Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Biochem Biophys Res Commun ; 582: 1-7, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34678590

ABSTRACT

In early stage of diabetes, insulin secretion from pancreatic ß-cells is increased to deal with the elevated blood glucose. Previous studies have reported that islet-produced carbon monoxide (CO) is associated with increased glucose-stimulated insulin secretion from ß-cells. However, this compensatory mechanism by which CO may act to enhance ß-cell function remain unclear. In this study, we revealed that CO promoted intracellular calcium ([Ca2+]i) elevation and glucose-stimulated insulin secretion (GSIS) from pancreatic ß-cells in leptin receptor deficient db/db mice but not in C57 mice. The stimulatory effects of CO on ß-cell function in db/db mice was blocked by inhibition of Phospholipase C (PLC) signaling pathway. We further demonstrated that CO triggered [Ca2+]i transients and enhanced GSIS in C57 islets when ß-cells overexpressed with PLCγ1 and PLCδ1, but not PLCß1. On the other hand, reducing PLCγ1 and PLCδ1 expressions in db/db islets dramatically attenuated the stimulatory effects of CO on ß-cell function, whereas interfering PLCß1 expression had no effects on CO-induced ß-cell function enhancement. Our findings showing that CO elevated [Ca2+]i and enhanced GSIS by activating PLC signaling through PLCγ1 and PLCδ1 isoforms in db/db pancreatic ß-cells may suggest an important mechanism by which CO promotes ß-cell function to prevent hyperglycemia. Our study may also provide new insights into the therapy for type II diabetes and offer a potential target for therapeutic applications of CO.


Subject(s)
Calcium/metabolism , Carbon Monoxide/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Phospholipase C delta/genetics , Phospholipase C gamma/genetics , Animals , Cell Line , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation , Glucose/metabolism , Glucose/pharmacology , Insulin/biosynthesis , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipase C beta/antagonists & inhibitors , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Phospholipase C delta/antagonists & inhibitors , Phospholipase C delta/metabolism , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Signal Transduction
2.
FEBS Open Bio ; 11(2): 435-445, 2021 02.
Article in English | MEDLINE | ID: mdl-33326693

ABSTRACT

Osteoarthritis (OA) is an age-related, chronic degenerative disease. With the increasing median age of the population, this disease has become an important public health problem. New, disease-modifying therapies are needed. A potential novel molecular target is phospholipase Cγ1 (PLCγ1), a critical enzyme with important functions including calcium signaling regulation and cell proliferation. In rat chondrocytes treated with IL-1ß (20 ng·mL-1 for 36 h), inhibition of PLCγ1 with U73122 (2 µm for 12 h) increased levels and expression of the cartilage matrix components Collagen2 and Aggrecan. This beneficial effect of PLCγ1 inhibition was counteracted by increased chondrocyte apoptosis and necroptosis, increased cell death, and increase levels of ROS, all potentially negative for OA. Combined treatment of IL-1ß + U73122-treated chondrocytes with inhibitors of apoptosis (Z-VAD, 10 µm) and necroptosis (Nec-1, 30 µm) enhanced the increases in levels and expression of Collagen2 and Aggrecan, and prevented the increases in cell death and ROS levels. These results suggest that PLCγ1 inhibition may be a viable approach for an OA therapy, if combined with targeted inhibition of chondrocyte apoptosis and necroptosis.


Subject(s)
Interleukin-1beta/immunology , Osteoarthritis/drug therapy , Phospholipase C gamma/antagonists & inhibitors , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/immunology , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/immunology , Chondrocytes/pathology , Disease Models, Animal , Drug Therapy, Combination/methods , Estrenes/pharmacology , Estrenes/therapeutic use , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Interleukin-1beta/metabolism , Necroptosis/drug effects , Necroptosis/immunology , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Osteoarthritis/immunology , Osteoarthritis/pathology , Phospholipase C gamma/metabolism , Primary Cell Culture , Pyrrolidinones/pharmacology , Pyrrolidinones/therapeutic use , Rats , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
3.
J Cell Mol Med ; 25(3): 1531-1545, 2021 02.
Article in English | MEDLINE | ID: mdl-33372388

ABSTRACT

Previous studies identified the involvement of phosphoinositide-specific phospholipase C (PLC) γ1 in some events of chondrocytes. This study aims to investigate whether and how PLCγ1 modulates autophagy to execute its role in osteoarthritis (OA) progression. Rat normal or human OA chondrocytes were pretreated with IL-1ß for mimicking or sustaining OA pathological condition. Using Western blotting, immunoprecipitation, qPCR, immunofluorescence and Dimethylmethylene blue assays, and ELISA and transmission electron microscope techniques, we found that PLCγ1 inhibitor U73122 enhanced Collagen II, Aggrecan and GAG levels, accompanied with increased LC3B-II/I ratio and decreased P62 expression level, whereas autophagy inhibitor Chloroquine partially diminished its effect. Meanwhile, U73122 dissociated Beclin1 from Beclin1-IP3R-Bcl-2 complex and blocked mTOR/ULK1 axis, in which the crosstalk between PLCγ1, AMPK, Erk and Akt were involved. Additionally, by haematoxylin and eosin, Safranin O/Fast green, and immunohistochemistry staining, we observed that intra-articular injection of Ad-shPLCγ1-1/2 significantly enhanced Collagen and Aggrecan levels, accompanied with increased LC3B and decreased P62 levels in a rat OA model induced by anterior cruciate ligament transection and medial meniscus resection. Consequently, PLCγ1 inhibition-driven autophagy conferred cartilage protection against OA through promoting ECM synthesis in OA chondrocytes in vivo and in vitro, involving the crosstalk between PLCγ1, AMPK, Erk and Akt.


Subject(s)
Autophagy , Chondrocytes/drug effects , Chondrocytes/metabolism , Interleukin-1beta/pharmacology , Osteoarthritis/etiology , Osteoarthritis/metabolism , Phospholipase C gamma/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Aged , Aged, 80 and over , Animals , Autophagy/drug effects , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Disease Susceptibility , Estrenes/pharmacology , Female , Humans , Male , Middle Aged , Osteoarthritis/pathology , Proto-Oncogene Proteins c-akt/metabolism , Pyrrolidinones/pharmacology , Rats , TOR Serine-Threonine Kinases/metabolism
4.
Oxid Med Cell Longev ; 2020: 6801587, 2020.
Article in English | MEDLINE | ID: mdl-33101590

ABSTRACT

Oxidative stress (OS) and neuronal apoptosis are major pathological processes after hypoxic-ischemic encephalopathy (HIE). Colony stimulating factor 1 (CSF1), binding to CSF1 receptor (CSF1R), has been shown to reduce neuronal loss after hypoxic-ischemia- (HI-) induced brain injury. In the present study, we hypothesized that CSF1 could alleviate OS-induced neuronal degeneration and apoptosis through the CSF1R/PLCG2/PKA/UCP2 signaling pathway in a rat model of HI. A total of 127 ten-day old Sprague Dawley rat pups were used. HI was induced by right common carotid artery ligation with subsequent exposure to hypoxia for 2.5 h. Exogenous recombinant human CSF1 (rh-CSF1) was administered intranasally at 1 h and 24 h after HI. The CSF1R inhibitor, BLZ945, or phospholipase C-gamma 2 (PLCG2) inhibitor, U73122, was injected intraperitoneally at 1 h before HI induction. Brain infarct volume measurement, cliff avoidance test, righting reflex test, double immunofluorescence staining, western blot assessment, 8-OHdG and MitoSOX staining, Fluoro-Jade C staining, and TUNEL staining were used. Our results indicated that the expressions of endogenous CSF1, CSF1R, p-CSF1R, p-PLCG2, p-PKA, and uncoupling protein2 (UCP2) were increased after HI. CSF1 and CSF1R were expressed in neurons and astrocytes. Rh-CSF1 treatment significantly attenuated neurological deficits, infarct volume, OS, neuronal apoptosis, and degeneration at 48 h after HI. Moreover, activation of CSF1R by rh-CSF1 significantly increased the brain tissue expressions of p-PLCG2, p-PKA, UCP2, and Bcl2/Bax ratio, but reduced the expression of cleaved caspase-3. The neuroprotective effects of rh-CSF1 were abolished by BLZ945 or U73122. These results suggested that rh-CSF1 treatment attenuated OS-induced neuronal degeneration and apoptosis after HI, at least in part, through the CSF1R/PLCG2/PKA/UCP2 signaling pathway. Rh-CSF1 may serve as therapeutic strategy against brain damage in patients with HIE.


Subject(s)
Apoptosis/drug effects , Hypoxia-Ischemia, Brain/pathology , Macrophage Colony-Stimulating Factor/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Benzothiazoles/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Humans , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Neurons/cytology , Neurons/metabolism , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Picolinic Acids/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Uncoupling Protein 2/metabolism , Up-Regulation/drug effects
5.
Cancer Res ; 80(24): 5491-5501, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33115803

ABSTRACT

Chromophobe renal cell carcinoma (chRCC) accounts for approximately 5% of all renal cancers and around 30% of chRCC cases have mutations in TP53. chRCC is poorly supported by microvessels and has markably lower glucose uptake than clear cell RCC and papillary RCC. Currently, the metabolic status and mechanisms by which this tumor adapts to nutrient-poor microenvironments remain to be investigated. In this study, we performed proteome and metabolome profiling of chRCC tumors and adjacent kidney tissues and identified major metabolic alterations in chRCC tumors, including the classical Warburg effect, the downregulation of gluconeogenesis and amino acid metabolism, and the upregulation of protein degradation and endocytosis. chRCC cells depended on extracellular macromolecules as an amino acid source by activating endocytosis to sustain cell proliferation and survival. Inhibition of the phospholipase C gamma 2 (PLCG2)/inositol 1,4,5-trisphosphate (IP3)/Ca2+/protein kinase C (PKC) pathway significantly impaired the activation of endocytosis for amino acid uptakes into chRCC cells. In chRCC, whole-exome sequencing revealed that TP53 mutations were not related to expression of PLCG2 and activation of endocytosis. Our study provides novel perspectives on metabolic rewiring in chRCC and identifies the PLCG2/IP3/Ca2+/PKC axis as a potential therapeutic target in patients with chRCC. SIGNIFICANCE: This study reveals macropinocytosis as an important process utilized by chRCC to gain extracellular nutrients in a p53-independent manner.


Subject(s)
Amino Acids/metabolism , Carcinoma, Renal Cell/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Endocytosis/drug effects , Kidney Neoplasms/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Boron Compounds/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Estrenes/pharmacology , Gluconeogenesis , Humans , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate/metabolism , Kidney Neoplasms/pathology , Maleimides/pharmacology , Metabolome , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proteome , Pyrrolidinones/pharmacology
6.
Cancer Res ; 80(22): 5051-5062, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32973083

ABSTRACT

Small cell lung cancer (SCLC) remains a recalcitrant disease where limited therapeutic options have not improved overall survival, and approved targeted therapies are lacking. Amplification of the tyrosine kinase receptor FGFR1 (fibroblast growth factor receptor 1) is one of the few actionable alterations found in the SCLC genome. However, efforts to develop targeted therapies for FGFR1-amplified SCLC are hindered by critical gaps in knowledge around the molecular origins and mediators of FGFR1-driven signaling as well as the physiologic impact of targeting FGFR1. Here we show that increased FGFR1 promotes tumorigenic progression in precancerous neuroendocrine cells and is required for SCLC development in vivo. Notably, Fgfr1 knockout suppressed tumor development in a mouse model lacking the retinoblastoma-like protein 2 (Rbl2) tumor suppressor gene but did not affect a model with wild-type Rbl2. In support of a functional interaction between these two genes, loss of RBL2 induced FGFR1 expression and restoration of RBL2 repressed it, suggesting a novel role for RBL2 as a regulator of FGFR1 in SCLC. Additionally, FGFR1 activated phospholipase C gamma 1 (PLCG1), whereas chemical inhibition of PLCG1 suppressed SCLC growth, implicating PLCG1 as an effector of FGFR1 signaling in SCLC. Collectively, this study uncovers mechanisms underlying FGFR1-driven SCLC that involve RBL2 upstream and PLCG1 downstream, thus providing potential biomarkers for anti-FGFR1 therapy. SIGNIFICANCE: This study identifies RBL2 and PLCG1 as critical components of amplified FGFR1 signaling in SCLC, thus representing potential targets for biomarker analysis and therapeutic development in this disease.


Subject(s)
Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , Phospholipase C gamma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Retinoblastoma-Like Protein p130/metabolism , Small Cell Lung Carcinoma/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Disease Progression , Female , Gene Deletion , Genes, Regulator , Genes, Retinoblastoma , Humans , Lung Neoplasms/etiology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Phospholipase C gamma/antagonists & inhibitors , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Retinoblastoma-Like Protein p130/genetics , Small Cell Lung Carcinoma/etiology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism
7.
Biochem Biophys Res Commun ; 532(2): 292-299, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32868075

ABSTRACT

Phosphatidylcholine-specific phospholipase Cγ1 (PLCγ1) is involved in regulating cell metabolism. However, little is known how PLCγ1 directs BMSC differentiation. Here, we investigated the role of PLCγ1 in rat BMSC differentiation into osteoblasts and chondrocytes. The results of Alizarin red and Alcian blue staining showed that PLCγ1 inhibitor U73122 significantly enhanced the mineralization capacity and proteoglycan deposition of BMSCs. The results of qPCR technique and Western blot analysis showed that long-term treatment of U73122 enhanced COL1A1 and OPG mRNA levels and Collagen 1A1, BMP2, and p-Smad1/5/9 protein levels and that short-term treatment of U73122 enhanced COL2A1 and SOX9 mRNA levels and Collagen 2, SOX9, Aggrecan, TGF-ß3, and p-Smad2/3 protein levels. Decreased p-mTOR and p-P38 contributed to enhanced osteogenic potentials of BMSCs and increased p-P38 contributed to enhanced chondrogenic potentials of BMSCs. The scaffold transplantation with U73122+BMSC was more efficacious than BMSC alone for osteochondral defect repair in a rat model. Therefore, suppressing PLCγ1 could improve the capacity to effectively use BMSCs for cell therapy of osteochondral defect.


Subject(s)
Chondrogenesis/physiology , Mesenchymal Stem Cells/cytology , Osteogenesis/physiology , Phospholipase C gamma/antagonists & inhibitors , Alginates , Animals , Cell Differentiation , Cells, Cultured , Chondrogenesis/drug effects , Estrenes/pharmacology , Hydrogels/administration & dosage , Hydrogels/chemistry , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Osteochondrosis/physiopathology , Osteochondrosis/therapy , Osteogenesis/drug effects , Patella/diagnostic imaging , Patella/pathology , Patella/surgery , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Pyrrolidinones/pharmacology , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Viruses ; 12(9)2020 08 24.
Article in English | MEDLINE | ID: mdl-32846937

ABSTRACT

Accumulating studies have shown that the epidermal growth factor receptor (EGFR) signaling pathway plays an essential role in mediating cellular entry of numerous viruses. In this study, we report that bovine herpesvirus 1 (BoHV-1) productive infection in both the human lung carcinoma cell line A549 and bovine kidney (MDBK) cells leads to activation of EGFR, as demonstrated by the increased phosphorylation of EGFR at Tyr1068 (Y1068), which in turn plays important roles in virus infection. A time-of-addition assay supported that virus replication at post-entry stages was affected by the EGFR specific inhibitor Gefitinib. Interestingly, both phospholipase C-γ1 (PLC-γ1) and Akt, canonical downstream effectors of EGFR, were activated following virus infection in A549 cells, while Gefitinib could inhibit the activation of PLC-γ1 but not Akt. In addition, virus titers in A549 cells was inhibited by chemical inhibition of PLC-γ1, but not by the inhibition of Akt. However, the Akt specific inhibitor Ly294002 could significantly reduce the virus titer in MDBK cells. Taken together, our data suggest that PLC-γ1 is stimulated in part through EGFR for efficient replication in A549 cells, whereas Akt can be stimulated by virus infection independent of EGFR, and is not essential for virus productive infection, indicating that Akt modulates BoHV-1 replication in a cell type-dependent manner. This study provides novel insights on how BoHV-1 infection activates EGFR signaling transduction to facilitate virus replication.


Subject(s)
ErbB Receptors/metabolism , Herpesvirus 1, Bovine/physiology , Signal Transduction , Virus Replication , A549 Cells , Animals , Cattle , Cell Line , ErbB Receptors/antagonists & inhibitors , Host-Pathogen Interactions , Humans , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Viral Load/drug effects , Virus Replication/drug effects
9.
J Leukoc Biol ; 108(3): 859-866, 2020 09.
Article in English | MEDLINE | ID: mdl-32480423

ABSTRACT

Mast cell activation through the high-affinity IgE receptor (FcεRI) plays a central role in allergic reactions. FcεRI-mediated activation triggers multiple signaling pathways leading to degranulation and synthesis of different inflammatory mediators. IgE-mediated mast cell activation can be modulated by different molecules, including several drugs. Herein, we investigated the immunomodulatory activity of the histone deacetylase inhibitor valproic acid (VPA) on IgE-mediated mast cell activation. To this end, bone marrow-derived mast cells (BMMC) were sensitized with IgE and treated with VPA followed by FcεRI cross-linking. The results indicated that VPA reduced mast cell IgE-dependent degranulation and cytokine release. VPA also induced a significant reduction in the cell surface expression of FcεRI and CD117, but not other mast cell surface molecules. Interestingly, VPA treatment inhibited the phosphorylation of PLCγ2, a key signaling molecule involved in IgE-mediated degranulation and cytokine secretion. However, VPA did not affect the phosphorylation of other key components of the FcεRI signaling pathway, such as Syk, Akt, ERK1/2, or p38. Altogether, our data demonstrate that VPA affects PLCγ2 phosphorylation, which in turn decreases IgE-mediated mast cell activation. These results suggest that VPA might be a key modulator of allergic reactions and might be a promising therapeutic candidate.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Immunoglobulin E/immunology , Mast Cells/drug effects , Phospholipase C gamma/antagonists & inhibitors , Receptors, IgE/drug effects , Valproic Acid/pharmacology , Animals , Cell Degranulation/drug effects , Down-Regulation/drug effects , Interleukin-13/metabolism , Interleukin-6/metabolism , Mast Cells/cytology , Mice , Phospholipase C gamma/physiology , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Int J Biol Sci ; 16(8): 1427-1440, 2020.
Article in English | MEDLINE | ID: mdl-32210730

ABSTRACT

Our previous studies indicated that phosphoinositide specific phospholipase Cγ1 (PLCγ1) was involved in autophagy induction in colon and hepatic carcinoma cells. However, whether and how PLCγ1 regulation in human lung adenocarcinoma is linked to autophagy remains unclear. Here, we assessed the protein expression of PLCγ1 in human lung adenocarcinoma tissue using immunohistochemistry assay and the relationship between PLCG1 and autophagy in The Cancer Genome Atlas Network (TCGA) using Spearman correlation analysis and GSEA software. Furthermore, the interaction between PLCγ1 and autophagy-related signal molecules was investigated in human lung adenocarcinoma A549 cells treated with different inhibitors or transduction with lentivirus-mediated PLCγ1 gene short-hairpin RNA (shRNA) vectors using MTT, clonogenicity, Transwell migration, RT-PCR, Caspase-3, mitochondrial transmembrane potential, and western blotting assays, as well as transmission electron microscope technique. Additionally, the effect of shRNA/PLCγ1 alone or combined with autophagic activator Lithium Chloride (LiCl) on tumor growth and metastasis was measured using immunohistochemistry and assays in A549 xenograft nude mouse model. The results showed that increased PLCγ1 expression occurred frequently in human lung adenocarcinoma tissue with higher grades of T in TNM staging classification. PLCγ1 significantly enriched in autophagic process and regulation, which negatively regulating autophagy was enriched in higher expression of PLCγ1. PLCγ1 inhibition partially reduced cell proliferation and migration of A549 cells, with an increased autophagic flux involving alterations of AMPKα, mTOR, and ERK levels. However, PLCγ1 inhibition-driven autophagy led to cell death without depending on Caspase-3 and RIP1. Additionally, the abrogation of PLCγ1 signaling by shRNA and combination with autophagic activator LiCl could efficaciously suppress tumor growth and metastasis in A549 xenograft nude mice, in combination with a decrease in P62 level. These findings collectively suggest that reduction of cell proliferation and migration by PLCγ1 inhibition could be partially attributed to PLCγ1 inhibition-driven autophagic cell death (ACD). It highlights the potential role of a combination between targeting PLCγ1 and autophagy pathway in anti-tumor therapy, which may be an efficacious new strategy to overcome the autophagy addition of tumor and acquired resistance to current therapy.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Autophagy/drug effects , Lung Neoplasms/drug therapy , Phosphatidylinositols/metabolism , Phospholipase C gamma/antagonists & inhibitors , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Autophagic Cell Death , Cell Line, Tumor , Cell Proliferation , DNA Damage , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genome, Human , HEK293 Cells , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Phospholipase C gamma/metabolism , RNA, Small Interfering/metabolism
11.
Arch Pharm Res ; 43(2): 224-232, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32016828

ABSTRACT

Nepetin derived from the flowers of Inula japonica, Inulae flos, has been reported to exert several biological activities, including anti-inflammatory responses. In this study, we evaluated the anti-allergic property of nepetin with its molecular mechanisms in bone marrow-derived mast cells (BMMC) and mice. In this in vitro study, we investigated the inhibitory effects of nepetin on degranulation and generation of leukotriene C4 (LTC4) and prostaglandin D2 (PGD2) in IgE/antigen (Ag)-stimulated BMMC. The effect of nepetin on passive cutaneous anaphylaxis (PCA) reaction was also studied in mice. Nepetin reduced degranulation and LTC4 generation in BMMC. The IgE/Ag-mediated signaling pathway demonstrated that nepetin suppressed intracellular Ca2+ level and activation of PLCγ1 and cPLA2. However, MAPKs were not affected by nepetin in BMMC. In addition, nepetin treatment reduced PGD2 production and suppressed cyclooxygenase-2 protein expression via the inhibition of the Akt and nuclear factor-κB signaling pathways. With respect to the local allergic response in vivo, oral administration of nepetin suppressed mast cell-dependent PCA reaction in a dose-dependent manner. The results of this study suggest that nepetin might have an anti-allergic potential related to mast cell-mediated inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biological Products/pharmacology , Flavones/pharmacology , Inula/chemistry , Leukotriene C4/antagonists & inhibitors , Prostaglandin D2/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Biological Products/chemistry , Biological Products/isolation & purification , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flavones/chemistry , Flavones/isolation & purification , Leukotriene C4/metabolism , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Molecular Structure , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Prostaglandin D2/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
12.
J Cell Mol Med ; 23(12): 8019-8024, 2019 12.
Article in English | MEDLINE | ID: mdl-31638328

ABSTRACT

To investigate the efficacy of sacral nerve stimulation (SNS) on nerve growth factor (NGF) mediated visceral sensitivity in normal rat and visceral hypersensitivity model rats. 120 male newborn rats were randomly divided into 6 groups: group A was normal model group; group B ~ F were all sensitized with acetic acid enema and grouped again. Group c2 was given NGF antagonist, d2 group was given NGF agonist, e2 group was given PI3K inhibitor, and f2 group was given PLC-γ inhibitor. After treatment, the expression of NGF, TrKA, PI3K, AKT, PLC-γ, NF-κB, TRPV1, pTRPV1 and intracellular Ca2+ content were detected. The expression of protein TRPV1 and pTRPV1 was increased, and Ca2+ was increased in the visceral hypersensitive group. NGF, TrKA in NGF antagonist group, PI3K, AKT, NF-κB in PI3K inhibitor group, PLC-γ in PLC-γ inhibitor group were all almost not expressed. The relative expression of NGF, TrKA, PI3K, AKT, PLC-γ and NF-κB in NGF antagonist group was lower than that in visceral hypersensitivity group and NGF activator group (P < .01). The relative expression of NGF, TrKA, PI3K and AKT mRNA in NGF antagonist group was lower than that in the normal model group (P < .01). There was no significant difference in the relative expression of PLC-γ and NF-κB mRNA (P > .05). The expression level of MAPK, ERK1 and ERK2 in visceral hypersensitivity group was higher than that in PI3K inhibitor group and PLC-γ inhibitor group. The normal group Ca2+ curve was flat, and the NGF agonist group had the highest Ca2+ curve peak. Calcium concentration in visceral hypersensitivity group was higher than that in PI3K inhibitor group and that in PLC-γ inhibitor group was higher than that in NGF antagonist group. The binding of TrkA receptor to NGF activates the MAPK/ERK pathway, the PI3K/Akt pathway and the PLC-γ pathway, causing changes in the fluidity of intracellular and extracellular Ca2+ , resulting in increased sensitivity of visceral tissues and organs.


Subject(s)
Colon/metabolism , Ganglia, Spinal/metabolism , Nerve Growth Factor/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Calcium/metabolism , Colon/cytology , Colon/drug effects , MAP Kinase Signaling System/genetics , Male , NF-kappa B/metabolism , Nerve Growth Factor/agonists , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factor/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, trkA/genetics , Receptor, trkA/metabolism , Sacrum/innervation , Signal Transduction/drug effects , Signal Transduction/genetics , TRPV Cation Channels/metabolism
13.
J Biol Chem ; 294(45): 16650-16662, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31537645

ABSTRACT

Calcium (Ca2+) signaling within the cell nucleus regulates specific cellular events such as gene transcription and cell proliferation. Nuclear and cytosolic Ca2+ levels can be independently regulated, and nuclear translocation of receptor tyrosine kinases (RTKs) is one way to locally activate signaling cascades within the nucleus. Nuclear RTKs, including the epidermal growth factor receptor (EGFR), are important for processes such as transcriptional regulation, DNA-damage repair, and cancer therapy resistance. RTKs can hydrolyze phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) within the nucleus, leading to Ca2+ release from the nucleoplasmic reticulum by inositol 1,4,5-trisphosphate receptors. PI(4,5)P2 hydrolysis is mediated by phospholipase C (PLC). However, it is unknown which nuclear PLC isoform is triggered by EGFR. Here, using subcellular fractionation, immunoblotting and fluorescence, siRNA-based gene knockdowns, and FRET-based biosensor reporter assays, we investigated the role of PLCδ4 in epidermal growth factor (EGF)-induced nuclear Ca2+ signaling and downstream events. We found that EGF-induced Ca2+ signals are inhibited when translocation of EGFR is impaired. Nuclear Ca2+ signals also were reduced by selectively buffering inositol 1,4,5-trisphosphate (InsP3) within the nucleus. EGF induced hydrolysis of nuclear PI(4,5)P2 by the intranuclear PLCδ4, rather than by PLCγ1. Moreover, protein kinase C, a downstream target of EGF, was active in the nucleus of stimulated cells. Furthermore, PLCδ4 and InsP3 modulated cell cycle progression by regulating the expression of cyclins A and B1. These results provide evidence that EGF-induced nuclear signaling is mediated by nuclear PLCδ4 and suggest new therapeutic targets to modulate the proliferative effects of this growth factor.


Subject(s)
Calcium Signaling/drug effects , Cell Nucleus/metabolism , Epidermal Growth Factor/pharmacology , Phospholipase C delta/metabolism , Cell Line , Cell Proliferation/drug effects , Clathrin Heavy Chains/antagonists & inhibitors , Clathrin Heavy Chains/genetics , Clathrin Heavy Chains/metabolism , Cyclin A/metabolism , Cyclin B1/metabolism , ErbB Receptors/metabolism , Humans , Hydrolysis , Inositol 1,4,5-Trisphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C delta/antagonists & inhibitors , Phospholipase C delta/genetics , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Protein Kinase C/metabolism , RNA Interference , RNA, Small Interfering/metabolism
14.
Int J Mol Sci ; 20(19)2019 Sep 23.
Article in English | MEDLINE | ID: mdl-31548507

ABSTRACT

The enzyme phospholipase C gamma 1 (PLCγ1) has been identified as a potential drug target of interest for various pathological conditions such as immune disorders, systemic lupus erythematosus, and cancers. Targeting its SH3 domain has been recognized as an efficient pharmacological approach for drug discovery against PLCγ1. Therefore, for the first time, a combination of various biophysical methods has been employed to shed light on the atomistic interactions between PLCγ1 and its known binding partners. Indeed, molecular modeling of PLCγ1 with SLP76 peptide and with previously reported inhibitors (ritonavir, anethole, daunorubicin, diflunisal, and rosiglitazone) facilitated the identification of the common critical residues (Gln805, Arg806, Asp808, Glu809, Asp825, Gly827, and Trp828) as well as the quantification of their interaction through binding energies calculations. These features are in agreement with previous experimental data. Such an in depth biophysical analysis of each complex provides an opportunity to identify new inhibitors through pharmacophore mapping, molecular docking and MD simulations. From such a systematic procedure, a total of seven compounds emerged as promising inhibitors, all characterized by a strong binding with PLCγ1 and a comparable or higher binding affinity to ritonavir (∆Gbind < -25 kcal/mol), one of the most potent inhibitor reported till now.


Subject(s)
Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Peptides/chemistry , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/chemistry , Enzyme Inhibitors/metabolism , Humans , Peptides/metabolism , Phospholipase C gamma/metabolism , Protein Binding , Protein Domains
15.
Life Sci ; 222: 29-35, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30826495

ABSTRACT

Recently, several plexins and semaphorins have been associated with osteoclastogenesis, a vital process for bone remodeling. Plexin-A2 is implicated in bone homeostasis, however, whether it plays a role in osteoclastogenesis and the underlying mechanism remain unknown. We show that plexin-A2 expression is upregulated during RANKL-induced osteoclastogenesis. In addition, the soluble Sema6A fused with IgG1 Fc region (Fc-Sema6A) interacts with plexin-A2 from cell lysates of osteoclasts, suggesting that plexin-A2 acts as a receptor of Sema6A in osteoclasts. Moreover, Sema6A treatment stimulates RANKL-induced osteoclastogenesis, and this effect is abolished when plexin-A2 is neutralized, which illustrates an indispensable role of plexin-A2 in mediating Sema6A effect on osteoclastogenesis. Mechanistically, Sema6A-plexin-A2 axis enhances RANKL-induced activation of PLCγ as well as downstream target NFATc1, one master transcriptional factor of osteoclastogenesis. Lastly, inhibition of PLCγ by pharmacological inhibitor U73122 abrogates Sema6A-stimulated NFATc1 activation and RANKL-induced osteoclastogenesis, thus demonstrating that the PLCγ-mediated NFATc1 activation accounts for the promotive role of Sema6A-plexin-A2 axis in RANKL-induced osteoclastogenesis. Taken together, this study uncovers a novel role of Sema6A and plexin-A2 in osteoclastogenesis, and also offers them as possible therapeutic targets in the intervention of osteolytic diseases.


Subject(s)
NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Osteogenesis/physiology , Phospholipase C gamma/metabolism , RANK Ligand/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Estrenes/pharmacology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/pharmacology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Phosphodiesterase Inhibitors/pharmacology , Phospholipase C gamma/antagonists & inhibitors , Pyrrolidinones/pharmacology , Semaphorins/pharmacology
16.
Cell Chem Biol ; 26(3): 400-410.e3, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30595532

ABSTRACT

Nerve growth factor elicits signaling outcomes by interacting with both its high-affinity receptor, TrkA, and its low-affinity receptor, p75NTR. Although these two receptors can regulate distinct cellular outcomes, they both activate the extracellular-signal-regulated kinase pathway upon nerve growth factor stimulation. To delineate TrkA subcircuits in PC12 cell differentiation, we developed an optogenetic system whereby light was used to specifically activate TrkA signaling in the absence of nerve growth factor. By using tyrosine mutants of the optogenetic TrkA in combination with pathway-specific pharmacological inhibition, we find that Y490 and Y785 each contributes to PC12 cell differentiation through the extracellular-signal-regulated kinase pathway in an additive manner. Optogenetic activation of TrkA eliminates the confounding effect of p75NTR and other potential off-target effects of the ligand. This approach can be generalized for the mechanistic study of other receptor-mediated signaling pathways.


Subject(s)
Cell Differentiation , Optogenetics , Protein-Tyrosine Kinases/metabolism , Animals , Cell Differentiation/radiation effects , Cell Membrane/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Light , Nerve Growth Factor/metabolism , PC12 Cells , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/genetics , Rats , Receptor, trkA/metabolism , Signal Transduction/radiation effects
17.
Arterioscler Thromb Vasc Biol ; 39(3): 496-512, 2019 03.
Article in English | MEDLINE | ID: mdl-30626204

ABSTRACT

Objective- Venous malformations (VMs) arise from developmental defects of the vasculature and are characterized by massively enlarged and tortuous venous channels. VMs grow commensurately leading to deformity, obstruction of vital structures, bleeding, and pain. Most VMs are associated with the activating mutation L914F in the endothelial cell (EC) tyrosine kinase receptor TIE2. Therapeutic options for VM are limited and ineffective while therapy with the mammalian target of rapamycin inhibitor rapamycin shows moderate efficacy. Here, we investigated novel therapeutic targets promoting VM regression. Approach and Results- We performed an unbiased screen of Food and Drug Administration-approved drugs in human umbilical vein ECs expressing the TIE2-L914F mutation (HUVEC-TIE2-L914F). Three ABL (Abelson) kinase inhibitors prevented cell proliferation of HUVEC-TIE2-L914F. Moreover, c-ABL, common target of these inhibitors, was highly phosphorylated in HUVEC-TIE2-L914F and VM patient-derived ECs with activating TIE2 mutations. Knockdown of c-ABL/ARG in HUVEC-TIE2-L914F reduced cell proliferation and vascularity of murine VM. Combination treatment with the ABL kinase inhibitor ponatinib and rapamycin caused VM regression in a xenograft model based on injection of HUVEC-TIE2-L914F. A reduced dose of this drug combination was effective in this VM murine model with minimal side effects. The drug combination was antiproliferative, enhanced cell apoptosis and vascular channel regression both in vivo and in a 3-dimensional fibrin gel assay. Conclusions- This is the first report of a combination therapy with ponatinib and rapamycin promoting regression of VM. Mechanistically, the drug combination enhanced AKT inhibition compared with single drug treatment and reduced PLCγ (phospholipase C) and ERK (extracellular signal-regulated kinase) activity.


Subject(s)
Imidazoles/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridazines/therapeutic use , Sirolimus/therapeutic use , Vascular Malformations/drug therapy , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cells, Cultured , Chemotaxis , Drug Evaluation, Preclinical , Drug Therapy, Combination , Heterografts , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Nude , Mutation, Missense , Phospholipase C gamma/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridazines/administration & dosage , Pyridazines/pharmacology , Receptor, TIE-2/genetics , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/pharmacology , Vascular Malformations/pathology
18.
Toxicol Appl Pharmacol ; 366: 46-53, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30684531

ABSTRACT

The classical mast cells degranulation pathway is mediated by FcεRI aggregation and varies in strength among subjects. Dehydroandrographolide (DA) is one of principal components of Andrographis paniculata (Burm.f.) Nees (family: Acanthaceae) and considered the main contributors of its therapeutic properties, such as anti-tumor. In this study, inhibition of IgE-mediated anaphylactic reactions and anti-inflammatory potential of DA were investigated. The anti-anaphylactic activity of DA was investigated using skin swelling and extravasation assays in vivo and mast cell degranulation assay in vitro. The release of cytokines was measured using ELISA kits. Human Phospho-Kinase Array kit and western blotting were used to explore the related molecular signaling pathways. DA inhibited IgE-mediated mast cell activation, including degranulation and release of cytokines in vitro. Moreover, DA reduced the degree of swelling and Evans blue exudation of mice paw in a dose-dependent manner by inhibiting mast cell degranulation. DA obviously reduced the concentrations of histamine, TNF-α, MCP-1, IL-8, IL-13, and IL-4 in mice serum and inhibited IgE-mediated anaphylactic reactions as a potential P-PLCγ inhibitor. Our study reveals that DA can inhibit allergic responses in vivo and in vitro, and it may be regarded as a novel P-PLCγ inhibitor for preventing mast cell-immediate and delayed allergic diseases.


Subject(s)
Anaphylaxis/drug therapy , Anti-Allergic Agents/pharmacology , Calcium Signaling/drug effects , Diterpenes/pharmacology , Histamine Release/drug effects , Immunoglobulin E/immunology , Mast Cells/drug effects , Anaphylaxis/immunology , Anaphylaxis/metabolism , Animals , Cell Degranulation/drug effects , Cell Line , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Humans , Male , Mast Cells/immunology , Mast Cells/metabolism , Mice, Inbred C57BL , Ovalbumin , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism
19.
Cell Physiol Biochem ; 48(5): 2011-2034, 2018.
Article in English | MEDLINE | ID: mdl-30099438

ABSTRACT

BACKGROUND/AIMS: Eggs of all animal species display intense cytoplasmic Ca2+ increases at fertilization. Previously, we reported that unfertilized eggs of Astropecten aranciacus exposed to an actin drug latrunculin A (LAT-A) exhibit similar Ca2+ waves and cortical flashes after 5-10 min time lag. Here, we have explored the molecular mechanisms underlying this unique phenomenon. METHODS: Starfish eggs were pretreated with various agents such as other actin drugs or inhibitors of phospholipase C (PLC), and the changes of the intracellular Ca2+ levels were monitored by use of Calcium Green in the presence or absence of LAT-A. The concomitant changes of the actin cytoskeleton were visualized with fluorescent F-actin probes in confocal microscopy. RESULTS: We have shown that the LAT-A-induced Ca2+ increases are related to the disassembly of actin flaments: i) not only LAT-A but also other agents depolymerizing F-actin (i.e. cytochalasin B and mycalolide B) induced similar Ca2+ increases, albeit with slightly lower efficiency; ii) drugs stabilizing F-actin (i.e. phalloidin and jasplakinolide) either blocked or significantly delayed the LAT-A-induced Ca2+ increases. Further studies utilizing pharmacological inhibitors of PLC (U-73122 and neomycin), dominant negative mutant of PLC-É£, specific sequestration of PIP2 (RFP-PH), InsP3 uncaging, and quantitation of endogenous InsP3 all indicated that LAT-A induces Ca2+ increases by stimulating PLC rather than sensitizing InsP3 receptors. In support of the idea, it bears emphasis that LAT-A timely increased intracellular contents of InsP3 with concomitant decrease of PIP2 levels in the plasma membrane. CONCLUSION: Taken together, our results suggest that suboolemmal actin filaments may serve as a scaffold for cell signaling and modulate the activity of the key enzyme involved in intracellular Ca2+ signaling.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium/metabolism , Cytosol/metabolism , Starfish/metabolism , Actin Cytoskeleton/chemistry , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cytochalasin B/pharmacology , Estrenes/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Microscopy, Confocal , Ovum/drug effects , Ovum/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Pyrrolidinones/pharmacology , Starfish/growth & development , Thiazolidines/pharmacology , src Homology Domains/genetics
20.
Cell Death Differ ; 25(10): 1796-1807, 2018 10.
Article in English | MEDLINE | ID: mdl-29899383

ABSTRACT

The endoplasmic reticulum stress sensor, the unfolded protein response (UPR), regulates intracellular protein homeostasis. While transient activation of the reactive UPR by unfolded protein is protective, prolonged and sustained activation of the reactive UPR triggers CHOP-mediated apoptosis. In the recently characterized, evolutionarily conserved anticipatory UPR, mitogenic hormones and other effectors pre-activate the UPR; how strong and sustained activation of the anticipatory UPR induces cell death was unknown. To characterize this cell death pathway, we used BHPI, a small molecule that activates the anticipatory UPR through estrogen receptor α (ERα) and induces death of ERα+ cancer cells. We show that sustained activation of the anticipatory UPR by BHPI kills cells by inducing depletion of intracellular ATP, resulting in classical necrosis phenotypes, including plasma membrane disruption and leakage of intracellular contents. Unlike reactive UPR activation, BHPI-induced hyperactivation of the anticipatory UPR does not induce apoptosis or sustained autophagy. BHPI does not induce CHOP protein or PARP cleavage, and two pan-caspase inhibitors, or Bcl2 overexpression, have no effect on BHPI-induced cell death. Moreover, BHPI does not increase expression of autophagy markers, or work through recently identified programmed-necrosis pathways, such as necroptosis. Opening of endoplasmic reticulum IP3R calcium channels stimulates cell swelling, cPLA2 activation, and arachidonic acid release. Notably, cPLA2 activation requires ATP depletion. Importantly, blocking rapid cell swelling or production of arachidonic acid does not prevent necrotic cell death. Rapid cell death is upstream of PERK activation and protein synthesis inhibition, and results from strong and sustained activation of early steps in the anticipatory UPR. Supporting a central role for ATP depletion, reversing ATP depletion blocks rapid cell death, and the onset of necrotic cell death is correlated with ATP depletion. Necrotic cell death initiated by strong and sustained activation of the anticipatory UPR is a newly discovered role of the UPR.


Subject(s)
Apoptosis , Unfolded Protein Response , Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arachidonic Acid/metabolism , Calcium Channels/metabolism , Cell Line , Cell Size/drug effects , Endoplasmic Reticulum/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Humans , Necrosis , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Phospholipases A2, Cytosolic/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL