Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Publication year range
1.
FASEB J ; 38(15): e23495, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39126242

ABSTRACT

Hepatic stellate cell (HSC) activation is the essential pathological process of liver fibrosis (LF). The molecular mechanisms regulating HSC activation and LF are incompletely understood. Here, we explored the effect of transcription factor SRY-related high mobility group box 7 (SOX7) on HSC activation and LF, and the underlying molecular mechanism. We found the expression levels of SOX7 were decreased in human and mouse fibrotic livers, particularly at the fibrotic foci. SOX7 was also downregulated in primary activated HSCs and TGF-ß1 stimulated LX-2 cells. SOX7 knockdown promoted activation and proliferation of LX-2 cells while inhibiting their apoptosis. On the other hand, overexpression of SOX7 suppressed the activation and proliferation of HSCs. Mechanistically, SOX7 attenuates HSC activation and LF by decreasing the expression of ß-catenin and phosphorylation of Smad2 and Smad3 induced by TGF-ß1. Furthermore, overexpression of SOX7 using AAV8-SOX7 mouse models ameliorated the extent of LF in response to CCl4 treatment in vivo. Collectively, SOX7 suppressed HSC activation and LF. Targeting SOX7, therefore, could be a potential novel strategy to protect against LF.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , SOXF Transcription Factors , Hepatic Stellate Cells/metabolism , Animals , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Humans , Male , SOXF Transcription Factors/metabolism , SOXF Transcription Factors/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cell Proliferation , Mice, Inbred C57BL , beta Catenin/metabolism , beta Catenin/genetics , Apoptosis , Smad2 Protein/metabolism , Smad2 Protein/genetics , Cell Line , Smad3 Protein/metabolism , Smad3 Protein/genetics
2.
Dev Dyn ; 253(4): 423-434, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37850829

ABSTRACT

BACKGROUND: Wnt/ß-catenin signaling plays a variety of roles in both the dental epithelium and mesenchyme at most stages of tooth development. In this study, we verified the roles of Hertwig's epithelial root sheath (HERS) breakdown in tooth root development. This breakdown results in formation of epithelial cell rests of Malassez (ERM). RESULTS: Following induction of ß-catenin stabilization in the epithelium of developing tooth at the moment of HERS breakdown, HERS failed to break down for ERM formation. HERS with stabilized ß-catenin was altered into a multicellular layer enveloping elongated root dentin with higher expression of junctional proteins such as Zo-1 and E-cadherin. Importantly, this impairment of HERS breakdown led to arrest of further root elongation. In addition, the portion of root dentin enveloped by the undissociated HERS remained in a hypomineralized state. The odontoblasts showed ectopically higher expression of pyrophosphate regulators including Ank and Npp1, whereas Tnap expression was unchanged. CONCLUSIONS: Our data suggest that Wnt/ß-catenin signaling is decreased in HERS for ERM formation during root development. Furthermore, ERM formation is important for further elongation and dentin mineralization of the tooth roots. These findings may provide new insight to understand the contribution of ERM to root formation.


Subject(s)
Tooth Root , beta Catenin , beta Catenin/genetics , beta Catenin/metabolism , Epithelial Cells/metabolism , Epithelium/metabolism , Odontogenesis , Cell Differentiation
3.
Genes Chromosomes Cancer ; 63(5): e23247, 2024 05.
Article in English | MEDLINE | ID: mdl-38757718

ABSTRACT

Superficial fibromas are a group of mesenchymal spindle cell lesions with pathomorphological heterogeneity and diverse molecular backgrounds. In part, they may be indicators of an underlying syndrome. Among the best-known entities of superficial fibromas is Gardner fibroma, a plaque-like benign tumor, which is associated with APC germline mutations and occurs in patients with familial adenomatosis polyposis (Gardner syndrome). Affected patients also have an increased risk to develop desmoid fibromatosis (DTF), a locally aggressive neoplasm of the deep soft tissue highly prone to local recurrences. Although a minority of DTFs occur in the syndromic context and harbor APC germline mutations, most frequently their underlying molecular aberration is a sporadic mutation in Exon 3 of the CTNNB1 gene. Up to date, a non-syndromic equivalent to Gardner fibroma carrying a CTNNB1 mutation has not been defined. Here, we present two cases of (sub-)cutaneous tumors with a hypocellular and collagen-rich Gardner fibroma-like appearance and pathogenic, somatic CTNNB1 mutations. We aim to differentiate these tumors from other fibromas according to their histological appearance, immunohistochemical staining profile and underlying somatic CTNNB1 mutations. Furthermore, we distinguish them from locally aggressive desmoid fibromatosis regarding their biological behavior, prognosis and indicated therapeutic strategies. Consequently, we call them CTNNB1-mutated superficial fibromas as a sporadic counterpart lesion to syndromic Gardner fibromas.


Subject(s)
Fibroma , beta Catenin , Humans , beta Catenin/genetics , Fibroma/genetics , Fibroma/pathology , Male , Female , Mutation , Middle Aged , Fibromatosis, Aggressive/genetics , Fibromatosis, Aggressive/pathology , Adult , Gardner Syndrome/genetics , Gardner Syndrome/pathology , Germ-Line Mutation
4.
Genesis ; 62(1): e23543, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37649322

ABSTRACT

Although epithelial-mesenchymal markers play an important role in prostate cancer (PC), further research is needed to better understand their utility in diagnosis, cancer progression prevention, and treatment resistance prediction. Our study included 111 PC patients who underwent transurethral resection, as well as 16 healthy controls. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to examine the expression of E-cadherin, ß-catenin, and Vimentin. We found that E-cadherin and ß-catenin were underexpressed in primary PC tissues. E-cadherin expression was found to be inversely associated with prostate-specific antigen progression (PSA-P; serum marker of progression; p = 0.01; |r| = 0.262). Furthermore, the underexpression of two markers, E-cadherin and ß-catenin, was found to be associated with advanced tumor stage and grade (p < 0.05). On the other hand, Vimentin was overexpressed in PC patients with a fold change of 2.141, and it was associated with the diagnosis, prognosis, and prediction of treatment resistance to androgen deprivation therapy (p = 0.002), abiraterone-acid (p = 0.001), and taxanes (p = 0.029). Moreover, the current study highlighted that poor survival could be significantly found in patients who progressed after primary surgery, did not use drugs, and expressed these genes aberrantly. In Cox regression multivariate analysis (p < 0.05), a positive correlation between the Vimentin marker and coronary heart disease in PC patients was identified (p = 0.034). In summary, the present study highlights the diagnostic (p < 0.001), prognostic (p < 0.001), and therapeutic potential of Vimentin in primary PC (p < 0.05), as well as its implications for cardiovascular disease. Furthermore, we confirm the potential prognostic value of E-cadherin and ß-catenin.


Subject(s)
Prostatic Neoplasms , beta Catenin , Male , Humans , beta Catenin/genetics , Vimentin/genetics , Vimentin/analysis , Vimentin/metabolism , Androgen Antagonists , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Cadherins/genetics , Epithelial-Mesenchymal Transition
5.
J Cell Mol Med ; 28(10): e18376, 2024 May.
Article in English | MEDLINE | ID: mdl-38780511

ABSTRACT

Taking into account homeostatic disorders resulting from arterial hypertension and the key importance of CacyBP/SIP, ß-catenin and endocannabinoids in the functioning of many organs, it was decided to assess the presence and distribution of CacyBP/SIP, ß-catenin, CB1 and CB2 in the adrenal glands of hypertensive rats of various aetiology. The study was conducted on the adrenal glands of rats with spontaneous and renovascular hypertension. The expression of CacyBP/SIP, ß-catenin, CB1 and CB2 was detected by immunohistochemistry and real-time PCR method. The results of the present study revealed both lower gene expression and immunoreactivity of CacyBP/SIP in the adrenal glands of all hypertensive groups compared to the normotensive rats. This study demonstrated a reduction in the immunoreactivity and expression of the ß-catenin, CB1 and CB2 genes in the adrenals of 2K1C rats. While in SHR, the reaction showing ß-catenin and CB1 was very weak or negative, and the expression of CB2 in the adrenal glands of these rats increased. The results of this study show, for the first time, marked differences in the expression of CacyBP/SIP, ß-catenin and CB1 and CB2 cannabinoid receptors in the adrenal glands of rats with primary (SHR) and secondary hypertension (2K1C).


Subject(s)
Adrenal Glands , Hypertension , Receptor, Cannabinoid, CB1 , Receptor, Cannabinoid, CB2 , beta Catenin , Animals , beta Catenin/metabolism , beta Catenin/genetics , Male , Hypertension/metabolism , Hypertension/genetics , Adrenal Glands/metabolism , Adrenal Glands/pathology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/metabolism , Receptor, Cannabinoid, CB2/genetics , Rats , Rats, Inbred SHR , Rats, Wistar , Immunohistochemistry , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/genetics , Hypertension, Renovascular/metabolism , Hypertension, Renovascular/genetics , Hypertension, Renovascular/pathology
6.
J Cell Physiol ; 239(6): e31245, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38497504

ABSTRACT

Parathyroid hormone (PTH) serves dual roles in bone metabolism, exhibiting both anabolic and catabolic effects. The anabolic properties of PTH have been utilized in the treatment of osteoporosis with proven efficacy in preventing fractures. Despite these benefits, PTH can be administered therapeutically for up to 2 years, and its use in patients with underlying malignancies remains a subject of ongoing debate. These considerations underscore the need for a more comprehensive understanding of the underlying mechanisms. p21-activated kinase 4 (PAK4) is involved in bone resorption and cancer-associated osteolysis; however, its role in osteoblast function and PTH action remains unknown. Therefore, in this study, we aimed to clarify the role of PAK4 in osteoblast function and its effects on PTH-induced anabolic activity. PAK4 enhanced MC3T3-E1 osteoblast viability and proliferation and upregulated cyclin D1 expression. PAK4 also augmented osteoblast differentiation, as indicated by increased mineralization found by alkaline phosphatase and Alizarin Red staining. Treatment with PTH (1-34), an active PTH fragment, stimulated PAK4 expression and phosphorylation in a protein kinase A-dependent manner. In addition, bone morphogenetic protein-2 (which is known to promote bone formation) increased phosphorylated PAK4 (p-PAK4) and PAK4 levels. PAK4 regulated the expression of both phosphorylated and total ß-catenin, which are critical for osteoblast proliferation and differentiation. Moreover, p-PAK4 directly interacted with ß-catenin, and disruption of ß-catenin's binding to T-cell factor impaired PAK4- and PTH-induced osteoblast differentiation. Our findings elucidate the effect of PAK4 on enhancing bone formation in osteoblasts and its pivotal role in the anabolic activity of PTH mediated through its interaction with ß-catenin. These insights improve the understanding of the mechanisms underlying PTH activity and should inform the development of more effective and safer osteoporosis treatments.


Subject(s)
Cell Differentiation , Cell Proliferation , Osteoblasts , Parathyroid Hormone , beta Catenin , p21-Activated Kinases , Animals , Humans , Mice , beta Catenin/metabolism , beta Catenin/genetics , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin D1/genetics , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Parathyroid Hormone/pharmacology , Parathyroid Hormone/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Cells, Cultured
7.
J Cell Physiol ; 239(4): e31183, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38348695

ABSTRACT

Osteogenic differentiation is important for fracture healing. Microfibrial-associated glycoprotein 2 (MAGP2) is found to function as a proangiogenic regulator in bone formation; however, its role in osteogenic differentiation during bone repair is not clear. Here, a mouse model of critical-sized femur fracture was constructed, and the adenovirus expressing MAGP2 was delivered into the fracture site. Mice with MAGP2 overexpression exhibited increased bone mineral density and bone volume fraction (BV/TV) at Day 14 postfracture. Within 7 days postfracture, overexpression of MAGP2 increased collagen I and II expression at the fracture callus, with increasing chondrogenesis. MAGP2 inhibited collagen II level but elevated collagen I by 14 days following fracture, accompanied by increased endochondral bone formation. In mouse osteoblast precursor MC3T3-E1 cells, MAGP2 treatment elevated the expression of osteoblastic factors (osterix, BGLAP and collagen I) and enhanced ALP activity and mineralization through activating ß-catenin signaling after osteogenic induction. Besides, MAGP2 could interact with lipoprotein receptor-related protein 5 (LRP5) and upregulated its expression. Promotion of osteogenic differentiation and ß-catenin activation mediated by MAGP2 was partially reversed by LRP5 knockdown. Interestingly, ß-catenin/transcription factor 4 (TCF4) increased MAGP2 expression probably by binding to MAGP2 promoter. These findings suggest that MAGP2 may interact with ß-catenin/TCF4 to enhance ß-catenin/TCF4's function and activate LRP5-activated ß-catenin signaling pathway, thus promoting osteogenic differentiation for fracture repair. mRNA sequencing identified the potential targets of MAGP2, providing novel insights into MAGP2 function and the directions for future research.


Subject(s)
Fractures, Bone , Osteogenesis , Animals , Mice , beta Catenin/genetics , beta Catenin/metabolism , Cell Differentiation/genetics , Collagen/metabolism , Fracture Healing , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Osteoblasts/metabolism , Wnt Signaling Pathway , Male , Mice, Inbred C57BL , Cell Line
8.
J Neurochem ; 168(4): 342-354, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37994470

ABSTRACT

Skeletal muscle fiber is a large syncytium with multiple and evenly distributed nuclei. Adult subsynaptic myonuclei beneath the neuromuscular junction (NMJ) express specific genes, the products of which coordinately function in the maintenance of the pre- and post-synaptic regions. However, the gene expression profiles that promote the NMJ formation during embryogenesis remain largely unexplored. We performed single-nucleus RNA sequencing (snRNA-seq) analysis of embryonic and neonatal mouse diaphragms, and found that each myonucleus had a distinct transcriptome pattern during the NMJ formation. Among the previously reported NMJ-constituting genes, Dok7, Chrna1, and Chrnd are specifically expressed in subsynaptic myonuclei at E18.5. In the E18.5 diaphragm, ca. 10.7% of the myonuclei express genes for the NMJ formation (Dok7, Chrna1, and Chrnd) together with four representative ß-catenin regulators (Amotl2, Ptprk, Fam53b, and Tcf7l2). Additionally, the temporal gene expression patterns of these seven genes are synchronized in differentiating C2C12 myoblasts. Amotl2 and Ptprk are expressed in the sarcoplasm, where ß-catenin serves as a structural protein to organize the membrane-anchored NMJ structure. In contrast, Fam53b and Tcf7l2 are expressed in the myonucleus, where ß-catenin serves as a transcriptional coactivator in Wnt/ß-catenin signaling at the NMJ. In C2C12 myotubes, knockdown of Amotl2 or Ptprk markedly, and that of Fam53b and Tcf7l2 less efficiently, impair the clustering of acetylcholine receptors. In contrast, knockdown of Fam53b and Tcf7l2, but not of Amotl2 or Ptprk, impairs the gene expression of Slit2 encoding an axonal attractant for motor neurons, which is required for the maturation of motor nerve terminal. Thus, Amotl2 and Ptprk exert different roles at the NM compared to Fam53b and Tcf7l2. Additionally, Wnt ligands originating from the spinal motor neurons and the perichondrium/chondrocyte are likely to work remotely on the subsynaptic nuclei and the myotendinous junctional nuclei, respectively. We conclude that snRNA-seq analysis of embryonic/neonatal diaphragms reveal a novel coordinated expression profile especially in the Wnt/ß-catenin signaling that regulate the formation of the embryonic NMJ.


Subject(s)
Transcriptome , beta Catenin , Mice , Animals , beta Catenin/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Wnt Signaling Pathway/genetics , RNA, Small Nuclear/metabolism , Embryonic Development , Muscle, Skeletal/metabolism , Receptors, Cholinergic/metabolism
9.
Eur J Neurosci ; 59(10): 2732-2747, 2024 May.
Article in English | MEDLINE | ID: mdl-38501537

ABSTRACT

Elevated serum homocysteine (Hcy) level is a risk factor for Alzheimer's disease (AD) and accelerates cell aging. However, the mechanism by which Hcy induces neuronal senescence remains largely unknown. In this study, we observed that Hcy significantly promoted senescence in neuroblastoma 2a (N2a) cells with elevated ß-catenin and Kelch-like ECH-associated protein 1 (KEAP1) levels. Intriguingly, Hcy promoted the interaction between KEAP1 and the Wilms tumor gene on the X chromosome (WTX) while hampering the ß-catenin-WTX interaction. Mechanistically, Hcy attenuated the methylation level of the KEAP1 promoter CpG island and activated KEAP1 transcription. However, a slow degradation rate rather than transcriptional activation contributed to the high level of ß-catenin. Hcy-upregulated KEAP1 competed with ß-catenin to bind to WTX. Knockdown of both ß-catenin and KEAP1 attenuated Hcy-induced senescence in N2a cells. Our data highlight a crucial role of the KEAP1-ß-catenin pathway in Hcy-induced neuronal-like senescence and uncover a promising target for AD treatment.


Subject(s)
Cellular Senescence , Homocysteine , Kelch-Like ECH-Associated Protein 1 , Neuroblastoma , Ubiquitination , beta Catenin , Animals , Mice , beta Catenin/metabolism , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/physiology , Homocysteine/pharmacology , Homocysteine/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Neuroblastoma/metabolism , Neurons/metabolism , Neurons/drug effects , Ubiquitination/drug effects
10.
J Transl Med ; 22(1): 133, 2024 02 03.
Article in English | MEDLINE | ID: mdl-38310229

ABSTRACT

BACKGROUND: Oxaliplatin resistance usually leads to therapeutic failure and poor prognosis in colorectal cancer (CRC), while the underlying mechanisms are not yet fully understood. Metabolic reprogramming is strongly linked to drug resistance, however, the role and mechanism of metabolic reprogramming in oxaliplatin resistance remain unclear. Here, we aim to explore the functions and mechanisms of purine metabolism on the oxaliplatin-induced apoptosis of CRC. METHODS: An oxaliplatin-resistant CRC cell line was generated, and untargeted metabolomics analysis was conducted. The inosine 5'-monophosphate dehydrogenase type II (IMPDH2) expression in CRC cell lines was determined by quantitative real-time polymerase chain reaction (qPCR) and western blotting analysis. The effects of IMPDH2 overexpression, knockdown and pharmacological inhibition on oxaliplatin resistance in CRC were assessed by flow cytometry analysis of cell apoptosis in vivo and in vitro. RESULTS: Metabolic analysis revealed that the levels of purine metabolites, especially guanosine monophosphate (GMP), were markedly elevated in oxaliplatin-resistant CRC cells. The accumulation of purine metabolites mainly arose from the upregulation of IMPDH2 expression. Gene set enrichment analysis (GSEA) indicated high IMPDH2 expression in CRC correlates with PURINE_METABOLISM and MULTIPLE-DRUG-RESISTANCE pathways. CRC cells with higher IMPDH2 expression were more resistant to oxaliplatin-induced apoptosis. Overexpression of IMPDH2 in CRC cells resulted in reduced cell death upon treatment with oxaliplatin, whereas knockdown of IMPDH2 led to increased sensitivity to oxaliplatin through influencing the activation of the Caspase 7/8/9 and PARP1 proteins on cell apoptosis. Targeted inhibition of IMPDH2 by mycophenolic acid (MPA) or mycophenolate mofetil (MMF) enhanced cell apoptosis in vitro and decreased in vivo tumour burden when combined with oxaliplatin treatment. Mechanistically, the Wnt/ß-catenin signalling was hyperactivated in oxaliplatin-resistant CRC cells, and a reciprocal positive regulatory mechanism existed between Wnt/ß-catenin and IMPDH2. Blocking the Wnt/ß-catenin pathway could resensitize resistant cells to oxaliplatin, which could be restored by the addition of GMP. CONCLUSIONS: IMPDH2 is a predictive biomarker and therapeutic target for oxaliplatin resistance in CRC.


Subject(s)
Colorectal Neoplasms , beta Catenin , Humans , Apoptosis , beta Catenin/metabolism , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Oxidoreductases/genetics , Oxidoreductases/metabolism , Wnt Signaling Pathway
11.
Histopathology ; 85(1): 75-80, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38530207

ABSTRACT

BACKGROUND: Testicular Leydig cell tumours (LCTs) are the most common type of sex cord-stromal tumour in men, representing 1%-3% of all testicular neoplasms. Among testicular sex cord-stromal tumours, CTNNB1 mutations and nuclear expression of ß-catenin have been typically associated with Sertoli cell tumour. Recent genomic analyses have shown that CTNNB1 variants are also identified in a subset of LCTs; however, the frequency and clinicopathologic associations of ß-catenin alterations remain incompletely understood in this tumour type. METHODS: In this study, we evaluated 32 LCTs (five malignant/metastasizing, 27 nonmetastasizing) using ß-catenin immunohistochemistry and DNA sequencing. RESULTS: Immunohistochemistry revealed focal or multifocal nuclear ß-catenin expression in 47% of the tumours. Diffuse nuclear ß-catenin expression (in >50% of the tumour cells) was not detected in any of the cases analysed herein. Comparison of ß-catenin-positive and ß-catenin-negative cases did not show significant differences in the frequency of adverse histopathologic findings or malignant clinical behaviour. DNA sequencing performed de novo on a subset of seven cases revealed the presence of exon 3 CTNNB1 variants in four of them (4/7, 57%), with variant allele frequencies (VAF) ranging from 7 to 33%. Two additional ß-catenin-positive cases that had been sequenced as part of a previous study harboured exon 3 CTNNB1 variants at VAF of 28% and 7%, respectively. CONCLUSION: These results demonstrate that ß-catenin alterations are relatively common in LCT, most likely occurring as subclonal events that are not enriched in cases with aggressive features. Further studies are needed to clarify the oncogenic role of ß-catenin in this tumour type.


Subject(s)
Immunohistochemistry , Leydig Cell Tumor , Testicular Neoplasms , beta Catenin , Humans , beta Catenin/genetics , beta Catenin/metabolism , Male , Testicular Neoplasms/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Leydig Cell Tumor/pathology , Leydig Cell Tumor/metabolism , Leydig Cell Tumor/genetics , Adult , Middle Aged , Aged , Young Adult , Adolescent , Mutation , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism
12.
Histopathology ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38922981

ABSTRACT

Adenoid ameloblastoma (AA) was recently recognised as a separate tumour type in the most recent World Health Organisation (WHO) classification of head and neck tumours. This decision has been considered controversial by several groups, who have described AA as a subtype of ameloblastoma, a hybrid odontogenic tumour or to fall within the spectrum of other recognised odontogenic tumours, including dentinogenic ghost cell tumour and adenomatoid odontogenic tumour. Here we review the reasons for the WHO decision to classify AA as a separate tumour type. We also critique molecular and histological findings from recent reports published since the WHO classification. While acknowledging that the classification of tumours is constantly evolving, the balance of current evidence suggests that AA should remain a distinct tumour type, and not a subtype of ameloblastoma, pending further molecular characterisation.

13.
Cell Biochem Funct ; 42(4): e4078, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38898665

ABSTRACT

Zinc finger proteins (ZNFs) play a significant role in the initiation and progression of tumors. Nevertheless, the specific contribution of ZNF610 to lung adenocarcinoma (LUAD) remains poorly understood. This study sought is to elucidate the role of ZNF610 in LUAD. Transcript data of LUAD were obtained from The Cancer Genome Atlas Program (TCGA) database and processed via R program. The expression of ZNF610 was assessed in various cell lines. To compare the proliferative capacity of cells with or without ZNF610 silencing, CCK8, cell colony formation assay, and Celigo label-free cell counting assay were employed. Furthermore, transwell migration and invasion assays were conducted to evaluate the migratory and invasive abilities of the cells. The expression levels of genes and proteins were assessed using quantitative real-time polymerase chain reaction (qRT-PCR) and western blot techniques. In different LUAD cells, the expression level of ZNF610 was found to be significantly higher in LUAD cells compared to MRC-5 and BASE-2B cells. Moreover, the silencing of ZNF610 resulted in a decrease in cell proliferation and migration abilities. Additionally, the apoptosis rate of cells increased upon silencing ZNF610. Notably, the proportion of cells in the G0/G1 phase increased, while the proportion of cells in the S phase decreased following ZNF610 silencing. Finally, ß-catenin and snail were identified as downstream targets of ZNF610 in cells. Our findings suggest that silencing ZNF610 could inhibit LUAD cell proliferation and migration, possibly through the downregulation of ß-catenin and snail.


Subject(s)
Adenocarcinoma of Lung , Cell Movement , Cell Proliferation , Lung Neoplasms , Humans , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Gene Silencing , Cell Line, Tumor , Apoptosis
14.
Orthod Craniofac Res ; 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39193636

ABSTRACT

OBJECTIVE: The effects of unilateral increased occlusal vertical dimension (iOVD) on bilateral craniofacial, mandibular and alveolar development in growing rats were investigated via cone-beam computed tomography (CBCT). The role of Wnt/ß-catenin signalling in this process was examined. MATERIALS AND METHODS: Forty-eight female Sprague-Dawley rats were randomly allocated into unilateral iOVD and sham groups. At 2, 4 and 8 weeks, the rats were scanned via CBCT to analyse cranial, maxillary, mandibular and dental morphology. Changes in temporomandibular joint (TMJ) cartilage histology and Wnt/ß-catenin signalling were assessed by histochemical and immunohistochemical staining and qRT-PCR. RESULTS: Dorsal cephalograms revealed that the mandible in the iOVD group tilted approximately 4° to the right. Unilateral iOVD had little effect on cranial and maxillary growth but inhibited mandibular growth (mandibular length and ramal height), especially on the deviated side (DS). Moreover, unilateral iOVD increased the length of the lower incisors and decreased the height of the molars on the DS. Unilateral iOVD induced bilateral osteoarthritis-like changes in the bilateral TMJ condylar cartilage and activated Wnt/ß-catenin signalling in the condylar cartilage, especially on the contralateral side (CLS). CONCLUSION: Occlusion with unilateral iOVD induced mandibular deviation, significantly inhibited mandibular growth and produced compensatory changes in the alveolar bone. In the iOVD group, the mandibular body length and ramal height were greater on the CLS than on the DS. Moreover, the greater ß-catenin protein expression in the TMJ condylar cartilage on the CLS than on the DS may account for the difference in asymmetrical mandibular development.

15.
Environ Toxicol ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38888371

ABSTRACT

Non-small cell lung cancer (NSCLC) is the primary inducer of cancer-related death worldwide. Asiaticoside (ATS) is a triterpenoid saponin that has been indicated to possess an antitumor activity in several malignancies. Nonetheless, its detailed functions in NSCLC remain unclarified. In this study, NSCLC cells were exposed to various doses of ATS. Functional experiments were employed to estimate the ATS effect on NSCLC cell behaviors. Western blotting was implemented for protein expression evaluation. A xenograft mouse model was established to assess the ATS effect on NSCLC in vivo. The results showed that ATS restrained NSCLC cell proliferation, cell cycle progression, migration, and invasiveness. ATS reversed TGF-ß-induced promotion in epithelial-mesenchymal transition (EMT). Mechanistically, ATS inhibited Wnt/ß-catenin signaling in NSCLC. Upregulating ß-catenin restored ATS-mediated suppression of NSCLC cell aggressiveness. Moreover, ATS administration repressed tumorigenesis in tumor-bearing mice. In conclusion, ATS represses growth and metastasis in NSCLC by blocking EMT via the inhibition of Wnt/ß-catenin signaling.

16.
Environ Toxicol ; 39(5): 3026-3039, 2024 May.
Article in English | MEDLINE | ID: mdl-38317508

ABSTRACT

Long noncoding RNAs have been reported to be involved in the development of breast cancer. LINC01572 was previously reported to promote the development of various tumors. However, the potential biological function of LINC01572 in breast cancer remains largely unknown. R language was used to perform bioinformatic analysis of The Cancer Genome Atlas data. The expression level of RNAs was examined by RT-qPCR. The effect of knocking down or overexpression LINC01572 in triple-negative breast cancer (TNBC) cell lines was evaluated by detecting cell proliferation, migrant action. RNA immunoprecipitation assay and RNA pull-down assay were performed to explore the regulatory relationship between LINC01572, EIF4A3, and ß-catenin. Bioinformatics analysis identifies LINC01572 as an oncogene of breast cancer. LINC01572 is over-expressed in TNBC tissues and cell lines, correlated with poor clinical prognosis in BC patients. Cell function studies confirmed that LINC01572 facilitated the proliferation and migration of TNBC cells in both vivo and vitro. Mechanistically, ß-catenin mRNA and EIF4A3 combine spatially to form a complex, LINC01572 helps transport this complex from the nucleus to the cytoplasm, thereby facilitating the translation of ß-catenin. Our findings confirm that LINC01572 acts as a tumor promoter and may act as a biomarker in TNBC. In addition, novel molecular regulatory relationships involving LINC01572/EIF4A3/ß-catenin are critical to the development of TNBC, which led to a new understanding of the mechanisms of TNBC progression and shows a new target for precision treatment for TNBC.


Subject(s)
MicroRNAs , Triple Negative Breast Neoplasms , Humans , beta Catenin/genetics , beta Catenin/metabolism , Triple Negative Breast Neoplasms/genetics , RNA, Messenger/genetics , Cell Line, Tumor , RNA , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism
17.
Drug Dev Res ; 85(2): e22178, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38528652

ABSTRACT

The wingless/integrase-1 (WNT) pathway involved in the pathogenesis of inflammatory airway diseases has recently generated considerable research interest. Montelukast, a leukotriene receptor antagonist, provides therapeutic benefits in allergic asthma involving eosinophils. We aimed to investigate the role of the WNT pathway in the therapeutic actions of montelukast (MT) in a mixed type of allergic-acute airway inflammation model induced by ovalbumin (OVA) and lipopolysaccharide (LPS) in mice. Female mice were sensitized with intraperitoneal OVA-Al(OH)3 administration in the initiation phase and intranasal OVA followed by LPS administration in the challenge phase. The mice were divided into eight groups: control, asthmatic, and control/asthmatic treated with XAV939 (inhibitor of the canonical WNT pathway), LGK-974 (inhibitor of the secretion of WNT ligands), or MT at different doses. The inhibition of the WNT pathway prevented tracheal 5-HT and bradykinin hyperreactivity, while only the inhibition of the canonical WNT pathway partially reduced 5-HT and bradykinin contractions compared to the inflammation group. Therefore, MT treatment hindered 5-HT and bradykinin hyperreactivity associated with airway inflammation. Furthermore, MT prevented the increases in the phosphorylated GSK-3ß and WNT5A levels, which had been induced by airway inflammation, in a dose-dependent manner. Conversely, the MT application caused a further increase in the fibronectin levels, while there was no significant alteration in the phosphorylation of the Smad-2 levels in the isolated lungs of the mice. The MT treatment reversed the increase in the mRNA expression levels of interleukin-17A. An increase in eosinophil and neutrophil counts was observed in bronchoalveolar lavage fluid samples obtained from the mice in the inflammation group, which was hampered by the MT treatment. The inhibition of the WNT pathway did not alter inflammatory cytokine expression or cell infiltration. The WNT pathway mediated the therapeutic effects of MT due to the inhibition of GSK-3ß phosphorylation as well as the reduction of WNT5A levels in a murine airway inflammation model.


Subject(s)
Acetates , Asthma , Cyclopropanes , Lipopolysaccharides , Quinolines , Sulfides , Mice , Female , Animals , Ovalbumin , Wnt Signaling Pathway , Glycogen Synthase Kinase 3 beta/metabolism , Serotonin/metabolism , Bradykinin/metabolism , Asthma/drug therapy , Lung/metabolism , Inflammation/metabolism , Mice, Inbred BALB C , Disease Models, Animal , Cytokines/metabolism
18.
EMBO J ; 38(2)2019 01 15.
Article in English | MEDLINE | ID: mdl-30425074

ABSTRACT

During canonical Wnt signalling, the activity of nuclear ß-catenin is largely mediated by the TCF/LEF family of transcription factors. To challenge this view, we used the CRISPR/Cas9 genome editing approach to generate HEK 293T cell clones lacking all four TCF/LEF genes. By performing unbiased whole transcriptome sequencing analysis, we found that a subset of ß-catenin transcriptional targets did not require TCF/LEF factors for their regulation. Consistent with this finding, we observed in a genome-wide analysis that ß-catenin occupied specific genomic regions in the absence of TCF/LEF Finally, we revealed the existence of a transcriptional activity of ß-catenin that specifically appears when TCF/LEF factors are absent, and refer to this as ß-catenin-GHOST response. Collectively, this study uncovers a previously neglected modus operandi of ß-catenin that bypasses the TCF/LEF transcription factors.


Subject(s)
Gene Expression Profiling/methods , TCF Transcription Factors/genetics , Transcription, Genetic , beta Catenin/metabolism , CRISPR-Cas Systems , Gene Editing , Gene Expression Regulation , HEK293 Cells , Humans , TCF Transcription Factors/metabolism , Exome Sequencing/methods , Wnt Signaling Pathway
19.
Molecules ; 26(21)2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34771159

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a chronic neurological illness that causes considerable cognitive impairment. Hepatic and renal dysfunction may worsen AD by disrupting ß-amyloid homeostasis at the periphery and by causing metabolic dysfunction. Wheatgrass (Triticum aestivum) has been shown to have antioxidant and anti-inflammatory properties. This work aims to study the effect of aluminum on neuronal cells, its consequences on the liver and kidneys, and the possible role of fluoxetine and wheatgrass juice in attenuating these pathological conditions. METHOD: Rats were divided into five groups. Control, AD (AlCl3), Fluoxetine (Fluoxetine and AlCl3), Wheatgrass (Wheatgrass and AlCl3), and combination group (fluoxetine, wheatgrass, and AlCl3). All groups were assigned daily to different treatments for five weeks. CONCLUSIONS: AlCl3 elevated liver and kidney enzymes, over-production of oxidative stress, and inflammatory markers. Besides, accumulation of tau protein and Aß, the elevation of ACHE and GSK-3ß, down-regulation of BDNF, and ß-catenin expression in the brain. Histopathological examinations of the liver, kidney, and brain confirmed this toxicity, while treating AD groups with fluoxetine, wheatgrass, or a combination alleviates toxic insults. CONCLUSION: Fluoxetine and wheatgrass combination demonstrated a more significant neuroprotective impact in treating AD than fluoxetine alone and has protective effects on liver and kidney tissues.


Subject(s)
Alzheimer Disease/drug therapy , Fluoxetine/pharmacology , Kidney/drug effects , Liver/drug effects , Neuroprotective Agents/pharmacology , Triticum/chemistry , Aluminum Chloride/antagonists & inhibitors , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Animals , Kidney/pathology , Liver/pathology , Male , Rats , Rats, Wistar
20.
Mol Cell Biochem ; 470(1-2): 145-155, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32440841

ABSTRACT

Randall's plaque (RP) serves as a nidus on which idiopathic calcium oxalate stones form. Renal interstitial mineralization may be the cause underlying RP, and recent studies demonstrated the similarities between the interstitial mineralization and ectopic calcification. The present study aimed to investigate whether human renal interstitial fibroblasts (hRIFs) could form calcification under osteogenic conditions, and whether long non-coding RNA H19 participated in regulating osteogenic differentiation of hRIFs through Wnt-ß-catenin pathway. HRIFs were isolated and induced for osteogenic differentiation under osteogenic conditions. Runx2, OCN, alkaline phosphatase (ALP) activity, and the mineralized nodule formation were used to assess the osteogenic phenotype. Molecule expressions were determined by qRT-PCR, immunofluorescence staining, and western blot. The mineralized nodules were assessed by Alizarin red staining. Compared to the normal renal papillary tissue, Runx2, OCN, and H19 were significantly upregulated in RP. After hRIFs were induced with osteogenic medium, osteogenic markers (Runx2, OCN and ALP), ß-catenin and H19 were significantly upregulated, and the mineralized nodules are formed. Additionally, overexpression of H19 promoted the osteogenic phenotype of hRIFs and increased the expression of ß-catenin, whereas knock-down of H19 or XAV939 (inhibitor of Wnt-ß-catenin signaling pathway) significantly repressed the osteogenic phenotype of hRIFs and decreased the ß-catenin. Moreover, XAV939 was shown to abolish the osteogenic differentiation of hRIFs promoted by H19. The study demonstrated that ectopic calcification partly participated in the formation of RP, and H19 promoted osteogenic differentiation of hRIFs by activating Wnt-ß-catenin pathway, which shed new light on the molecular mechanism of the RP formation.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Fibroblasts/cytology , Osteogenesis , RNA, Long Noncoding/metabolism , Wnt Signaling Pathway , Cell Differentiation , Humans , Kidney/cytology , Mesenchymal Stem Cells/metabolism , Prevalence
SELECTION OF CITATIONS
SEARCH DETAIL