Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.840
Filter
1.
Int Heart J ; 65(3): 475-486, 2024.
Article in English | MEDLINE | ID: mdl-38825493

ABSTRACT

This study aimed to investigate the molecular mechanisms underlying the protective effects of cyclooxygenase (cox) inhibitors against myocardial hypertrophy.Rat H9c2 cardiomyocytes were induced by mechanical stretching. SD rats underwent transverse aortic constriction to induce pressure overload myocardial hypertrophy. Rats were subjected to echocardiography and tail arterial pressure in 12W. qPCR and western blot were used to detect the expression of Notch-related signaling. The inflammatory factors were tested by ELISA in serum, heart tissue, and cell culture supernatant.Compared with control, levels of pro-inflammatory cytokines IL-6, TNF-α, and IL-1ß were increased and anti-inflammatory cytokine IL-10 was reduced in myocardial tissues and serum of rat models. Levels of Notch1 and Hes1 were reduced in myocardial tissues. However, cox inhibitor treatment (aspirin and celecoxib), the improvement of exacerbated myocardial hypertrophy, fibrosis, dysfunction, and inflammation was parallel to the activation of Notch1/Hes1 pathway. Moreover, in vitro experiments showed that, in cardiomyocyte H9c2 cells, application of ~20% mechanical stretching activated inflammatory mediators (IL-6, TNF-α, and IL-1ß) and hypertrophic markers (ANP and BNP). Moreover, expression levels of Notch1 and Hes1 were decreased. These changes were effectively alleviated by aspirin and celecoxib.Cox inhibitors may protect heart from hypertrophy and inflammation possibly via the Notch1/Hes1 signaling pathway.


Subject(s)
Aspirin , Celecoxib , Myocytes, Cardiac , Rats, Sprague-Dawley , Receptor, Notch1 , Signal Transduction , Transcription Factor HES-1 , Animals , Receptor, Notch1/metabolism , Rats , Transcription Factor HES-1/metabolism , Signal Transduction/drug effects , Celecoxib/pharmacology , Aspirin/pharmacology , Aspirin/therapeutic use , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Cardiomegaly/etiology , Disease Models, Animal
2.
Cell Commun Signal ; 22(1): 256, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38705997

ABSTRACT

BACKGROUND: Melanoma is a highly heterogeneous cancer, in which frequent changes in activation of signaling pathways lead to a high adaptability to ever changing tumor microenvironments. The elucidation of cancer specific signaling pathways is of great importance, as demonstrated by the inhibitor of the common BrafV600E mutation PLX4032 in melanoma treatment. We therefore investigated signaling pathways that were influenced by neurotrophin NRN1, which has been shown to be upregulated in melanoma. METHODS: Using a cell culture model system with an NRN1 overexpression, we investigated the influence of NRN1 on melanoma cells' functionality and signaling. We employed real time cell analysis and spheroid formation assays, while for investigation of molecular mechanisms we used a kinase phosphorylation kit as well as promotor activity analysis followed by mRNA and protein analysis. RESULTS: We revealed that NRN1 interacts directly with the cleaved intracellular domain (NICD) of Notch1 and Notch3, causing a potential retention of NICD in the cytoplasm and thereby reducing the expression of its direct downstream target Hes1. This leads to decreased sequestration of JAK and STAT3 in a Hes1-driven phosphorylation complex. Consequently, our data shows less phosphorylation of STAT3 while presenting an accumulation of total protein levels of STAT3 in association with NRN1 overexpression. The potential of the STAT3 signaling pathway to act in both a tumor suppressive and oncogenic manner led us to investigate specific downstream targets - namely Vegf A, Mdr1, cMet - which were found to be upregulated under oncogenic levels of NRN1. CONCLUSIONS: In summary, we were able to show that NRN1 links oncogenic signaling events between Notch and STAT3 in melanoma. We also suggest that in future research more attention should be payed to cellular regulation of signaling molecules outside of the classically known phosphorylation events.


Subject(s)
Melanoma , Neuropeptides , STAT3 Transcription Factor , Signal Transduction , Humans , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Melanoma/metabolism , Melanoma/genetics , Melanoma/pathology , Phosphorylation , Protein Binding , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Receptor, Notch3/metabolism , Receptor, Notch3/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics
3.
Cardiovasc Toxicol ; 24(6): 587-597, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38691303

ABSTRACT

Vascular lesions frequently arise as complication in patients diagnosed with diabetes mellitus (DM). Presently, percutaneous coronary intervention (PCI) and antithrombotic therapy serve as primary treatments. However, in-stent restenosis persists as a challenging clinical issue following PCI, lacking sustained and effective treatment. Linarin (LN) exhibits diverse pharmacological activities and is regarded as a potential drug for treating various diseases, including DM. But its specific role in restenosis after vascular injury in DM patients remains unclear. A rat model of diabetes-related restenosis was established to evaluate the role of LN on neointimal hyperplasia. Vascular smooth muscle cells (VSMCs) stimulated by high glucose (HG, 30 mM) underwent LN treatment. Additionally, an overexpression plasmid of A disintegrin and metalloproteinases (ADAM10) was constructed to transfect VSMCs. We employed CCK-8, Brdu, wound-healing scratch, and transwell migration assays to evaluate the proliferation and migration of VSMCs. Furthermore, western blot and immunofluorescence assays were utilized to investigate the expressions of ADAM10 and the downstream Notch signaling pathway in vivo and in vitro models. LN notably alleviated intimal hyperplasia after vascular injury in DM rats and reduced the protein expression of ADAM10, alongside its downstream Notch1 signaling pathway-related proteins (Notch1, NICD and Hes1) in rat carotid artery tissues. LN effectively suppressed the proliferation and migration of VSMCs induced by HG, downregulating the protein expression of ADAM10, Notch1, NICD and Hes1. Moreover, our findings indicated that ADAM10 overexpression significantly reversed LN's effects on proliferation, migration, and the expression of Notch1 signaling pathway-related proteins in HG-treated VSMCs. LN demonstrates potential therapeutic efficacy in addressing restenosis after diabetic-related vascular injury, with the ADAM10 mediated Notch signaling pathway playing a pivotal role.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Carotid Artery Injuries , Cell Movement , Cell Proliferation , Diabetes Mellitus, Experimental , Membrane Proteins , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Neointima , Rats, Sprague-Dawley , Signal Transduction , Animals , ADAM10 Protein/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/enzymology , Cell Movement/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/enzymology , Cell Proliferation/drug effects , Male , Membrane Proteins/metabolism , Membrane Proteins/genetics , Amyloid Precursor Protein Secretases/metabolism , Cells, Cultured , Carotid Artery Injuries/pathology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/enzymology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Hyperplasia , Receptors, Notch/metabolism , Receptor, Notch1/metabolism , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Disease Models, Animal , Rats , Coronary Restenosis/pathology , Coronary Restenosis/etiology , Coronary Restenosis/metabolism , Coronary Restenosis/prevention & control
4.
Zhongguo Zhong Yao Za Zhi ; 49(7): 1741-1748, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38812186

ABSTRACT

Ulcerative colitis(UC) is one of the common gastrointestinal diseases worldwide. In recent years, the incidence of UC has been continuously increasing, seriously threatening the health of people globally. It thus has become an urgent problem that needs to be addressed. There is research evidence that intestinal mucosal barrier dysfunction, including changes in intestinal stem cell secretion lineage, mucosal layer damage, disruption of cell junctions, overactive immune function, and imbalanced gut microbiota, is an important pathogenic factor and molecular basis of UC. The Notch signaling pathway is a highly conserved signaling pathway in eukaryotes during evolution, which transmits signals through cell connections between adjacent cells, affecting a series of processes such as cell proliferation, differentiation, development, migration, and apoptosis. Therefore, the Notch signaling pathway can regulate intestinal stem cells, CD4~+T cells, innate lymphoid cells(ILCs), macrophages(MØ), and intestinal microbiota and thus affect the chemical, physical, immune, and biological mucosal barriers of the intestinal mucosa. Its function is extensive and unique, different from those signaling pathways that mainly focus on anti-inflammatory and antioxidant stress. It can explain the therapeutic effects of traditional Chinese medicine from different perspectives. This article reviewed the role of the Notch1 signaling pathway in the pathogenesis of UC and the relevant literature on the targeted prevention and treatment of UC with traditional Chinese medicine, so as to provide new targets and theoretical support for further research on the effective prevention and treatment of UC.


Subject(s)
Colitis, Ulcerative , Receptor, Notch1 , Signal Transduction , Humans , Signal Transduction/drug effects , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Animals , Drugs, Chinese Herbal/pharmacology , Medicine, Chinese Traditional
5.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791169

ABSTRACT

The Notch pathway is a key cancer driver and is important in tumor progression. Early research suggested that Notch activity was highly dependent on the expression of the intracellular cleaved domain of Notch-1 (NICD). However, recent insights into Notch signaling reveal the presence of Notch pathway signatures, which may vary depending on different cancer types and tumor microenvironments. Herein, we perform a comprehensive investigation of the Notch signaling pathway in adult T-cell leukemia (ATL) primary patient samples. Using gene arrays, we demonstrate that the Notch pathway is constitutively activated in ATL patient samples. Furthermore, the activation of Notch in ATL cells remains elevated irrespective of the presence of activating mutations in Notch itself or its repressor, FBXW7, and that ATL cells are dependent upon Notch-1 expression for proliferation and survival. We demonstrate that ATL cells exhibit the expression of pivotal Notch-related genes, including notch-1, hes1, c-myc, H19, and hes4, thereby defining a critical Notch signature associated with ATL disease. Finally, we demonstrate that lncRNA H19 is highly expressed in ATL patient samples and ATL cells and contributes to Notch signaling activation. Collectively, our results shed further light on the Notch pathway in ATL leukemia and reveal new therapeutic approaches to inhibit Notch activation in ATL cells.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell , MicroRNAs , RNA, Long Noncoding , Signal Transduction , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Gene Expression Regulation, Leukemic , Receptors, Notch/metabolism , Receptors, Notch/genetics , Cell Proliferation/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , F-Box-WD Repeat-Containing Protein 7/genetics , Gene Expression Regulation, Neoplastic , Adult
6.
Int J Mol Sci ; 25(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38791441

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive cardiopulmonary disease characterized by pathologic vascular remodeling of small pulmonary arteries. Endothelial dysfunction in advanced PAH is associated with proliferation, apoptosis resistance, and endothelial to mesenchymal transition (EndoMT) due to aberrant signaling. DLL4, a cell membrane associated NOTCH ligand, plays a pivotal role maintaining vascular integrity. Inhibition of DLL4 has been associated with the development of pulmonary hypertension, but the mechanism is incompletely understood. Here we report that BMPR2 silencing in pulmonary artery endothelial cells (PAECs) activated AKT and suppressed the expression of DLL4. Consistent with these in vitro findings, increased AKT activation and reduced DLL4 expression was found in the small pulmonary arteries of patients with PAH. Increased NOTCH1 activation through exogenous DLL4 blocked AKT activation, decreased proliferation and reversed EndoMT. Exogenous and overexpression of DLL4 induced BMPR2 and PPRE promoter activity, and BMPR2 and PPARG mRNA in idiopathic PAH (IPAH) ECs. PPARγ, a nuclear receptor associated with EC homeostasis, suppressed by BMPR2 loss was induced and activated by DLL4/NOTCH1 signaling in both BMPR2-silenced and IPAH ECs, reversing aberrant phenotypic changes, in part through AKT inhibition. Directly blocking AKT or restoring DLL4/NOTCH1/PPARγ signaling may be beneficial in preventing or reversing the pathologic vascular remodeling of PAH.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II , Endothelial Cells , PPAR gamma , Proto-Oncogene Proteins c-akt , Pulmonary Artery , Receptor, Notch1 , Signal Transduction , Humans , Proto-Oncogene Proteins c-akt/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Endothelial Cells/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/pathology , Male , Cell Proliferation , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Female , Cells, Cultured
7.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731894

ABSTRACT

Triptolide is a natural compound in herbal remedies with anti-inflammatory and anti-proliferative properties. We studied its effects on critical signaling processes within the cell, including Notch1 and STAT3 signaling. Our research showed that triptolide reduces cancer cell proliferation by decreasing the expression of downstream targets of these signals. The levels of each signal-related protein and mRNA were analyzed using Western blot and qPCR methods. Interestingly, inhibiting one signal with a single inhibitor alone did not significantly reduce cancer cell proliferation. Instead, MTT assays showed that the simultaneous inhibition of Notch1 and STAT3 signaling reduced cell proliferation. The effect of triptolide was similar to a combination treatment with inhibitors for both signals. When we conducted a study on the impact of triptolide on zebrafish larvae, we found that it inhibited muscle development and interfered with muscle cell proliferation, as evidenced by differences in the staining of myosin heavy chain and F-actin proteins in confocal fluorescence microscopy. Additionally, we noticed that inhibiting a single type of signaling did not lead to any significant muscle defects. This implies that triptolide obstructs multiple signals simultaneously, including Notch1 and STAT3, during muscle development. Chemotherapy is commonly used to treat cancer, but it may cause muscle loss due to drug-related adverse reactions or other complex mechanisms. Our study suggests that anticancer agents like triptolide, inhibiting essential signaling pathways including Notch1 and STAT3 signaling, may cause muscle atrophy through anti-proliferative activity.


Subject(s)
Cell Proliferation , Diterpenes , Epoxy Compounds , Phenanthrenes , Receptor, Notch1 , STAT3 Transcription Factor , Animals , Humans , Cell Line, Tumor , Cell Proliferation/drug effects , Diterpenes/pharmacology , Epoxy Compounds/pharmacology , Phenanthrenes/pharmacology , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Receptors, Notch/metabolism , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism , Zebrafish , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
8.
J Coll Physicians Surg Pak ; 34(5): 527-532, 2024 May.
Article in English | MEDLINE | ID: mdl-38720211

ABSTRACT

OBJECTIVE: To develop an intervention based on Notch-1 signalling pathway blockade by investigating the potential application of the neurogenic locus notch homologue protein 1(Notch-1) signalling pathway as a key regulator of chronic inflammation and adipogenesis in the treatment of hepatic insulin resistance (HIR). STUDY DESIGN: Experimental study. Place and Duration of the Study: Animal Laboratory of the Fourth Hospital of Hebei Medical University, Shijiazhuang, China, from April 2021 to June 2022. METHODOLOGY: HIR models were established in Notch-1WT and Notch-1MAC-KO mice by high fat diet (HFD) for 16 weeks. Haematoxylin and eosin (HE) staining and oil red O (ORO) staining were used to detect inflammatory infiltration and lipid accumulation in each group. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of TNF-α and IL-6. Free fatty acid (FFA) and total cholesterol (TC) were measured with relevant kits. Moreover, real-time quantitative polymerase chain reaction (PCR) was performed to detect the relative expressions of F4/80, Mcp1, and CD11b in hepatic tissues. Mass spectrometry was used to analyse the levels of triglyceride (TG), diacylglycerol (DAG) and conformite europeenne (CE) in liver tissue. Western blotting was used to detect the expression of related proteins. RESULTS: Specific knockdown of Notch-1 in macrophages decreases the relative fluorescence intensity of CD68 and attenuates inflammatory infiltration and lipid degeneration. There was no difference in plasma levels of FFA and TG. Specific knockdown of Notch-1 in macrophages decreases the expression of F4/80, Mcp1, and CD11b, as well as the levels of TG, DAG, CE, IL-6, and TNF-α. CONCLUSION: Specific knockout of Notch-1 in macrophages may reduce HIR by inhibiting the IRE1α-XBP1 signalling pathway. KEY WORDS: Hepatic insulin resistance, Macrophages, Notch-1, IRE1α, XBP1.


Subject(s)
Diet, High-Fat , Insulin Resistance , Macrophages , Mice, Knockout , Protein Serine-Threonine Kinases , Receptor, Notch1 , Signal Transduction , Animals , Mice , Disease Models, Animal , Endoribonucleases/metabolism , Endoribonucleases/genetics , Insulin Resistance/physiology , Liver/metabolism , Macrophages/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics
9.
Toxicol Appl Pharmacol ; 487: 116969, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744347

ABSTRACT

Cysteine and glycine-rich protein 2 (CSRP2) is expressed differently in numerous cancers and plays a key role in carcinogenesis. However, the role of CSRP2 in glioma is unknown. This study sought to determine the expression profile and clinical significance of CSRP2 in glioma and explore its biological functions and mechanisms via lentivirus-mediated CSRP2 silencing experiments. Increased CSRP2 was frequently observed in gliomas, which was associated with clinicopathological characteristics and an unfavourable prognosis. Decreasing CSRP2 led to the suppression of malignant proliferation, metastasis and stemness in glioma cells while causing hypersensitivity to chemotherapeutic drugs. Mechanistic investigations revealed that CSRP2 plays a role in mediating the Notch signalling cascade. Silencing CSRP2 decreased the levels of Notch1, cleaved Notch1, HES1 and HEY1, suppressing the Notch signalling cascade. Reactivation of Notch markedly diminished the tumour-inhibiting effects of CSRP2 silencing on the malignant phenotypes of glioma cells. Notably, CSRP2-silencing glioma cells exhibited reduced potential in the formation of xenografts in nude mice in vivo, which was associated with an impaired Notch signalling cascade. These results showed that CSRP2 is overexpressed in glioma and has a crucial role in sustaining the malignant phenotypes of glioma, suggesting that targeting CSRP2 could be a promising strategy for glioma treatment.


Subject(s)
Glioma , Mice, Nude , Signal Transduction , Humans , Glioma/pathology , Glioma/metabolism , Glioma/genetics , Animals , Cell Line, Tumor , Mice , Male , Cell Proliferation , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Female , Phenotype , Receptors, Notch/metabolism , Receptors, Notch/genetics , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Mice, Inbred BALB C , Middle Aged , Gene Expression Regulation, Neoplastic , Xenograft Model Antitumor Assays
10.
Arch Microbiol ; 206(6): 249, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38713385

ABSTRACT

Escherichia coli (E. coli) can induce severe clinical bovine mastitis, which is to blame for large losses experienced by dairy farms. Macrophage polarization into various states is in response to pathogen infections. Lycopene, a naturally occurring hydrocarbon carotenoid, relieved inflammation by controlling M1/M2 status of macrophages. Thus, we wanted to explore the effect of lycopene on polarization states of macrophages in E. coli-induced mastitis. Macrophages were cultivated with lycopene for 24, before E. coli inoculation for 6 h. Lycopene (0.5 µmol/L) significantly enhanced cell viabilities and significantly reduced lactic dehydrogenase (LDH) levels in macrophages, whereas 2 and 3 µmol/L lycopene significantly enhanced LDH activities. Lycopene treatment significantly reduced the increase in LDH release, iNOS, CD86, TNF-α, IL-1ß and phosphatase and tensin homolog (PTEN) expressions in E. coli group. 0.5 µmol/L lycopene significantly increased E. coli-induced downregulation of CD206, arginase I (ARG1), indoleamine 2,3-dioxygenase (IDO), chitinase 3-like 3 (YM1), PI3K, AKT, p-AKT, mammalian target of rapamycin (mTOR), p-mTOR, jumonji domain-containing protein-3 (JMJD3) and interferon regulatory factor 4 (IRF4) levels. Moreover, Ginkgolic acid C17:1 (a specific PTEN inhibitor), 740YPDGFR (a specific PI3K activator), SC79 (a specific AKT activator) or CHPG sodium salt (a specific NF-κB activator) significantly decreased CD206, AGR1, IDO and YM1 expressions in lycopene and E. coli-treated macrophages. Therefore, lycopene increased M2 macrophages via inhibiting NOTCH1-PI3K-mTOR-NF-κB-JMJD3-IRF4 pathway in response to E. coli infection in macrophages. These results contribute to revealing the pathogenesis of E. coli-caused bovine mastitis, providing the new angle of the prevention and management of mastitis.


Subject(s)
Escherichia coli Infections , Escherichia coli , Lycopene , Macrophages , Signal Transduction , Animals , Cattle , Female , Mice , Cell Line , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Lycopene/pharmacology , Macrophages/drug effects , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Mastitis, Bovine/microbiology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
11.
FASEB J ; 38(9): e23640, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38690715

ABSTRACT

Osteoarthritis (OA) is the main cause of cartilage damage and disability. This study explored the biological function of S-phase kinase-associated protein 2 (SKP2) and Kruppel-like factor 11 (KLF11) in OA progression and its underlying mechanisms. C28/I2 chondrocytes were stimulated with IL-1ß to mimic OA in vitro. We found that SKP2, Jumonji domain-containing protein D3 (JMJD3), and Notch receptor 1 (NOTCH1) were upregulated, while KLF11 was downregulated in IL-1ß-stimulated chondrocytes. SKP2/JMJD3 silencing or KLF11 overexpression repressed apoptosis and extracellular matrix (ECM) degradation in chondrocytes. Mechanistically, SKP2 triggered the ubiquitination and degradation of KLF11 to transcriptionally activate JMJD3, which resulted in activation of NOTCH1 through inhibiting H3K27me3. What's more, the in vivo study found that KLF11 overexpression delayed OA development in rats via restraining apoptosis and maintaining the balance of ECM metabolism. Taken together, ubiquitination and degradation of KLF11 regulated by SKP2 contributed to OA progression by activation of JMJD3/NOTCH1 pathway. Our findings provide promising therapeutic targets for OA.


Subject(s)
Chondrocytes , Jumonji Domain-Containing Histone Demethylases , Osteoarthritis , Receptor, Notch1 , S-Phase Kinase-Associated Proteins , Ubiquitination , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Animals , S-Phase Kinase-Associated Proteins/metabolism , S-Phase Kinase-Associated Proteins/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/genetics , Rats , Chondrocytes/metabolism , Chondrocytes/pathology , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Signal Transduction , Rats, Sprague-Dawley , Humans , Apoptosis , Repressor Proteins/metabolism , Repressor Proteins/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics
12.
Mol Biol Rep ; 51(1): 507, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622406

ABSTRACT

BACKGROUND: Our previous research has demonstrated that hypoxic preconditioning (HPC) can improve spatial learning and memory abilities in adult mice. Adult hippocampal neurogenesis has been associated with learning and memory. The Neurogenic locus notch homolog protein (Notch) was involved in adult hippocampal neurogenesis, as well as in learning and memory. It is currently unclear whether the Notch pathway regulates hippocampal neuroregeneration by modifying the DNA methylation status of the Notch gene following HPC. METHOD: The HPC animal model and cell model were established through repeated hypoxia exposure using mice and the mouse hippocampal neuronal cell line HT22. Step-down test was conducted on HPC mice. Real-time PCR and Western blot analysis were used to assess the mRNA and protein expression levels of Notch1 and hairy and enhancer of split1 (HES1). The presence of BrdU-positive cells and Notch1 expression in the hippocampal dental gyrus (DG) were examined with confocal microscopy. The methylation status of the Notch1 was analyzed using methylation-specific PCR (MS-PCR). HT22 cells were employed to elucidate the impact of HPC on Notch1 in vitro. RESULTS: HPC significantly improved the step-down test performance of mice with elevated levels of mRNA and protein expression of Notch1 and HES1 (P < 0.05). The intensities of the Notch1 signal in the control group, the H group and the HPC group were 2.62 ± 0.57 × 107, 2.87 ± 0.84 × 107, and 3.32 ± 0.14 × 107, respectively, and the number of BrdU (+) cells in the hippocampal DG were 1.83 ± 0.54, 3.71 ± 0.64, and 7.29 ± 0.68 respectively. Compared with that in C and H group, the intensity of the Notch1 signal and the number of BrdU (+) cells increased significantly in HPC group (P < 0.05). The methylation levels of the Notch1 promoter 0.82 ± 0.03, 0.65 ± 0.03, and 0.60 ± 0.02 in the C, H, and HPC groups, respectively. The methylation levels of Notch1 decreased significantly (P < 0.05). The effect of HPC on HT22 cells exhibited similarities to that observed in the hippocampus. CONCLUSION: HPC may confer neuroprotection by activating the Notch1 signaling pathway and regulating its methylation level, resulting in the regeneration of hippocampal neurons.


Subject(s)
DNA Methylation , Hippocampus , Mice , Animals , DNA Methylation/genetics , Bromodeoxyuridine/metabolism , Hippocampus/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Receptors, Notch/metabolism , RNA, Messenger/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism
13.
PeerJ ; 12: e17222, 2024.
Article in English | MEDLINE | ID: mdl-38650654

ABSTRACT

Targeting tumor angiogenesis is an important approach in advanced tumor therapy. Here we investigated the effect of the suppressor of variegation 3-9 homolog 1 (SUV39H1) on tumor angiogenesis in oral squamous cell carcinoma (OSCC). The GEPIA database was used to analyze the expression of SUV39H1 in various cancer tissues. The expression of SUV39H1 in OSCC was detected by immunohistochemistry, and the correlation between SUV39H1 and Notch1 and microvascular density (MVD) was analyzed. The effect of SUV39H1 inhibition on OSCC was investigated in vivo by chaetocin treatment. The migration and tube formation of vascular endothelial cells by conditioned culture-medium of different treatments of oral squamous cell cells were measured. The transcriptional level of SUV39H1 is elevated in various cancer tissues. The transcription level of SUV39H1 in head and neck squamous cell carcinoma was significantly higher than that in control. Immunohistochemistry result showed increased SUV39H1 expression in OSCC, which was significantly correlated with T staging. The expression of SUV39H1 was significantly correlated with Notch1 and CD31. In vivo experiment chaetocin treatment significantly inhibit the growth of tumor, and reduce SUV39H1, Notch1, CD31 expression. The decreased expression of SUV39H1 in OSCC cells lead to the decreased expression of Notch1 and VEGF proteins, as well as the decreased migration and tube formation ability of vascular endothelial cells. Inhibition of Notch1 further enhance this effect. Our results suggest inhibition of SUV39H1 may affect angiogenesis by regulating Notch1 expression. This study provides a foundation for SUV39H1 as a potential therapeutic target for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Methyltransferases , Mouth Neoplasms , Neovascularization, Pathologic , Receptor, Notch1 , Repressor Proteins , Humans , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/drug therapy , Mouth Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Animals , Repressor Proteins/metabolism , Repressor Proteins/genetics , Methyltransferases/metabolism , Methyltransferases/antagonists & inhibitors , Methyltransferases/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/blood supply , Cell Line, Tumor , Mice , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Angiogenesis
14.
J Stroke Cerebrovasc Dis ; 33(6): 106578, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636320

ABSTRACT

BACKGROUND: Notch1 signaling inhibiton with N-[N-(3,5-difluorophenacetyl)-1-alanyl]-S-phenylglycine t-butylester] (DAPT) treatment could promote brain recovery and the intervention effect is different between striatum (STR) and cortex (CTX), which might be accounted for different changes of glial activities, but the in-depth mechanism is still unknown. The purpose of this study was to identify whether DAPT could modulate microglial subtype shifts and astroglial-endfeet aquaporin-4 (AQP4) mediated waste solute drainage. METHODS: Sprague-Dawley rats (n=10) were subjected to 90min of middle cerebral artery occlusion (MCAO) and were treated with DAPT (n=5) or act as control with no treatment (n=5). Two groups of rats underwent MRI scans at 24h and 4 week, and sacrificed at 4 week after stroke for immunofluorescence (IF). RESULTS: Compared with control rats, MRI data showed structural recovery in ipsilateral STR but not CTX. And IF showed decreased pro-inflammatory M1 microglia and increased anti-inflammatory M2 microglia in striatal lesion core and peri-lesions of STR, CTX. Meanwhile, IF showed decreased AQP4 polarity in ischemic brain tissue, however, AQP4 polarity in striatal peri-lesions of DAPT treated rats was higher than that in control rats but shows no difference in cortical peri-lesions between control and treated rats. CONCLUSIONS: The present study indicated that DAPT could promote protective microglia subtype shift and striatal astrocyte mediated waste solute drainage, that the later might be the major contributor of waste solute metabolism and one of the accounts for discrepant recovery of STR and CTX.


Subject(s)
Aquaporin 4 , Astrocytes , Dipeptides , Disease Models, Animal , Infarction, Middle Cerebral Artery , Microglia , Rats, Sprague-Dawley , Receptor, Notch1 , Recovery of Function , Signal Transduction , Animals , Aquaporin 4/metabolism , Receptor, Notch1/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Male , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Dipeptides/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Corpus Striatum/pathology , Time Factors , Neuroprotective Agents/pharmacology , Ischemic Stroke/metabolism , Ischemic Stroke/drug therapy , Ischemic Stroke/physiopathology , Ischemic Stroke/pathology
15.
Clin Immunol ; 263: 110203, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38575046

ABSTRACT

Langerhans cell histiocytosis (LCH) is characterized by an expansion and accumulation of pathological histiocytes expressing langerin (CD207) and CD1a in different organs under an inflammatory milieu. The origin of pathognomonic precursors of LCH is widely debated, but monocytes and pre-dendritic cells (pre-DC) play a significant role. Remarkably, we found an expansion of AXLhigh cells in the CD11c+ subset of patients with active LCH, which also express the pathognomonic CD207 and CD1a. Moreover, we obtained a monocyte-derived LC-like (mo-LC-like) expressing high levels of AXL when treated with inflammatory cytokine, or plasma of patients with active disease. Intriguingly, inhibiting the mTOR pathway at the initial stages of monocyte differentiation to LC-like fosters the pathognomonic LCH program, highly increasing CD207 levels, together with NOTCH1 induction. We define here that AXLhigh could also be taken as a strong pathognomonic marker for LCH, and the release of Langerin and NOTCH1 expression depends on the inhibition of the mTOR pathway.


Subject(s)
Antigens, CD , Axl Receptor Tyrosine Kinase , Histiocytosis, Langerhans-Cell , Lectins, C-Type , Mannose-Binding Lectins , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , TOR Serine-Threonine Kinases , Humans , Histiocytosis, Langerhans-Cell/metabolism , TOR Serine-Threonine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Antigens, CD/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Mannose-Binding Lectins/metabolism , Lectins, C-Type/metabolism , Male , Myeloid Cells/metabolism , Biomarkers , Female , Adolescent , Receptor, Notch1/metabolism , Antigens, CD1/metabolism , Child , Monocytes/metabolism , Monocytes/immunology , Adult , Child, Preschool , Signal Transduction , Cell Differentiation
16.
Immunity ; 57(5): 1124-1140.e9, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38636522

ABSTRACT

Signaling through Notch receptors intrinsically regulates tumor cell development and growth. Here, we studied the role of the Notch ligand Jagged2 on immune evasion in non-small cell lung cancer (NSCLC). Higher expression of JAG2 in NSCLC negatively correlated with survival. In NSCLC pre-clinical models, deletion of Jag2, but not Jag1, in cancer cells attenuated tumor growth and activated protective anti-tumor T cell responses. Jag2-/- lung tumors exhibited higher frequencies of macrophages that expressed immunostimulatory mediators and triggered T cell-dependent anti-tumor immunity. Mechanistically, Jag2 ablation promoted Nr4a-mediated induction of Notch ligands DLL1/4 on cancer cells. DLL1/4-initiated Notch1/2 signaling in macrophages induced the expression of transcription factor IRF4 and macrophage immunostimulatory functionality. IRF4 expression was required for the anti-tumor effects of Jag2 deletion in lung tumors. Antibody targeting of Jagged2 inhibited tumor growth and activated IRF4-driven macrophage-mediated anti-tumor immunity. Thus, Jagged2 orchestrates immunosuppressive systems in NSCLC that can be overcome to incite macrophage-mediated anti-tumor immunity.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Interferon Regulatory Factors , Jagged-2 Protein , Lung Neoplasms , Mice, Knockout , Tumor-Associated Macrophages , Jagged-2 Protein/metabolism , Jagged-2 Protein/genetics , Jagged-2 Protein/immunology , Animals , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Mice , Humans , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Signal Transduction , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Mice, Inbred C57BL , Receptors, Notch/metabolism , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Macrophages/immunology , Macrophages/metabolism , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Tumor Escape/immunology
17.
Am J Physiol Endocrinol Metab ; 326(6): E832-E841, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38656129

ABSTRACT

Thyroid dysgenesis (TD) is the common pathogenic mechanism of congenital hypothyroidism (CH). In addition, known pathogenic genes are limited to those that are directly involved in thyroid development. To identify additional candidate pathogenetic genes, we performed forward genetic screening for TD in zebrafish, followed by positional cloning. The candidate gene was confirmed in vitro using the Nthy-ori 3.1 cell line and in vivo using a zebrafish model organism. We obtained a novel zebrafish line with thyroid dysgenesis and identified the candidate pathogenetic mutation TATA-box binding protein associated Factor 1 (taf1) by positional cloning. Further molecular studies revealed that taf1 was needed for the proliferation of thyroid follicular cells by binding to the NOTCH1 promoter region. Knockdown of TAF1 impaired the proliferation and maturation of thyroid cells, thereby leading to thyroid dysplasia. This study showed that TAF1 promoted Notch signaling and that this association played a pivotal role in thyroid development.NEW & NOTEWORTHY In our study, we obtained a novel zebrafish line with thyroid dysgenesis (TD) and identified the candidate pathogenetic mutation TATA-box binding protein associated Factor 1 (taf1). Further researches revealed that taf1 was required for thyroid follicular cells by binding to the NOTCH1 promoter region. Our findings revealed a novel role of TAF1 in thyroid morphogenesis.


Subject(s)
Cell Proliferation , Signal Transduction , TATA-Binding Protein Associated Factors , Thyroid Gland , Transcription Factor TFIID , Zebrafish , Animals , Zebrafish/genetics , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Signal Transduction/genetics , Cell Proliferation/genetics , Thyroid Gland/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Thyroid Dysgenesis/genetics , Thyroid Dysgenesis/metabolism , Humans , Histone Acetyltransferases
18.
J Ethnopharmacol ; 330: 118225, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38670408

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Neuroinflammation is involved in the pathogenesis of depression disorder by activating microglia cells, increasing proinflammatory cytokines, effecting serotonin synthesis and metabolism, and neuronal apoptosis and neurogenesis. Arjunolic acid (ARG) is a triterpenoid derived from the fruits of Akebia trifoliata for treating psychiatric disorders in TCM clinic, which exhibits anti-inflammatory and neuroprotective effects. However, its anti-depressive effect and underlying mechanism are unknown. AIM OF THE STUDY: The aim of this study is to explore the effect of arjunolic acid on depression and its possible mechanisms. METHODS: Intraperitoneal injection of LPS in mice and LPS stimulated-BV2 microglia were utilized to set up in vivo and in vitro models. Behavioral tests, H&E staining and ELISA were employed to evaluate the effect of arjunolic acid on depression. RT-qPCR, immunofluorescence, molecular docking and Western blot were performed to elucidate the molecular mechanisms. RESULTS: Arjunolic acid dramatically ameliorated depressive behavior in LPS-induced mice. The levels of BDNF and 5-HT in the hippocampus of the mice were increased, while the number of iNOS + IBA1+ cells in the brain were decreased and Arg1+IBA1+ positive cells were increased after arjunolic acid treatment. In addition, arjunolic acid promoted the polarization of BV2 microglia from M1 to M2 type. Notably, drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA) and molecular docking technologies identified SIRT1 as the target of arjunolic acid. Moreover, after SIRT1 inhibition by using EX-527, the effects of arjunolic acid on ameliorating LPS-induced depressive behavior in mice and promoting M2 Microglia polarization were blocked. In addition, arjunolic acid activated AMPK and decreased Notch1 expression, however, inhibition of AMPK, the effect of arjunolic acid on the downregulation of Notch1 expression were weaken. CONCLUSIONS: This study elucidates that arjunolic acid suppressed neuroinflammation through modulating the SIRT1/AMPK/Notch1 signaling pathway. Our study demonstrates that arjunolic acid might serve as a potiential anti-depressant.


Subject(s)
Depression , Lipopolysaccharides , Microglia , Signal Transduction , Animals , Male , Mice , AMP-Activated Protein Kinases/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Cell Line , Depression/drug therapy , Depression/chemically induced , Depression/metabolism , Lipopolysaccharides/toxicity , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Molecular Docking Simulation , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Sirtuin 1/metabolism , Triterpenes/pharmacology , Triterpenes/therapeutic use
19.
Cancer Cell ; 42(5): 904-914.e9, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38579724

ABSTRACT

A subset of patients with IDH-mutant glioma respond to inhibitors of mutant IDH (IDHi), yet the molecular underpinnings of such responses are not understood. Here, we profiled by single-cell or single-nucleus RNA-sequencing three IDH-mutant oligodendrogliomas from patients who derived clinical benefit from IDHi. Importantly, the tissues were sampled on-drug, four weeks from treatment initiation. We further integrate our findings with analysis of single-cell and bulk transcriptomes from independent cohorts and experimental models. We find that IDHi treatment induces a robust differentiation toward the astrocytic lineage, accompanied by a depletion of stem-like cells and a reduction of cell proliferation. Furthermore, mutations in NOTCH1 are associated with decreased astrocytic differentiation and may limit the response to IDHi. Our study highlights the differentiating potential of IDHi on the cellular hierarchies that drive oligodendrogliomas and suggests a genetic modifier that may improve patient stratification.


Subject(s)
Brain Neoplasms , Cell Differentiation , Isocitrate Dehydrogenase , Mutation , Oligodendroglioma , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Oligodendroglioma/drug therapy , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/antagonists & inhibitors , Humans , Cell Differentiation/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Cell Lineage/drug effects , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Cell Proliferation/drug effects , Animals , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Mice , Single-Cell Analysis/methods
20.
Int J Mol Sci ; 25(6)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38542260

ABSTRACT

Notch signaling is involved in the prevention of cell differentiation and cell fate in various organs, including the lungs. We aimed to determine the transcriptomic and protein expression of Notch receptors, their ligands, and related transcription factors in stable COPD. The expression and localization of Notch receptors, their ligands, and related transcription factors were measured in bronchial biopsies of individuals with stable mild/moderate (MCOPD) (n = 18) or severe/very severe (SCOPD) (n = 16) COPD, control smokers (CSs) (n = 13), and control nonsmokers (CNSs) (n = 11), and in the lung parenchyma of those with MCOPD (n = 13), CSs (n = 10), and CNSs (n = 10) using immunohistochemistry, ELISA tests, and transcriptome analyses. In the bronchial biopsies, Notch4 and HES7 significantly increased in the lamina propria of those with SCOPD compared to those with MCOPD, CSs, and CNSs. In the peripheral lung bronchiolar epithelium, Notch1 significantly increased in those with MCOPD and CSs compared to CNSs. ELISA tests of lung parenchyma homogenates showed significantly increased Notch2 in those with MCOPD compared to CSs and CNSs. Transcriptomic data in lung parenchyma showed increased DLL4 and HES1 mRNA levels in those with MCOPD and CSs compared to CNSs. These data show the increased expression of the Notch pathway in the lungs of those with stable COPD. These alterations may play a role in impairing the regenerative-reparative responses of diseased bronchioles and lung parenchyma.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Cell Differentiation/genetics , Receptor, Notch1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...