Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273
Filter
1.
Journal of Central South University(Medical Sciences) ; (12): 641-647, 2023.
Article in English | WPRIM | ID: wpr-982333

ABSTRACT

OBJECTIVES@#Application of ultrashort wave (USW) to rats with cerebral ischemia and reperfusion injury could inhibit the decrease of expression of secretory pathway Ca2+-ATPase 1 (SPCA1), an important participant in Golgi stress, reduce the damage of Golgi apparatus and the apoptosis of neuronal cells, thereby alleviating cerebral ischemia-reperfusion injury. This study aims to investigate the effect of USW on oxygen-glucose deprivation/reperfusion (OGD/R) injury and the expression of SPCA1 at the cellular level.@*METHODS@#N2a cells were randomly divided into a control (Con) group, an OGD/R group, and an USW group. The cells in the Con group were cultured without exposure to OGD. The cells in the OGD/R group were treated with OGD/R. The cells in the USW group were treated with USW after OGD/R. Cell morphology was observed under the inverted phase-contrast optical microscope, cell activity was detected by cell counting kit-8 (CCK-8), apoptosis was detected by flow cytometry, and SPCA1 expression was detected by Western blotting.@*RESULTS@#Most of the cells in the Con group showed spindle shape with a clear outline and good adhesion. In the OGD/R group, cells were wrinkled, with blurred outline, poor adhesion, and lots of suspended dead cells appeared; compared with the OGD/R group, the cell morphology and adherence were improved, with clearer outlines and fewer dead cells in the USW group. Compared with the Con group, the OGD/R group showed decreased cell activity, increased apoptotic rate, and down-regulating SPCA1 expression with significant differences (all P<0.001); compared with the OGD/R group, the USW group showed increased cell activity, decreased apoptotic rate, and up-regulating SPCA1 expression with significant differences (P<0.01 or P<0.001).@*CONCLUSIONS@#USW alleviates the injury of cellular OGD/R, and its protective effect may be related to its up-regulation of SPCA1 expression.


Subject(s)
Animals , Rats , Apoptosis , Brain Ischemia , Glucose/metabolism , Oxygen/metabolism , Reperfusion Injury/metabolism , Transcriptional Activation , Up-Regulation , Calcium-Transporting ATPases/metabolism
2.
Journal of Central South University(Medical Sciences) ; (12): 333-344, 2021.
Article in English | WPRIM | ID: wpr-880664

ABSTRACT

OBJECTIVES@#To analyze the expressions and distributions of hypoxia-inducible factor-1α (HIF-1α), CD147, and glucose transporter 1 (GLUT1) in epidermis from psoriasis vulgaris and normal people, and to explore the associations among these proteins and their roles in hypoxic HaCaT cell line.@*METHODS@#The expression levels of HIF-1α, CD147, and GLUT1 were determined by immunohistochemistry staining in skin biopsies from 48 psoriasis vularis patients and 33 healthy subjects. Cobalt chloride (CoCl@*RESULTS@#HIF-1α, CD147, and GLUT1 were highly expressed and the glycolytic capacity was increased in lesions of psoriasis vulgaris; HIF-1α upregulated the expression of CD147 and GLUT1, increased the lactate production and decreased the ATP level in CoCl@*CONCLUSIONS@#Glycolytic capacity increases in the injured keratinocytes of psoriasis vulgaris, suggesting that HIF-1α, CD147, and GLUT1 are associated with glycolysis, which can be considered as the promising targets for psoriasis therapy.


Subject(s)
Humans , Basigin , Glucose Transporter Type 1 , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Psoriasis/genetics , Transcriptional Activation , Up-Regulation
3.
Biol. Res ; 53: 25, 2020. tab, graf
Article in English | LILACS | ID: biblio-1124210

ABSTRACT

BACKGROUND: Hypoxia inducible factor-1 (HIF-1) is considered as the most activated transcriptional factor in response to low oxygen level or hypoxia. HIF-1 binds the hypoxia response element (HRE) sequence in the promoter of different genes, mainly through the bHLH domain and activates the transcription of genes, especially those involved in angiogenesis and EMT. Considering the critical role of bHLH in binding HIF-1 to the HRE sequence, we hypothesized that bHLH could be a promising candidate to be targeted in hypoxia condition. METHODS: We inserted an inhibitory bHLH (ibHLH) domain in a pIRES2-EGFP vector and transfected HEK293T cells with either the control vector or the designed construct. The ibHLH domain consisted of bHLH domains of both HIF-1a and Arnt, capable of competing with HIF-1 in binding to HRE sequences. The transfected cells were then treated with 200 µM of cobalt chloride (CoCl2) for 48 h to induce hypoxia. Real-time PCR and western blot were performed to evaluate the effect of ibHLH on the genes and proteins involved in angiogenesis and EMT. RESULTS: Hypoxia was successfully induced in the HEK293T cell line as the gene expression of VEGF, vimentin, and ß-catenin were significantly increased after treatment of untransfected HEK293T cells with 200 µM CoCl2. The gene expression of VEGF, vimentin, and ß-catenin and protein level of ß-catenin were significantly decreased in the cells transfected with either control or ibHLH vectors in hypoxia. However, ibHLH failed to be effective on these genes and the protein level of ß-catenin, when compared to the control vector. We also observed that overexpression of ibHLH had more inhibitory effect on gene and protein expression of N-cadherin compared to the control vector. However, it was not statistically significant. CONCLUSION: bHLH has been reported to be an important domain involved in the DNA binding activity of HIF. However, we found that targeting this domain is not sufficient to inhibit the endogenous HIF-1 transcriptional activity. Further studies about the function of critical domains of HIF-1 are necessary for developing a specific HIF-1 inhibitor.


Subject(s)
Humans , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism , Gene Expression , Transcriptional Activation/genetics , Blotting, Western , Basic Helix-Loop-Helix Transcription Factors/genetics , Hypoxia-Inducible Factor 1/genetics , HEK293 Cells , Real-Time Polymerase Chain Reaction , Hypoxia/genetics
5.
Braz. j. med. biol. res ; 53(6): e9346, 2020. graf
Article in English | LILACS, ColecionaSUS | ID: biblio-1132516

ABSTRACT

Atherosclerosis (AS) is a common vascular disease, which can cause apoptosis of vascular endothelial cells. Notoginsenoside R1 (NGR1) is considered an anti-AS drug. MicroRNAs (miRNAs) are believed to play a vital role in cell apoptosis and angiogenesis. This study aimed to explore the mechanism of NGR1 for treating AS through miRNAs. Flow cytometry was used to detect the apoptosis rate. The levels of inflammatory cytokines interleukin (IL)-6 and IL-1β were detected using ELISA. Reactive oxygen species (ROS) and malondialdehyde (MDA) levels were measured using corresponding assay kits. Quantitative real-time polymerase chain reaction (qRT-PCR) assay was performed to detect miR-221-3p expression. Dual-luciferase reporter and RNA immunoprecipitation assays were carried out to examine the relationship between miR-221-3p and toll-like receptors 4 (TLR4). Also, western blot analysis was performed to determine the levels of TLR4 and nuclear factor kappa B (NF-κB) signaling pathway-related proteins. Oxidized low-density lipoprotein (ox-LDL) induced human umbilical vein endothelial cells (HUVECs) apoptosis, inflammation, and oxidative stress. NGR1 alleviated the negative effect of ox-LDL through promoting the expression of miR-221-3p in HUVECs. TLR4 was a target of miR-221-3p, and its overexpression could reverse the inhibition effects of miR-221-3p on apoptosis, inflammation, and oxidative stress. NGR1 improved miR-221-3p expression to inhibit the activation of the TLR4/NF-κB pathway in ox-LDL-treated HUVECs. NGR1 decreased ox-LDL-induced HUVECs apoptosis, inflammation, and oxidative stress through increasing miR-221-3p expression, thereby inhibiting the activation of the TLR4/NF-κB pathway. This study of the mechanism of NGR1 provided a more theoretical basis for the treatment of AS.


Subject(s)
Humans , Apoptosis/drug effects , Oxidative Stress/drug effects , Ginsenosides/pharmacology , MicroRNAs/adverse effects , Human Umbilical Vein Endothelial Cells/drug effects , Inflammation/metabolism , Lipoproteins, LDL/metabolism , Enzyme-Linked Immunosorbent Assay , Signal Transduction , Transcriptional Activation , Up-Regulation , Blotting, Western , NF-kappa B/antagonists & inhibitors , Reactive Oxygen Species , MicroRNAs/metabolism , Immunoprecipitation , Toll-Like Receptor 4/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/metabolism , Real-Time Polymerase Chain Reaction
6.
The Korean Journal of Physiology and Pharmacology ; : 53-68, 2020.
Article in English | WPRIM | ID: wpr-787138

ABSTRACT

The purpose of this study was to characterize the genetic contribution to endothelial adaptation to exercise training. Vasoreactivity was assessed in aortas from four inbred mouse strains (129S1, B6, NON, and SJL) after 4 weeks of moderate intensity continuous exercise training (MOD), high intensity interval training (HIT) or in sedentary controls (SED). Intrinsic variations in endothelium-dependent vasorelaxation (EDR) to acetylcholine (ACh) as well as vasocontractile responses were observed across SED groups. For responses to exercise training, there was a significant interaction between mouse strain and training intensity on EDR. Exercise training had no effect on EDR in aortas from 129S1 and B6 mice. In NON, EDR was improved in aortas from MOD and HIT compared with respective SED, accompanied by diminished responses to PE in those groups. Interestingly, EDR was impaired in aorta from SJL HIT compared with SED. The transcriptional activation of endothelial genes was also influenced by the interaction between mouse strain and training intensity. The number of genes altered by HIT was greater than MOD, and there was little overlap between genes altered by HIT and MOD. HIT was associated with gene pathways for inflammatory responses. NON MOD genes showed enrichment for vessel growth pathways. These findings indicate that exercise training has non-uniform effects on endothelial function and transcriptional activation of endothelial genes depending on the interaction between genetic background and training intensity.


Subject(s)
Animals , Mice , Acetylcholine , Aorta , Endothelium , Gene Expression Profiling , Genetic Background , Mice, Inbred Strains , Transcriptional Activation , Vasodilation
7.
Braz. j. med. biol. res ; 52(10): e8631, 2019. tab, graf
Article in English | LILACS | ID: biblio-1039247

ABSTRACT

The long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3), a tumor suppressor, is critical for the carcinogenesis and progression of different cancers, including hepatocellular carcinoma (HCC). To date, the roles of lncRNA MEG3 in HCC are not well illustrated. Therefore, this study used western blot and qRT-PCR to evaluate the expression of MEG3, miR-9-5p, and Sex determining Region Y-related HMG-box 11 (SOX11) in HCC tissues and cell lines. RNA pull-down and luciferase reporter assay were used to evaluate these molecular interactions. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and flow cytometry detected the viability and apoptosis of HCC cells, respectively. The results showed that MEG3 and SOX11 were poorly expressed but miR-9-5p was highly expressed in HCC. The expression levels of these molecules suggested a negative correlation between MEG3 and miR-9-5p and a positive correlation with SOX11, confirmed by Pearson's correlation analysis and biology experiments. Furthermore, MEG3 could combine with miR-9-5p, and SOX11 was a direct target of miR-9-5p. Moreover, MEG3 over-expression promoted cell apoptosis and growth inhibition in HCC cells through sponging miR-9-5p to up-regulate SOX11. Therefore, the interactions among MEG3, miR-9-5p, and SOX11 might offer a novel insight for understanding HCC pathogeny and provide potential diagnostic markers and therapeutic targets for HCC.


Subject(s)
Humans , Male , Female , Middle Aged , Carcinoma, Hepatocellular/genetics , MicroRNAs/genetics , SOXC Transcription Factors/genetics , RNA, Long Noncoding/genetics , Liver Neoplasms/genetics , Transfection , Gene Expression Regulation, Neoplastic , Transcriptional Activation , Up-Regulation , Apoptosis/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , MicroRNAs/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , SOXC Transcription Factors/metabolism , Real-Time Polymerase Chain Reaction , RNA, Long Noncoding/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasm Staging
8.
Biol. Res ; 52: 20, 2019. tab, graf
Article in English | LILACS | ID: biblio-1011422

ABSTRACT

BACKGROUND: Histone acetylation is an important epigenetic modification that regulates gene activity in response to stress. Histone acetylation levels are reversibly regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs). The imperative roles of HDACs in gene transcription, transcriptional regulation, growth and responses to stressful environment have been widely investigated in Arabidopsis. However, data regarding HDACs in kenaf crop has not been disclosed yet. RESULTS: In this study, six HDACs genes (HcHDA2, HcHDA6, HcHDA8, HcHDA9, HcHDA19, and HcSRT2) were isolated and characterized. Phylogenetic tree revealed that these HcHDACs shared high degree of sequence homology with those of Gossypium arboreum. Subcellular localization analysis showed that GFP-tagged HcHDA2 and HcHDA8 were predominantly localized in the nucleus, HcHDA6 and HcHDA19 in nucleus and cytosol. The HcHDA9 was found in both nucleus and plasma membranes. Real-time quantitative PCR showed that the six HcHDACs genes were expressed with distinct expression patterns across plant tissues. Furthermore, we determined differential accumulation of HcHDACs transcripts under salt and drought treatments, indicating that these enzymes may participate in the biological process under stress in kenaf. Finally, we showed that the levels of histone H3 and H4 acetylation were modulated by salt and drought stress in kenaf. CONCLUSIONS: We have isolated and characterized six HDACs genes from kenaf. These data showed that HDACs are imperative players for growth and development as well abiotic stress responses in kenaf.


Subject(s)
Stress, Physiological/physiology , Hibiscus/enzymology , Histone Acetyltransferases/physiology , Droughts , Histone Deacetylases/physiology , Transcriptional Activation/physiology , Cloning, Molecular , Hibiscus/growth & development , Hibiscus/physiology , Real-Time Polymerase Chain Reaction
9.
Braz. j. med. biol. res ; 52(7): e8381, 2019. tab, graf
Article in English | LILACS | ID: biblio-1011592

ABSTRACT

Experiments were conducted to determine if the follicle-stimulating hormone (FSH) receptor binding inhibitor (FRBI) impacts the expression levels of AT-rich interactive domain-containing protein 1A (ARID1A) and phosphatase and tensin homolog (PTEN) in ovaries and blood, as well as expressions of follicle-stimulating hormone cognate receptor (FSHR) gene and proteins. Mice in FRBI-10, FRBI-20, FRBI-30, and FRBI-40 groups were intramuscularly injected with 10, 20, 30, and 40 mg FRBI/kg, respectively, for five consecutive days. Western blotting and qRT-PCR were utilized to determine expression levels of ARID1A and PTEN proteins and mRNAs. Serum ARID1A and PTEN concentrations of the FRBI-40 group were higher than the control group (CG) and FSH group (P<0.05). FSHR mRNA levels of FRBI-20, FRBI-30, and FRBI-40 groups were lower than that of CG and FSH groups on day 15 (P<0.05 or P<0.01). Expression levels of FSHR proteins of FRBI-30 and FRBI-40 groups were lower than those of CG and FSH groups (P<0.05). Levels of ARID1A and PTEN proteins of the FRBI-30 group were greater than CG on days 20 and 30 (P<0.05). FRBI doses had significant positive correlations to levels of ARID1A and PTEN proteins. Additionally, ARID1A and PTEN had negative correlations to FSHR mRNAs and proteins. A high dose of FRBI could promote the expression levels of ARID1A and PTEN proteins in ovarian tissues. FRBI increased serum concentrations of ARID1A and PTEN. However, FRBI depressed expression levels of FSHR mRNAs and proteins in mouse ovaries.


Subject(s)
Animals , Female , Rabbits , Ovarian Neoplasms/metabolism , Receptors, FSH/antagonists & inhibitors , Nuclear Proteins/blood , DNA-Binding Proteins/metabolism , PTEN Phosphohydrolase/blood , Follicle Stimulating Hormone/metabolism , Phosphorylation , Transcription Factors , Nuclear Proteins/metabolism , Transcriptional Activation/genetics , Up-Regulation , Blotting, Western , DNA-Binding Proteins/blood , PTEN Phosphohydrolase/metabolism , Real-Time Polymerase Chain Reaction
10.
Chinese Journal of Biotechnology ; (12): 1126-1134, 2019.
Article in Chinese | WPRIM | ID: wpr-771815

ABSTRACT

Human bocavirus 1 (HBoV1) non-structural protein NS1 is a multifunctional protein important for virus replication and induction of apoptosis in host cell. To better understand the function of the NS1 protein, it is urgent to address reducing the toxicity of NS1 to host cells. In the present study, we established a stable cell line that regulates expression of NS1 of HBoV1. The recombinant lentivirus plasmid containing a regulatable promoter fused with ns1 gene was constructed and transfected into HEK 293T cells using transfection reagent. The HEK 293T cell lines stably expressing NS1-100 and NS1-70 proteins were established by screening resistant cells with puromycin and inducing NS1 expression with doxycycline. The expression of NS1 protein was determined by fluorescent labeling protein and Western blotting. HBoV1 promoter was transfected into stably expressing NS1 cell line and its trans-transcriptional activity was analyzed. The results showed that NS1 protein was expressed stably in the established cell lines and had a strong activation activity on the HBoV1 promoter driving luciferase gene. Taken together, this study provides a solid basis for further research on the function of NS1 and the pathogenesis of human bocavirus.


Subject(s)
Human bocavirus , Promoter Regions, Genetic , Transcriptional Activation , Viral Nonstructural Proteins , Virus Replication
11.
Neuroscience Bulletin ; (6): 15-24, 2019.
Article in English | WPRIM | ID: wpr-775480

ABSTRACT

Fenofibrate, an agonist for peroxisome proliferator-activated receptor alpha (PPAR-α), lowers blood pressure, but whether this action is mediated via baroreflex afferents has not been elucidated. In this study, the distribution of PPAR-α and PPAR-γ was assessed in the nodose ganglion (NG) and the nucleus of the solitary tract (NTS). Hypertension induced by drinking high fructose (HFD) was reduced, along with complete restoration of impaired baroreceptor sensitivity, by chronic treatment with fenofibrate. The molecular data also showed that both PPAR-α and PPAR-γ were dramatically up-regulated in the NG and NTS of the HFD group. Expression of the downstream signaling molecule of PPAR-α, the mitochondrial uncoupling protein 2 (UCP2), was up-regulated in the baroreflex afferent pathway under similar experimental conditions, along with amelioration of reduced superoxide dismutase activity and increased superoxide in HFD rats. These results suggest that chronic treatment with fenofibrate plays a crucial role in the neural control of blood pressure by improving baroreflex afferent function due at least partially to PPAR-mediated up-regulation of UCP2 expression and reduction of oxidative stress.


Subject(s)
Animals , Male , Afferent Pathways , Antihypertensive Agents , Pharmacology , Baroreflex , Blood Pressure , Fenofibrate , Pharmacology , Oxidative Stress , PPAR gamma , Metabolism , Rats, Sprague-Dawley , Signal Transduction , Transcriptional Activation , Uncoupling Protein 2 , Metabolism , Up-Regulation
12.
Journal of Korean Medical Science ; : e100-2019.
Article in English | WPRIM | ID: wpr-764935

ABSTRACT

BACKGROUND: Increased expression of MDR1 gene is one of the major mechanisms responsible for multidrug resistance in cancer cells. Two alternative promoters, upstream and downstream, are responsible for transcription of MDR1 gene in the human. However, the molecular mechanism regarding the transactivation of MDR1 upstream promoter (USP) has not been determined. METHODS: Dual-luciferase reporter gene assays were used to assess the effect of Nkx-2.5 on MDR1 USP activity using reporter plasmids for human MDR1 USP and its mutants. MDR1 mRNA level was examined by quantitative real-time PCR. The direct binding of Nkx-2.5 to the USP of MDR1 was evaluated by promoter enzyme immunoassays and chromatin immunoprecipitation assays.


Subject(s)
Humans , Breast Neoplasms , Breast , Chromatin Immunoprecipitation , Drug Resistance, Multiple , Genes, Reporter , Immunoassay , Immunoenzyme Techniques , Phenotype , Plasmids , Real-Time Polymerase Chain Reaction , RNA, Messenger , Transcriptional Activation
13.
Journal of Bone Metabolism ; : 3-12, 2019.
Article in English | WPRIM | ID: wpr-740481

ABSTRACT

Vitamin D (VD) is essential for bone health, and VD or its analogues are widely used in clinics to ameliorate bone loss. The targets and mode of VD anti-osteoporotic actions appear to be different from those of other classes of drugs modulating bone remodeling. VD exerts its biological activities through the nuclear VD receptor (VDR)-mediated transcriptional regulation of target mRNA and non-coding RNA genes. VD-induced gene regulation involves epigenetic modifications of chromatin conformation at the target loci as well as reconfiguration of higher-order chromosomal organization through VDR-mediated recruitment of various regulatory factors. Enhancer RNAs (eRNA), a class of non-coding enhancer-derived RNAs, have recently emerged as VDR target gene candidates that act through reorganization of chromatin looping to induce enhancer-promoter interaction in activation of mRNA-encoding genes. This review outlines the molecular mechanisms of VD actions mediated by the VDR and suggests novel function of eRNAs in VDR transactivation.


Subject(s)
Bone Remodeling , Chromatin , Epigenomics , Metabolism , Receptors, Calcitriol , RNA , RNA, Messenger , RNA, Untranslated , Transcriptional Activation , Vitamin D , Vitamins
14.
Cancer Research and Treatment ; : 576-592, 2019.
Article in English | WPRIM | ID: wpr-763136

ABSTRACT

PURPOSE: Although the interferon α (IFNα) signaling and the paired-like homeodomain transcription factor 2 (PITX2) have both been implicated in the progression of breast cancer (BCa), it remains obscure whether these two pathways act in a coordinated manner. We therefore aimed to elucidate the expression and function of PITX2 during the pathogenesis of endocrine resistance in BCa. MATERIALS AND METHODS: PITX2 expression was assessed in BCa tissues using quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunohistochemistry and in experimentally induced letrozole-resistant BCa cells using RT-qPCR and immunoblotting. Effects of PITX2 deregulation on BCa progression was determined by assessing MTT, apoptosis and xenograft model. Finally, using multiple assays, the transcriptional regulation of interferon-inducible transmembrane protein 1 (IFITM1) by PITX2 was studied at both molecular and functional levels. RESULTS: PITX2 expression was induced in letrozole-resistant BCa tissues and cells, and PITX2 induction by IFNα signaling powerfully protected BCa cells against letrozole insult and potentiated letrozole-resistance. Mechanistically, PITX2 enhanced IFNα-induced AKT activation by transactivating the transcription of IFITM1, thus rendering BCa cells unresponsive to letrozoleelicited cell death. Additionally, ablation of IFITM1 expression using siRNA substantially abolished IFNα-elicited AKT phosphorylation, even in the presence of PITX2 overexpression, thus sensitizing BCa cells to letrozole treatment. CONCLUSION: These results demonstrate that constitutive upregulation of PITX2/IFITM1 cascade is an intrinsic adaptive mechanism during the pathogenesis of letrozole-resistance, and modulation of PITX2/IFITM1 level using different genetic and pharmacological means would thus have a novel therapeutic potential against letrozole resistance in BCa.


Subject(s)
Apoptosis , Breast Neoplasms , Breast , Cell Death , Heterografts , Immunoblotting , Immunohistochemistry , Interferons , Phosphorylation , Polymerase Chain Reaction , Reverse Transcription , RNA, Small Interfering , Transcription Factors , Transcriptional Activation , Up-Regulation
15.
International Journal of Oral Science ; (4): 27-27, 2018.
Article in English | WPRIM | ID: wpr-772289

ABSTRACT

The interplay between mechanoresponses and a broad range of fundamental biological processes, such as cell cycle progression, growth and differentiation, has been extensively investigated. However, metabolic regulation in mechanobiology remains largely unexplored. Here, we identified glucose transporter 1 (GLUT1)-the primary glucose transporter in various cells-as a novel mechanosensitive gene in orthodontic tooth movement (OTM). Using an in vivo rat OTM model, we demonstrated the specific induction of Glut1 proteins on the compressive side of a physically strained periodontal ligament. This transcriptional activation could be recapitulated in in vitro cultured human periodontal ligament cells (PDLCs), showing a time- and dose-dependent mechanoresponse. Importantly, application of GLUT1 specific inhibitor WZB117 greatly suppressed the efficiency of orthodontic tooth movement in a mouse OTM model, and this reduction was associated with a decline in osteoclastic activities. A mechanistic study suggested that GLUT1 inhibition affected the receptor activator for nuclear factor-κ B Ligand (RANKL)/osteoprotegerin (OPG) system by impairing compressive force-mediated RANKL upregulation. Consistently, pretreatment of PDLCs with WZB117 severely impeded the osteoclastic differentiation of co-cultured RAW264.7 cells. Further biochemical analysis indicated mutual regulation between GLUT1 and the MEK/ERK cascade to relay potential communication between glucose uptake and mechanical stress response. Together, these cross-species experiments revealed the transcriptional activation of GLUT1 as a novel and conserved linkage between metabolism and bone remodelling.


Subject(s)
Animals , Humans , Mice , Rats , Biomechanical Phenomena , Blotting, Western , Bone Remodeling , Cells, Cultured , Glucose Transporter Type 1 , Genetics , Hydroxybenzoates , Pharmacology , Immunohistochemistry , MAP Kinase Signaling System , Mice, Inbred C57BL , Osteoprotegerin , Metabolism , Periodontal Ligament , Cell Biology , RANK Ligand , Metabolism , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tooth Movement Techniques , Transcriptional Activation
16.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 41-52, 2018.
Article in English | WPRIM | ID: wpr-812430

ABSTRACT

The aims of the present study were to evaluate the effects of puerarin on angiotensin II-induced cardiac fibroblast proliferation and to explore the molecular mechanisms of action. Considering the role of HO in nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation, we hypothesized that modulating catalase activity would be a potential target in regulating the redox-sensitive pathways. Our results showed that the activation of Rac1 was dependent on the levels of intracellular HO. Puerarin blocked the phosphorylation of extracellular regulated protein kinases (ERK)1/2, abolished activator protein (AP)-1 binding activity, and eventually attenuated cardiac fibroblast proliferation through the inhibition of HO-dependent Rac1 activation. Further studies revealed that angiotensin II treatment resulted in decreased catalase protein expression and enzyme activity, which was disrupted by puerarin via the upregulation of catalase protein expression at the transcriptional level and the prolonged protein degradation. These findings indicated that the anti-proliferation mechanism of puerarin was mainly through blocking angiontensin II-triggered downregulation of catalase expression and HO-dependent Rac1 activation.


Subject(s)
Animals , Mice , Angiotensin II , Pharmacology , Angiotensin II Type 1 Receptor Blockers , Pharmacology , Animals, Newborn , Catalase , Genetics , Metabolism , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases , Metabolism , Fibroblasts , Gene Expression Regulation , Heart , Hydrogen Peroxide , Metabolism , Pharmacology , Isoflavones , Pharmacology , Myocardium , Cell Biology , Metabolism , NADPH Oxidases , Metabolism , Neuropeptides , Metabolism , Signal Transduction , Transcription Factor AP-1 , Metabolism , Transcriptional Activation , rac1 GTP-Binding Protein , Metabolism
17.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 330-338, 2018.
Article in English | WPRIM | ID: wpr-812398

ABSTRACT

Fibroblast-like synoviocytes (FLS) play a pivotal role in Rheumatoid arthritis (RA) pathogenesis through aggressive migration and invasion. Madecassoside (Madec), a triterpenoid saponin present in Centella asiatica herbs, has a potent anti-inflammatory effect. In the present study, Madec exerted an obvious therapeutic effect in reversing the histological lesions in adjuvant-induced arthritis (AIA) rats. To recognize the anti-rheumatoid potentials of Madec, we further investigated whether Madec interfered with FLS invasion and metalloproteinase (MMP) expression. In cultures of primary FLS isolated from the AIA rats, Madec (10 and 30 μmol·L) was proven to considerably inhibit migration and invasion of FLS induced by interleukin 1β (IL-1β), but exhibiting no obvious effect on cell proliferation. Madec repressed IL-1β-triggered FLS invasion by prohibiting the expression of MMP-13. Additionally, Madec suppressed MMP-13 transcription via inhibiting the MMP-13 promoter-binding activity of NF-κB. Our results further showed that Madec down-regulated the translocation and phosphorylation of NF-κB as demonstrated by Western blotting and immunofluorescence assays. In conclusion, our results suggest that Madec exerts anti-RA activity via inhibiting the NF-κB/MMP-13 pathway.


Subject(s)
Animals , Rats , Antirheumatic Agents , Chemistry , Pharmacology , Therapeutic Uses , Arthritis, Experimental , Drug Therapy , Pathology , Cell Movement , Cell Nucleus , Metabolism , Cells, Cultured , Gene Expression Regulation, Enzymologic , Interleukin-1beta , Pharmacology , Matrix Metalloproteinase 13 , Genetics , NF-kappa B , Genetics , Metabolism , Phosphorylation , Protein Transport , Signal Transduction , Synoviocytes , Metabolism , Transcriptional Activation , Triterpenes , Chemistry , Pharmacology , Therapeutic Uses
18.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 41-52, 2018.
Article in English | WPRIM | ID: wpr-773640

ABSTRACT

The aims of the present study were to evaluate the effects of puerarin on angiotensin II-induced cardiac fibroblast proliferation and to explore the molecular mechanisms of action. Considering the role of HO in nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation, we hypothesized that modulating catalase activity would be a potential target in regulating the redox-sensitive pathways. Our results showed that the activation of Rac1 was dependent on the levels of intracellular HO. Puerarin blocked the phosphorylation of extracellular regulated protein kinases (ERK)1/2, abolished activator protein (AP)-1 binding activity, and eventually attenuated cardiac fibroblast proliferation through the inhibition of HO-dependent Rac1 activation. Further studies revealed that angiotensin II treatment resulted in decreased catalase protein expression and enzyme activity, which was disrupted by puerarin via the upregulation of catalase protein expression at the transcriptional level and the prolonged protein degradation. These findings indicated that the anti-proliferation mechanism of puerarin was mainly through blocking angiontensin II-triggered downregulation of catalase expression and HO-dependent Rac1 activation.


Subject(s)
Animals , Mice , Angiotensin II , Pharmacology , Angiotensin II Type 1 Receptor Blockers , Pharmacology , Animals, Newborn , Catalase , Genetics , Metabolism , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases , Metabolism , Fibroblasts , Gene Expression Regulation , Heart , Hydrogen Peroxide , Metabolism , Pharmacology , Isoflavones , Pharmacology , Myocardium , Cell Biology , Metabolism , NADPH Oxidases , Metabolism , Neuropeptides , Metabolism , Signal Transduction , Transcription Factor AP-1 , Metabolism , Transcriptional Activation , rac1 GTP-Binding Protein , Metabolism
19.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 330-338, 2018.
Article in English | WPRIM | ID: wpr-773609

ABSTRACT

Fibroblast-like synoviocytes (FLS) play a pivotal role in Rheumatoid arthritis (RA) pathogenesis through aggressive migration and invasion. Madecassoside (Madec), a triterpenoid saponin present in Centella asiatica herbs, has a potent anti-inflammatory effect. In the present study, Madec exerted an obvious therapeutic effect in reversing the histological lesions in adjuvant-induced arthritis (AIA) rats. To recognize the anti-rheumatoid potentials of Madec, we further investigated whether Madec interfered with FLS invasion and metalloproteinase (MMP) expression. In cultures of primary FLS isolated from the AIA rats, Madec (10 and 30 μmol·L) was proven to considerably inhibit migration and invasion of FLS induced by interleukin 1β (IL-1β), but exhibiting no obvious effect on cell proliferation. Madec repressed IL-1β-triggered FLS invasion by prohibiting the expression of MMP-13. Additionally, Madec suppressed MMP-13 transcription via inhibiting the MMP-13 promoter-binding activity of NF-κB. Our results further showed that Madec down-regulated the translocation and phosphorylation of NF-κB as demonstrated by Western blotting and immunofluorescence assays. In conclusion, our results suggest that Madec exerts anti-RA activity via inhibiting the NF-κB/MMP-13 pathway.


Subject(s)
Animals , Rats , Antirheumatic Agents , Chemistry , Pharmacology , Therapeutic Uses , Arthritis, Experimental , Drug Therapy , Pathology , Cell Movement , Cell Nucleus , Metabolism , Cells, Cultured , Gene Expression Regulation, Enzymologic , Interleukin-1beta , Pharmacology , Matrix Metalloproteinase 13 , Genetics , NF-kappa B , Genetics , Metabolism , Phosphorylation , Protein Transport , Signal Transduction , Synoviocytes , Metabolism , Transcriptional Activation , Triterpenes , Chemistry , Pharmacology , Therapeutic Uses
20.
Journal of Cancer Prevention ; : 109-116, 2018.
Article in English | WPRIM | ID: wpr-740109

ABSTRACT

PIM (proviral integration site for moloney murine leukemia virus) kinase plays a key role as an oncogene in various cancers including myeloma, leukemia, prostate and breast cancers. The aberrant expression and/or activation of PIM kinases in various cancers follow an isoform-specific pattern. While PIM1 is predominantly expressed in hematological and solid tumors, PIM2 and PIM3 are largely expressed in leukemia and solid tumors, respectively. All of PIM kinases cause transcriptional activation of genes involved in cell survival and cell cycle progression in cancer. A variety of pro-tumorigenic signaling molecules, such as MYC, p21(Cip1/Waf1)/p27(kip1), CDC25, Notch1 and BAD have been identified as the downstream targets of PIM kinases. So far, three kinds of adenosine triphosphate-competitive PIM inhibitors, SGI-1776, AZD1208, and LGH447 have been in clinical trials for the treatment of acute myelogenous leukemia, prostate cancer, lymphoma, or multiple myeloma. This review sheds light on the signaling pathways involved in the PIM kinase regulation and current status of developing PIM kinase inhibitors as clinical success in combating human cancer.


Subject(s)
Humans , Adenosine , Breast , Cell Cycle , Cell Survival , Leukemia , Leukemia, Myeloid, Acute , Lymphoma , Multiple Myeloma , Oncogenes , Phosphotransferases , Prostate , Prostatic Neoplasms , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL