Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cancer Sci ; 112(10): 4208-4219, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34363714

ABSTRACT

Previous studies reported the critical role of the brefeldin A-inhibited guanine nucleotide exchange protein 3-prohibitin 2 (BIG3-PHB2) complex in modulating estrogen signaling activation in breast cancer cells, yet its pathophysiological roles in osteosarcoma (OS) cells remain elusive. Here, we report a novel function of BIG3-PHB2 in OS malignancy. BIG3-PHB2 complexes were localized mainly in mitochondria in OS cells, unlike in estrogen-dependent breast cancer cells. Depletion of endogenous BIG3 expression by small interfering RNA (siRNA) treatment led to significant inhibition of OS cell growth. Disruption of BIG3-PHB2 complex formation by treatment with specific peptide inhibitor also resulted in significant dose-dependent suppression of OS cell growth, migration, and invasion resulting from G2/M-phase arrest and in PARP cleavage, ultimately leading to PARP-1/apoptosis-inducing factor (AIF) pathway activation-dependent apoptosis in OS cells. Subsequent proteomic and bioinformatic pathway analyses revealed that disruption of the BIG3-PHB2 complex might lead to downregulation of inner mitochondrial membrane protein complex activity. Our findings indicate that the mitochondrial BIG3-PHB2 complex might regulate PARP-1/AIF pathway-dependent apoptosis during OS cell proliferation and progression and that disruption of this complex may be a promising therapeutic strategy for OS.


Subject(s)
Bone Neoplasms/pathology , Cell Proliferation/physiology , Cell Survival/physiology , Guanine Nucleotide Exchange Factors/physiology , Mitochondria/metabolism , Osteosarcoma/pathology , Repressor Proteins/physiology , Animals , Apoptosis/physiology , Apoptosis Inducing Factor/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell-Penetrating Peptides/pharmacology , Databases, Factual , Down-Regulation , G2 Phase Cell Cycle Checkpoints , Gene Silencing , Guanine Nucleotide Exchange Factors/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Humans , M Phase Cell Cycle Checkpoints , Membrane Proteins/metabolism , Mice , Mice, Nude , Mitochondrial Membranes/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Osteosarcoma/metabolism , Osteosarcoma/therapy , Poly (ADP-Ribose) Polymerase-1/metabolism , Prohibitins , RNA, Small Interfering/metabolism , Repressor Proteins/drug effects , Repressor Proteins/metabolism
2.
J Hum Genet ; 66(9): 927-935, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34285339

ABSTRACT

Breast cancer is a heterogeneous disease that develops through a multistep process via the accumulation of genetic/epigenetic alterations in various cancer-related genes. Current treatment options for breast cancer patients include surgery, radiotherapy, and chemotherapy including conventional cytotoxic and molecular-targeted anticancer drugs for each intrinsic subtype, such as endocrine therapy and antihuman epidermal growth factor receptor 2 (HER2) therapy. However, these therapies often fail to prevent recurrence and metastasis due to resistance. Overall, understanding the molecular mechanisms of breast carcinogenesis and progression will help to establish therapeutic modalities to improve treatment. The recent development of comprehensive omics technologies has led to the discovery of driver genes, including oncogenes and tumor-suppressor genes, contributing to the development of molecular-targeted anticancer drugs. Here, we review the development of anticancer drugs targeting cancer-specific functional therapeutic targets, namely, MELK (maternal embryonic leucine zipper kinase), TOPK (T-lymphokine-activated killer cell-originated protein kinase), and BIG3 (brefeldin A-inhibited guanine nucleotide-exchange protein 3), as identified through comprehensive breast cancer transcriptomics.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Drug Discovery , Genomics , Breast Neoplasms/genetics , Female , Humans
3.
Biochem Biophys Res Commun ; 518(1): 183-189, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31421830

ABSTRACT

Brefeldin A-inhibited guanine nucleotide-exchange protein 3 (BIG3) interacts with and inhibits the tumor suppressor function of prohibitin-2 (PHB2), and recent in vivo studies have demonstrated that the BIG3-PHB2 interaction is a promising target for breast cancer therapy. However, little biophysical characterization on BIG3 and its interaction with PHB2 has been reported. Here we compared the calculated 8-class secondary structure of the N-terminal domains of BIG family proteins and identified a loop region unique to BIG3. Our biophysical characterization demonstrated that this loop region significantly affects the colloidal and thermodynamic stability of BIG3 and the thermodynamic and kinetic profile of its interaction with PHB2. These results establish a model for the BIG3-PHB2 interaction and an entry for drug discovery for breast cancer.


Subject(s)
Biophysical Phenomena , Breast Neoplasms/metabolism , Conserved Sequence , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Colloids/chemistry , Female , Humans , Kinetics , Models, Biological , Prohibitins , Protein Binding , Protein Domains , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/metabolism , Structure-Activity Relationship , Temperature
4.
Cancer Sci ; 108(4): 785-794, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28122154

ABSTRACT

Xanthohumol (XN), a simple prenylated chalcone, can be isolated from hops and has the potential to be a cancer chemopreventive agent against several human tumor cell lines. We previously identified valosin-containing protein (VCP) as a target of XN; VCP can also play crucial roles in cancer progression and prognosis. Therefore, we investigated the molecular mechanisms governing the contribution of VCP to the antitumor activity of XN. Several human tumor cell lines were treated with XN to investigate which human tumor cell lines are sensitive to XN. Several cell lines exhibited high sensitivity to XN both in vitro and in vivo. shRNA screening and bioinformatics analysis identified that the inhibition of the adenylate cyclase (AC) pathway synergistically facilitated apoptosis induced by VCP inhibition. These results suggest that there is crosstalk between the AC pathway and VCP function, and targeting both VCP and the AC pathway is a potential chemotherapeutic strategy for a subset of tumor cells.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Neoplasms/drug therapy , Propiophenones/pharmacology , Xenograft Model Antitumor Assays , A549 Cells , Adenosine Triphosphatases/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics , Survivin , Valosin Containing Protein
5.
Cancer Sci ; 106(5): 550-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25736224

ABSTRACT

Our previous studies demonstrated that specific inhibition of the BIG3-PHB2 complex, which is a critical modulator in estrogen (E2) signaling, using ERAP, a dominant negative peptide inhibitor, leads to suppression of E2-dependent estrogen receptor (ER) alpha activation through the reactivation of the tumor suppressive activity of PHB2. Here, we report that ERAP has significant suppressive effects against synergistic activation caused by the crosstalk between E2 and growth factors associated with intrinsic or acquired resistance to anti-estrogen tamoxifen in breast cancer cells. Intrinsic PHB2 released from BIG3 by ERAP effectively disrupted each interaction of membrane-associated ERα and insulin-like growth factor 1 receptor beta (IGF-1Rß), EGFR, PI3K or human epidermal growth factor 2 (HER2) in the presence of E2 and the growth factors IGF or EGF, followed by inhibited the activation of IGF-1Rß, EGFR or HER2, and reduced Akt, MAPK and ERα phosphorylation levels, resulting in significant suppression of proliferation of ERα-positive breast cancer cells in vitro and in vivo. More importantly, combined treatment with ERAP and tamoxifen led to a synergistic suppression of signaling that was activated by crosstalk between E2 and growth factors or HER2 amplification. Taken together, our findings suggest that the specific inhibition of BIG3-PHB2 is a novel potential therapeutic approach for the treatment of tamoxifen-resistant breast cancers activated by the crosstalk between E2 and growth factor signaling, especially in premenopausal women.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell-Penetrating Peptides/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Repressor Proteins/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/metabolism , Female , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice, Nude , Molecular Targeted Therapy , Phosphorylation/drug effects , Prohibitins , Receptor, ErbB-2/metabolism , Signal Transduction , Tamoxifen/pharmacology , Xenograft Model Antitumor Assays
6.
Cell Rep ; 42(5): 112479, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37178686

ABSTRACT

CREB/ATF transcription factor OASIS/CREB3L1 is upregulated in long-term-cultured astrocytes undergoing cell-cycle arrest due to loss of DNA integrity by repeated replication. However, the roles of OASIS in the cell cycle remain unexplored. We find that OASIS arrests the cell cycle at G2/M phase after DNA damage via direct induction of p21. Cell-cycle arrest by OASIS is dominant in astrocytes and osteoblasts, but not in fibroblasts, which are dependent on p53. In a brain injury model, Oasis-/- reactive astrocytes surrounding the lesion core show sustained growth and inhibition of cell-cycle arrest, resulting in prolonged gliosis. We find that some glioma patients exhibit low expression of OASIS due to high methylation of its promoter. Specific removal of this hypermethylation in glioblastomas transplanted into nude mice by epigenomic engineering suppresses the tumorigenesis. These findings suggest OASIS as a critical cell-cycle inhibitor with potential to act as a tumor suppressor.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Tumor Suppressor Protein p53 , Mice , Animals , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Mice, Nude , Cell Cycle Checkpoints , Activating Transcription Factors/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism
7.
Eur J Immunol ; 40(4): 1205-17, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20101614

ABSTRACT

Simultaneous stimulation with antigen and adenosine in mast cells induces a synergistic degranulation response at a low antigen dose that is insufficient to cause secretion by itself. This kind of stimulation is thought to be relevant to the immediate asthmatic response upon bronchial challenge with low-dose allergen. In this context, FcepsilonRI- and adenosine receptor-mediated signalings cooperate to increase degranulation in mast cells. In the present study, we prepared mast cells that have mutations (Y219F/Y225F/Y229F) in three tyrosine residues of the FcepsilonRI beta-chain (FcRbeta)-ITAM in order to elucidate the molecular mechanisms of degranulation response synergistically elicited by costimulation with low-dose antigen and adenosine. Introduction of mutations in the FcRbeta-ITAM abolished the synergistic degranulation response. Upon costimulation with low-dose antigen and adenosine, tyrosine phosphorylation of Grb2-associated binder 2, which is located upstream of PI3K-signaling, was significantly increased, but severely diminished in FcRbeta-ITAM mutant cells. These findings indicate that FcRbeta acts as a critical element in mast cell synergistic degranulation response through FcepsilonRI and adenosine receptors, and that PI3K-signaling through FcRbeta-ITAM is a crucial participant in augmentation of FcepsilonRI-mediated degranulation by adenosine.


Subject(s)
Cell Degranulation/physiology , Mast Cells/physiology , Phosphatidylinositol 3-Kinases/physiology , Receptor Cross-Talk/physiology , Receptors, IgE/physiology , Receptors, Purinergic P1/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Adenosine/pharmacology , Animals , Calcium Signaling/physiology , Immunoglobulin E/immunology , Mice , Mice, Inbred Strains , Mice, Knockout , Mutagenesis, Site-Directed , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/analysis , Protein Processing, Post-Translational , Protein Structure, Tertiary , Receptors, IgE/genetics , Recombinant Fusion Proteins/metabolism
8.
J Immunol ; 181(8): 5414-24, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832698

ABSTRACT

Src homology region 2 domain-containing phosphatase 1 (SHP-1), a cytoplasmic protein tyrosine phosphatase, plays an important role for the regulation of signaling from various hematopoietic cell receptors. Although SHP-1 is shown to be a negative signal modulator in mast cells, its precise molecular mechanisms are not well defined. To elucidate how SHP-1 regulates mast cell signaling, we established bone marrow-derived mast cells from SHP-1-deficient motheaten and wild-type mice and analyzed downstream signals induced by cross-linking of high affinity IgE receptor, Fc epsilonRI. Upon Fc epsilonRI ligation, motheaten-derived bone marrow-derived mast cells showed enhanced tyrosine phosphorylation of Src homology region 2 domain-containing leukocyte protein of 76 kDa (SLP-76) and linker for activation of T cells, activation of mitogen-activated protein kinases and gene transcription and production of cytokine. Because the activity of Syk, responsible for the phosphorylation of SLP-76 and linker for activation of T cells, is comparable irrespective of SHP-1, both molecules might be substrates of SHP-1 in mast cells. Interestingly, the absence of SHP-1 expression disrupted the association between SLP-76 and phospholipase Cgamma, which resulted in the decreased phospholipase Cgamma phosphorylation, calcium mobilization, and degranulation. Collectively, these results suggest that SHP-1 regulates Fc epsilonRI-induced downstream signaling events both negatively and positively by functioning as a protein tyrosine phosphatase and as an adaptor protein contributing to the formation of signaling complex, respectively.


Subject(s)
Cell Degranulation/immunology , Gene Expression Regulation, Enzymologic/immunology , Mast Cells/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Receptors, IgE/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium/immunology , Calcium/metabolism , Cell Degranulation/genetics , Extracellular Signal-Regulated MAP Kinases , Gene Expression Regulation, Enzymologic/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mast Cells/enzymology , Mice , Mice, Mutant Strains , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Receptors, IgE/genetics , Receptors, IgE/metabolism , Signal Transduction/genetics , Syk Kinase , T-Lymphocytes/enzymology , T-Lymphocytes/immunology
9.
Int J Oncol ; 56(2): 581-595, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31894262

ABSTRACT

Polypeptide N­acetylgalactosaminyltransferase 6 (GALNT6), which is involved in the initiation of O­glycosylation, has been reported to play crucial roles in mammary carcinogenesis through binding to several substrates; however, its biological roles in mediating growth­promoting effects remain unknown. The present study demonstrated a crucial pathophysiological role of GALNT6 through its O­glycosylation of lectin galactoside­binding soluble 3 binding protein (LGALS3BP), a secreted growth­promoting glycoprotein, in breast cancer growth. The Cancer Genome Atlas data analysis revealed that high expression levels of GALNT6 were significantly associated with poor prognosis of breast cancer. GALNT6 O­glycosylated LGALS3BP in breast cancer cells, whereas knockdown of GALNT6 by siRNA led to the inhibition of both the O­glycosylation and secretion of LGALS3BP, resulting in the suppression of breast cancer cell growth. Notably, LGALS3BP is potentially O­glycosylated at three sites (T556, T571 and S582) by GALNT6, thereby promoting autocrine cell growth, whereas the expression of LGALS3BP with three Ala substitutions (T556A, T571A and S582A) in cells drastically reduced GALNT6­dependent LGALS3BP O­glycosylation and secretion, resulting in suppression of autocrine growth­promoting effect. The findings of the present study suggest that the GALNT6­LGALS3BP axis is crucial for breast cancer cell proliferation and may be a therapeutic target and biomarker for mammary tumors.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , N-Acetylgalactosaminyltransferases/metabolism , Amino Acid Substitution , Antigens, Neoplasm/genetics , Autocrine Communication , Biomarkers, Tumor/genetics , Breast/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation/genetics , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycosylation , Humans , N-Acetylgalactosaminyltransferases/genetics , RNA, Small Interfering/metabolism , Polypeptide N-acetylgalactosaminyltransferase
10.
Biochim Biophys Acta ; 1783(5): 789-802, 2008 May.
Article in English | MEDLINE | ID: mdl-18178162

ABSTRACT

Earlier studies have demonstrated that mast cells produce reactive oxygen species (ROS), which play a role in regulating Ca(2+) influx, while in other cell types ROS are produced in a Ca(2+)-dependent manner. We sought to determine whether ROS are produced downstream of the extracellular Ca(2+) entry in mast cells. Thapsigargin (TG), a receptor-independent agonist, could evoke a robust burst of intracellular ROS. However, this response was distinct from the antigen-induced burst of ROS with respect to time course and dependence on Ca(2+) and phosphatidylinositol-3-kinase (PI3K). The antigen-induced ROS generation occurred immediately, while the TG-induced ROS generation occurred with a significant lag time (~2 min). Antigen but not TG caused extracellular release of superoxide (O(2)(*-))/hydrogen peroxide (H(2)O(2)), which was blocked by diphenyleneiodonium, apocynin, and wortmannin. A capacitative Ca(2+) entry resulted in the generation of O(2)(*-) in the mitochondria in a PI3K-independent manner. Blockade of ROS generation inhibited TG-induced mediator release. Finally, when used together, antigen and TG evoked the release of leukotriene C(4), tumor necrosis factor-alpha, and interleukin-13 as well as ROS generation synergistically. These results suggest that ROS produced upstream of Ca(2+) influx by NADPH oxidase and downstream of Ca(2+) influx by the mitochondria regulate the proinflammatory response of mast cells.


Subject(s)
Calcium/metabolism , Inflammation Mediators/metabolism , Mast Cells/immunology , Mitochondria/metabolism , NADPH Oxidases/physiology , Reactive Oxygen Species/metabolism , Animals , Antigens/immunology , Cell Line , Cells, Cultured , Hydrogen Peroxide/metabolism , Mast Cells/drug effects , Mast Cells/enzymology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Superoxides/metabolism , Thapsigargin/pharmacology
11.
Biochim Biophys Acta ; 1783(5): 924-34, 2008 May.
Article in English | MEDLINE | ID: mdl-18302939

ABSTRACT

Galectin-1 and galectin-3 are the most ubiquitously expressed members of the galectin family and more importantly, these two molecules are shown to have opposite effects on pro-inflammatory responses and/or apoptosis depending on the cell type. Herein, we demonstrate for the first time that galectin-3 induces mast cell apoptosis. Mast cells expressed substantial levels of galectin-3 and galectin-1 and to a lesser extent the receptor for advanced glycation end products (RAGE) on their surfaces. Treatment of cells with galectin-3 at concentrations of > or =100 nM for 18-44 h resulted in cell death by apoptosis. Galectin-3-induced apoptosis was completely prevented by lactose, neutralizing antibody to RAGE, and the caspase-3 inhibitor z-DEVD-fmk. Galectin-3-induced apoptosis was also completely abolished by dithiothreitol and superoxide dismutase, but not inhibited by catalase. Moreover, galectin-3 but not galectin-1 induced the release of superoxide, which was blocked by lactose, anti-RAGE, and dithiothreitol. Finally, galectin-3-induced apoptosis was blocked by bongkrekic acid, an antagonist of the mitochondrial permeability transition pore (PTP), while atractyloside, an agonist of the PTP, greatly facilitated galectin-1-induced apoptosis. These data suggest that galectin-3 induces oxidative stress, PTP opening, and the caspase-dependent death pathway by binding to putative surface receptors including RAGE via the carbohydrate recognition domain.


Subject(s)
Apoptosis , Galectin 1/metabolism , Galectin 3/metabolism , Mast Cells/immunology , Mitochondrial Membrane Transport Proteins/metabolism , Oxidative Stress , Animals , Cell Line , Cells, Cultured , Galectin 1/pharmacology , Galectin 3/pharmacology , Lactose/pharmacology , Mast Cells/drug effects , Mice , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore , Oxidation-Reduction , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Sulfhydryl Compounds/chemistry
12.
Biochim Biophys Acta ; 1783(12): 2332-43, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18822320

ABSTRACT

Advanced glycation end products (AGEs) accumulate during aging and to higher extents under pathological conditions such as diabetes. Since we previously showed that mast cells expressed the AGE-binding protein, receptor for AGEs (RAGE) on their cell surface, we examined whether AGE affected mast cell survival. Herein, we demonstrate that mast cells undergo apoptosis in response to AGE. Glycated albumin (GA), an AGE, but not stimulation with the high-affinity IgE receptor (FcepsilonRI), can induce mast cell death, as measured by annexin V/propidium iodide double-staining. GA (> or =0.1 mg/ml) exhibited this pro-apoptotic activity in a concentration-dependent manner. GA and FcepsilonRI stimulation increased the cytosolic Ca(2+) levels to a similar extent, whereas GA, but not FcepsilonRI stimulation, caused mitochondrial Ca(2+) overload and membrane potential collapse, resulting in mitochondrial integrity disruption, cytochrome c release and caspase-3/7 activation. In addition, GA, but not FcepsilonRI stimulation, induced extracellular release of superoxide from mitochondria, and this release played a key role in the disruption of Ca(2+) homeostasis. Knockdown of RAGE expression using small interfering RNA abolished GA-induced apoptosis, mitochondrial Ca(2+) overload, and superoxide release, demonstrating that RAGE mediates the GA-induced mitochondrial death pathway. AGE-induced mast cell apoptosis may contribute to the immunocompromised and inflammatory conditions.


Subject(s)
Apoptosis/drug effects , Glycation End Products, Advanced/pharmacology , Mast Cells/pathology , Mitochondria/drug effects , Serum Albumin/pharmacology , Superoxides/metabolism , Animals , Bone Marrow/metabolism , Calcium/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cells, Cultured , Cytochromes c/metabolism , Enzyme Activation/drug effects , Glycosylation , Humans , Leukemia, Basophilic, Acute/metabolism , Leukemia, Basophilic, Acute/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Receptor for Advanced Glycation End Products , Receptors, IgG/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Glycated Serum Albumin
13.
Clin Immunol ; 131(1): 145-56, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19144570

ABSTRACT

Aspirin is a well-known nonsteroidal anti-inflammatory drug (NSAID) that may potentiate some acute allergies and causes adverse immunological reactions collectively referred to as aspirin intolerance. Aspirin intolerance is accompanied by increased leukotriene (LT) synthesis, and high levels of serum IgE are a risk factor for NSAID sensitivity. Here we demonstrate that aspirin modulates LTC(4) secretion in mast cells. Therapeutic levels of aspirin and salicylates (or=0.3 mM dose-dependently reduced Ca(2+) store emptying and Ca(2+) release-activated Ca(2+) channel activation. Instead, aspirin facilitated a dihydropyridine receptor-mediated Ca(2+) influx, resulting in increased LTC(4) secretion. This novel action of aspirin may play roles in exacerbation of immediate allergy and aspirin intolerance.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Immunoglobulin E/immunology , Leukotriene C4/immunology , Mast Cells/drug effects , Salicylates/pharmacology , Animals , Blotting, Western , Cell Line , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorometry , Leukotriene C4/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C3H , Mitogen-Activated Protein Kinase 3/metabolism , Phospholipases A2/metabolism , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Leukoc Biol ; 83(5): 1218-29, 2008 May.
Article in English | MEDLINE | ID: mdl-18285401

ABSTRACT

NO is known to suppress mast cell activation, but the role of NO in mast cell survival is unclear. Ligation of the high-affinity receptor for IgE (FcepsilonRI) resulted in NO production in mast cells within minutes. This NO production was largely dependent on NO synthase (NOS) activity and extracellular Ca(2+). The NO production required an aggregation of FcepsilonRI and was accompanied by increased phosphorylation of endothelial NOS (eNOS) at Ser1177 and Akt at Ser473. The phosphorylation of eNOS and Akt and the production of NO were abolished by the PI-3K inhibitor wortmannin. Although thapsigargin (TG) induced NO production as well, this response occurred with a considerable lag time (>10 min) and was independent of FcepsilonRI aggregation and PI-3K and NOS activity. Mast cells underwent apoptosis in response to TG but not upon FcepsilonRI ligation. However, when the NOS-dependent NO production was blocked, FcepsilonRI ligation caused sizable apoptosis, substantial mitochondrial cytochrome c release, caspase-3/7 activation, and collapse of the mitochondrial membrane potential, all of which were inhibited by the caspase-3 inhibitor z-Asp-Glu-Val-Asp-fluoromethylketone. The data suggest that the NO produced by the PI-3K-Akt-eNOS pathway is involved in protecting mast cells from cell death.


Subject(s)
Mast Cells/cytology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Death , Cell Line, Tumor , Cell Survival , Leukemia, Basophilic, Acute , Mast Cells/enzymology , Mast Cells/pathology , Mitochondria/pathology , Nitric Oxide/biosynthesis , Rats
15.
Clin Immunol ; 129(1): 155-62, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18700185

ABSTRACT

Besides mast cells and basophils, the high-affinity IgE Fc receptor (FcepsilonRI) is exclusively expressed on certain FcalphaR (IgA Fc receptor)-expressing immune cells such as neutrophils in allergic patients. Transfected rat basophilic leukemia cell line (RBL-2H3) co-expressing FcepsilonRI and FcalphaR was analyzed for effects of simultaneous receptor engagement by their specific antibodies on degranulation and signaling. Whereas supraoptimal FcepsilonRI engagement decreased degranulation, which is known as a bell-shaped dose-response curve, such inhibitory effect was not observed with FcalphaR engagement. However, simultaneous engagement of FcepsilonRI and FcalphaR showed that supraoptimal FcepsilonRI engagement down-regulates FcalphaR-mediated degranulation. This inhibition was associated with extensive phosphorylation of inositol polyphosphate 5'-phosphatase SHIP1 and FcepsilonRIbeta, and reversed by adding actin-depolymerizing drug, latrunculin B. The results suggest an endogenous mechanism by which FcalphaR functionality is down-regulated in an 'allergic environment' where FcepsilonRI is co-expressed and extensively cross-linked on FcalphaR-expressing effector cells.


Subject(s)
Antigens, CD/metabolism , Cell Degranulation , Phosphoric Monoester Hydrolases/metabolism , Receptors, Fc/metabolism , Receptors, IgE/metabolism , Actins/metabolism , Animals , Antigens, CD/immunology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Degranulation/drug effects , Cell Line, Tumor , Down-Regulation , Humans , Inositol Polyphosphate 5-Phosphatases , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/immunology , Phosphorylation , Rats , Receptors, Fc/immunology , Receptors, IgE/immunology , Thiazolidines/pharmacology
16.
Life Sci ; 83(7-8): 242-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18634805

ABSTRACT

Adenosine has been shown to enhance mast cell degranulation when added together with an antigen. Such augmentation of mast cell activation is relevant to exacerbation of allergic asthma symptoms. Na-Tosyl-Phe chloromethyl ketone (TPCK) is a chymotrypsine-like chymase inhibitor, which has anti-inflammatory properties. In this study, we investigated the effects of TPCK on mast cell synergistic degranulation induced by antigen and adenosine. Here, we report that TPCK almost completely suppressed enhanced degranulation by inhibiting granule movement. Consistent with this, intraperitoneal administration of TPCK resulted in significant amelioration of passive cutaneous anaphylaxis in mice. Furthermore, we demonstrated that TPCK completely inhibited Thr308 phosphorylation of protein kinase B in mast cells stimulated with antigen and adenosine. These results provide a novel action of TPCK for the prevention of mast cell degranulation induced by antigen and adenosine.


Subject(s)
Adenosine/pharmacology , Antigens/pharmacology , Cell Degranulation/drug effects , Mast Cells/immunology , Serine Proteinase Inhibitors/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacokinetics , Vasodilator Agents/pharmacology , Adenosine/agonists , Adenosine/immunology , Anaphylaxis/enzymology , Anaphylaxis/immunology , Animals , Antigens/immunology , Cell Degranulation/immunology , Cells, Cultured , Chymases/antagonists & inhibitors , Chymases/immunology , Mast Cells/enzymology , Mice , Secretory Vesicles , Vasodilator Agents/agonists , Vasodilator Agents/immunology
17.
Cancer Res ; 78(9): 2233-2247, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29440146

ABSTRACT

The p53 and EGFR pathways are frequently altered in bladder cancer, yet their contributions to its progression remain elusive. Here we report that DEAD box polypeptide 31 (DDX31) plays a critical role in the multistep progression of muscle-invasive bladder cancer (MIBC) through its sequential interactions with mutant p53 (mutp53) and EGFR. In early MIBC cells, nuclear DDX31-bound mutp53/SP1 enhanced mutp53 transcriptional activation, leading to migration and invasion of MIBC. Cytoplasmic DDX31 also bound EGFR and phospho-nucleolin in advanced MIBC, leading to EGFR-Akt signaling activation. High expression of both cytoplasmic DDX31 and p53 proteins correlated with poor prognosis in patients with MIBC, and blocking the DDX31/NCL interaction resulted in downregulation of EGFR/Akt signaling, eliciting an in vivo antitumor effect against bladder cancer. These findings reveal that DDX31 cooperates with mutp53 and EGFR to promote progression of MIBC, and inhibition of DDX31/NCL formation may lead to potential treatment strategies for advanced MIBC.Significance: DDX31 cooperates with mutp53 and EGFR to promote progression of muscle invasive bladder cancer. Cancer Res; 78(9); 2233-47. ©2018 AACR.


Subject(s)
DEAD-box RNA Helicases/metabolism , ErbB Receptors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Mutation , Neoplasm Invasiveness , Prognosis , Protein Binding , Transcription, Genetic , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
18.
Int J Oncol ; 52(5): 1539-1558, 2018 May.
Article in English | MEDLINE | ID: mdl-29512727

ABSTRACT

Triple-negative breast cancer (TNBC), defined as breast cancer lacking estrogen- and progesterone­receptor expression and human epidermal growth factor receptor 2 (HER2) amplification, is a heterogeneous disease. RNA-sequencing analysis of 15 TNBC specimens and The Cancer Genome Atlas-TNBC dataset analysis identified the frequent downregulation of leucine-rich repeat-containing 26 (LRRC26), which negatively regulates nuclear factor-κB (NF-κB) signaling, in TNBC tissues. Quantitative polymerase chain reaction and bisulfite pyrosequencing analyses revealed that LRRC26 was frequently silenced in TNBC tissues and cell lines as a result of promoter methylation. LRRC26 expression was restored by 5-aza-2'-deoxycytidine (5'-aza-dC) treatment in HCC1937 TNBC cells, which lack LRRC26 expression. Notably, small interfering RNA-mediated knockdown of LRRC26 expression significantly enhanced the anchorage-independent growth, invasion and migration of HCC70 cells, whereas ectopic overexpression of LRRC26 in BT20 cells suppressed their invasion and migration. Conversely, neither knockdown nor overexpression of LRRC26 had an effect on cell viability in the absence of tumor necrosis factor-α (TNF-α) stimulation. Meanwhile, overexpression of LRRC26 caused the reduction of TNF-α-mediated NF-κB luciferase reporter activity, whereas depleting LRRC26 expression resulted in the upregulation of TNF-α-mediated NF-κB downstream genes [interleukin-6 (IL-6), IL-8 and C-X-C motif chemokine ligand-1]. Taken together, these findings demonstrate that LRRC26 is frequently downregulated in TNBC due to DNA methylation and that it suppresses the TNF-α-independent anchorage-independent growth, invasion and migration of TNBC cells. Loss of LRRC26 function may be a critical event in the aggressiveness of TNBC cells through a TNF-α/NF-κB-independent mechanism.

19.
J Leukoc Biol ; 79(3): 508-18, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16365155

ABSTRACT

An increase in intracellular Ca2+ ([Ca2+]i) is necessary for mast cell exocytosis, but there is controversy over the requirement for Ca2+ in the extracellular medium. Here, we demonstrate that mitochondrial function is a critical determinant of Ca2+ dependence. In the presence of extracellular Ca2+, mitochondrial metabolic inhibitors, including rotenone, antimycin A, and the protonophore carbonyl cyanide p-trifluoromethoxyphenylhydrazone (FCCP), significantly reduced degranulation induced by immunoglobulin E (IgE) antigen or by thapsigargin, as measured by beta-hexosaminidase release. In the absence of extracellular Ca2+; however, antimycin A and FCCP, but not rotenone, enhanced, rather than reduced, degranulation to a maximum of 76% of that observed in the presence of extracellular Ca2+. This enhancement of extracellular, Ca2+-independent degranulation was concomitant with a rapid collapse of the mitochondrial transmembrane potential. Mitochondrial depolarization did not enhance degranulation induced by thapsigargin, irrespective of the presence or absence of extracellular Ca2+. IgE antigen was more effective than thapsigargin as an inducer of [Ca2+]i release, and mitochondrial depolarization augmented IgE-mediated but not thapsigargin-induced Ca2+ store release and mitochondrial Ca2+ ([Ca2+]m) release. Finally, atractyloside and bongkrekic acid [an agonist and an antagonist, respectively, of the mitochondrial permeability transition pore (mPTP)], respectively, augmented and reduced IgE-mediated Ca2+ store release, [Ca2+]m release, and/or degranulation, whereas they had no effects on thapsigargin-induced Ca2+ store release. These data suggest that the mPTP is involved in the regulation of Ca2+ signaling, thereby affecting the mode of mast cell degranulation. This finding may shed light on a new role for mitochondria in the regulation of mast cell activation.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Cell Degranulation/physiology , Cytoplasmic Granules/metabolism , Mast Cells/metabolism , Mitochondria/metabolism , Animals , Antimycin A/pharmacology , Atractyloside/pharmacology , Bongkrekic Acid/pharmacology , Calcium Signaling/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Degranulation/immunology , Cell Line, Tumor , Cytoplasmic Granules/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Enzyme Inhibitors/pharmacology , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/pharmacology , Ion Channels/drug effects , Ion Channels/physiology , Mast Cells/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Rats , Rotenone/pharmacology , Thapsigargin/pharmacology , beta-N-Acetylhexosaminidases/drug effects , beta-N-Acetylhexosaminidases/metabolism
20.
Nat Commun ; 8: 15427, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28555617

ABSTRACT

Approximately 70% of breast cancer cells express oestrogen receptor alpha (ERα). Previous studies have shown that the Brefeldin A-inhibited guanine nucleotide-exchange protein 3-prohibitin 2 (BIG3-PHB2) complex has a crucial role in these cells. However, it remains unclear how BIG3 regulates the suppressive activity of PHB2. Here we demonstrate that BIG3 functions as an A-kinase anchoring protein that binds protein kinase A (PKA) and the α isoform of the catalytic subunit of protein phosphatase 1 (PP1Cα), thereby dephosphorylating and inactivating PHB2. E2-induced PKA-mediated phosphorylation of BIG3-S305 and -S1208 serves to enhance PP1Cα activity, resulting in E2/ERα signalling activation via PHB2 inactivation due to PHB2-S39 dephosphorylation. Furthermore, an analysis of independent cohorts of ERα-positive breast cancers patients reveal that both BIG3 overexpression and PHB2-S39 dephosphorylation are strongly associated with poor prognosis. This is the first demonstration of the mechanism of E2/ERα signalling activation via the BIG3-PKA-PP1Cα tri-complex in breast cancer cells.


Subject(s)
A Kinase Anchor Proteins/metabolism , Breast Neoplasms/metabolism , Estrogens/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Amino Acid Sequence , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Models, Biological , Phosphorylation , Phosphoserine/metabolism , Prognosis , Prohibitins , Protein Phosphatase 1/metabolism , Protein Subunits/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL