Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 90
1.
J Pharmacol Exp Ther ; 389(2): 208-218, 2024 Apr 18.
Article En | MEDLINE | ID: mdl-38453525

Renal fibrosis is distinguished by the abnormal deposition of extracellular matrix and progressive loss of nephron function, with a lack of effective treatment options in clinical practice. In this study, we discovered that the Beclin-1-derived peptide MP1 significantly inhibits the abnormal expression of fibrosis and epithelial-mesenchymal transition-related markers, including α-smooth muscle actin, fibronectin, collagen I, matrix metallopeptidase 2, Snail1, and vimentin both in vitro and in vivo. H&E staining was employed to evaluate renal function, while serum creatinine (Scr) and blood urea nitrogen (BUN) were used as main indices to assess pathologic changes in the obstructed kidney. The results demonstrated that daily treatment with MP1 during the 14-day experiment significantly alleviated renal dysfunction and changes in Scr and BUN in mice with unilateral ureteral obstruction. Mechanistic research revealed that MP1 was found to have a significant inhibitory effect on the expression of crucial components involved in both the Wnt/ß-catenin and transforming growth factor (TGF)-ß/Smad pathways, including ß-catenin, C-Myc, cyclin D1, TGF-ß1, and p-Smad/Smad. However, MP1 exhibited no significant impact on either the LC3II/LC3I ratio or P62 levels. These findings indicate that MP1 improves renal physiologic function and mitigates the fibrosis progression by inhibiting the Wnt/ß-catenin pathway. Our study suggests that MP1 represents a promising and novel candidate drug precursor for the treatment of renal fibrosis. SIGNIFICANCE STATEMENT: This study indicated that the Beclin-1-derived peptide MP1 effectively mitigated renal fibrosis induced by unilateral ureteral obstruction through inhibiting the Wnt/ß-catenin pathway and transforming growth factor-ß/Smad pathway, thereby improving renal physiological function. Importantly, unlike other Beclin-1-derived peptides, MP1 exhibited no significant impact on autophagy in normal cells. MP1 represents a promising and novel candidate drug precursor for the treatment of renal fibrosis focusing on Beclin-1 derivatives and Wnt/ß-catenin pathway.


Kidney Diseases , Prodrugs , Ureteral Obstruction , Animals , Mice , Beclin-1/metabolism , Beclin-1/pharmacology , beta Catenin/metabolism , beta Catenin/pharmacology , Fibrosis , Kidney , Kidney Diseases/drug therapy , Kidney Diseases/prevention & control , Kidney Diseases/metabolism , Prodrugs/pharmacology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factors/metabolism , Transforming Growth Factors/pharmacology , Ureteral Obstruction/complications , Ureteral Obstruction/drug therapy , Ureteral Obstruction/metabolism
2.
Discov Med ; 36(181): 323-331, 2024 Feb.
Article En | MEDLINE | ID: mdl-38409837

BACKGROUND: Childhood asthma is a chronic inflammatory disease of the respiratory tract characterized by bronchial inflammation, airway hyperresponsiveness, airflow disorder, and obstruction. Secreted frizzled-related protein 5 (SFRP5) may be associated with respiratory inflammatory diseases. This study investigated the effect of SFRP5 on human airway smooth muscle cells (HASMCs) to provide new ideas for treating asthma. METHODS: A total of 30 children with asthma and 30 children who had a physical examination at the same time were selected and divided into asthma and healthy groups. Serum SFRP5 levels were determined by enzyme-linked immunosorbent assay (ELISA) and real-time quantitative polymerase chain reaction (RT-qPCR). Lipofectamine 2000™ regent was used to transfect the SFRP5 overexpression plasmid (pc-SFRP5) or corresponding negative control (pc-NC) into HASMCs. HASMCs were treated with 10 µg/L platelet-derived growth factor-BB (PDGF-BB), which is an inducer to mimic the asthma-like condition at the cellular level of childhood asthma. HASMCs were divided into control, PDGF-BB (PDGF-BB treatment), PDGF-BB+pc-NC (pc-NC transfection and PDGF-BB treatment), and PDGF-BB+pc-SFRP5 (pc-SFRP5 transfection and PDGF-BB treatment) groups. Cell proliferation was measured by 5-ethynyl-2'-deoxyuridine (EdU) and cell counting kit-8 (CCK-8) assay. Cell migration was detected by Transwell assay. The protein expression was detected by western blot. RESULTS: Serum SFRP5 expression in the asthmatic group was decreased versus the healthy group (p < 0.0001). Induction of PDGF-BB decreased SFRP5 expression in HASMCs (p < 0.01). SFRP5 expression in the pc-SFRP5 group was increased (p < 0.01). The proliferation and migration of HASMCs increased after PDGF-BB treatment (p < 0.001, p < 0.0001), indicating that the asthma model was successfully inducted in vitro. Moreover, the expression of ß-catenin, cellular-myelocytomatosis viral oncogene (c-Myc), and cyclinD1 proteins in HASMCs increased after PDGF-BB treatment (p < 0.0001). SFRP5 overexpression partly inhibited PDGF-BB-induced proliferation, migration, and expressions of ß-catenin, c-Myc, and cyclinD proteins in HASMCs (p < 0.01, p < 0.001, p < 0.0001). CONCLUSIONS: Serum SFRP5 expression decreases in children with asthma. SFRP5 overexpression partially inhibits PDGF-BB-induced HASMC proliferation and migration by regulating the wingless-type mouse mammary tumor virus (MMTV) integration site family (Wnt)/ß-catenin pathway.


Asthma , beta Catenin , Animals , Child , Mice , Humans , Becaplermin/metabolism , Becaplermin/pharmacology , beta Catenin/metabolism , beta Catenin/pharmacology , Wnt Signaling Pathway/genetics , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Cell Proliferation/genetics , Lung/metabolism , Cell Movement , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Cells, Cultured , Adaptor Proteins, Signal Transducing/metabolism
3.
Photochem Photobiol Sci ; 23(3): 463-478, 2024 Mar.
Article En | MEDLINE | ID: mdl-38326693

UVB radiation significantly threatens skin health, contributing to wrinkle formation and an elevated risk of skin cancer. This study aimed to explore bioactive compounds with potential UVB-protective properties. Using in silico analysis, we chose compounds to reduce binding energy with matrix metalloproteinase-1 (MMP1). Piperitoside, procyanidin C1, and mulberrofuran E emerged as promising candidates through this computational screening process. We investigated the UVB-protective efficacy of the selected compounds and underlying mechanisms in human immortalized keratinocytes (HaCaT). We also investigated the molecular pathways implicated in their action, focusing on the transforming growth factor (TGF)-ß and wingless-related integration site (Wnt)/ß-catenin signaling pathways. In UVB-exposed HaCaT cells (100 mJ/cm2 for 30 min), piperitoside, procyanidin C1, and mulberrofuran E significantly reduced reactive oxygen species (ROS) and lipid peroxides, coupled with an augmentation of collagen expression. These compounds suppressed MMP1, tumor necrosis factor-alpha (TNF-α), and inducible nitric oxide synthase (iNOS) expression, while they concurrently enhanced collagen-1 (COL1A1), ß-catenin (CTNNB1), and superoxide dismutase type-1 (SOD1) expression. Furthermore, Wnt/ß-catenin inhibitors, when administered subsequently, partially counteracted the reduction in MMP1 expression and alleviated inflammatory and oxidative stress markers induced by the bioactive compounds. In conclusion, piperitoside, procyanidin C1, and mulberrofuran E protected against UVB-induced damage in HaCaT cells by inhibiting MMP1 expression and elevating ß-catenin expression. Consequently, these bioactive compounds emerge as promising preventive agents for UVB-induced skin damage, promoting skin health.


Matrix Metalloproteinase 1 , Skin Aging , Wnt Signaling Pathway , Humans , beta Catenin/metabolism , beta Catenin/pharmacology , Cell Line , Collagen/pharmacology , Keratinocytes/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/pharmacology , Reactive Oxygen Species/metabolism , Ultraviolet Rays
4.
Scand Cardiovasc J ; 58(1): 2295785, 2024 Dec.
Article En | MEDLINE | ID: mdl-38164796

Objective. Myocardial fibrosis (MF) is a common manifestation of end-stage cardiovascular diseases. Triptolide (TP) provides protection against cardiovascular diseases. This study was to explore the functional mechanism of TP in MF rats via the Wnt/ß-catenin pathway. Methods. The MF rat model was established via subcutaneous injection of isoproterenol (ISO) and treated with low/medium/high doses of TP (L-TP/M-TP/H-TP) or Wnt agonist BML-284. Cardiac function was examined by echocardiography. Pathological changes of myocardial tissues were observed by HE and Masson staining. Col-I/Col-III/Vimentin/α-SMA levels were detected by immunohistochemistry, RT-qPCR, and Western blot. Collagen volume fraction content was measured. Expression levels of the Wnt/ß-catenin pathway-related proteins (ß-catenin/c-myc/Cyclin D1) were detected by Western blot. Rat cardiac fibroblasts were utilized for in vitro validation experiments. Results. MF rats had enlarged left ventricle, decreased systolic and diastolic function and cardiac dysfunction, elevated collagen fiber distribution, collagen volume fraction and hydroxyproline content. Levels of Col-I/Col-III/Vimentin/α-SMA, and protein levels of ß-catenin/c-myc/Cyclin D1 were increased in MF rats. The Wnt/ß-catenin pathway was activated in the myocardial tissues of MF rats. TP treatment alleviated impairments of cardiac function and myocardial tissuepathological injury, decreased collagen fibers, collagen volume fraction, Col-I, Col-III, α-SMA and Vimentin levels, HYP content, inhibited Wnt/ß-catenin pathway, with H-TP showing the most significant effects. Wnt agonist BML-284 antagonized the inhibitive effect of TP on MF. TP inhibited the Wnt/ß-catenin pathway to repress the proliferation and differentiation of mouse cardiac fibroblasts in vitro. Conclusions. TP was found to ameliorate ISO-induced MF in rats by inhibiting the Wnt/ß-catenin pathway.


Cardiomyopathies , Cardiovascular Diseases , Mice , Rats , Animals , Wnt Signaling Pathway , beta Catenin/metabolism , beta Catenin/pharmacology , Cyclin D1/metabolism , Cyclin D1/pharmacology , Vimentin/metabolism , Vimentin/pharmacology , Rats, Sprague-Dawley , Fibrosis , Collagen/pharmacology
5.
Sci Total Environ ; 918: 170410, 2024 Mar 25.
Article En | MEDLINE | ID: mdl-38280596

Heat stress (HS) is a critical challenge in broilers due to the high metabolic rate and lack of sweat glands. Results from this study show that implementing a cyclic chronic HS (34 °C for 7 h/d) to finisher broilers decreased the diversity of cecal microbiota and impaired intestinal barrier, resulting in gut leak and decreased body weight (both P < 0.05). These alterations might be related to inflammatory outbursts and the retarded proliferation of intestinal epithelial cells (IECs) according to the transcriptome analysis. Considering the potential beneficial properties of Lactobacillus on intestinal development and function, the protective effects of Lactobacillus rhamnosus (L. rhamnosus) on the intestine were investigated under HS conditions in this study. Orally supplemented L. rhamnosus improved the composition of cecal microbiota and upregulated the transcription of tight junction proteins in both duodenum and jejunum, with a consequent suppression in intestinal gene expressions of pro-inflammatory cytokines and facilitation in digestive capability. Meanwhile, the jejunal villus height of the birds that received L. rhamnosus was significantly higher compared with those treated with the broth (P < 0.05). The expression abundances of genes related to IECs proliferation and differentiation were increased by L. rhamnosus, along with upregulated mRNA levels of Wnt3a and ß-catenin in jejunum. In addition, L. rhamnosus attenuated enterocyte apoptosis as indicated by decreased caspase-3 and caspase-9 gene expressions. The results indicated that oral administration with L. rhamnosus mitigated HS-induced dysfunction by promoting intestinal development and epithelial maturation in broilers and that the effects of L. rhamnosus might be dependent of Wnt/ß-catenin signaling.


Gastrointestinal Microbiome , Heat Stress Disorders , Animals , Chickens , beta Catenin/pharmacology , Lactobacillus , Dietary Supplements/analysis , Heat-Shock Response , Animal Feed/analysis
6.
Curr Med Chem ; 31(11): 1315-1331, 2024.
Article En | MEDLINE | ID: mdl-37031385

In recent years, substantial advances have been made in cancer treatment modalities. Yet, within the last three decades, neither cancer incidence nor the cancer-induced mortality rate has changed. Available anti-cancer chemotherapeutics possess remarkably restricted effectiveness and often have severe adverse effects. Hence, the identification of novel pharmaceutical agents that do not exhibit these major disadvantages is imperative. Melatonin, an important endogenous molecule synthesized and secreted by the pineal gland, is a promising chemical agent that has been comprehensively assessed over the last decades for its anti-inflammatory and anti-cancer properties. Melatonin is reportedly a significant inhibitor of cancer initiation, progression, and metastasis. The anti-- cancer potential of melatonin is principally mediated by reversing the up-regulated amounts of different transcription factors, growth factors, inflammatory cytokines, protein kinases, and other oncogenic agents. Also, melatonin often has signifcant inhibitory effects on cancer cell proliferation through either promoting apoptosis or inducing cell cycle arrest. The current review provides an insight into melatonin-induced effects against various human cancers with a particular focus on the regulation of Wnt/ß-catenin signaling pathway.


Melatonin , Neoplasms , Humans , Melatonin/pharmacology , Melatonin/therapeutic use , Wnt Signaling Pathway , Neoplasms/pathology , Cell Proliferation , Intercellular Signaling Peptides and Proteins , Apoptosis , beta Catenin/metabolism , beta Catenin/pharmacology , Cell Line, Tumor
7.
Tissue Eng Part B Rev ; 30(2): 270-283, 2024 Apr.
Article En | MEDLINE | ID: mdl-37795571

Background and Aims: Recent research has focused on developing nanoparticle and nanotopography-based technologies for bone regeneration. The Wingless-related integration site (Wnt) signaling pathway has been shown to play a vital role in this process, in particular in osteogenic differentiation and proliferation. The exact mechanisms by which nanoparticles and nanotopographies activate the Wnt signaling pathway, however, are not fully understood. This review aimed to elucidate the mechanisms by which nanoscale technologies activate the Wnt signaling pathway during bone regeneration. Methods: The terms "Wnt," "bone," and "nano*" were searched on PubMed and Ovid with no date limit. Only original research articles related to Wnt signaling and bone regeneration in the context of nanotopographies, nanoparticles, or scaffolds with nanotopographies/nanoparticles were reviewed. Results: The primary mechanism by which nanoparticles activated the Wnt pathway was by internalization through the endocytic pathway or diffusion through the cell membrane, leading to accumulation of nonphosphorylated ß-catenin in the cytoplasm and subsequently downstream osteogenic signaling (e.g., upregulation of runt-related transcription factor 2 [RUNX2]). The specific size of the nanoparticles and the process of endocytosis itself has been shown to modulate the Wnt-ß-catenin pathway. Nanotopographies were shown to directly activate frizzled receptors, initiating Wnt/ß-catenin signaling. Additional studies showed nanotopographies to activate the Wnt/calcium (Wnt/Ca2+)-dependent and Wnt/planar cell polarity pathways through nuclear factor of activated T cells, and α5ß1 integrin stimulation. Finally, scaffolds containing nanotopographies/nanoparticles were found to induce Wnt signaling through a combination of ion release (e.g., lithium, boron, lanthanum, and icariin), which inhibited glycogen synthase kinase 3 beta (GSK-3ß) activity, and through similar mechanisms to the nanotopographies. Conclusion: This review concludes that nanoparticles and nanotopographies cause Wnt activation through several different mechanisms, specific to the size, shape, and structure of the nanoparticles or nanotopographies. Endocytosis-related mechanisms, integrin signaling and ion release were the major mechanisms identified across nanoparticles, nanotopographies, and scaffolds, respectively. Knowledge of these mechanisms will help develop more effective targeted nanoscale technologies for bone regeneration. Impact statement Nanoparticles and nanotopographies can activate the Wingless-related integration site (Wnt) signaling pathway, which is essential for bone regeneration. This review has identified that activation is due to endocytosis, integrin signaling and ion release, depending on the size, shape, and structure of the nanoparticles or nanotopographies. By identifying and further understanding these mechanisms, more effective nanoscale technologies that target the Wnt signaling pathway can be developed. These technologies can be used for the treatment of nonunion bone fractures, a major clinical challenge, with the potential to improve the quality of life of millions of patients around the world.


Nanoparticles , Wnt Signaling Pathway , Humans , Osteogenesis , beta Catenin/metabolism , beta Catenin/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/pharmacology , Quality of Life , Bone Regeneration , Cell Differentiation , Nanoparticles/chemistry , Integrins , Cells, Cultured
8.
Int Wound J ; 21(4): e14589, 2024 Apr.
Article En | MEDLINE | ID: mdl-38135901

This study focused on unravelling the role of PCAT-1 in wound-healing process, particularly its impact on regenerative and osteogenic abilities of mesenchymal stem cells (MSCs). We delved into how PCAT-1 regulates mitochondrial oxidative phosphorylation (OXPHOS) and interacts with pivotal molecular pathways, especially ß-catenin and PKM2, using human bone marrow-derived MSCs. MSCs were cultured under specific conditions and PCAT-1 expression was modified through transfection. We thoroughly assessed several critical parameters: MSC proliferation, mitochondrial functionality, ATP production and expression of wound healing and osteogenic differentiation markers. Further, we evaluated alkaline phosphatase (ALP) activity and mineral deposition, essential for bone healing. Our findings revealed that overexpressing PCAT-1 significantly reduced MSC proliferation, hampered mitochondrial performance and lowered ATP levels, suggesting the clear inhibitory effect of PCAT-1 on these vital wound-healing processes. Additionally, PCAT-1 overexpression notably decreased ALP activity and calcium accumulation in MSCs, crucial for effective bone regeneration. This overexpression also led to the reduction in osteogenic marker expression, indicating suppression of osteogenic differentiation, essential in wound-healing scenarios. Moreover, our study uncovered a direct interaction between PCAT-1 and the PKM2/ß-catenin pathway, where PCAT-1 overexpression intensified PKM2 activity while inhibiting ß-catenin, thereby adversely affecting osteogenesis. This research thus highlights PCAT-1's significant role in impairing wound healing, offering insights into the molecular mechanisms that may guide future therapeutic strategies for enhancing wound repair and bone regeneration.


Mitochondrial Diseases , beta Catenin , Humans , beta Catenin/metabolism , beta Catenin/pharmacology , Osteogenesis/physiology , Wound Healing , Bone Marrow Cells/metabolism , Adenosine Triphosphate/pharmacology , Cell Differentiation/physiology , Cells, Cultured
9.
Stem Cells Transl Med ; 13(3): 255-267, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38159248

BACKGROUND: Mesenchymal stem cells (MSCs) have been widely studied to alleviate acute lung injury (ALI) due to their paracrine function. However, the microenvironment of inflammatory outbreaks significantly restricted the factors secreted from MSCs like keratinocyte growth factor (KGF). KGF is a growth factor with tissue-repaired ability. Is there a better therapeutic prospect for MSCs in combination with compounds that promote their paracrine function? Through compound screening, we screened out isoxazole-9 (ISX-9) to promote MSCs derived KGF secretion and investigated the underlying mechanisms of action. METHODS: Compounds that promote KGF secretion were screened by a dual-luciferase reporter gene assay. The TMT isotope labeling quantitative technique was used to detect the differential proteins upon ISX-9 administrated to MSCs. The expressions of NGFR, ERK, TAU, and ß-catenin were detected by Western blot. In the ALI model, we measured the inflammatory changes by HE staining, SOD content detection, RT-qPCR, immunofluorescence, etc. The influence of ISX-9 on the residence time of MSCs transplantation was explored by optical in vivo imaging. RESULTS: We found out that ISX-9 can promote the expression of KGF in MSCs. ISX-9 acted on the membrane receptor protein NGFR, upregulated phosphorylation of downstream signaling proteins ERK and TAU, downregulated phosphorylation of ß-catenin, and accelerated ß-catenin into the nucleus to further increase the expression of KGF. In the ALI model, combined ISX-9 with MSCs treatments upgraded the expression of KGF in the lung, and enhanced the effect of MSCs in reducing inflammation and repairing lung damage compared with MSCs alone. CONCLUSIONS: ISX-9 facilitated the secretion of KGF from MSCs both in vivo and in vitro. The combination of ISX-9 with MSCs enhanced the paracrine function and anti-inflammatory effect of MSCs compared with MSCs applied alone in ALI. ISX-9 played a contributive role in the transplantation of MSCs for the treatment of ALI.


Acute Lung Injury , Isoxazoles , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Thiophenes , Humans , beta Catenin/metabolism , beta Catenin/pharmacology , Fibroblast Growth Factor 7/metabolism , Fibroblast Growth Factor 7/pharmacology , Acute Lung Injury/therapy , Mesenchymal Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism
10.
J Hazard Mater ; 464: 132917, 2024 02 15.
Article En | MEDLINE | ID: mdl-37979429

Zearalenone (ZEA) poses severe reproductive toxicity to both humans and animals. Scutellarin has been demonstrated to rescue ZEA-induced apoptosis in mouse ovarian granulosa cells (GCs), but its specific targets remain unclear. In the present study, the potential targets of scutellarin were determined to clarify the mechanisms of scutellarin against ZEA-induced ovarian damage. 287 targets of scutellarin in mouse ovarian GCs were obtained by magnetic nano-probe-based fishing assay and liquid chromatography-tandem mass spectrometry. Wnt5a had the lowest binding free energy with scutellarin at - 8.3 kcal/mol. QRT-PCR and western blot showed that scutellarin significantly increased the Wnt5a and ß-catenin expression compared with the ZEA-treated group, and cleaved-caspase-3 expression was significantly increased in the scutellarin-treated group after interfering with the expression of Wnt5a. The affinity constant (KD) of Wnt5a and scutellarin was 1.7 × 10-5 M. The pull-down assay also demonstrated that scutellarin could specifically bind to Wnt5a protein. Molecular docking results showed that scutellarin could form hydrogen bonds with TRY52, GLN56, and SER90 on Wnt5a protein, and western blot assay confirmed SER90 was an important site for the binding. Scutellarin significantly increased Wnt5a and ß-catenin expression and decreased cleaved-caspase-3 expression in ovarian tissues of mice. In conclusion, scutellarin exerted anti-apoptotic effects on ZEA-induced mouse ovarian GCs by targeting Wnt5a.


Zearalenone , Humans , Female , Mice , Animals , Zearalenone/toxicity , Wnt-5a Protein/metabolism , Wnt-5a Protein/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , beta Catenin/metabolism , beta Catenin/pharmacology , Granulosa Cells/metabolism , Molecular Docking Simulation , Apoptosis
11.
Pestic Biochem Physiol ; 197: 105649, 2023 Dec.
Article En | MEDLINE | ID: mdl-38072524

Thiram is a plant fungicide, its excessive use has exceeded the required environmental standards. It causes tibial dyschondroplasia (TD) in broilers which is a common metabolic disease that affects the growth plate of tibia bone. It has been studied that many microRNAs (miRNAs) are involved in the differentiation of chondrocytes however, their specific roles and mechanisms have not been fully investigated. The selected features of tibial chondrocytes of broilers were studied in this experiment which included the expression of miR-181b-1-3p and the genes related to WIF1/Wnt/ß-catenin pathway in chondrocytes through qRT-PCR, western blot and immunofluorescence. The correlation between miR-181b-1-3p and WIF1 was determined by dual luciferase reporter gene assay whereas, the role of miR-181b-1-3p and WIF1/Wnt/ß-catenin in chondrocyte differentiation was determined by mimics and inhibitor transfection experiments. Results revealed that thiram exposure resulted in decreased expression of miR-181b-1-3p and increased expression of WIF1 in chondrocytes. A negative correlation was also observed between miR-181b-1-3p and WIF1. After overexpression of miR-181b-1-3p, the expression of ACAN, ß-catenin and Col2a1 increased but the expression of GSK-3ß decreased. It was observed that inhibition of WIF1 increased the expression of ALP, ß-catenin, Col2a1 and ACAN but decreased the expression of GSK-3ß. It is concluded that miR-181b-1-3p can reverse the inhibitory effect of thiram on cartilage proliferation and differentiation by inhibiting WIF1 expression and activating Wnt/ß-catenin signaling pathway. This study provides a new molecular target for the early diagnosis and possible treatment of TD in broilers.


MicroRNAs , Osteochondrodysplasias , Animals , Chondrocytes/metabolism , Chickens/genetics , Chickens/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Osteochondrodysplasias/genetics , Osteochondrodysplasias/veterinary , Osteochondrodysplasias/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Thiram , Tibia/metabolism , MicroRNAs/genetics , Cell Proliferation/genetics
12.
AIDS Res Ther ; 20(1): 82, 2023 11 19.
Article En | MEDLINE | ID: mdl-37981694

BACKGROUND: Human immunodeficiency virus (HIV) infection is associated with an elevated incidence of cervical cancer, and accelerated disease progression, but the underlying mechanisms are not well understood. This study aimed to investigate the relationship between HIV infection and epithelial-mesenchymal transition (EMT) in cervical cancer. METHODS: Tissue samples from HIV-positive and negative patients with cervical intraepithelial neoplasia (CIN) and cervical cancer were analyzed for EMT-related proteins. Human cervical cancer SiHa cells were treated with HIV Tat and gp120 proteins to test their effects on EMT, migration, and invasion. RESULTS: HIV-positive patients had lower E-cadherin and cytokeratin, and higher N-cadherin and vimentin levels than HIV-negative patients. HIV Tat and gp120 proteins induced EMT, migration, and invasion in SiHa cells. Transcriptome sequencing analysis revealed that, compared to the control group, the protein-treated group showed upregulation of 22 genes and downregulation of 77 genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed the involvement of the Wnt signaling pathway in EMT. Further analysis of gene expression related to this pathway revealed upregulation of DVL1, TCF7, KRT17, and VMAC, while GSK3ß, SFRP2, and CDH1 were downregulated. Immunofluorescence assay demonstrated that HIVgp120 and Tat proteins treatment induced elevated ß-catenin expression with nuclear accumulation in SiHa cells. CONCLUSIONS: The treatment of SiHa cells with HIV Tat and gp120 proteins induces EMT and activates the Wnt/ß-catenin pathway, suggesting that the Wnt/ß-catenin pathway may play a crucial role in promoting EMT progression in cervical lesion tissues of HIV-infected patients.


HIV Infections , Uterine Cervical Neoplasms , Female , Humans , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Cell Line, Tumor , Gene Products, tat/pharmacology , Epithelial-Mesenchymal Transition/physiology
13.
Endokrynol Pol ; 74(5): 544-552, 2023.
Article En | MEDLINE | ID: mdl-37779373

INTRODUCTION: Recent studies have shown that a decline in isletß cells quality is due to ß-cell dedifferentiation, not only ß-cell apoptosis. Angiotensin (1-7) [Ang(1-7)] could attenuate high glucose-induced apoptosis and dedifferentiation of pancreaticß cells by combining with MAS receptors. However, the mechanism of such action has not been elucidated. Recent studies have revealed that Wnt/ß-catenin and forkhead box transcription factor O1 (FoxO1) are associated with ß-cell dedifferentiation. Our study aims to explore whether the effects of Ang(1-7)on islet b cell dedifferentiation are mediated through the Wnt/ß-catenin/FoxO1 pathway. MATERIAL AND METHODS: Isletß cells were divided into 6 groups: a control group, a high-glucose group, high glucose with Ang(1-7) group, high-glucose with Ang(1-7) and A779 group, high-glucose with angiotensin(1-7) and CHIR99021 group, and high-glucose with CHIR99021 group. A779 is a kind of MAS receptor antagonist that blocks the action of Ang(1-7), and CHIR99021 is a Wnt pathway activator. The morphology of pancreaticß cells was observed in each group after 48 hours of intervention. ß-cell insulin secretory function and expressions of relevant factors were measured. RESULTS: Compared with the control group, the cell morphology became degraded in the high-glucose group and the capability of insulin secretion was reduced. Meanwhile, the expressions of matureß cells markers [pancreatic and duodenal homeobox 1 (Pdx1) and MAF BZIP transcription factor A (MafA)] were reduced, while the expressions of endocrine progenitor cells makers [octamer-binding transcription factor 4 (Oct4) and Nanog] were increased. The addition of CHIR99021 resulted in profound deep destruction ofß cells compared with the high-glucose group. However, such changes were dramatically reversed following the treatment of Ang(1-7). The addition of A779 significantly inhibited the improvement caused by Ang(1-7). CONCLUSION: Ang(1-7) can effectively reverseß cell dedifferentiation through Wnt/ß-catenin/FoxO1 pathway. It might be a new strategy for preventing and treating diabetes.


Glucose , Insulin-Secreting Cells , Humans , Glucose/pharmacology , Glucose/metabolism , Wnt Signaling Pathway , Cell Dedifferentiation , beta Catenin/metabolism , beta Catenin/pharmacology , Insulin-Secreting Cells/metabolism
14.
Fitoterapia ; 171: 105684, 2023 Dec.
Article En | MEDLINE | ID: mdl-37751799

Houttuynia cordata (Thunb.), an important medicinal plant of Northeast India, Korea, and China, is used to treat various ailments and for anticancer research. Knowing its traditional practices, we are interested in the mode-of-action of HCT on HepG2 to co-relate the traditional practice with modern drug therapeutics. UPLC-Q-ToF-Ms analysis of HCT reveals identification of 14 metabolites. Network pharmacology analysis of the 14 compounds showed interaction with 232 different targets with their potential involvement in hepatocellular carcinoma. Whole extracts impart cytotoxicity on variety of cell lines including HepG2. There was a significant morphological alteration in treated HepG2 cells due to impairment of cytoskeletal components like ß and γ- tubulin. Arrest at G1-S checkpoint was clearly indicated downregulation of Cyclin D1. The root extracts actuated apoptosis in HepG2 as evident from altered mitochondrial membrane potential, Annexin V- FITC, BrdU-PI, AO/EtBr assays, and modulations of apoptotic protein expression but without ROS generation. Whole extracts caused abrogation of epithelial to mesenchymal transition with repression of Snail, N-Cadherin, Vimentin, MMP-9, and upregulation of Pan-Cadherin. Pathway analysis found GSK-3ß in Wnt/ß-Catenin signaling cascade to be involved through Hepatocellular carcinoma (hsa05225) pathway. The GSK-3ß/ß-Catenin/PDL-1 signaling was found to be inhibited with the downregulation of pathway components. This was further confirmed by application of EGF, an inducer of the GSK-3ß/ß-Catenin pathway that neutralized the effect of Houttuynia cordata (Thunb.) root extract on the said pathway. Network pharmacology analysis also confirms the synergy network with botanical-bioactive-target-disease which showed Kaempferol to have the highest degree of association with the said pathway.


Carcinoma, Hepatocellular , Houttuynia , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/pharmacology , Houttuynia/metabolism , Cell Line, Tumor , beta Catenin/metabolism , beta Catenin/pharmacology , Tandem Mass Spectrometry , Epithelial-Mesenchymal Transition , Cell Proliferation , Molecular Structure , Wnt Signaling Pathway , Liver Neoplasms/drug therapy , Apoptosis
15.
BMC Med ; 21(1): 366, 2023 09 25.
Article En | MEDLINE | ID: mdl-37743483

BACKGROUND: Multitarget tyrosine kinase inhibitors (mTKIs) such as Regorafenib and Sorafenib have already been approved for the treatment of many solid tumours. However, the efficacy of mTKIs in colorectal cancer (CRC) is limited; the underlined mechanism remains largely elusive. Our study was aimed to find out the resistance mechanism of mTKIs in CRC. METHODS: RNA sequencing was used to identify the expression of Activin A receptor-like type 1 (ACVRL1) under the treatment of mTKIs. Gain/loss-of-function experiments were performed to assess the biological function of ACVRL1 in resistance to mTKIs. The underlying mechanisms of ACVRL1-mediated mTKI resistance were investigated by using liquid chromatography-mass spectrometry assays (LC-MS), co-immunoprecipitation assays (Co-IP), chromatin immunoprecipitation assays, ubiquitination assays, dual luciferase reporter assays, etc. RESULTS: RNA sequencing identified the activation of ACVRL1 under the treatment of mTKIs in CRC cells. ACVRL1 knockdown and overexpression significantly affects the sensitivity of CRC cells to mTKIs both in vitro and vivo. Mechanistically, we found the ß-catenin/TCF-1-KCNQ1OT1/miR-7-5p axis mediated the activation of ACVRL1. Furthermore, LC-MS assays indicated the interaction between ACVRL1 and glutathione peroxidase 2(GPX2) protein. IP assay defined ACVRL1 truncation (282-503aa) could be responsible for interacting with GPX2, and rescue experiments with ACVRL1 truncations confirmed the importance of this interaction in driving mTKI resistance. Co-IP assays confirmed that ACVRL1 associates with ubiquitin-specific peptidase 15(USP15) which directly deubiquinates GPX2 at the K187(K, lysine) site, leading to the accumulation of GPX2 protein. Rescue experiments performed with the lysine mutants in GPX2 CRISPR knockout cell model confirmed the importance of GPX2 K187 mutant. As a result, the increased ROS clearance and decreased cell apoptosis eventually lead to mTKI resistance in CRC. CONCLUSIONS: Our results demonstrate that the Wnt/ß-catenin/KCNQ1OT1/miR-7-5p/ACVRL1/GPX2 biological axis plays a vital role in CRC, targeting which may be an effective approach for overcoming mTKI resistance.


Colorectal Neoplasms , MicroRNAs , Humans , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Lysine/genetics , Lysine/metabolism , Lysine/pharmacology , MicroRNAs/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Peroxidase/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Activin Receptors, Type II/pharmacology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/pharmacology
16.
Funct Integr Genomics ; 23(4): 314, 2023 Sep 30.
Article En | MEDLINE | ID: mdl-37777676

The dysfunction of blood-brain barrier (BBB) plays a pivotal role in brain injury and subsequent neurological deficits of ischemic stroke. The current study aimed to examine the potential correlation between p53 inhibition and the neuroprotective effect of on the BBB. Rat middle cerebral artery occlusion and reperfusion model (MCAO/R) and oxygen-glucose deprivation/re-oxygenation model (OGD/R) were employed to simulate cerebral ischemia-reperfusion (CI/R) injury occurrence in vivo and in vitro. mNSS and TTC staining were applied to evaluate neurological deficits and brain infarct volumes. Evans blue (EB) staining was carried out to examine the permeability of BBB. RT-qPCR and Western blot to examine the mRNA and protein levels. Cell viabilities were detected by CCK-8. Flow cytometry and ELISA assay were employed to examine apoptosis and neuroinflammation levels. TEER value and sodium fluorescein were carried out to explore the permeability of HBMEC cells. PFT-α inhibited P53 and promoted the expression of ß-catenin and cyclin D1, which were reversed by DKK1. PFT-α inhibited neurological deficits, brain infarct volume, neuroinflammation, apoptosis, and BBB integrity than the MCAO/R rats; however, this inhibition was reversed by DKK1. PFT-α promoted OGD/R-induced cell viability in NSCs, and suppressed inflammation and apoptosis, but DKK1 weakened the effect of PFT-α. PFT-α increased OGD/R-induced TEER values in cerebrovascular endothelial cells, inhibited sodium fluorescein permeability, and increased the mRNA levels of tight junction protein, but they were all attenuated by DKK1. PFT-α protects the BBB after acute ischemic stroke via the Wnt/ß-catenin pathway, which in turn improves neurological function.


Ischemic Stroke , Reperfusion Injury , Wnt Signaling Pathway , Animals , Rats , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Blood-Brain Barrier/metabolism , Brain Infarction/metabolism , Endothelial Cells/metabolism , Fluorescein/metabolism , Fluorescein/pharmacology , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Neuroinflammatory Diseases , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics , Wnt Signaling Pathway/drug effects
17.
J Oral Biosci ; 65(4): 347-355, 2023 12.
Article En | MEDLINE | ID: mdl-37625505

OBJECTIVES: This study aimed to explore the role and specific mechanism of the cholesterol-lowering drug simvastatin in inhibiting oral squamous cell carcinoma (OSCC). METHODS: The proliferation, apoptosis, and migration levels of OSCC cells were detected by CCK8, quantitative real-time polymerase chain reaction, Western blot, colony formation, TdT-mediated dUTP Nick-End Labeling assay, and wound healing assay. The inhibitory effect of simvastatin in vivo was detected by a mouse xenograft tumor model. Immunohistochemistry and immunofluorescence staining were used to assess the KLF2 and ß-catenin expressions in cells and tissues. RESULTS: KLF2 expression in OSCC cells and tissues was downregulated. The addition of KLF2 inducer, GGTI298, inhibited the proliferation and migration of OSCC cells. Simvastatin played a role in inhibiting the proliferation and promoting the apoptosis of OSCC cells. Moreover, it inhibited ß-catenin expression and promoted KLF2 expression in OSCC cells. KLF2 siRNA reversed the effect of simvastatin on the proliferation and apoptosis of OSCC cells. CONCLUSIONS: KLF2, as a tumor suppressor gene, may be an important marker for diagnosing and treating OSCC. Simvastatin inhibits the progression of OSCC by regulating the KLF2 signal.


Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Animals , Mice , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Squamous Cell Carcinoma of Head and Neck , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Simvastatin/pharmacology , Simvastatin/therapeutic use , Cell Line, Tumor , Cell Proliferation/genetics , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Apoptosis/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/pharmacology
18.
Chin J Dent Res ; 26(2): 69-75, 2023 06 23.
Article En | MEDLINE | ID: mdl-37395518

OBJECTIVE: To investigate the potential effect of small molecule nitazoxanide (NTZ) on the osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). METHODS: Cell counting Kit-8 assay was used to examine the effect of NTZ on proliferation of BMSCs. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot analysis were used to measure the expression of osteogenic and adipogenic marker gene. Alkaline phosphatase (ALP) staining and activity assay and Alizarin Red S (ARS) staining were used to investigate the effect of NTZ on osteogenesis. Oil red O (ORO) staining assay was used to assess the impact of NTZ on adipogenesis. RESULTS: NTZ significantly suppressed the osteogenic differentiation but promoted the adipogenic differentiation of BMSCs. Mechanistically, NTZ regulated osteogenic/adipogenic differentiation of BMSCs by inhibiting the Wnt/ß-catenin signalling pathway. The addition of Wnt/ß-catenin signalling pathway activator, lithium chloride, could reverse the effect of NTZ on BMSCs. CONCLUSION: NTZ affected osteogenic and adipogenic differentiation of BMSCs with the involvement of Wnt/ß-catenin signalling pathway. This finding expanded the understanding of NTZ pharmacology and indicated that NTZ might have an adverse effect on bone homeostasis.


Adipogenesis , Mesenchymal Stem Cells , Osteogenesis/genetics , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Cell Differentiation , Coloring Agents , Cells, Cultured
19.
Clin Exp Immunol ; 214(1): 120-129, 2023 12 11.
Article En | MEDLINE | ID: mdl-37402316

Excessive inflammatory injury is the main cause of the incidence of severe neonatal pneumonia (NP) and associated deaths. Although dickkopf-3 (DKK3) exhibits anti-inflammatory activity in numerous pathological processes, its role in NP is still unknown. In this study, human embryonic lung WI-38 and MRC-5 cells were treated with lipopolysaccharide (LPS) to induce inflammatory injury of NP in vitro. The expression of DKK3 was downregulated in LPS-stimulated WI-38 and MRC-5 cells. DKK3 overexpression decreased LPS-induced inhibition of cell viability, and reduced LPS-induced apoptosis of WI-38 and MRC-5 cells. DKK3 overexpression also reduced LPS-induced production of pro-inflammatory factors such as ROS, IL-6, MCP-1, and TNF-α. Nuclear respiratory factors 1 (NRF1) knockdown was found to upregulate DKK3 and inactivate the GSK-3ß/ß-catenin pathway in LPS-injured WI-38 and MRC-5 cells. NRF1 knockdown also suppressed LPS-induced inhibition on cell viability, repressed LPS-induced apoptosis, and inhibited the accumulation of ROS, IL-6, MCP-1, and TNF-α in LPS-injured WI-38 and MRC-5 cells. DKK3 knockdown or re-activation of the GSK-3ß/ß-catenin pathway reversed the inhibitory effects of NRF1 knockdown on LPS-induced inflammatory injury. In conclusion, NRF1 knockdown can alleviate LPS-triggered inflammatory injury by regulating DKK3 and the GSK-3ß/ß-catenin pathway.


Pneumonia , Signal Transduction , Infant, Newborn , Humans , Lipopolysaccharides , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/pharmacology , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Reactive Oxygen Species/metabolism , Apoptosis/genetics , Adaptor Proteins, Signal Transducing/genetics
20.
J Integr Med ; 21(5): 474-486, 2023 09.
Article En | MEDLINE | ID: mdl-37453868

OBJECTIVE: Jiedu Recipe (JR), a Chinese herbal remedy, has been shown to prolong overall survival time and decrease recurrence and metastasis rates in patients with hepatocellular carcinoma (HCC). This work investigated the mechanism of JR in HCC treatment. METHODS: The chemical constituents of JR were detected using liquid chromatography-mass spectrometry. The potential anti-HCC mechanism of JR was screened using network pharmacology and messenger ribonucleic acid (mRNA) microarray chip assay, followed by experimental validation in human HCC cells (SMMC-7721 and Huh7) in vitro and a nude mouse subcutaneous transplantation model of HCC in vivo. HCC cell characteristics of proliferation, migration and invasion under hypoxic setting were investigated using thiazolyl blue tetrazolium bromide, wound healing and Transwell assays, respectively. Image-iT™ Hypoxia Reagent was added to reveal hypoxic conditions. Stem cell sphere formation assay was used to detect the stemness. Epithelial-mesenchymal transition (EMT) markers like E-cadherin, vimentin and α-smooth muscle actin, and pluripotent transcription factors including nanog homeobox, octamer-binding transcription factor 4, and sex-determining region Y box protein 2 were analyzed using Western blotting and real-time polymerase chain reaction. Western blot was performed to ascertain the anti-HCC effect of JR under hypoxia involving the Wnt/ß-catenin pathway. RESULTS: According to network pharmacology and mRNA microarray chip analysis, JR may potentially act on hypoxia and inhibit the Wnt/ß-catenin pathway. In vitro and in vivo experiments showed that JR significantly decreased hypoxia, and suppressed HCC cell features of proliferation, migration and invasion; furthermore, the hypoxia-induced increases in EMT and stemness marker expression in HCC cells were inhibited by JR. Results based on the co-administration of JR and an agonist (LiCl) or inhibitor (IWR-1-endo) verified that JR suppressed HCC cancer stem-like properties under hypoxia by blocking the Wnt/ß-catenin pathway. CONCLUSION: JR exerts potent anti-HCC effects by inhibiting cancer stemness via abating the Wnt/ß-catenin pathway under hypoxic conditions. Please cite this article as: Guo BJ, Ruan Y, Wang YJ, Xiao CL, Zhong ZP, Cheng BB, Du J, Li B, Gu W, Yin ZF. Jiedu Recipe, a compound Chinese herbal medicine, inhibits cancer stemness in hepatocellular carcinoma via Wnt/ß-catenin pathway under hypoxia. J Integr Med. 2023; 21(5): 474-486.


Carcinoma, Hepatocellular , Drugs, Chinese Herbal , Liver Neoplasms , Animals , Mice , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , RNA, Messenger/therapeutic use , Cell Line, Tumor , Cell Proliferation , Cell Movement , Gene Expression Regulation, Neoplastic
...