Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48.465
Filter
1.
J Clin Invest ; 134(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087471

ABSTRACT

Glomerular visceral epithelial cells (i.e., podocytes) are an essential component of the tripartite glomerular filtration barrier. Healthy podocytes are terminally differentiated cells with limited replicative capacity; however, inappropriate cell cycle reentry can be induced in podocytes by various injurious stimuli. In this issue of the JCI, Yamaguchi et al. report on a somatic mosaic gain-of-function mutation in the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic α subunit (p110α, encoded by PIK3CA). The study reveals that activating mutations of p110α can drive podocyte proliferation in PIK3CA-related overgrowth syndrome (PROS). They also showed that selective, small-molecule inhibitors of p110 may be useful for the treatment of proliferative glomerulonephritis.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Phosphatidylinositol 3-Kinases , Podocytes , Humans , Podocytes/pathology , Podocytes/metabolism , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Cell Proliferation , Mice , Mutation , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/pathology , Glomerulonephritis, Membranoproliferative/immunology
2.
J Nanobiotechnology ; 22(1): 460, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090717

ABSTRACT

BACKGROUND: Nanoplastics (NPs) are emerging pollutants that pose risks to living organisms. Recent findings have unveiled the reproductive harm caused by polystyrene nanoparticles (PS-NPs) in female animals, yet the intricate mechanism remains incompletely understood. Under this research, we investigated whether sustained exposure to PS-NPs at certain concentrations in vivo can enter oocytes through the zona pellucida or through other routes that affect female reproduction. RESULTS: We show that PS-NPs disrupted ovarian functions and decreased oocyte quality, which may be a contributing factor to lower female fertility in mice. RNA sequencing of mouse ovaries illustrated that the PI3K-AKT signaling pathway emerged as the predominant environmental information processing pathway responding to PS-NPs. Western blotting results of ovaries in vivo and cells in vitro showed that PS-NPs deactivated PI3K-AKT signaling pathway by down-regulating the expression of PI3K and reducing AKT phosphorylation at the protein level, PI3K-AKT signaling pathway which was accompanied by the activation of autophagy and apoptosis and the disruption of steroidogenesis in granulosa cells. Since PS-NPs penetrate granulosa cells but not oocytes, we examined whether PS-NPs indirectly affect oocyte quality through granulosa cells using a granulosa cell-oocyte coculture system. Preincubation of granulosa cells with PS-NPs causes granulosa cell dysfunction, resulting in a decrease in the quality of the cocultured oocytes that can be reversed by the addition of 17ß-estradiol. CONCLUSIONS: This study provides findings on how PS-NPs impact ovarian function and include transcriptome sequencing analysis of ovarian tissue. The study demonstrates that PS-NPs impair oocyte quality by altering the functioning of ovarian granulosa cells. Therefore, it is necessary to focus on the research on the effects of PS-NPs on female reproduction and the related methods that may mitigate their toxicity.


Subject(s)
Granulosa Cells , Nanoparticles , Oocytes , Polystyrenes , Signal Transduction , Animals , Female , Mice , Apoptosis/drug effects , Autophagy/drug effects , Fertility/drug effects , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Nanoparticles/toxicity , Oocytes/drug effects , Oocytes/metabolism , Ovary/drug effects , Ovary/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Polystyrenes/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
3.
PLoS One ; 19(8): e0308051, 2024.
Article in English | MEDLINE | ID: mdl-39093890

ABSTRACT

Preclinical models that replicate patient tumours as closely as possible are crucial for translational cancer research. While in vitro cancer models have many advantages in assessing tumour response therapy, in vivo systems are essential to enable evaluation of the role of the tumour cell extrinsic factors, such as the tumour microenvironment and host immune system. The requirement for a functional immune system is particularly important given the current focus on immunotherapies. Therefore, we set out to generate an immunocompetent, transplantable model of colorectal cancer suitable for in vivo assessment of immune-based therapeutic approaches. Intestinal tumours from a genetically engineered mouse model, driven by expression of a Pik3ca mutation and loss of Apc, were transplanted into wild type C57BL/6 host mice and subsequently passaged to form a novel syngeneic transplant model of colorectal cancer. Our work confirms the potential to develop a panel of mouse syngeneic grafts, akin to human PDX panels, from different genetically engineered, or carcinogen-induced, mouse models. Such panels would allow the in vivo testing of new pharmaceutical and immunotherapeutic treatment approaches across a range of tumours with a variety of genetic driver mutations.


Subject(s)
Adenocarcinoma , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Mice, Inbred C57BL , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Mice , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Adenomatous Polyposis Coli Protein/genetics , Phosphatidylinositol 3-Kinases/metabolism , Transplantation, Isogeneic , Mutation , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology
4.
J Cell Mol Med ; 28(15): e18544, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39098996

ABSTRACT

Peripheral nerve defect are common clinical problem caused by trauma or other diseases, often leading to the loss of sensory and motor function in patients. Autologous nerve transplantation has been the gold standard for repairing peripheral nerve defects, but its clinical application is limited due to insufficient donor tissue. In recent years, the application of tissue engineering methods to synthesize nerve conduits for treating peripheral nerve defect has become a current research focus. This study introduces a novel approach for treating peripheral nerve defects using a tissue-engineered PLCL/SF/NGF@TA-PPy-RGD conduit. The conduit was fabricated by combining electrospun PLCL/SF with an NGF-loaded conductive TA-PPy-RGD gel. The gel, synthesized from RGD-modified tannic acid (TA) and polypyrrole (PPy), provides growth anchor points for nerve cells. In vitro results showed that this hybrid conduit could enhance PC12 cell proliferation, migration, and reduce apoptosis under oxidative stress. Furthermore, the conduit activated the PI3K/AKT signalling pathway in PC12 cells. In a rat model of sciatic nerve defect, the PLCL/SF/NGF@TA-PPy-RGD conduit significantly improved motor function, gastrocnemius muscle function, and myelin sheath axon thickness, comparable to autologous nerve transplantation. It also promoted angiogenesis around the nerve defect. This study suggests that PLCL/SF/NGF@TA-PPy-RGD conduits provide a conducive environment for nerve regeneration, offering a new strategy for peripheral nerve defect treatment, this study provided theoretical basis and new strategies for the research and treatment of peripheral nerve defect.


Subject(s)
Hydrogels , Nerve Growth Factor , Nerve Regeneration , Oligopeptides , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Sciatic Nerve , Signal Transduction , Animals , Nerve Regeneration/drug effects , Rats , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , PC12 Cells , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Oligopeptides/pharmacology , Oligopeptides/chemistry , Hydrogels/chemistry , Nerve Growth Factor/pharmacology , Nerve Growth Factor/metabolism , Rats, Sprague-Dawley , Male , Cell Proliferation/drug effects , Apoptosis/drug effects , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Polymers/chemistry
5.
Int J Nanomedicine ; 19: 7851-7870, 2024.
Article in English | MEDLINE | ID: mdl-39105098

ABSTRACT

Background: Inhibiting ROS overproduction is considered a very effective strategy for the treatment of peripheral nerve injuries, and Se has a remarkable antioxidant effect; however, since the difference between the effective concentration of Se and the toxic dose is not large, we synthesized a nanomaterial that can release Se slowly so that it can be used more effectively. Methods: Se@SiO2 NPs were synthesized using a mixture of Cu2-x Se nanocrystals, and the mechanism of action of Se@SiO2 NPs was initially explored by performing sequencing, immunofluorescence staining and Western blotting of cellular experiments. The mechanism of action of Se@SiO2 NPs was further determined by performing behavioral assays after animal experiments and by sampling the material for histological staining, immunofluorescence staining, and ELISA. The effects, mechanisms and biocompatibility of Se@SiO2 NPs for peripheral nerve regeneration were determined. Results: Porous Se@SiO2 was successfully synthesized, had good particle properties, and could release Se slowly. CCK-8 experiments revealed that the optimal experimental doses were 100 µM H2O2 and 200 µg/mL Se@SiO2, and RNA-seq revealed that porous Se@SiO2 was associated with cell proliferation, apoptosis, and the PI3K/AKT pathway. WB showed that porous Se@SiO2 could increase the expression of cell proliferation antigens (PCNA and S100) and antiapoptotic proteins (Bcl-2), decrease the expression of proapoptotic proteins (Bax), and increase the expression of antioxidative stress proteins (Nrf2, HO-1, and SOD2). EdU cell proliferation and ROS fluorescence assays showed that porous Se@SiO2 promoted cell proliferation and reduced ROS levels. The therapeutic effect of LY294002 (a PI3K/AKT pathway inhibitor) was decreased significantly and its effect was lost when it was added simultaneously with porous Se@SiO2. Animal experiments revealed that the regenerated nerve fiber density, myelin thickness, axon area, gastrocnemius muscle wet-to-weight ratio, myofiber area, sciatic nerve function index (SFI), CMAP, apoptotic cell ratio, and levels of antioxidative stress proteins and anti-inflammatory factors were increased following the administration of porous Se@SiO2. The levels of oxidative stress proteins and anti-inflammatory factors were significantly greater in the Se@SiO2 group than in the PNI group, and the effect of LY294002 was decreased significantly and was lost when it was added simultaneously with porous Se@SiO2. Conclusion: Se@SiO2 NPs are promising, economical and effective Se-releasing nanomaterials that can effectively reduce ROS production, inhibit apoptosis and promote cell proliferation after nerve injury via the PI3K/AKT pathway, ultimately accelerating nerve regeneration. These findings could be used to design new, promising drugs for the treatment of peripheral nerve injury.


Subject(s)
Cell Proliferation , Nerve Regeneration , Peripheral Nerve Injuries , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Selenium , Signal Transduction , Silicon Dioxide , Animals , Selenium/chemistry , Selenium/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Peripheral Nerve Injuries/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Nerve Regeneration/drug effects , Cell Proliferation/drug effects , Rats , Apoptosis/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Nanoparticles/chemistry , Male , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/chemistry , Rats, Sprague-Dawley , Oxidative Stress/drug effects , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Schwann Cells/drug effects , Schwann Cells/metabolism
6.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3857-3867, 2024 Jul.
Article in Chinese | MEDLINE | ID: mdl-39099359

ABSTRACT

The study investigated the protective effect and mechanism of 2-phenylethyl-beta-glucopyranoside(Phe) from Huaizhong No.1 Rehmannia glutinosa on hypoxic pulmonary hypertension(PH), aiming to provide a theoretical basis for clinical treatment of PAH. Male C57BL/6N mice were randomly divided into normal group, model group, positive drug(bosentan, 100 mg·kg~(-1)) group, and low-and high-dose Phe groups(20 and 40 mg·kg~(-1)). Except for the normal group, all other groups were continuously subjected to model induction in a 10% hypoxic environment for 5 weeks, with oral administration for 14 days starting from the 3rd week. The cardiopulmonary function, right ventricular pressure, cough and asthma index, lung injury, cell apoptosis, oxidative stress-related indicators, immune cells, and phosphatidylinositol 3-kinase(PI3K)/protein kinase B(Akt)/mammalian target of rapamycin(mTOR)/hypoxic inducible factor 1α(HIF-1α) pathway-related proteins or mRNA levels were examined. Furthermore, hypoxia-induced pulmonary arterial smooth muscle cell(PASMC) were used to further explore the mechanism of Phe intervention in PH combined with PI3K ago-nist(740Y-P). The results showed that Phe significantly improved the cardiopulmonary function of mice with PH, decreased right ventricular pressure, cough and asthma index, and lung injury, reduced cell apoptosis, oxidative stress-related indicators, and nuclear levels of phosphorylated Akt(p-Akt) and phosphorylated mTOR(p-mTOR), inhibited the expression levels of HIF-1α and PI3K mRNA and proteins, and maintained the immune cell homeostasis in mice. Further mechanistic studies revealed that Phe significantly reduced the viability and migration ability of hypoxia-induced PASMC, decreased the expression of HIF-1α and PI3K proteins and nuc-lear levels of p-Akt and p-mTOR, and this effect was blocked by 740Y-P. Therefore, it is inferred that Phe may exert anti-PH effects by alleviating the imbalance of oxidative stress and apoptosis in lung tissues and regulating immune levels, and its mechanism may be related to the regulation of the PI3K/Akt/mTOR/HIF-1α pathway. This study is expected to provide drug references and research ideas for the treatment of PH.


Subject(s)
Glucosides , Hypertension, Pulmonary , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rehmannia , TOR Serine-Threonine Kinases , Animals , Male , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Mice , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Rehmannia/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Glucosides/pharmacology , Hypoxia/drug therapy , Hypoxia/physiopathology , Hypoxia/metabolism , Signal Transduction/drug effects , Humans , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/chemistry , Apoptosis/drug effects
7.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3878-3886, 2024 Jul.
Article in Chinese | MEDLINE | ID: mdl-39099361

ABSTRACT

To investigate the mechanism by which Peitu Yifei Granules inhibit idiopathic pulmonary fibrosis(IPF) in rats, fifty specific-pathogen-free(SPF) grade male Wistar rats were randomly divided into blank group and modeling group. IPF was induced in the modeling group rats by tracheal infusion of 5 mg·kg~(-1) bleomycin(BLM) and then randomly divided into model group, pirfenidone group, and high-dose, medium-dose, and low-dose groups treated with Peitu Yifei Granules. After 24 hours of modeling, the treatment groups received intragastric administration of either Peitu Yifei Granules or pirfenidone as a positive control drug; meanwhile, the model group received an equal volume of normal saline. After 21 days of treatment administration, lung tissue samples were collected for analysis. Pathological changes in lung tissues were assessed using hematoxylin-eosin(HE) staining and Masson's trichrome staining. The expression levels of protein kinase B(Akt), mammalian target of rapamycin(mTOR), their phosphorylated forms, and sequestosome 1(p62) were determined through Western blot(WB). Fluorescent quantitative real-time polymerase chain reaction(RT-qPCR) was used to measure messenger ribonucleic acid(mRNA) expression levels of Beclin-1, microtubule-associated proteins 1A/1B light chain 3B(LC3B), and p62. Immunohistochemistry was performed to assess protein expression levels of Beclin-1 and LC3B in lung tissue samples. RESULTS:: demonstrated that lung tissue structure appeared normal without significant collagen deposition in the blank group rats. In contrast, rats from the model group exhibited thickened alveolar septa along with evident inflammatory changes and collagen deposition. Compared to the model group rats, those treated with Peitu Yifei Granules or pirfenidone showed significantly improved lung tissue structure with reduced inflammation and collagen deposition observed histologically. Furthermore, compared with those of the blank group, the expressions of p62 and its mRNA, p-Akt and p-mTOR protein in lung tissues of the model group were significantly increased, while Beclin-1, LC3B and their mRNA levels were significantly decreased. Compared with those of the model group, the expressions of p62 and its mRNA, p-Akt and p-mTOR in lung tissues of the pirfenidone group and Peitu Yifei Granules high-dose and medium-dose groups were significantly decreased, while Beclin-1, LC3B and their mRNA expressions were significantly increased. The above results indicate that Peitu Yifei Granules can improve autophagy levels in lung tissues by inhibiting the phosphoinositide 3-kinase(PI3K)/Akt/mTOR signaling pathway and delay the development of IPF disease.


Subject(s)
Autophagy , Drugs, Chinese Herbal , Idiopathic Pulmonary Fibrosis , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Wistar , Signal Transduction , TOR Serine-Threonine Kinases , Animals , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Male , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Rats , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/administration & dosage , Autophagy/drug effects , Signal Transduction/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Lung/drug effects , Lung/metabolism , Lung/pathology , Humans
8.
J Transl Med ; 22(1): 717, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095789

ABSTRACT

BACKGROUND: The global prevalence of autoimmune hepatitis (AIH) is increasing due in part to the lack of effective pharmacotherapies. Growing evidence suggests that fibroblast growth factor 4 (FGF4) is crucial for diverse aspects of liver pathophysiology. However, its role in AIH remains unknown. Therefore, we investigated whether FGF4 can regulate M1 macrophage and thereby help treat liver inflammation in AIH. METHODS: We obtained transcriptome-sequencing and clinical data for patients with AIH. Mice were injected with concanavalin A to induce experimental autoimmune hepatitis (EAH). The mechanism of action of FGF4 was examined using macrophage cell lines and bone marrow-derived macrophages. RESULTS: We observed higher expression of markers associated with M1 and M2 macrophages in patients with AIH than that in individuals without AIH. EAH mice showed greater M1-macrophage polarization than control mice. The expression of M1-macrophage markers correlated positively with FGF4 expression. The loss of hepatic Fgf4 aggravated hepatic inflammation by increasing the abundance of M1 macrophages. In contrast, the pharmacological administration of FGF4 mitigated hepatic inflammation by reducing M1-macrophage levels. The efficacy of FGF4 treatment was compromised following the in vivo clearance of macrophage populations. Mechanistically, FGF4 treatment activated the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT)-signal pathway in macrophages, which led to reduced M1 macrophages and hepatic inflammation. CONCLUSION: We identified FGF4 as a novel M1/M2 macrophage-phenotype regulator that acts through the PI3K-AKT-signaling pathway, suggesting that FGF4 may represent a novel target for treating inflammation in patients with AIH.


Subject(s)
Cell Polarity , Fibroblast Growth Factor 4 , Hepatitis, Autoimmune , Inflammation , Macrophages , Mice, Inbred C57BL , Animals , Female , Humans , Male , Mice , Cell Polarity/drug effects , Disease Models, Animal , Fibroblast Growth Factor 4/metabolism , Hepatitis, Autoimmune/pathology , Hepatitis, Autoimmune/metabolism , Inflammation/pathology , Liver/pathology , Liver/metabolism , Liver/drug effects , Macrophage Activation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
9.
Cell Commun Signal ; 22(1): 392, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118068

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a cellular process in embryonic development, wound healing, organ fibrosis, and cancer metastasis. Previously, we and others have reported that proinflammatory cytokine interleukin-1ß (IL-1ß) induces EMT. However, the exact mechanisms, especially the signal transduction pathways, underlying IL-1ß-mediated EMT are not yet completely understood. Here, we found that IL-1ß stimulation leads to the partial EMT-like phenotype in human lung epithelial A549 cells, including the gain of mesenchymal marker (vimentin) and high migratory potential, without the complete loss of epithelial marker (E-cadherin). IL-1ß-mediated partial EMT induction was repressed by PI3K inhibitor LY294002, indicating that the PI3K/AKT pathway plays a significant role in the induction. In addition, ERK1/2 inhibitor FR180204 markedly inhibited the IL-1ß-mediated partial EMT induction, demonstrating that the MEK/ERK pathway was also involved in the induction. Furthermore, we found that the activation of the PI3K/AKT and MEK/ERK pathways occurred downstream of the epidermal growth factor receptor (EGFR) pathway and the IL-1 receptor (IL-1R) pathway, respectively. Our findings suggest that the PI3K/AKT and MEK/ERK pathways coordinately promote the IL-1ß-mediated partial EMT induction. The inhibition of not one but both pathways is expected yield clinical benefits by preventing partial EMT-related disorders such as organ fibrosis and cancer metastasis.


Subject(s)
Epithelial-Mesenchymal Transition , Interleukin-1beta , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Humans , Interleukin-1beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , A549 Cells , ErbB Receptors/metabolism
10.
Int J Med Sci ; 21(10): 1814-1823, 2024.
Article in English | MEDLINE | ID: mdl-39113885

ABSTRACT

Background: BMS-1166, a PD-1/PD-L1 inhibitor, inhibits the binding of PD-L1 to PD-1, restores T cell function, and enhances tumor immune response. However, mutations in the tumor suppressor or impaired cellular signaling pathways may also lead to cellular transformation. In this study, the SW480 and SW480R cell lines were used as the model to elucidate the treatment with BMS-1166, BEZ235, and their combination. Methods: MTT and colony-formation assays were used to evaluate cell proliferation. Wound-healing assay was used to assess cell migration. Cell cycle and apoptosis were analyzed by flow cytometry. The phosphorylation level of the key kinases in the PI3K/Akt/mTOR and MAPK pathways, PD-L1, and the protein levels related to the proliferation, migration, and apoptosis were assessed using western blotting. Results: BEZ235 enhanced BMS-1166-mediated cell proliferation and migration inhibition in SW480 and SW480R cells and promoted apoptosis. Interestingly, the downregulation of the negative regulator PTEN raised the PD-L1 level, which was abolished by the inhibition of Akt. BMS-1166 promoted PI3K, Akt, mTOR, and Erk phosphorylation. However, the combination of BEZ235 with BMS-1166 suppressed the expression of PI3K, p-Akt, p-mTOR, and p-Erk in SW480 and SW480R cells compared to BMS-1166 or BEZ235 single treatment by inhibiting the binding of PD1 to PD-L1. Conclusions: PD-1 binds to PD-L1 and activates the PI3K/mTOR and MAPK pathways, which might be the molecular mechanism of acquired resistance of CRC to BMS-1166. The combination of the two drugs inhibited the phosphorylation of PI3K, Akt, and Erk in the PI3K/mTOR and MAPK pathway, i.e., BEZ235 enhanced the BMS-1166 treatment effect by blocking the PI3K/mTOR pathway and interfering with the crosstalk of the MAPK pathway. Therefore, these findings provide a theoretical basis for BMS-1166 combined with BEZ235 in the trial treatment of colorectal cancer.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Colorectal Neoplasms , Imidazoles , Phosphoinositide-3 Kinase Inhibitors , Quinolines , TOR Serine-Threonine Kinases , Humans , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Drug Synergism , Imidazoles/pharmacology , MTOR Inhibitors/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors
11.
Mol Cancer ; 23(1): 159, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107843

ABSTRACT

Circular RNA (circRNA) is thought to mediate the occurrence and development of human cancer and usually acts as a tiny RNA (miRNA) sponge to regulate downstream gene expression. However, it is not clear whether and how circACVR2A (hsa_circ_0001073) is involved in the progression of HCC. The purpose of this study is to clarify the potential role and molecular mechanism of circACVR2A in regulating the progression of hepatocellular carcinoma cells (HCC). The abundance of related proteins in circACVR2A, microRNA (miR511-5p) and PI3K-Akt signaling pathway was determined by quantitative reverse transcriptase polymerase chain reaction (RT-PCR) or Western blotting. Cell viability, invasion and apoptosis were analyzed by CCK-8, Transwell analysis and Tunel staining, respectively. The interaction between circACVR2A and microRNA was evaluated by double luciferase reporter gene assay. The results showed that circACVR2A was highly expressed in hepatocellular carcinoma cell lines. Our in vivo and in vitro data showed that circACVR2A promoted the proliferation, migration and invasion of HCC. In terms of mechanism, we found that circACVR2A can directly interact with miR511-5p and act as a miRNA sponge to regulate the expression of related proteins in PI3K-Akt signaling pathway.In HCC, circACVR2A can mediate miR-511-5p/mRNA network to activate PI3K signal pathway. This shows that the molecular regulatory network with circACVR2A as the core is a new potential target for diagnosis and treatment of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Liver Neoplasms , MicroRNAs , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , RNA, Circular , Signal Transduction , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , MicroRNAs/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , RNA, Circular/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Movement/genetics , Animals , Cell Line, Tumor , Mice , Apoptosis/genetics , Disease Progression , Male
12.
Theranostics ; 14(11): 4278-4296, 2024.
Article in English | MEDLINE | ID: mdl-39113800

ABSTRACT

Background: Ulcerative colitis (UC) is an intestinal inflammatory disease that is strongly associated with mitochondrial damage and dysfunction as well as mitophagy and lacks of satisfactory treatments. Hair follicle mesenchymal stem cell (HF-MSC)-derived exosomes owe benefit effectiveness on inflammatory therapies. Hypoxia-preconditioned HF-MSCs exhibit enhanced proliferation and migration abilities, and their exosomes exert stronger effects than normal exosomes. However, the therapeutic function of Hy-Exos in UC is unknown. Methods: The inflammation model was established with LPS-treated MODE-K cells, and the mouse UC model was established by dextran sulfate sodium (DSS) administration. The therapeutic effects of HF-MSC-derived exosomes (Exos) and hypoxia-preconditioned HF-MSC-derived exosomes (Hy-Exos) were compared in vitro and in vivo. Immunofluorescence staining and western blotting were used to explore the effects of Hy-Exos on mitochondrial function, mitochondrial fission and fusion and mitophagy. MiRNA sequencing analysis was applied to investigate the differences in components between Exos and Hy-Exos. Results: Hy-Exos had a better therapeutic effect on LPS-treated MODE-K cells and DSS-induced UC mice. Hy-Exos promoted colonic tight junction proteins expression, suppressed the oxidative stress response, and reduced UC-related inflammatory injury. Hy-Exos may exert these effects via miR-214-3p-mediated inhibition of the PI3K/AKT/mTOR signaling pathway, maintenance of mitochondrial dynamic stability, alleviation of mitochondrial dysfunction and enhancement of mitophagy. Conclusion: This study revealed a vital role for Hy-Exos in suppressing inflammatory progression in UC and suggested that miR-214-3p is a potential critical target for Hy-Exos in alleviating UC.


Subject(s)
Colitis, Ulcerative , Disease Models, Animal , Exosomes , Hair Follicle , Mesenchymal Stem Cells , Mitophagy , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/therapy , Colitis, Ulcerative/pathology , Mesenchymal Stem Cells/metabolism , Exosomes/metabolism , Mice , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Hair Follicle/metabolism , Dextran Sulfate , Male , Mitochondria/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Humans
13.
BMC Cancer ; 24(1): 1060, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39192214

ABSTRACT

BACKGROUND: It is challenging to improve the effects of chemotherapy and reduce its adverse impact on the ovaries. Our previous study suggested that the combination of galaxamide could enhance the antitumor effect of cisplatin (CIS) in HeLa cell xenograft mice. However, their potential effects on ovarian tissues remain unknown. METHODS: The Hela tumor-bearing female BALB/c mice model was established and randomly divided into three groups: control group (PBS group), CIS group (0.3 mg/kg CIS group) and galaxamide group (0.3 mg/kg CIS + 3 mg/kg galaxamide-treated group). The serum sex hormones levels, ovarian morphology, functional and molecular characterisation were determined and compared with those of the control group. RESULTS: The hormonal effects indicated premature ovarian insufficiency (POI) associated with CIS-induced tumor-bearing mice. CIS induces the apoptosis in primordial and developing follicles and subsequently increases follicular atresia, eventually leading to follicle loss. After cotreatment, galaxamide significantly increased anti-Mullerian hormone (AMH) and follicle-stimulating hormone receptor (FSHR) expression and prevented the CIS-induced PI3K pathway, which triggers follicle activation, apoptosis or atresia. CONCLUSION: These findings demonstrate that galaxamide could attenuate CIS-induced follicle loss by acting on the PI3K signaling pathway by stimulating AMH and/or FSHR and thus provides promising therapeutic options for patients with cervical cancer.


Subject(s)
Cisplatin , Phosphatidylinositol 3-Kinases , Primary Ovarian Insufficiency , Signal Transduction , Animals , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/metabolism , Female , Humans , Mice , Cisplatin/adverse effects , Signal Transduction/drug effects , HeLa Cells , Phosphatidylinositol 3-Kinases/metabolism , Mice, Inbred BALB C , Apoptosis/drug effects , Xenograft Model Antitumor Assays , Anti-Mullerian Hormone/blood , Anti-Mullerian Hormone/metabolism , Antineoplastic Agents/pharmacology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Receptors, FSH/metabolism , Receptors, FSH/genetics
14.
Proc Natl Acad Sci U S A ; 121(34): e2320257121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39150784

ABSTRACT

Lysosomal degradation pathways coordinate the clearance of superfluous and damaged cellular components. Compromised lysosomal degradation is a hallmark of many degenerative diseases, including lysosomal storage diseases (LSDs), which are caused by loss-of-function mutations within both alleles of a lysosomal hydrolase, leading to lysosomal substrate accumulation. Gaucher's disease, characterized by <15% of normal glucocerebrosidase function, is the most common LSD and is a prominent risk factor for developing Parkinson's disease. Here, we show that either of two structurally distinct small molecules that modulate PIKfyve activity, identified in a high-throughput cellular lipid droplet clearance screen, can improve glucocerebrosidase function in Gaucher patient-derived fibroblasts through an MiT/TFE transcription factor that promotes lysosomal gene translation. An integrated stress response (ISR) antagonist used in combination with a PIKfyve modulator further improves cellular glucocerebrosidase activity, likely because ISR signaling appears to also be slightly activated by treatment by either small molecule at the higher doses employed. This strategy of combining a PIKfyve modulator with an ISR inhibitor improves mutant lysosomal hydrolase function in cellular models of additional LSD.


Subject(s)
Fibroblasts , Glucosylceramidase , Lysosomal Storage Diseases , Lysosomes , Phosphatidylinositol 3-Kinases , Humans , Phosphatidylinositol 3-Kinases/metabolism , Lysosomes/metabolism , Lysosomes/drug effects , Glucosylceramidase/metabolism , Glucosylceramidase/genetics , Fibroblasts/metabolism , Fibroblasts/drug effects , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/metabolism , Gaucher Disease/drug therapy , Gaucher Disease/genetics , Gaucher Disease/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology
15.
Cells ; 13(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39195232

ABSTRACT

From birth to adulthood, the mammalian heart grows primarily through increasing cardiomyocyte (CM) size, which is known as maturational hypertrophic growth. The Hippo-YAP signaling pathway is well known for regulating heart development and regeneration, but its roles in CM maturational hypertrophy have not been clearly addressed. Vestigial-like 4 (VGLL4) is a crucial component of the Hippo-YAP pathway, and it functions as a suppressor of YAP/TAZ, the terminal transcriptional effectors of this signaling pathway. To develop an in vitro model for studying CM maturational hypertrophy, we compared the biological effects of T3 (triiodothyronine), Dex (dexamethasone), and T3/Dex in cultured neonatal rat ventricular myocytes (NRVMs). The T3/Dex combination treatment stimulated greater maturational hypertrophy than either the T3 or Dex single treatment. Using T3/Dex treatment of NRVMs as an in vitro model, we found that activation of VGLL4 suppressed CM maturational hypertrophy. In the postnatal heart, activation of VGLL4 suppressed heart growth, impaired heart function, and decreased CM size. On the molecular level, activation of VGLL4 inhibited the PI3K-AKT pathway, and disrupting VGLL4 and TEAD interaction abolished this inhibition. In conclusion, our data suggest that VGLL4 suppresses CM maturational hypertrophy by inhibiting the YAP/TAZ-TEAD complex and its downstream activation of the PI3K-AKT pathway.


Subject(s)
Cardiomegaly , Myocytes, Cardiac , Transcription Factors , Animals , Rats , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Dexamethasone/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transcription Factors/metabolism , Triiodothyronine/pharmacology , YAP-Signaling Proteins/metabolism
16.
Cells ; 13(16)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39195262

ABSTRACT

A key aspect of preeclampsia pathophysiology is the reduced invasiveness of trophoblasts and the impairment of spiral artery remodelling. Understanding the causes of altered trophoblast function is critical to understand the development of preeclampsia. B7-H4, a checkpoint molecule, controls a wide range of processes, including T-cell activation, cytokine release, and tumour progression. Our previous findings indicated that B7-H4 levels are elevated in both maternal blood and placental villous tissue during the early stages of preeclampsia. Here, we investigated the function of B7-H4 in trophoblast physiology. Recombinant B7-H4 protein was used to treat human SGHPL-5 extravillous trophoblast cells. Biological functions were investigated using MTT, wound healing, and transwell assays. Signalling pathways were analysed by immunoblotting and immunofluorescence. The functionality of B7-H4 was further confirmed by immunoblotting and immunohistochemical analysis in placental tissues from control and preeclamptic patients following therapeutic plasma exchange (TPE) or standard of care treatment. This study showed that B7-H4 inhibited the proliferation, migration, and invasion capacities of SGHPL-5 extravillous cells while promoting apoptosis by downregulating the PI3K/Akt/STAT3 signalling pathway. These results were consistently confirmed in placental tissues from preterm controls compared to early-onset preeclamptic placental tissues from patients treated with standard of care or TPE treatment. B7-H4 may play a role in the development of preeclampsia by inhibiting essential functions of extravillous trophoblast cells during placental development. One possible mechanism by which TPE improves pregnancy outcomes in preeclampsia is through the elimination of B7-H4 amongst other factors.


Subject(s)
Cell Movement , Cell Proliferation , Pre-Eclampsia , Signal Transduction , Trophoblasts , V-Set Domain-Containing T-Cell Activation Inhibitor 1 , Humans , Pre-Eclampsia/pathology , Pre-Eclampsia/metabolism , Female , Trophoblasts/metabolism , Trophoblasts/pathology , Pregnancy , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Apoptosis , Cell Line , Proto-Oncogene Proteins c-akt/metabolism , Immunologic Factors/metabolism , STAT3 Transcription Factor/metabolism , Placenta/metabolism , Adult , Phosphatidylinositol 3-Kinases/metabolism , Extravillous Trophoblasts
17.
Int J Mol Sci ; 25(16)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39201413

ABSTRACT

High-Protein Mulberry is a novel strain of mulberry. High-Protein Mulberry leaves (HPM) were the subject of this study, which aimed to investigate its efficacy and underlying mechanisms in modulating glucose and lipid metabolism. A six-week intervention using db/db mice was carried out to assess the effects of HPM on serum lipid levels, liver function, and insulin (INS) levels. qRT-PCR and Western Blotting were employed to measure key RNA and protein expressions in the PI3K/Akt and PPARα/CPT-1 pathways. UHPLC-MS and the Kjeldahl method were utilized to analyze the component content and total protein. Additionally, network pharmacology was employed to predict regulatory mechanism differences between HPM and Traditional Mulberry leaves. The results of the study revealed significant improvements in fasting blood glucose, glucose tolerance, and insulin resistance in mice treated with HPM. HPM notably reduced serum levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and INS, while increasing high-density lipoprotein cholesterol (HDL-C) levels. The treatment also effectively mitigated liver fatty lesions, inflammatory infiltration, and islet atrophy. HPM activation of the PI3K/Akt/PPARα/CPT-1 pathway suggested its pivotal role in the regulation of glucose and lipid metabolism. With its rich composition and pharmacodynamic material basis, HPM displayed a greater number of targets associated with glucose and lipid metabolism pathways, underscoring the need for further research into its potential therapeutic applications.


Subject(s)
Lipid Metabolism , Morus , PPAR alpha , Phosphatidylinositol 3-Kinases , Plant Leaves , Proto-Oncogene Proteins c-akt , Signal Transduction , Morus/chemistry , Morus/metabolism , Animals , PPAR alpha/metabolism , PPAR alpha/genetics , Mice , Lipid Metabolism/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Plant Leaves/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Male , Glucose/metabolism , Insulin Resistance , Blood Glucose/metabolism , Plant Extracts/pharmacology , Liver/metabolism , Liver/drug effects , Carnitine O-Palmitoyltransferase
18.
Int J Mol Sci ; 25(16)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39201464

ABSTRACT

Glucocorticoid receptor (GR) overexpression has been linked to increased tumour aggressiveness and treatment resistance. GR antagonists have been shown to enhance treatment effectiveness. Emerging research has investigated mifepristone, a GR antagonist, as an anticancer agent with limited research in the context of oral cancer. This study investigated the effect of mifepristone at micromolar (µM) concentrations of 1, 5, 10 and 20 on the proliferation and migration of oral cancer cells, at 24 and 48 h. Scratch and scatter assays were utilised to assess cell migration, MTT assays were used to measure cell proliferation, Western blotting was used to investigate the expression of GR and the activation of underlying Phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and mitogen-activated protein kinase (MAPK) signalling pathways, and immunofluorescence (IF) was used to determine the localisation of proteins in HaCaT (immortalised human skin keratinocytes), TYS (oral adeno squamous cell carcinoma), and SAS-H1 cells (squamous cell carcinoma of human tongue). Mifepristone resulted in a dose-dependent reduction in the proliferation of HaCaT, TYS, and SAS-H1 cells. Mifepristone at a concentration of 20 µM effectively reduced collective migration and scattering of oral cancer cells, consistent with the suppression of the PI3K-Akt and MAPK signalling pathways, and reduced expression of N-Cadherin. An elongated cell morphology was, however, observed, which may be linked to the localisation pattern of E-Cadherin in response to mifepristone. Overall, this study found that a high concentration of mifepristone was effective in the suppression of migration and proliferation of oral cancer cells via the inhibition of PI3K-Akt and MAPK signalling pathways. Further investigation is needed to define its impact on epithelial-mesenchymal transition (EMT) markers.


Subject(s)
Cell Movement , Cell Proliferation , Mifepristone , Mouth Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Mifepristone/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/drug therapy , Cell Line, Tumor , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Receptors, Glucocorticoid/metabolism , Phosphatidylinositol 3-Kinases/metabolism , MAP Kinase Signaling System/drug effects
19.
Sci Rep ; 14(1): 18321, 2024 08 07.
Article in English | MEDLINE | ID: mdl-39112614

ABSTRACT

The team aimed to explore the possible functional significance of M6A regulation in Pan-programmed cell death (PCD) among patients with bladder cancer (BLCA). In BLCA patients, the analysis was conducted on the13 patterns of programmed cell death (PCD) and the regulation of M6A. Transcriptome, genomics, and clinical data were collected from TCGA-BLCA, GEO32548, and IMvigor210. Consensus clustering analysis, functional enrichment analysis, and other prognostic tools were used to validate the Pan-PCD. Finally, in vitro experiments and transcription sequencing were performed to understand the potential influence of the PI3K pathway on Pan-PCD in BLCA patients. Diverse PCD patterns were simultaneously activated, and M6A regulators exhibited significant variability in bladder malignant tissues. The machine learning algorithm established an 8-gene M6A-related Pan-PCD signature. This signature was validated in three independent datasets, and BLCA patients with higher risk scores had worse prognosis. An unsupervised clustering approach identified activated and suppressed Pan-PCD subgroups of BLCA patients, with distinct responses to immunotherapy and drug sensitivity. In addition, the PI3K pathway was identified as a key mechanism for various forms of programmed cell death, encompassing apoptosis, pyroptosis, autophagy, and cell death dependent on lysosomes. This research revealed that the Pan-PCD model was a more promising approach for BLCA patients under M6A regulation. A new signature from M6A-related Pan-PCD was proposed, with prognostic value for survival or drug sensitivity. The PI3K pathway was a key mechanism for multiple PCDs in BLCA patients.


Subject(s)
Apoptosis , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/mortality , Prognosis , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Transcriptome , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Autophagy/genetics , Signal Transduction , Gene Expression Profiling , Male
20.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39125841

ABSTRACT

Glioblastoma multiforme is the most common and fatal brain tumor among human cancers. Ceramide (Cer) and Sphingosine 1-phosphate (S1P) have emerged as bioeffector molecules that control several biological processes involved in both cancer development and resistance. Cer acts as a tumor suppressor, inhibiting cancer progression, promoting apoptosis, enhancing immunotherapy and sensitizing cells to chemotherapy. In contrast, S1P functions as an onco-promoter molecule, increasing proliferation, survival, invasiveness, and resistance to drug-induced apoptosis. The pro-survival PI3K/Akt pathway is a recognized downstream target of S1P, and we have previously demonstrated that in glioma cells it also improves Cer transport and metabolism towards complex sphingolipids in glioma cells. Here, we first examined the possibility that, in T98G glioma cells, S1P may regulate Cer metabolism through PI3K/Akt signaling. Our research showed that exogenous S1P increases the rate of vesicular trafficking of Cer from the endoplasmic reticulum (ER) to the Golgi apparatus through S1P receptor-mediated activation of the PI3K/Akt pathway. Interestingly, the effect of S1P results in cell protection against toxicity arising from Cer accumulation in the ER, highlighting the role of S1P as a survival factor to escape from the Cer-generating cell death response.


Subject(s)
Cell Survival , Ceramides , Endoplasmic Reticulum , Glioma , Golgi Apparatus , Lysophospholipids , Proto-Oncogene Proteins c-akt , Signal Transduction , Sphingosine , Humans , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Ceramides/metabolism , Ceramides/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/drug effects , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Cell Survival/drug effects , Glioma/metabolism , Glioma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL