliferation and metastasis of various types of cancer. Hippo pathway is one of the pathways that regulate tissue development and carcinogenesis. Dysregulation of this pathway plays a vital role in the progression of cancer. This study aimed to evaluate the possible correlation between miR-762, the Hippo signaling pathway, TWIST1, and SMAD3 in patients with lung cancer, as well as patients with chronic inflammatory diseases. The relative expression of miR-762, MST1, LATS2, YAP, TWIST1, and SMAD3 was determined in 50 lung cancer patients, 30 patients with chronic inflammatory diseases, and 20 healthy volunteers by real-time PCR. The levels of YAP protein and neuron-specific enolase were estimated by ELISA and electrochemiluminescence immunoassay, respectively. Compared to the control group, miR-762, YAP, TWIST1, and SMAD3 expression were significantly upregulated in lung cancer patients and chronic inflammatory patients, except SMAD3 was significantly downregulated in chronic inflammatory patients. MST1, LATS2, and YAP protein were significantly downregulated in all patients. MiR-762 has a significant negative correlation with MST1, LATS2, and YAP protein in lung cancer patients and with MST1 and LATS2 in chronic inflammatory patients. MiR-762 may be involved in the induction of malignant behaviors in lung cancer through suppression of the Hippo pathway. MiR-762, MST1, LATS2, YAP mRNA and protein, TWIST1, and SMAD3 may be effective diagnostic biomarkers in both lung cancer patients and chronic inflammatory patients. High YAP, TWIST1, SMA3 expression, and NSE level are associated with a favorable prognosis for lung cancer.


Subject(s)
Lung Neoplasms , MicroRNAs , Humans , Hippo Signaling Pathway , Signal Transduction , Lung Neoplasms/genetics , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Chronic Disease , Cell Proliferation/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
Oncogene ; 43(19): 1431-1444, 2024 May.
Article in English | MEDLINE | ID: mdl-38485737

ABSTRACT

MET amplification/mutations are important targetable oncogenic drivers in NSCLC, however, acquired resistance is inevitable and the majority of patients with targetable MET alterations fail to respond to MET tyrosine kinase inhibitors (TKIs). Furthermore, MET amplification is among the most common mediators of TKI resistance. As such, novel therapies to target MET pathway and overcome MET TKI resistance are clearly needed. Here we show that the epithelial-mesenchymal transition (EMT) transcription factor, TWIST1 is a key downstream mediator of HGF/MET induced resistance through suppression of p27 and targeting TWIST1 can overcome resistance. We found that TWIST1 is overexpressed at the time of TKI resistance in multiple MET-dependent TKI acquired resistance PDX models. We have shown for the first time that MET directly stabilized the TWIST protein leading to TKI resistance and that TWIST1 was required for MET-driven lung tumorigenesis as well as could induce MET TKI resistance when overexpressed. TWIST1 mediated MET TKI resistance through suppression of p27 expression and genetic or pharmacologic inhibition of TWIST1 overcame TKI resistance in vitro and in vivo. Our findings suggest that targeting TWIST1 may be an effective therapeutic strategy to overcome resistance in MET-driven NSCLC as well as in other oncogene driven subtypes in which MET amplification is the resistance mechanism.


Subject(s)
Drug Resistance, Neoplasm , Hepatocyte Growth Factor , Lung Neoplasms , Nuclear Proteins , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-met , Twist-Related Protein 1 , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Oncogenes/genetics , Xenograft Model Antitumor Assays
16.
J Cutan Pathol ; 51(3): 232-238, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37932931

ABSTRACT

BACKGROUND: Diagnosis of early mycosis fungoides (eMF) is challenging and often delayed as many of its clinical and histopathologic features may mimic various benign inflammatory dermatoses (BIDs). The products of the thymocyte selection-associated high mobility group box (TOX), twist family BHLH transcription factor 1 (TWIST1), signal transducer and activator of transcription 4 (STAT4), and special AT-rich sequence-binding protein 1 (SATB1) genes function as transcription factors and are involved in the pathogenesis of MF. OBJECTIVES: We aim to determine the diagnostic value of TOX, TWIST1, STAT4, and SATB1 protein expressions in eMF. METHODS: This non-randomized, controlled, prospective analytic study was conducted by performing immunohistochemistry staining with TOX, TWIST1, STAT4, and SATB1 polyclonal antibodies in lesional skin biopsies of eMF and BID patients. Nuclear staining of lymphocytes was compared between eMF and BIDs, and the capacity of these antibodies to predict eMF was determined. RESULTS: Immunostainings with anti-TWIST1 showed an increase in protein expression (p = 0.003) and showed a decrease with anti-SATB1 antibodies in eMF compared to BIDs (p = 0.005) while anti-TOX and anti-STAT4 antibodies did not exhibit significant differences (p = 0.384; p = 0.150). Receiver operating characteristic analysis showed that immunohistochemical evaluations of TWIST1 and SATB1 protein expressions can differentiate eMF (area under the curve [AUC]: 0.728, 95% confidence interval [CI]: 0.605-0.851, p = 0.002; AUC: 0.686, 95% CI: 0.565-0.807, p = 0.013). CONCLUSIONS: TWIST1 and SATB1 are potential diagnostic markers for the histologic diagnosis of eMF.


Subject(s)
Matrix Attachment Region Binding Proteins , Mycosis Fungoides , Skin Neoplasms , Humans , Matrix Attachment Region Binding Proteins/metabolism , Mycosis Fungoides/pathology , Nuclear Proteins/metabolism , Prospective Studies , Skin Neoplasms/pathology , STAT4 Transcription Factor/metabolism , Twist-Related Protein 1/metabolism
17.
Cell Death Dis ; 14(12): 822, 2023 12 13.
Article in English | MEDLINE | ID: mdl-38092725

ABSTRACT

Jagged1 (JAG1) is a Notch ligand that correlates with tumor progression. Not limited to its function as a ligand, JAG1 can be cleaved, and its intracellular domain translocates to the nucleus, where it functions as a transcriptional cofactor. Previously, we showed that JAG1 intracellular domain (JICD1) forms a protein complex with DDX17/SMAD3/TGIF2. However, the molecular mechanisms underlying JICD1-mediated tumor aggressiveness remains unclear. Here, we demonstrate that JICD1 enhances the invasive phenotypes of glioblastoma cells by transcriptionally activating epithelial-to-mesenchymal transition (EMT)-related genes, especially TWIST1. The inhibition of TWIST1 reduced JICD1-driven tumor aggressiveness. Although SMAD3 is an important component of transforming growth factor (TGF)-ß signaling, the JICD1/SMAD3 transcriptional complex was shown to govern brain tumor invasion independent of TGF-ß signaling. Moreover, JICD1-TWIST1-MMP2 and MMP9 axes were significantly correlated with clinical outcome of glioblastoma patients. Collectively, we identified the JICD1/SMAD3-TWIST1 axis as a novel inducer of invasive phenotypes in cancer cells.


Subject(s)
Glioblastoma , Humans , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Glioblastoma/genetics , Homeodomain Proteins/metabolism , Ligands , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
18.
PLoS Biol ; 21(12): e3002446, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38134227

ABSTRACT

Tumor metastasis is the major cause of breast cancer morbidity and mortality. It has been reported that the F-box protein FBXO3 functions as an E3 ubiquitin ligase in regulating various biological processes, including host autoimmune, antiviral innate immunity, and inflammatory response. However, the role of FBXO3 in tumor metastasis remains elusive. We have previously shown that ΔNp63α is a common inhibitory target in oncogene-induced cell motility and tumor metastasis. In this study, we show that FBXO3 plays a vital role in PI3K-mediated breast cancer metastasis independent of its E3 ligase activity and ΔNp63α in breast cancer cells and in mouse. FBXO3 can bind to and stabilize USP4, leading to Twist1 protein stabilization and increased breast cancer cell migration and tumor metastasis. Mechanistically, FBXO3 disrupts the interaction between USP4 and aspartyl aminopeptidase (DNPEP), thereby protecting USP4 from DNPEP-mediated degradation. Furthermore, p110αH1047R facilitates the phosphorylation and stabilization of FBXO3 in an ERK1-dependent manner. Knockdown of either FBXO3 or USP4 leads to significant inhibition of PI3K-induced breast cancer metastasis. Clinically, elevated expression of p110α/FBXO3/USP4/Twist1 is associated with poor overall survival (OS) and recurrence-free survival (RFS) of breast cancer patients. Taken together, this study reveals that the FBXO3-USP4-Twist1 axis is pivotal in PI3K-mediated breast tumor metastasis and that FBXO3/USP4 may be potential therapeutic targets for breast cancer treatment.


Subject(s)
Breast Neoplasms , Melanoma , Skin Neoplasms , Animals , Female , Humans , Mice , Breast Neoplasms/metabolism , Cell Line, Tumor , Phosphatidylinositol 3-Kinases/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
19.
Int J Mol Sci ; 24(22)2023 Nov 14.
Article in English | MEDLINE | ID: mdl-38003483

ABSTRACT

Cyclin-dependent kinase (CDK) 4/6 inhibitors have significantly improved progression-free survival in hormone-receptor-positive (HR+), human-epidermal-growth-factor-receptor-type-2-negative (HER2-) metastatic luminal breast cancer (mLBC). Several studies have shown that in patients with endocrine-sensitive or endocrine-resistant LBC, the addition of CDK4/6 inhibitors to endocrine therapy significantly prolongs progression-free survival. However, the percentage of patients who are unresponsive or refractory to these therapies is as high as 40%, and no reliable and reproducible biomarkers have been validated to select a priori responders or refractory patients. The selection of mutant clones in the target oncoprotein is the main cause of resistance. Other mechanisms such as oncogene amplification/overexpression or mutations in other pathways have been described in several models. In this study, we focused on palbociclib, a selective CDK4/6 inhibitor. We generated a human MCF-7 luminal breast cancer cell line that was able to survive and proliferate at different concentrations of palbociclib and also showed cross-resistance to abemaciclib. The resistant cell line was characterized via RNA sequencing and was found to strongly activate the epithelial-to-mesenchymal transition. Among the top deregulated genes, we found a dramatic downregulation of the CDK4 inhibitor CDKN2B and an upregulation of the TWIST1 transcription factor. TWIST1 was further validated as a target for the reversal of palbociclib resistance. This study provides new relevant information about the mechanisms of resistance to CDK4/6 inhibitors and suggests potential new markers for patients' follow-up care during treatment.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Up-Regulation , Cyclin-Dependent Kinase 4 , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Progression-Free Survival , Cyclin-Dependent Kinase 6 , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
20.
Sci Adv ; 9(45): eadi2387, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37948519

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) have been evaluated in >1500 clinical trials, but outcomes remain suboptimal because of knowledge gaps in quality attributes that confer potency. We show that TWIST1 directly represses TSG6 expression that TWIST1 and TSG6 are inversely correlated across bone marrow-derived MSC (BM-MSC) donor cohorts and predict interdonor differences in their proangiogenic, anti-inflammatory, and immune suppressive activity in vitro and in sterile inflammation and autoimmune type 1 diabetes preclinical models. Transcript profiling of TWIST1HiTSG6Low versus TWISTLowTSG6Hi BM-MSCs revealed previously unidentified roles for TWIST1/TSG6 in regulating cellular oxidative stress and TGF-ß2 in modulating TSG6 expression and anti-inflammatory activity. TWIST1 and TSG6 levels also correlate to donor stature and predict differences in iPSC-derived MSC quality attributes. These results validate TWIST1 and TSG6 as biomarkers that predict interdonor differences in potency across laboratories and assay platforms, thereby providing a means to manufacture MSC products tailored to specific diseases.


Subject(s)
Mesenchymal Stem Cells , Humans , Anti-Inflammatory Agents/pharmacology , Biomarkers/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation , Immunologic Factors/metabolism , Mesenchymal Stem Cells/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL