Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 323
Filter
1.
Commun Biol ; 7(1): 1290, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384976

ABSTRACT

High salt conditions and subsequent hyperosmolarity are injurious cellular stresses that can activate immune signaling. Nuclear factor of activated T-cells 5 (NFAT5) is an essential transcription factor that induces osmoprotective genes such as aldose reductase (AR) and betaine-GABA transporter 1 (BGT1). High salt stress-mediated NFAT5 activation is also reported to accelerate the inflammatory response and autoimmune diseases. However, the systemic regulation of NFAT5 remains unclear. Here, we performed a genome-wide siRNA screen to comprehensively identify the regulators of NFAT5. We monitored NFAT5 nuclear translocation and identified one of the Notch signaling effectors, Hairy and enhancer of split-1 (HES1), as a positive regulator of NFAT5. HES1 was induced by high salinity via ERK signaling and facilitated NFAT5 recruitment to its target promoter region, resulting in the proper induction of osmoprotective genes and cytoprotection under high salt stress. These findings suggest that, though HES1 is well known as a transcriptional repressor, it positively regulates NFAT5-dependent transcription in the context of a high salinity/hyperosmotic response.


Subject(s)
Transcription Factor HES-1 , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Humans , Salt Stress , Animals , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Mice , DNA/metabolism , DNA/genetics , Gene Expression Regulation/drug effects , HEK293 Cells , Protein Binding , Transcription Factors
2.
Int J Mol Med ; 54(6)2024 Dec.
Article in English | MEDLINE | ID: mdl-39364745

ABSTRACT

The early restoration of hemodynamics/reperfusion in acute myocardial infarction (AMI) is an effective therapeutic strategy to reduce sudden death and improve patient prognosis. However, reperfusion induces additional cardiomyocyte damage and cardiac tissue dysfunction. In this context, turmeric­derived curcumin (Cur) has been shown to exhibit a protective effect against myocardial ischemia/reperfusion injury (I/RI). The molecular mechanism of its activity, however, remains unclear. The current study investigated the protective effect of Cur and its molecular mechanism via in vitro experiments. The Cell Counting Kit­8 and lactate dehydrogenase (LDH) assay kit were used to assess the cell viability and cytotoxicity. The contents of malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase, glutathione (GSH)/glutathione disulfide (GSSG), total iron, ferrous iron, caspase­3 and reactive oxygen species (ROS) were measured using an appropriate kit. Western blotting was used to detect the expression of relevant proteins. The levels of apoptosis, mitochondrial permeability transition pore (MPTP) opening, and mitochondrial membrane potential (MMP) were detected by flow cytometry. The study findings indicated that anoxia/reoxygenation (A/R) injury significantly decreased cell viability, increased in LDH and caspase­3 activities, induced ferroptosis, increased apoptosis and overactivated autophagy. However, pretreatment with Cur or ferrostatin­1 (Fer­1, a ferroptosis inhibitor) significantly increased A/R­reduced cell viability, SOD, glutathione peroxidase activity, GSH/GSSH ratio and HES1 and glutathione peroxidase 4 protein expression; attenuated A/R­induced LDH, MDA, total iron, ferrous iron, prostaglandin­endoperoxide synthase 2 protein expression and prevented ROS overproduction and MMP loss. In addition, Cur inhibited caspase­3 activity, upregulated the Bcl­2/Bax ratio, reduced apoptotic cell number and inhibited MPTP over­opening. Furthermore, Cur increased P62, LC3II/I, NDUFB8 and UQCRC2 expression and upregulated the p­AMPK/AMPK ratio. However, erastin (a ferroptosis activator), pAD/HES1­short hairpin RNA, rapamycin (an autophagy activator) and Compound C (an AMPK inhibitor) blocked the protective effect of Cur. In conclusion, Cur pretreatment inhibited ferroptosis, autophagy overactivation and oxidative stress; improved mitochondrial dysfunction; maintained energy homeostasis; attenuated apoptosis; and ultimately protected the myocardium from A/R injury via increased HES1 expression.


Subject(s)
Apoptosis , Autophagy , Curcumin , Ferroptosis , Myocardial Reperfusion Injury , Transcription Factor HES-1 , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Ferroptosis/drug effects , Apoptosis/drug effects , Autophagy/drug effects , Curcumin/pharmacology , Transcription Factor HES-1/metabolism , Animals , Cell Survival/drug effects , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Humans , Oxidative Stress/drug effects
3.
Development ; 151(18)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39315665

ABSTRACT

The intricate dynamics of Hes expression across diverse cell types in the developing vertebrate embryonic tail have remained elusive. To address this, we have developed an endogenously tagged Hes1-Achilles mouse line, enabling precise quantification of dynamics at the single-cell resolution across various tissues. Our findings reveal striking disparities in Hes1 dynamics between presomitic mesoderm (PSM) and preneural tube (pre-NT) cells. While pre-NT cells display variable, low-amplitude oscillations, PSM cells exhibit synchronized, high-amplitude oscillations. Upon the induction of differentiation, the oscillation amplitude increases in pre-NT cells. Additionally, our study of Notch inhibition on Hes1 oscillations unveils distinct responses in PSM and pre-NT cells, corresponding to differential Notch ligand expression dynamics. These findings suggest the involvement of separate mechanisms driving Hes1 oscillations. Thus, Hes1 demonstrates dynamic behaviour across adjacent tissues of the embryonic tail, yet the varying oscillation parameters imply differences in the information that can be transmitted by these dynamics.


Subject(s)
Embryo, Mammalian , Gene Expression Regulation, Developmental , Mesoderm , Single-Cell Analysis , Transcription Factor HES-1 , Animals , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Mice , Mesoderm/metabolism , Mesoderm/cytology , Mesoderm/embryology , Embryo, Mammalian/metabolism , Receptors, Notch/metabolism , Cell Differentiation , Body Patterning , Somites/metabolism , Somites/embryology , Embryonic Development/genetics , Tail/embryology
4.
Eur J Med Res ; 29(1): 472, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342366

ABSTRACT

This study investigated the possible mechanisms of microRNA-124a on the differentiation of bone marrow mesenchymal stem cells (BMSCs) and its underlying mechanism. ß-Thiol ethanol induced Notch1 mRNA expression, microRNA-124a inhibitor reduced the effects of ß-thiol ethanol on Notch1 mRNA expression in BMSCs. Baicalin induced Hes1 mRNA expression, and microRNA-124a inhibitor reduced the effects of baicalin on Hes1 mRNA expression in BMSCs. Si-Notch1 suppressed Hes1 mRNA expression in BMSCs. Baicalin increased the effects of Notch1 on Hes1 mRNA expression in BMSCs. Si-Notch1 increased cell growth of BMSCs. Baicalin reduced the effects of si-Notch1 on cell growth and the differentiation of BMSCs. We demonstrated that microRNA-124a promoted the differentiation of BMSCs into neurons through Notch/Hes1 signal pathway.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , MicroRNAs , Neurons , Receptor, Notch1 , Signal Transduction , Transcription Factor HES-1 , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction/drug effects , Cell Differentiation/drug effects , Animals , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Neurons/metabolism , Neurons/cytology , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Flavonoids/pharmacology , Cell Proliferation/drug effects , Rats , Cells, Cultured , Receptors, Notch/metabolism , Receptors, Notch/genetics , Rats, Sprague-Dawley
5.
Development ; 151(18)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39324411

ABSTRACT

Hes proteins are transcription factors that are dynamically expressed during embryonic development, but it remains unclear how the oscillations in Hes expression differ across cell types during development. In this new study, Ina Sonnen and colleagues find that the Notch-driven Hes1 oscillation dynamics in the mouse embryonic tail are cell-type specific. To find out more about the story behind the paper, we caught up with first author Yasmine el Azhar and corresponding author Ina Sonnen, Group Leader at the Hubrecht Institute, The Netherlands.


Subject(s)
Developmental Biology , Animals , Mice , Embryonic Development/genetics , History, 21st Century , Humans , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Gene Expression Regulation, Developmental , History, 20th Century
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1620-1630, 2024 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-39276059

ABSTRACT

OBJECTIVE: To explore the inhibitory effect ORY-1001, a lysine-specific histone demethylase 1 (LSD1) inhibitor, on growth of glioblastoma (GBM) and the underlying mechanism. METHODS: We analyzed LSD1 expressions in GBM and normal brain tissues based on data from TCGA and HPA databases. Female BALB/c mouse models bearing xenografts derived from U87 cells or cells with lentivirus-mediated LSD1 silencing or Notch overexpression were treated with saline or 400 µg/kg ORY-1001 by gavage every 7 days, and GBM formation and survival time of the mice were recorded. The effect of ORY-1001 on GBM cell viability was assessed, and its effect on LSD1 expression was analyzed with Western blotting. The genes and pathways associated with LSD1 were analyzed using bioinformatics methods. Western blotting and qRT-PCR were used to detect Notch/HES1 pathway expression after LSD1 silencing and ORY-1001 treatment. The impact of ORY-1001 on viability of U87 cells with Notch1 silencing or overexpression was assessed, and the regulatory effects of ORY-1001 on Notch/HES1 pathway were analyzed using chromatin immunoprecipitation assay. RESULTS: A high expression of LSD1 in GBM was negatively correlated with patient survival (P < 0.001). ORY-1001 and LSD1 silencing obviously reduced tumor burden and prolonged the survival time of GBM-bearing mice. ORY-1001 treatment significantly inhibited the viability and dose-dependently decreased LSD1 expression in GBM cells, and such inhibitory effect of ORY-1001 was attenuated by LSD1 silencing. The Notch pathway enriched the differential genes related to LSD1, and Notch/HES1 pathway expression was significantly down-regulated after LSD1 silencing and ORY-1001 treatment. Notch1 overexpression significantly attenuated the anti-tumor effect of ORY-1001 on GBM. Mechanistically, ORY-1001 disrupted the interaction between LSD1 and the Notch pathway target genes including Notch3, HES1 and CR2. CONCLUSION: ORY-1001 down-regulates the Notch/HES1 pathway by inhibiting LSD1 expression to suppress the growth of GBM in mice.


Subject(s)
Cell Proliferation , Glioblastoma , Histone Demethylases , Mice, Inbred BALB C , Transcription Factor HES-1 , Histone Demethylases/metabolism , Histone Demethylases/genetics , Animals , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Mice , Cell Line, Tumor , Female , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Humans , Cell Proliferation/drug effects , Signal Transduction , Receptors, Notch/metabolism , Down-Regulation , Brain Neoplasms/metabolism , Brain Neoplasms/pathology
7.
Immunopharmacol Immunotoxicol ; 46(5): 703-714, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39134472

ABSTRACT

BACKGROUND: Gremlin1 is a multifunctional protein whose expression is demonstrated to be involved in a series of physiology and pathological processes. The association between Gremlin1 and apcial periodontitis (AP) has been established. M1-polarized macrophages are crucial immune cells that exacerbate the progression of apical periodontal inflammatory response, but the function of Gremlin1 during macrophages activation in periapical lesions is still unclear. This study attempts to explore the regulatory effects of Gremlin1 on macrophage polarization on apical periodontitis microenviroment. METHODS: Clinical specimens were used to determine the expression of Gremlin1 in periapical tissues by immunohistochemical (IHC) staining. Then, the disease models of periapical inflammation in rats were established, and adenovirus- associated virus (AAVs) was used to blockade Gremlin1 expression. Lentivirus carrying sh-Gremlin1 particles were used to transfect THP-1 induced M1-subtype macrophages. To assess the expression of associated molecules, Western blot, immunofluorescence staining were performed. RESULTS: Gremlin1 was significantly up-regulated in the periapical tissues of subjects with AP as identified by IHC staining, and positively correlated with levels of M1 macrophage-associated genes. Rats AP model with inhibition of Gremlin1 in periapical lesions exhibited limited infiltration of macrophages and decreased expression of M1 macrophage-related genes in periapical lesions. Furthermore, Gremlin1 blockade substantially decreased the Notch1/Hes1 signaling pathway activation level. The in vitro experiments confirmed the above results. CONCLUSION: Taken together, current study illustrated that the Gremlin1 suppression in periapical lesions inhibited M1 macrophage polarization through Notch1/Hes1 axis. Moreover, Gremlin1 may act as a potential candidate in the treatment of AP.


Subject(s)
Intercellular Signaling Peptides and Proteins , Macrophages , Periapical Periodontitis , Receptor, Notch1 , Signal Transduction , Transcription Factor HES-1 , Animals , Periapical Periodontitis/pathology , Periapical Periodontitis/metabolism , Periapical Periodontitis/immunology , Receptor, Notch1/metabolism , Humans , Macrophages/metabolism , Macrophages/immunology , Rats , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Female , Rats, Sprague-Dawley , THP-1 Cells , Macrophage Activation/drug effects , Disease Models, Animal
8.
Int J Mol Sci ; 25(16)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39201457

ABSTRACT

Uremic toxins cause bone disorders in patients with chronic kidney disease (CKD). These disorders are characterized by low turnover osteodystrophy and impaired bone formation in the early stages of CKD. Evidence indicates that the aryl hydrocarbon receptor (AhR) mediates signals that suppress early osteogenic differentiation in bone marrow mesenchymal stem cells (BMSCs). However, whether the AhR mediates the effects of indoxyl sulfate (IS), a uremic toxin, on BMSC osteogenesis remains unclear. We investigated whether IS affects osteogenesis through the AhR/Hes1 pathway. Expression levels of osteogenesis genes (Runx2, Bmp2, Alp, and Oc), AhR, and Hes1 were measured in mouse BMSCs (D1 cells). At concentrations of 2-50 µM, IS significantly reduced mineralization, particularly in the early stages of BMSC osteogenesis. Furthermore, IS significantly downregulated the expression of Runx2, Bmp2, Oc, and Alp. Notably, this downregulation could be prevented using an AhR antagonist and through Ahr knockdown. Mechanistically, IS induced the expression of Hes1 through AhR signaling, thereby suppressing the transcription of Runx2 and Bmp2. Our findings suggest that IS inhibits early osteogenesis of BMSCs through the AhR/Hes1 pathway, thus suppressing the transcription of Runx2 and Bmp2. Our findings may guide new therapeutic strategies against CKD-related bone disorders.


Subject(s)
Indican , Mesenchymal Stem Cells , Osteogenesis , Receptors, Aryl Hydrocarbon , Signal Transduction , Transcription Factor HES-1 , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Animals , Osteogenesis/drug effects , Mice , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Signal Transduction/drug effects , Cell Differentiation/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Basic Helix-Loop-Helix Transcription Factors
9.
J Comp Neurol ; 532(7): e25648, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38958676

ABSTRACT

In this study, we investigated recurrent copy number variations (CNVs) in the 19p12 locus, which are associated with neurodevelopmental disorders. The two genes in this locus, ZNF675 and ZNF681, arose via gene duplication in primates, and their presence in several pathological CNVs in the human population suggests that either or both of these genes are required for normal human brain development. ZNF675 and ZNF681 are members of the Krüppel-associated box zinc finger (KZNF) protein family, a class of transcriptional repressors important for epigenetic silencing of specific genomic regions. About 170 primate-specific KZNFs are present in the human genome. Although KZNFs are primarily associated with repressing retrotransposon-derived DNA, evidence is emerging that they can be co-opted for other gene regulatory processes. We show that genetic deletion of ZNF675 causes developmental defects in cortical organoids, and our data suggest that part of the observed neurodevelopmental phenotype is mediated by a gene regulatory role of ZNF675 on the promoter of the neurodevelopmental gene Hes family BHLH transcription factor 1 (HES1). We also find evidence for the recently evolved regulation of genes involved in neurological disorders, microcephalin 1 and sestrin 3. We show that ZNF675 interferes with HES1 auto-inhibition, a process essential for the maintenance of neural progenitors. As a striking example of how some KZNFs have integrated into preexisting gene expression networks, these findings suggest the emergence of ZNF675 has caused a change in the balance of HES1 autoregulation. The association of ZNF675 CNV with human developmental disorders and ZNF675-mediated regulation of neurodevelopmental genes suggests that it evolved into an important factor for human brain development.


Subject(s)
Primates , Transcription Factor HES-1 , Humans , Animals , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Primates/genetics , Homeostasis/physiology , Homeostasis/genetics , DNA Copy Number Variations/genetics , Mice , Biological Evolution , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
10.
Stem Cell Res Ther ; 15(1): 235, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075526

ABSTRACT

BACKGROUND: Hematopoiesis in mammal is a complex and highly regulated process in which hematopoietic stem cells (HSCs) give rise to all types of differentiated blood cells. Previous studies have shown that hairy and enhancer of split (HES) repressors are essential regulators of adult HSC development downstream of Notch signaling. METHODS: In this study, we investigated the role of HES1, a member of HES family, in fetal hematopoiesis using an embryonic hematopoietic specific Hes1 conditional knockout mouse model by using phenotypic flow cytometry, histopathology analysis, and functional in vitro colony forming unit (CFU) assay and in vivo bone marrow transplant (BMT) assay. RESULTS: We found that loss of Hes1 in early embryonic stage leads to smaller embryos and fetal livers, decreases hematopoietic stem progenitor cell (HSPC) pool, results in defective multi-lineage differentiation. Functionally, fetal hematopoietic cells deficient for Hes1 exhibit reduced in vitro progenitor activity and compromised in vivo repopulation capacity in the transplanted recipients. Further analysis shows that fetal hematopoiesis defects in Hes1fl/flFlt3Cre embryos are resulted from decreased proliferation and elevated apoptosis, associated with de-repressed HES1 targets, p27 and PTEN in Hes1-KO fetal HSPCs. Finally, pharmacological inhibition of p27 or PTEN improves fetal HSPCs function both in vitro and in vivo. CONCLUSION: Together, our findings reveal a previously unappreciated role for HES1 in regulating fetal hematopoiesis, and provide new insight into the differences between fetal and adult HSC maintenance.


Subject(s)
Fetus , Hematopoiesis , Hematopoietic Stem Cells , Mice, Knockout , Transcription Factor HES-1 , Animals , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Mice , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Fetus/cytology , Fetus/metabolism , Cell Differentiation , Apoptosis , Cell Proliferation , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Signal Transduction , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics
11.
J Cell Biol ; 223(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38958606

ABSTRACT

Calorie restriction increases lifespan. Among the tissue-specific protective effects of calorie restriction, the impact on the gastrointestinal tract remains unclear. We report increased numbers of chromogranin A-positive (+), including orexigenic ghrelin+ cells, in the stomach of calorie-restricted mice. This effect was accompanied by increased Notch target Hes1 and Notch ligand Jag1 and was reversed by blocking Notch with DAPT, a gamma-secretase inhibitor. Primary cultures and genetically modified reporter mice show that increased endocrine cell abundance is due to altered Lgr5+ stem and Neurog3+ endocrine progenitor cell proliferation. Different from the intestine, calorie restriction decreased gastric Lgr5+ stem cells, while increasing a FOXO1/Neurog3+ subpopulation of endocrine progenitors in a Notch-dependent manner. Further, activation of FOXO1 was sufficient to promote endocrine cell differentiation independent of Notch. The Notch inhibitor PF-03084014 or ghrelin receptor antagonist GHRP-6 reversed the phenotypic effects of calorie restriction in mice. Tirzepatide additionally expanded ghrelin+ cells in mice. In summary, calorie restriction promotes Notch-dependent, FOXO1-regulated gastric endocrine cell differentiation.


Subject(s)
Caloric Restriction , Forkhead Box Protein O1 , Ghrelin , Receptors, Notch , Signal Transduction , Animals , Ghrelin/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Receptors, Notch/metabolism , Receptors, Notch/genetics , Mice , Cell Differentiation , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Cell Proliferation , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Gastric Mucosa/metabolism , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Male , Stomach
12.
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 , Cardiomegaly , Celecoxib , Myocytes, Cardiac , Animals , Rats , Aspirin/pharmacology , Aspirin/therapeutic use , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Cardiomegaly/etiology , Celecoxib/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Disease Models, Animal , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Rats, Sprague-Dawley , Receptor, Notch1/drug effects , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Transcription Factor HES-1/drug effects , Transcription Factor HES-1/metabolism
13.
Cell Biochem Biophys ; 82(3): 2523-2532, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38913282

ABSTRACT

The occurrence of myocardial ischemia/reperfusion injury is commonly observed during cardiac surgery; however, there remains a dearth of effective therapeutic strategies to mitigate this injury. The a disintegrin and metallopeptidase domain 10 (ADAM10) is a transmembrane protein anchored on the cell membrane surface, and its precise mechanism of action in myocardial ischemia/reperfusion injury remains incompletely understood. This study aims to investigate the impact of ADAM10 on cardiomyocyte injury induced by hypoxia/reoxygenation (H/R) and elucidate the underlying mechanisms. The ADAM10 overexpression plasmid was transfected into H9c2 cells, which were subsequently treated with the Notch signaling pathway inhibitor DAPT and cultured under H/R conditions. Cell proliferation activity was assessed using the CCK-8 assay. The levels of LDH, SOD, and MDA were quantified through colorimetric analysis. The levels of ROS and the rate of apoptosis were measured using flow cytometry. The morphological changes in the nucleus of H9c2 cells were observed by employing Hoechst 33258 staining. The mRNA expression levels of ADAM10, Notch1, NICD, and Hes1 in H9c2 cells were determined using qRT-PCR. The expressions of Notch signaling pathway and apoptosis-related proteins were analyzed by Western blot. Overexpression of ADAM10 provided protection to H9c2 cells against injury induced by H/R, leading to an increase in SOD levels and alleviation of oxidative stress caused by the accumulation of ROS and the decrease of SOD activity. Meanwhile, overexpression of ADAM10 inhibited apoptosis in H9c2 cells exposed to H/R by regulating the expression of apoptosis-related proteins, such as Bax, Bcl-2 and Cleaved-caspase-3. Additionally, overexpression of ADAM10 facilitated the activation of the Notch1 signaling pathway in H9c2 cells exposed to H/R by upregulating the protein expression of Notch1, NICD, and Hes1. However, the protective effect of ADAM10 on H/R-induced H9c2 cells was partially reversed by DAPT. Our findings demonstrate that ADAM10 exerts protective effects in H/R-induced H9c2 cells by suppressing oxidative stress and apoptosis via the activation of the Notch signaling pathway.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Apoptosis , Cell Hypoxia , Membrane Proteins , Myocytes, Cardiac , Signal Transduction , Transcription Factor HES-1 , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Animals , Rats , Cell Line , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Receptors, Notch/metabolism , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Reactive Oxygen Species/metabolism , Cell Proliferation , Dipeptides/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Oxidative Stress , Diamines/pharmacology , Superoxide Dismutase/metabolism , Superoxide Dismutase/genetics
14.
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
15.
Cell Mol Life Sci ; 81(1): 205, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38703204

ABSTRACT

BACKGROUND: Exposure to chronic psychological stress (CPS) is a risk factor for thrombotic cardiocerebrovascular diseases (CCVDs). The expression and activity of the cysteine cathepsin K (CTSK) are upregulated in stressed cardiovascular tissues, and we investigated whether CTSK is involved in chronic stress-related thrombosis, focusing on stress serum-induced endothelial apoptosis. METHODS AND RESULTS: Eight-week-old wild-type male mice (CTSK+/+) randomly divided to non-stress and 3-week restraint stress groups received a left carotid artery iron chloride3 (FeCl3)-induced thrombosis injury for biological and morphological evaluations at specific timepoints. On day 21 post-stress/injury, the stress had enhanced the arterial thrombi weights and lengths, in addition to harmful alterations of plasma ADAMTS13, von Willebrand factor, and plasminogen activation inhibitor-1, plus injured-artery endothelial loss and CTSK protein/mRNA expression. The stressed CTSK+/+ mice had increased levels of injured arterial cleaved Notch1, Hes1, cleaved caspase8, matrix metalloproteinase-9/-2, angiotensin type 1 receptor, galactin3, p16IN4A, p22phox, gp91phox, intracellular adhesion molecule-1, TNF-α, MCP-1, and TLR-4 proteins and/or genes. Pharmacological and genetic inhibitions of CTSK ameliorated the stress-induced thrombus formation and the observed molecular and morphological changes. In cultured HUVECs, CTSK overexpression and silencing respectively increased and mitigated stressed-serum- and H2O2-induced apoptosis associated with apoptosis-related protein changes. Recombinant human CTSK degraded γ-secretase substrate in a dose-dependent manor and activated Notch1 and Hes1 expression upregulation. CONCLUSIONS: CTSK appeared to contribute to stress-related thrombosis in mice subjected to FeCl3 stress, possibly via the modulation of vascular inflammation, oxidative production and apoptosis, suggesting that CTSK could be an effective therapeutic target for CPS-related thrombotic events in patients with CCVDs.


Subject(s)
Apoptosis , Cathepsin K , Chlorides , Disease Models, Animal , Ferric Compounds , Thrombosis , Animals , Humans , Male , Mice , ADAMTS13 Protein/metabolism , ADAMTS13 Protein/genetics , Cathepsin K/metabolism , Cathepsin K/genetics , Chlorides/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/genetics , Stress, Psychological/complications , Stress, Psychological/metabolism , Thrombosis/metabolism , Thrombosis/pathology , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics
16.
Food Chem Toxicol ; 189: 114724, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734200

ABSTRACT

Notch signaling regulates cartilage formation and homeostasis. Kashin-Beck Disease (KBD), an endemic osteochondropathy, is characterized by severe cartilage degradation. The etiology of KBD is related to the exposure of HT-2 toxin, a mycotoxin and primary metabolite of T-2 toxin. This study aims to explore the role of HT-2 toxin in the Notch signaling regulation and extracellular matrix (ECM) metabolism of hiPSCs-Chondrocytes. Immunohistochemistry and qRT-PCR were employed to investigate the expression of Notch pathway molecules in KBD articular cartilage and primary chondrocytes. hiPSCs-Chondrocytes, derived from hiPSCs, were treated with 100 ng/mL HT-2 toxin and the γ-secretase inhibitor (DAPT) for 48h, respectively. The markers related to the Notch signaling pathway and ECM were assessed using qRT-PCR and Western blot. Notch pathway dysregulation was prominent in KBD cartilage. HT-2 toxin exposure caused cytotoxicity in hiPSCs-Chondrocytes, and activated Notch signaling by increasing the mRNA and protein levels of NOTCH1 and HES1. HT-2 toxin also upregulated ECM catabolic enzymes and downregulated ECM components (COL2A1 and ACAN), indicating ECM degradation. DAPT-mediated Notch signaling inhibition suppressed the mRNA and protein level of ADAMTS5 expression while enhancing ECM component expression in hiPSCs-Chondrocytes. This study suggests that HT-2 toxin may induce ECM degradation in hiPSCs-Chondrocytes through activating Notch signaling.


Subject(s)
Chondrocytes , Extracellular Matrix , Induced Pluripotent Stem Cells , Receptors, Notch , Signal Transduction , T-2 Toxin , Humans , Signal Transduction/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , T-2 Toxin/toxicity , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Receptors, Notch/metabolism , Receptors, Notch/genetics , Kashin-Beck Disease/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Cells, Cultured
17.
Int J Mol Sci ; 25(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38791273

ABSTRACT

The HMG-domain containing transcription factor Sox10 plays a crucial role in regulating Schwann cell survival and differentiation and is expressed throughout the entire Schwann cell lineage. While its importance in peripheral myelination is well established, little is known about its role in the early stages of Schwann cell development. In a search for direct target genes of Sox10 in Schwann cell precursors, the transcriptional co-repressor Tle4 was identified. At least two regions upstream of the Tle4 gene appear involved in mediating the Sox10-dependent activation. Once induced, Tle4 works in tandem with the bHLH transcriptional repressor Hes1 and exerts a dual inhibitory effect on Sox10 by preventing the Sox10 protein from transcriptionally activating maturation genes and by suppressing Sox10 expression through known enhancers of the gene. This mechanism establishes a regulatory barrier that prevents premature activation of factors involved in differentiation and myelin formation by Sox10 in immature Schwann cells. The identification of Tle4 as a critical downstream target of Sox10 sheds light on the gene regulatory network in the early phases of Schwann cell development. It unravels an elaborate regulatory circuitry that fine-tunes the timing and extent of Schwann cell differentiation and myelin gene expression.


Subject(s)
Cell Differentiation , DNA-Binding Proteins , SOXE Transcription Factors , Schwann Cells , Animals , Humans , Mice , Rats , Cell Differentiation/genetics , Co-Repressor Proteins/metabolism , Co-Repressor Proteins/genetics , Feedback, Physiological , Gene Expression Regulation , Myelin Sheath/metabolism , Schwann Cells/metabolism , Schwann Cells/cytology , SOXE Transcription Factors/metabolism , SOXE Transcription Factors/genetics , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , DNA-Binding Proteins/metabolism
18.
Dev Cell ; 59(15): 1913-1923.e6, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38772376

ABSTRACT

Neural stem cells (NSCs) differentiate into neuron-fated intermediate progenitor cells (IPCs) via cell division. Although differentiation from NSCs to IPCs is a discrete process, recent transcriptome analyses identified a continuous transcriptional trajectory during this process, raising the question of how to reconcile these contradictory observations. In mouse NSCs, Hes1 expression oscillates, regulating the oscillatory expression of the proneural gene Neurog2, while Hes1 expression disappears in IPCs. Thus, the transition from Hes1 oscillation to suppression is involved in the differentiation of NSCs to IPCs. Here, we found that Neurog2 oscillations induce the accumulation of Tbr2, which suppresses Hes1 expression, generating an IPC-like gene expression state in NSCs. In the absence of Tbr2, Hes1 expression is up-regulated, decreasing the formation of IPCs. These results indicate that the Neurog2-Tbr2 axis forms a continuous transcriptional trajectory to an IPC-like neurogenic state in NSCs, which then differentiate into IPCs via cell division.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Nerve Tissue Proteins , Neural Stem Cells , Neurogenesis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Animals , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Mice , Neurogenesis/genetics , Gene Expression Regulation, Developmental , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Neurons/metabolism , Neurons/cytology , T-Box Domain Proteins
19.
Pflugers Arch ; 476(8): 1279-1288, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38772920

ABSTRACT

Phosphate homeostasis is vital for many biological processes and disruptions in circulating levels can be detrimental. While the mechanisms behind FGF23 regulation have been regularly studied, the role of extracellular phosphate sensing and its impact on fibroblast growth factor 23 (FGF23) expression remains unclear. This study aimed to investigate the involvement of reactive oxygen species (ROS), silent information regulator 1 (SIRT1), and Hairy and Enhancer of Split-1 (HES1) in regulating FGF23 in FGF23 expressing MC3T3-E1 cells. MC3T3-E1 cells treated with ß-glycerophosphate (BGP) resulted in increased Fgf23 expression. Inhibition of ROS formation by inhibition of NADPH oxidase, which is essential for ROS production, did not affect this response to BGP, suggesting ROS is not involved in this process. Moreover, treatment with tert-butyl hydroperoxide (TBHP), a ROS-inducing agent, did not increase Fgf23 expression. This suggests that ROS machinery is not involved in FGF23 stimulation as previously suggested. Nonetheless, inhibition of SIRT1 using Ex527 eliminated the Fgf23 response to BGP, indicating its involvement in FGF23 regulation after BGP treatment. Indeed, activation of SIRT1 using SRT1720 increased Fgf23 expression. Moreover, transcription factor Hes1 was upregulated by BGP treatment, which was diminished when cells were treated with Ex527 implying it is also regulated through SIRT1. These findings suggest the existence of an upstream SIRT1-HES1 axis in the regulation of FGF23 by phosphate, though we were unable to find a role for ROS in this process. Further research should provide insights into phosphate homeostasis and potential therapeutic targets for phosphate-related disorders.


Subject(s)
Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Glycerophosphates , Reactive Oxygen Species , Sirtuin 1 , Animals , Sirtuin 1/metabolism , Sirtuin 1/genetics , Glycerophosphates/pharmacology , Glycerophosphates/metabolism , Fibroblast Growth Factors/metabolism , Mice , Fibroblast Growth Factor-23/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Cell Line , Heterocyclic Compounds, 4 or More Rings/pharmacology , Carbazoles/pharmacology
20.
Mol Oncol ; 18(6): 1510-1530, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459621

ABSTRACT

The transcription factor receptor-interacting protein 140 (RIP140) regulates intestinal homeostasis and tumorigenesis through Wnt signaling. In this study, we investigated its effect on the Notch/HES1 signaling pathway. In colorectal cancer (CRC) cell lines, RIP140 positively regulated HES1 gene expression at the transcriptional level via a recombining binding protein suppressor of hairless (RBPJ)/neurogenic locus notch homolog protein 1 (NICD)-mediated mechanism. In support of these in vitro data, RIP140 and HES1 expression significantly correlated in mouse intestine and in a cohort of CRC samples, thus supporting the positive regulation of HES1 gene expression by RIP140. Interestingly, when the Notch pathway is fully activated, RIP140 exerted a strong inhibition of HES1 gene transcription controlled by the level of HES1 itself. Moreover, RIP140 directly interacts with HES1 and reversed its mitogenic activity in human CRC cells. In line with this observation, HES1 levels were associated with a better patient survival only when tumors expressed high levels of RIP140. Our data identify RIP140 as a key regulator of the Notch/HES1 signaling pathway, with a dual effect on HES1 gene expression at the transcriptional level and a strong impact on colon cancer cell proliferation.


Subject(s)
Cell Proliferation , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Nuclear Receptor Interacting Protein 1 , Transcription Factor HES-1 , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Nuclear Receptor Interacting Protein 1/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Signal Transduction , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL