Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Hum Exp Toxicol ; 40(2): 369-379, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32856486

ABSTRACT

OBJECTIVE: To explore the potential function of MDM2-mediated Notch/hes1 signaling pathway in cisplatin-induced renal injury. METHODS: The acute renal injury models of mice after intraperitoneal injection of cisplatin in vivo, and the apoptotic models of human renal tubular epithelial (HK-2) cells induced by cisplatin in vitro, were conducted respectively. The renal function-related parameters were measured. The renal tissue pathological changes and apoptosis were observed by PAS staining and TUNEL staining, respectively. Cell viability and apoptosis were detected by MTT and flow cytometry. Notch/hes1 pathway-related proteins were tested by Western blotting. RESULTS: After mice injected by cisplatin, the levels of Cr, BUN, urine cystatin C, urine NGAL and urine ACR were increased and GFR was decreased with the elevation of renal tubular injury scores, the upregulation of the expressions of MDM2, N1ICD, Hes1 and Cleaved caspase-3, as well as the enhancement of cell apoptosis accompanying decreased ratio of Bcl-2/Bax. However, these cisplatin-induced renal injuries of mice have been improved by MDM2 inhibition. Besides, the declined viability, increased cytotoxicity, and enhanced apoptosis were observed in cisplatin-induced HK-2 cells, with the activated Notch/hes1 pathway. Notably, the phenomenon was alleviated in cisplatin-induced HK-2 cells transfected with MDM2 shRNA, but was severer in those co-treated with AdMDM2. Moreover, Notch1 siRNA can reverse the injury of AdMDM2 on HK-2 cells. CONCLUSION: Inhibiting MDM2 could reduce cell apoptosis through blocking Notch/hes1 signaling pathway, thus alleviating the acute renal injury caused by cisplatin.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney Diseases/chemically induced , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Receptor, Notch1/antagonists & inhibitors , Transcription Factor HES-1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blood Urea Nitrogen , Cell Line , Cell Survival/drug effects , Creatinine/analysis , Glomerular Filtration Rate/drug effects , Humans , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Male , Mice, Inbred C57BL , Proto-Oncogene Proteins c-mdm2/genetics , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Transcription Factor HES-1/metabolism
2.
Virology ; 542: 54-62, 2020 03.
Article in English | MEDLINE | ID: mdl-32056668

ABSTRACT

Intergenic region of begomovirus genome is vital to virus replication and viral gene transcription in plants. Previous studies have reported that Tomato yellow leaf curl China virus (TYLCCNV), a begomovirus, is able to accumulate and transcribe in its whitefly vector. However, the viral and host components that participate in begomovirus transcription in whiteflies are hitherto unknown. Using a yeast one-hybrid system, we identified >50 whitefly proteins that interacted with TYLCCNV intergenic region. Dual luciferase analysis revealed that one of the identified proteins, the hairy and enhancer of split homolog-1 (HES1), specifically bound to CACGTG motif in TYLCCNV intergenic region. Silencing HES1 decreased viral transcription, accumulation and transmission. These results demonstrate that the interactions between whitefly proteins and the intergenic region of TYLCCNV may contribute to viral transcription in the whitefly vector. Our findings offer valuable clues for the research and development of novel strategies to interfere with begomovirus transmission.


Subject(s)
Begomovirus/genetics , Hemiptera/metabolism , Hemiptera/virology , Insect Proteins/metabolism , Transcription Factor HES-1/metabolism , Animals , Begomovirus/pathogenicity , Begomovirus/physiology , DNA, Intergenic , Gene Knockdown Techniques , Genome, Viral , Hemiptera/genetics , Host Microbial Interactions/genetics , Insect Proteins/antagonists & inhibitors , Insect Proteins/genetics , Insect Vectors/genetics , Insect Vectors/metabolism , Insect Vectors/virology , Solanum lycopersicum/virology , Plant Diseases/virology , Protein Binding , Nicotiana/virology , Transcription Factor HES-1/antagonists & inhibitors , Transcription Factor HES-1/genetics , Transcription, Genetic , Two-Hybrid System Techniques
3.
J Cell Physiol ; 235(3): 2643-2654, 2020 03.
Article in English | MEDLINE | ID: mdl-31517391

ABSTRACT

Cancer stem cells are undifferentiated cancer cells that have self-renewal ability, a high tumorigenic activity, and a multilineage differentiation potential. MicroRNAs play a critical role in regulating gene expression during carcinogenesis. Here, we investigated the role of miR-7 and the mechanism by which it is dysregulated in gastric cancer stem cells (GCSCs). The stem cell marker, CD44, was used to sort GCSCs by fluorescence-activated cell sorting. We found that CD44 (+) cells have higher invasiveness and form more number of sphere colonies than CD44 (-) cells. Quantitative real-time polymerase chain reaction (PCR) revealed that the miR-7-5p expression was remarkably downregulated in GCSCs but was significantly increased in the methionine-deprived medium. The downregulation of miR-7-5p results from the increased DNA methylation in the promoter region using the methylation-specific PCR. Overexpression of miR-7-5p reduced the formation of colony and decreased the invasion of GCSCs through targeting Smo and Hes1 and subsequent repressing Notch and Hedgehog signaling pathways in vitro. Notably, upregulating miR-7-5p inhibited the growth of tumor in the xenograft model. Hence, these data demonstrated that miR-7-5p represses GCSC invasion through inhibition of Smo and Hes1, which provides a potential therapeutic target of gastric cancer treatment.


Subject(s)
DNA Methylation/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Smoothened Receptor/antagonists & inhibitors , Stomach Neoplasms/pathology , Transcription Factor HES-1/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Hyaluronan Receptors/analysis , Mice , Mice, Inbred BALB C , Mice, Nude , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Smoothened Receptor/genetics , Transcription Factor HES-1/genetics , Xenograft Model Antitumor Assays
4.
J Biol Chem ; 294(31): 11741-11750, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31186352

ABSTRACT

The Notch receptor is a key mediator of developmental programs and cell-fate decisions. Imbalanced Notch signaling leads to developmental disorders and cancer. To fully characterize the Notch signaling pathway and exploit it in novel therapeutic interventions, a comprehensive view on the regulation and requirements of Notch signaling is needed. Notch is regulated at different levels, ranging from ligand binding, stability to endocytosis. Using an array of different techniques, including reporter gene assays, immunocytochemistry, and ChIP-qPCR we show here, to the best of our knowledge for the first time, regulation of Notch signaling at the level of the nuclear pore. We found that the nuclear pore protein Nup214 (nucleoporin 214) and its interaction partner Nup88 negatively regulate Notch signaling in vitro and in vivo in zebrafish. In mammalian cells, loss of Nup88/214 inhibited nuclear export of recombination signal-binding protein for immunoglobulin κJ region (RBP-J), the DNA-binding component of the Notch pathway. This inhibition increased binding of RBP-J to its cognate promoter regions, resulting in increased downstream Notch signaling. Interestingly, we also found that NUP214 fusion proteins, causative for certain cases of T-cell acute lymphatic leukemia, potentially contribute to tumorigenesis via a Notch-dependent mechanism. In summary, the nuclear pore components Nup88/214 suppress Notch signaling in vitro, and in zebrafish, nuclear RBP-J levels are rate-limiting factors for Notch signaling in mammalian cells, and regulation of nucleocytoplasmic transport of RBP-J may contribute to fine-tuning Notch activity in cells.


Subject(s)
Nuclear Pore Complex Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Morpholinos/genetics , Morpholinos/metabolism , Nuclear Pore Complex Proteins/antagonists & inhibitors , Nuclear Pore Complex Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Notch/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factor HES-1/antagonists & inhibitors , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Cell Physiol Biochem ; 49(6): 2333-2347, 2018.
Article in English | MEDLINE | ID: mdl-30261495

ABSTRACT

BACKGROUND/AIMS: Little is known how miR-203 is involved in epidermal stem cells (ESCs) differentiation and scar formation. METHODS: We first used luciferase assay to determine the interaction of miR-203 with the 3'-UTR in regulation of Hes1 expression. We then used flow cytometry to analyze the effects of miR-203 expression on the differentiation of ESCs to MFB by determination of CK15 ratio and α-SMA. To confirm the results of flow cytometry analysis, we used Western blot to examine the expression of α-SMA, Collagen I (Col I), and Collagen III (Col III), as well as the expression of Notch1, Jagged1, and Hes1 in ESCs after the treatment of pre-miR-203 or anti-miR-203. Finally, we examined the effects local anti-miR-203 treatment on would closure and scar formation using a mouse skin wound model. RESULTS: Pre-miR-203 treatment increased ESCs differentiation to MFB cells, as indicated by decreased CK15 ratio and increased MFB biomarkers. This phenomenon was reversed by overexpression of Hes1 in ESCs. In addition, skin incision increased expression of miR-203 in wound tissue. Local treatment of anti-miR-203 could accelerate wound closure and reduce scar formation in vivo, which was associated with increased re-epithelialization, skin attachment regeneration, and collagen reassignment. Finally, we confirmed that anti-miR-203 treatment could inhibit ESCs differentiation in vivo via increasing Hesl expression. CONCLUSION: Taken together, our results suggested that overexpression of miR-203 in ESCs after skin wound may be a critical mechanism underlying the scar formation.


Subject(s)
Cicatrix/prevention & control , MicroRNAs/metabolism , Transcription Factor HES-1/metabolism , Wound Healing , 3' Untranslated Regions , Actins/metabolism , Animals , Antagomirs/metabolism , Cell Differentiation , Cicatrix/pathology , Epidermal Cells , Female , Hyperplasia/pathology , Keratin-15/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myofibroblasts/cytology , Myofibroblasts/metabolism , Skin/pathology , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factor HES-1/antagonists & inhibitors , Transcription Factor HES-1/genetics
6.
J Biol Chem ; 293(21): 8295-8296, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802140

ABSTRACT

Notch signaling plays critical roles in cancer progression, motivating efforts to identify inhibitors of this pathway. Perron et al. report a small-molecule screen intended to discover compounds that could interfere with the downstream transcription factor HES1. Target validation of their compounds unexpectedly identified a novel HES1-interacting protein, prohibitin 2. This highlights a new mechanism to block Notch signaling and prompting further exploration of HES1 biology.


Subject(s)
Antineoplastic Agents/pharmacology , High-Throughput Screening Assays , Neoplasms/drug therapy , Protein Multimerization/drug effects , Receptors, Notch/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Transcription Factor HES-1/antagonists & inhibitors , Humans , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
7.
J Biol Chem ; 293(21): 8285-8294, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29523683

ABSTRACT

The transcription factor Hes family basic helix-loop-helix transcription factor 1 (Hes1) is a downstream effector of Notch signaling and plays a crucial role in orchestrating developmental processes during the embryonic stage. However, its aberrant signaling in adulthood is linked to the pathogenesis of cancer. In the present study, we report the discovery of small organic molecules (JI051 and JI130) that impair the ability of Hes1 to repress transcription. Hes1 interacts with the transcriptional corepressor transducing-like enhancer of split 1 (TLE1) via an interaction domain comprising two tryptophan residues, prompting us to search a chemical library of 1,800 small molecules enriched for indole-like π-electron-rich pharmacophores for a compound that blocks Hes1-mediated transcriptional repression. This screening identified a lead compound whose extensive chemical modification to improve potency yielded JI051, which inhibited HEK293 cell proliferation with an EC50 of 0.3 µm Unexpectedly, using immunomagnetic isolation and nanoscale LC-MS/MS, we found that JI051 does not bind TLE1 but instead interacts with prohibitin 2 (PHB2), a cancer-associated protein chaperone. We also found that JI051 stabilizes PHB2's interaction with Hes1 outside the nucleus, inducing G2/M cell-cycle arrest. Of note, JI051 dose-dependently reduced cell growth of the human pancreatic cancer cell line MIA PaCa-2, and JI130 treatment significantly reduced tumor volume in a murine pancreatic tumor xenograft model. These results suggest a previously unrecognized role for PHB2 in the regulation of Hes1 and may inform potential strategies for managing pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , High-Throughput Screening Assays , Pancreatic Neoplasms/drug therapy , Repressor Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Transcription Factor HES-1/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Cycle , Cell Differentiation , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prohibitins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
PLoS One ; 13(2): e0193037, 2018.
Article in English | MEDLINE | ID: mdl-29447233

ABSTRACT

Notch signaling pathway is involved in many physiological and pathological processes. The γ-secretase inhibitor DAPT inhibits Notch signaling pathway and promotes nerve regeneration after cerebral ischemia. However, neuroprotective effects of DAPT against acute craniocerebral injury remain unclear. In this study, we established rat model of acute craniocerebral injury, and found that with the increase of damage grade, the expression of Notch and downstream protein Hes1 and Hes5 expression gradually increased. After the administration of DAPT, the expression of Notch, Hes1 and Hes5 was inhibited, apoptosis and oxidative stress decreased, neurological function and cognitive function improved. These results suggest that Notch signaling can be used as an indicator to assess the severity of post-traumatic brain injury. Notch inhibitor DAPT can reduce oxidative stress and apoptosis after acute craniocerebral injury, and is a potential drug for the treatment of acute craniocerebral injury.


Subject(s)
Craniocerebral Trauma/prevention & control , Diamines/pharmacology , Neuroprotective Agents/pharmacology , Receptors, Notch/antagonists & inhibitors , Thiazoles/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/prevention & control , Craniocerebral Trauma/pathology , Craniocerebral Trauma/physiopathology , Disease Models, Animal , Down-Regulation/drug effects , Male , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/drug effects , Transcription Factor HES-1/antagonists & inhibitors , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
9.
J Ethnopharmacol ; 208: 165-173, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28694103

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Vasculogenic mimicry (VM) has been suggested to be present in various malignant tumors and associated with tumor nutrition supply and metastasis, leading to poor prognosis of patients. Notch1 has been demonstrated to contribute to VM formation in hepathocellular carcinoma (HCC). Celastrus orbiculatus extract (COE), a mixture of 11 terpenoids isolated from the Chinese Herb Celastrus orbiculatus Vine, has been suggested to be effective in cancer treatment. AIM OF THE STUDY: In the current study, experiments were carried out to examine the effect of COE on VM formation and HCC tumor growth both in vitro and in vivo. MATERIALS AND METHODS: CCK-8 assay and Nikon live-work station were used to observe the viability of malignant cells treated with COE. Cell invasion was examined using Transwell. Matrigel was used to establish a 3-D culture condition for VM formation. Changes of mRNA and protein expression were examined by RT-PCR and Western Blot respectively. Tumor growth in vivo was monitored using in vivo fluorescence imaging device. PAS-CD34 dual staining and electron microscopy were used to observe VM formation. Immunohistochemical staining (IHC) was used to examine Notch1 and Hes1 expression in tumor tissues. RESULTS: Results showed that COE can inhibit HCC cells proliferation and invasion in a concentration-dependent manner. VM formation induced by TGF-ß1 was blocked by COE. In mouse xenograft model, COE inhibited tumor growth and VM formation. Both in vitro and in vivo studies showed that COE can downregulate expression of Notch1 and Hes1. CONCLUSION: The current results indicate that COE can inhibit VM formation and HCC tumor growth by downregulating Notch1 signaling. This study demonstrates that COE is superior to other anti-angiogenesis agents and can be considered as a promising candidate in HCC treatment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Hepatocellular/metabolism , Celastrus , Liver Neoplasms/metabolism , Plant Extracts/pharmacology , Receptor, Notch1/antagonists & inhibitors , Angiogenesis Inhibitors/therapeutic use , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Neovascularization, Pathologic/drug therapy , Plant Extracts/therapeutic use , Receptor, Notch1/metabolism , Transcription Factor HES-1/antagonists & inhibitors , Transcription Factor HES-1/metabolism , Transforming Growth Factor beta1/pharmacology
10.
Mol Ther ; 25(7): 1718-1729, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28624262

ABSTRACT

Inhibition of Notch signaling via systemic drug administration triggers conversion of white adipocytes into beige adipocytes (browning) and reduces adiposity. However, translation of this discovery into clinical practice is challenged by potential off-target side effects and lack of control over the location and temporal extent of beige adipocyte biogenesis. Here, we demonstrate an alternative approach to stimulate browning using nanoparticles (NPs) composed of FDA-approved poly(lactide-co-glycolide) that enable sustained local release of a Notch inhibitor (dibenzazepine, DBZ). These DBZ-loaded NPs support rapid cellular internalization and inhibit Notch signaling in adipocytes. Importantly, focal injection of these NPs into the inguinal white adipose tissue depots of diet-induced obese mice results in localized NP retention and browning of adipocytes, consequently improving the glucose homeostasis and attenuating body-weight gain of the treated mice. These findings offer new avenues to develop a potential therapeutic strategy for clinical treatment of obesity and its associated metabolic syndrome.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Anti-Obesity Agents/pharmacology , Dibenzazepines/pharmacology , Nanoparticles/chemistry , Obesity/drug therapy , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Anti-Obesity Agents/chemistry , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Dibenzazepines/chemistry , Drug Carriers , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gene Expression Regulation , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Lactic Acid/chemistry , Lactic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Nanoparticles/metabolism , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/agonists , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Polyglycolic Acid/chemistry , Polyglycolic Acid/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Signal Transduction , Transcription Factor HES-1/antagonists & inhibitors , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Iodothyronine Deiodinase Type II
11.
Oncotarget ; 8(11): 17873-17886, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28157712

ABSTRACT

The invasive and lethal nature of Glioblastoma multiforme (GBM) necessitates the continuous identification of molecular targets and search of efficacious therapies to inhibit GBM growth. The GBM resistance to chemotherapy and radiation it is attributed to the existence of a rare fraction of cancer stem cells (CSC) that we have identified within the tumor core and in peritumor tissue of GBM. Since Notch1 pathway is a potential therapeutic target in brain cancer, earlier we highlighted that pharmacological inhibition of Notch1 signalling by γ-secretase inhibitor-X (GSI-X), reduced cell growth of some c-CSC than to their respective p-CSC, but produced negligible effects on cell cycle distribution, apoptosis and cell invasion. In the current study, we assessed the effects of Hes1-targeted shRNA, a Notch1 gene target, specifically on GBM CSC refractory to GSI-X. Depletion of Hes1 protein induces major changes in cell morphology, cell growth rate and in the invasive ability of shHes1-CSC in response to growth factor EGF. shHes1-CSC show a decrease of the stemness marker Nestin concurrently to a marked increase of neuronal marker MAP2 compared to pLKO.1-CSC. Those effects correlated with repression of EGFR protein and modulation of Stat3 phosphorylation at Y705 and S727 residues. In the last decade Stat3 has gained attention as therapeutic target in cancer but there is not yet any approved Stat3-based glioma therapy. Herein, we report that exposure to a Stat3/5 inhibitor, induced apoptosis either in shHes1-CSC or control cells. Taken together, Hes1 seems to be a favorable target but not sufficient itself to target GBM efficaciously, therefore a possible pharmacological intervention should provide for the use of anti-Stat3/5 drugs either alone or in combination regimen.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Receptor, Notch1/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/antagonists & inhibitors , Transcription Factor HES-1/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Apoptosis/drug effects , Benzimidazoles/pharmacology , Brain Neoplasms/pathology , Carbamates/pharmacology , Cell Differentiation/genetics , Cell Proliferation/genetics , Dipeptides/pharmacology , ErbB Receptors/antagonists & inhibitors , Glioblastoma/pathology , Humans , Microtubule-Associated Proteins/metabolism , Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/metabolism , Phosphorylation , Piperidines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Transcription Factor HES-1/genetics , Tumor Suppressor Proteins/metabolism
12.
Reprod Fertil Dev ; 28(6): 700-12, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25344626

ABSTRACT

The growth of oocytes and the development of follicles require certain pathways involved in cell proliferation and survival, such as the phosphatidylinositol 3-kinase (PI3K) pathway and the Notch signalling pathway. The aim of the present study was to investigate the interaction between Notch and the PI3K/AKT signalling pathways and their effects on primordial follicle recruitment. When the Notch pathway was inhibited by L-685,458 or N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycinet-butyl ester (DAPT) in vitro, the expression of genes in the pathway and the percentage of oocytes in growing follicles decreased significantly in mouse ovaries. By 2 days postpartum, ovaries exposed to DAPT, short interference (si) RNA against Notch1 or siRNA against Hairy and enhancer of split-1 (Hes1) had significantly decreased expression of HES1, the target protein of the Notch signalling pathway. In contrast, expression of phosphatase and tensin homologue (Pten), a negative regulator of the AKT signalling pathway, was increased significantly. Co immunoprecipitation (Co-IP) revealed an interaction between HES1 and PTEN. In addition, inhibition of the Notch signalling pathway suppressed AKT phosphorylation and the proliferation of granulosa cells. In conclusion, the recruitment of primordial follicles was affected by the proliferation of granulosa cells and regulation of the interaction between the Notch and PI3K/AKT signalling pathways.


Subject(s)
Gene Expression Regulation, Developmental , Oogenesis , Ovarian Follicle/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cell Communication/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Granulosa Cells/cytology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Mice , Oogenesis/drug effects , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , PTEN Phosphohydrolase/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction/drug effects , Tissue Culture Techniques , Transcription Factor HES-1/antagonists & inhibitors , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...