Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 12(9)2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32825566

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively is able to increase apoptosis in cancer cells as agent with minimum toxicity to noncancerous cells. However, all cancer cells are not sensitive to TRAIL-induced apoptosis. In this study, we showed the sub-lethal concentrations of a lysosomotropic autophagy inhibitor, IITZ-01, sensitizes cancer cells (renal, lung, and breast carcinoma) to TRAIL-induced apoptosis through DR5 upregulation and survivin downregulation through ubiquitin-proteasome pathway. Knockdown of DR5 or overexpression of survivin inhibited combined treatment with IITZ-01 and TRAIL-induced apoptosis. IITZ-01 downregulated protein expression of Cbl, ubiquitin E3 ligase, and decreased expression level of Cbl markedly led to increase DR5 protein expression and TRAIL sensitivity. Moreover, IITZ-01 decreased expression level of survivin protein via downregulation of deubiquitinase ubiquitin-specific protease 9X (USP9X) expression. Taken together, these results provide the first evidence that IITZ-01 enhances TRAIL-mediated apoptosis through DR5 stabilization by downregulation of Cbl and USP9X-dependent survivin ubiquitination and degradation in renal carcinoma cells.

2.
Biomed Rep ; 9(3): 241-246, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30271600

ABSTRACT

Members of the Ras superfamily of small G-proteins serve as molecular switches of intracellular signaling pathways. Rac2 is a Rho subfamily GTPase switch that is specifically expressed in hematopoietic cells and regulates AKT activation in cell signaling. Ras activating protein-like 3 (RASAL3) is the recently identified Ras GTPase activating protein (GAP) that is also specifically expressed in hematopoietic cells and stimulates p21ras GTPase activity. The restricted expression of both Rac2 and RASAL3 suggests that they may serve critical roles in hematopoietic cell signaling. Here in the present study demonstrates that the catalytic domain of RASAL3 may also be able to interact with Rac2 and stimulate its GTPase activity in vitro. By contrast, p50 rhoGAP molecules did not markedly affect Rac2 GTPase activity, but did accelerate the activity of other Rho GTPases, including Rac1, RhoA and Cdc42. Collectively, the present results indicate, seemingly for the first time, that GAP activity for Rac2 is regulated by the RasGAP family protein, RASAL3.

3.
Asian-Australas J Anim Sci ; 31(12): 1897-1902, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30056668

ABSTRACT

OBJECTIVE: This study was conducted to evaluate the effect of homo or hetero fermentative inoculants on fermentation quality and aerobic stability of sweet potato vine (SPV) silage containing Italian ryegrass hay as moisture absorbent. METHODS: The SPV was harvested at 15% dry matter, mixed with Italian ryegrass hay at 1:1 ratio on a fresh weight basis, and chopped to 3 to 5 cm length. After then, the chopped forage mixture was ensiled into 20-L mini silos in quadruplicate for 7, 48, and 100 days after application of microbial inoculants at 1.2×105 colony forming units (cfu)/g of forage following: no inoculant (CON), Lactobacillus plantarum as a homo fermentative (LP), Lactobacillus buchneri as a hetero fermentative (LB), and mixture of LP and LB at 1:1 ratio as a combo fermentative (MIX). RESULTS: The LP and MIX silages had lowest pH (p<0.001) on 7 and 48 days, while MIX and CON silages had greatest lactate concentrations (p<0.05) on 7 and 48 days, respectively. Acetate concentrations were highest (p<0.01) in LB and MIX silages on 7 days, and in LB silage on 48 days, while lactate to acetate ratios were lowest (p<0.001) in LB silages. The chemical compositions and nutrient digestibility of silage ensiled for 100 days was not affected by inoculants. On 100 days of ensiling, LB silage had lowest (p<0.01) lactate concentration and lactate to acetate ratio, but highest acetate concentration. Aerobic stability was highest (p<0.001) in LB silage followed in MIX silage. On contrast, LB silage had lowest (p<0.05) lactic acid bacteria and mold. CONCLUSION: The results indicated that application of LB solely had a better effect on aerobic stability than not only LP, but also MIX. However, LP application did not show beneficial effects from the viewpoints of fermentation quality and aerobic stability compared to CON.

4.
Int J Mol Sci ; 19(5)2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29702597

ABSTRACT

Corosolic acid is one of the pentacyclic triterpenoids isolated from Lagerstroemia speciose and has been reported to exhibit anti-cancer and anti-proliferative activities in various cancer cells. In the present study, we investigated the molecular mechanisms of corosolic acid in cancer cell death. Corosolic acid induces a decrease of cell viability and an increase of cell cytotoxicity in human renal carcinoma Caki cells. Corosolic acid-induced cell death is not inhibited by apoptosis inhibitor (z-VAD-fmk, a pan-caspase inhibitor), necroptosis inhibitor (necrostatin-1), or ferroptosis inhibitors (ferrostatin-1 and deferoxamine (DFO)). Furthermore, corosolic acid significantly induces reactive oxygen species (ROS) levels, but antioxidants (N-acetyl-l-cysteine (NAC) and trolox) do not inhibit corosolic acid-induced cell death. Interestingly, corosolic acid induces lipid oxidation, and α-tocopherol markedly prevents corosolic acid-induced lipid peroxidation and cell death. Anti-chemotherapeutic effects of α-tocopherol are dependent on inhibition of lipid oxidation rather than inhibition of ROS production. In addition, corosolic acid induces non-apoptotic cell death in other renal cancer (ACHN and A498), breast cancer (MDA-MB231), and hepatocellular carcinoma (SK-Hep1 and Huh7) cells, and α-tocopherol markedly inhibits corosolic acid-induced cell death. Therefore, our results suggest that corosolic acid induces non-apoptotic cell death in cancer cells through the increase of lipid peroxidation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Renal Cell/pathology , Cell Death/drug effects , Kidney Neoplasms/pathology , Lipid Peroxidation/drug effects , Reactive Oxygen Species/metabolism , Triterpenes/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Lagerstroemia/chemistry , alpha-Tocopherol/pharmacology
5.
ACS Appl Mater Interfaces ; 7(36): 20438-46, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26305487

ABSTRACT

We present a simple and industrially accessible method of producing liquid crystalline lipid nanoparticles with various internal structures based on phytantriol, Pluronic F127, and vitamin E acetate. Bilayer vesicles were produced when an ethanolic solution dissolving the lipid components was mixed with deionized water. After the evaporation of ethanol from the aqueous mixture, vesicles were transformed into lipid-filled liquid crystalline nanoparticles with well-defined internal structures such as hexagonal lattices (mostly inverted cubic Pn3m), lined or coiled pattern (inverted hexagonal H2), and disordered structure (inverse microemulsion, L2), depending on the compositions. Further studies suggested that their internal structures were also affected by temperature. The internal structures were characterized from cryo-TEM and small-angle X-ray scattering results. Microcalorimetry studies were performed to investigate the degree of molecular ordering/crystallinity of lipid components within the nanostructures. From the comparative studies, we demonstrated the present method could produce the lipid nanoparticles with similar characteristics to those made from a conventional method. More importantly, the production only requires simple tools for mixing and ethanol evaporation and it is possible to produce 10 kg or so per batch of aqueous lipid nanoparticles dispersions, enabling the large-scale production of the liquid crystalline nanoparticles for various biomedical applications.


Subject(s)
Lipids/chemistry , Liquid Crystals/chemistry , Nanostructures/chemistry , Fatty Alcohols/chemistry , Nanoparticles/chemistry , Poloxamer/chemistry , Scattering, Small Angle , Temperature , Vitamin E/chemistry , X-Ray Diffraction
6.
Chem Biol Interact ; 211: 36-43, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24440808

ABSTRACT

Silibinin, an effective anti-cancer and chemopreventive agent, has been shown to exert multiple effects on cancer cells, including inhibition of both cell proliferation and migration. However, the molecular mechanisms responsible for these effects are not fully understood. We observed that silibinin significantly induced the expression of the non-steroidal anti-inflammatory drug-activated gene-1 (NAG-1) in both p53 wild-type and p53-null cancer cell lines, suggesting that silibinin-induced NAG-1 up-regulation is p53-independent manner. Silibinin up-regulates early growth response-1 (EGR-1) expression. The ectopic expression of EGR-1 significantly increased NAG-1 promoter activity and NAG-1 protein expression in a dose-dependent manner. Furthermore, down-regulation of EGR-1 expression using siRNA markedly reduced silibinin-mediated NAG-1 expression, suggesting that the expression of EGR-1 is critical for silibinin-induced NAG-1 expression. We also observed that reactive oxygen species (ROS) are generated by silibinin; however, ROS did not affect silibinin-induced NAG-1 expression and apoptosis. In addition, we demonstrated that the mitogen-activated protein kinase (MAP kinase) signal transduction pathway is involved in silibinin-induced NAG-1 expression. Inhibitors of p38 MAP kinase (SB203580) attenuated silibinin-induced NAG-1 expression. Furthermore, we found that siRNA-mediated knockdown of NAG-1 attenuated silibinin-induced apoptosis. Collectively, the results of this study demonstrate for the first time that up-regulation of NAG-1 contributes to silibinin-induced apoptosis in cancer cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Early Growth Response Protein 1/metabolism , Growth Differentiation Factor 15/genetics , Silymarin/pharmacology , Up-Regulation/drug effects , Animals , Caco-2 Cells , Cell Line , Cell Line, Tumor , Growth Differentiation Factor 15/metabolism , HCT116 Cells , HT29 Cells , Humans , Mice , Silybin
7.
Exp Mol Med ; 45: e19, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23598593

ABSTRACT

New colchicine analogs have been synthesized with the aim of developing stronger potential anticancer activities. Among the analogs, CT20126 has been previously reported to show immunosuppressive activities. Here, we report that CT20126 also shows potential anticancer effects via an unusual mechanism: the modulation of microtubule integrity and cell cycle arrest at the G2/M phase before apoptosis. When we treated COS-7 cells with CT20126 (5 µM), the normal thread-like microtubules were disrupted into tubulin dimers within 10 min and thereafter repolymerized into short, thick filaments. In contrast, cells treated with the same concentration of colchicine exhibited microtubule depolymerization after 20 min and never underwent repolymerization. Furthermore, optical density (OD) analysis (350 nm) with purified tubulin showed that CT20126 had a higher repolymerizing activity than that of Taxol, a potent microtubule-polymerizing agent. These results suggest that the effects of CT20126 on microtubule integrity differ from those of colchicine: the analog first destabilizes microtubules and then stabilizes the disrupted tubulins into short, thick polymers. Furthermore, CT20126 induced a greater level of apoptotic activity in Jurkat T cells than colchicine (assessed by G2/M arrest, caspase-3 activation and cell sorting). At 20 nM, CT20126 induced 47% apoptosis among Jurkat T cells, whereas colchicine induced only 33% apoptosis. Our results suggest that the colchicine analog CT20126 can potently induce apoptosis by disrupting microtubule integrity in a manner that differs from that of colchicine or Taxol.


Subject(s)
Apoptosis/drug effects , Colchicine/analogs & derivatives , Microtubules/metabolism , Tubulin Modulators/pharmacology , Acetylation/drug effects , Animals , COS Cells , Caspase 3/metabolism , Cattle , Cell Division/drug effects , Chlorocebus aethiops , Colchicine/chemistry , Colchicine/pharmacology , Enzyme Activation/drug effects , G2 Phase/drug effects , Humans , Jurkat Cells , Poly(ADP-ribose) Polymerases/metabolism , Tubulin/metabolism , Tubulin Modulators/chemistry
8.
J Microbiol Biotechnol ; 22(12): 1629-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23221524

ABSTRACT

Previously, we demonstrated that the erythropoietin receptor (EpoR) is present on fibroblasts, where it regulates focal contact. Here, we assessed whether this action of EpoR is involved in the reduced cell adhesion observed in colonocytes exposed to Clostridium difficile toxin A. EpoR was present and functionally active in cells of the human colonic epithelial cell line HT29 and epithelial cells of human colon tissues. Toxin A significantly decreased activating phosphorylations of EpoR and its downstream signaling molecules JAK-2 (Janus kinase 2) and STAT5 (signal transducer and activator of transcription 5). In vitro kinase assays confirmed that toxin A inhibited JAK 2 kinase activity. Pharmacological inhibition of JAK2 (with AG490) abrogated activating phosphorylations of EpoR and also decreased focal contacts in association with inactivation of paxillin, an essential focal adhesion molecule. In addition, AG490 treatment significantly decreased expression of occludin (a tight junction molecule) and tight junction levels. Taken together, these data suggest that inhibition of JAK2 by toxin A in colonocytes causes inactivation of EpoR, thereby enhancing the inhibition of focal contact formation and loss of tight junctions known to be associated with the enzymatic activity of toxin A.


Subject(s)
Bacterial Toxins/pharmacology , Enterotoxins/pharmacology , Focal Adhesions/drug effects , Janus Kinase 2/antagonists & inhibitors , Receptors, Erythropoietin/antagonists & inhibitors , Signal Transduction/drug effects , Analysis of Variance , Caco-2 Cells , Cell Adhesion/drug effects , Cells, Cultured , Colorectal Neoplasms , Enzyme Activation/drug effects , Focal Adhesions/metabolism , Humans , Janus Kinase 2/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Receptors, Erythropoietin/metabolism , Tight Junctions/metabolism
9.
J Pept Sci ; 18(10): 650-6, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22969062

ABSTRACT

We recently demonstrated that the insect peptide CopA3 (LLCIALRKK), a disulfide-linked dimeric peptide, exerts antimicrobial and anti-inflammatory activities in a mouse colitis model. Here, we examined whether CopA3 inhibited activation of macrophages by LPS. Exposure of an unseparated mouse peritoneal cell population or isolated peritoneal macrophages to LPS markedly increased secretion of IL-6 and TNF-α; these effects were significantly inhibited by CopA3 treatment. The inhibitory effect of CopA3 was also evident in murine macrophage cell line, RAW 264.7. Western blotting revealed that LPS-induced activation of STAT1 and STAT5 in macrophages was significantly inhibited by CopA3. Inhibition of JAK (STAT1/STAT5 kinase) with AG490 markedly reduced the production of IL-6 and TNF-α in macrophages. Collectively, these observations suggest that CopA3 inhibits macrophage activation by inhibiting activating phosphorylations of the transcription factors, STAT1 and STAT5, and blocking subsequent production of IL-6 and TNF-α and indicate that CopA3 may be useful as an immune-modulating agent.


Subject(s)
Insect Proteins/pharmacology , Insecta/chemistry , Lipopolysaccharides/antagonists & inhibitors , Macrophage Activation/drug effects , Macrophages/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Insect Proteins/chemical synthesis , Insect Proteins/chemistry , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Mice , Phosphorylation , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
10.
J Microbiol Biotechnol ; 22(1): 50-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22297219

ABSTRACT

Phospholipase C-γl (PLC-γl) expression is associated with cellular transformation. Notably, PLC-gamma is up-regulated in colorectal cancer tissue and breast carcinoma. Because exotoxins released by Clostridium botulinum have been shown to induce apoptosis and promote growth arrest in various cancer cell lines, we examined here the potential of Clostridium difficile toxin A to selectively induce apoptosis in cells transformed by PLC-γl overexpression. We found that PLC-γl-transformed cells, but not vectortransformed (control) cells, were highly sensitive to C. difficile toxin A-induced apoptosis and mitotic inhibition. Moreover, expression of the proapoptotic Bcl2 family member, Bim, and activation of caspase-3 were significantly up-regulated by toxin A in PLC-γl-transformed cells. Toxin A-induced cell rounding and paxillin dephosphorylation were also significantly higher in PLC-γl-transformed cells than in control cells. These findings suggest that C. difficile toxin A may have potential as an anticancer agent against colorectal cancers and breast carcinomas in which PLC-γl is highly up-regulated.


Subject(s)
Apoptosis , Bacterial Toxins/toxicity , Cell Transformation, Neoplastic , Enterotoxins/toxicity , Fibroblasts/drug effects , Mitosis , Phospholipase C gamma/biosynthesis , Animals , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Profiling , Phospholipase C gamma/genetics , Rats
11.
BMB Rep ; 45(2): 85-90, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22360885

ABSTRACT

Our previous study demonstrated that CopA3, a disulfide dimer of the coprisin peptide analogue (LLCIALRKK), has antibacterial activity. In this study, we assessed whether CopA3 caused cellular toxicity in various mammalian cell lines. CopA3 selectively caused a marked decrease in cell viability in Jurkat T, U937, and AML-2 cells (human leukemia cells), but was not cytotoxic to Caki or Hela cells. Fragmentation of DNA, a marker of apoptosis, was also confirmed in the leukemia cell lines, but not in the other cells. CopA3-induced apoptosis in leukemia cells was mediated by apoptosis inducing factor (AIF), indicating induction of a caspase-independent signaling pathway.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Insect Proteins/toxicity , Amino Acid Sequence , Animals , Apoptosis Inducing Factor/metabolism , Cell Line, Tumor , Coleoptera/metabolism , HeLa Cells , Humans , Insect Proteins/chemical synthesis , Insect Proteins/therapeutic use , Jurkat Cells , Leukemia/drug therapy , Leukemia/metabolism , Signal Transduction
12.
J Microbiol Biotechnol ; 22(2): 170-5, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22370345

ABSTRACT

Clostridium difficile toxin A glucosylates Rho family proteins, resulting in actin filament disaggregation and cell rounding in cultured colonocytes. Given that the cellular toxicity of toxin A is dependent on its receptor binding and subsequent entry into the cell, we herein sought to identify additional colonocyte proteins that might bind to toxin A following its internalization. Our results revealed that toxin A interacted with ERK1 and ERK2 in two human colonocyte cell lines (NCM460 and HT29). A GST-pulldown assay also showed that toxin A can directly bind to ERK1 and ERK2. In NCM460 cells exposed to PMA (an ERK1/2 activator), the phosphorylation of ERK1/2 did not affect the interaction between toxin A and ERK1/2. However, an in vitro kinase assay showed that the direct binding of toxin A to ERK1 or ERK2 inhibited their kinase activities. These results suggest a new molecular mechanism for the cellular toxicity seen in cells exposed to toxin A.


Subject(s)
Bacterial Toxins/metabolism , Clostridioides difficile/pathogenicity , Enterotoxins/metabolism , Enzyme Inhibitors/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Cell Line , Epithelial Cells/drug effects , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Binding
14.
Exp Mol Med ; 43(3): 153-60, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21339697

ABSTRACT

Phosphatidylinositol phosphates (PtdInsPs) are ubiquitous membrane phospholipids that play diverse roles in cell growth and differentiation. To clarify the regulation mechanism acting on neurofilament light chain (NF-L) self assembly, we examined the effects of various PtdInsPs on this process. We found that PtdInsPs, including PI(4,5)P((2)), directly bind to the positively charged Arg(54) of murine NF-L, and this binding promotes NF-L self assembly in vitro. Mutant NF-L (R53A/R54A) proteins lacking binding affinity to PtdInsPs did not have the same effect, but the mutant NF-L proteins showed greater self assembly than the wild-type in the absence of any PtdInsP. These results collectively suggest that Arg(54) plays a pivotal role in NF-L self assembly by binding with PtdInsPs.


Subject(s)
Neurofilament Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Multimerization , Animals , Fluorescent Antibody Technique , Mice , Mutation/genetics , Neurofilament Proteins/genetics , Phospholipase C gamma/metabolism
15.
J Biol Chem ; 285(43): 32888-32896, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20696758

ABSTRACT

Clostridium difficile toxin A is known to cause actin disaggregation through the enzymatic inactivation of intracellular Rho proteins. Based on the rapid and severe cell rounding of toxin A-exposed cells, we speculated that toxin A may be involved in post-translational modification of tubulin, leading to microtubule instability. In the current study, we observed that toxin A strongly reduced α-tubulin acetylation in human colonocytes and mouse intestine. Fractionation analysis demonstrated that toxin A-induced α-tubulin deacetylation yielded monomeric tubulin, indicating the presence of microtubule depolymerization. Inhibition of the glucosyltransferase activity against Rho proteins of toxin A by UDP-2',3'-dialdehyde significantly abrogated toxin A-induced α-tubulin deacetylation. In colonocytes treated with trichostatin A (TSA), an inhibitor of the HDAC6 tubulin deacetylase, toxin A-induced α-tubulin deacetylation and loss of tight junction were completely blocked. Administration of TSA also attenuated proinflammatory cytokine production, mucosal damage, and epithelial cell apoptosis in mouse intestine exposed to toxin A. These results suggest that toxin A causes microtubule depolymerization by activation of HDAC6-mediated tubulin deacetylation. Indeed, blockage of HDAC6 by TSA markedly attenuates α-tubulin deacetylation, proinflammatory cytokine production, and mucosal damage in a toxin A-induced mouse enteritis model. Tubulin deacetylation is an important component of the intestinal inflammatory cascade following toxin A-mediated Rho inactivation in vitro and in vivo.


Subject(s)
Bacterial Toxins/toxicity , Enteritis/metabolism , Enterotoxins/toxicity , Histone Deacetylases/metabolism , Intestinal Mucosa/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational/drug effects , Tubulin/metabolism , Acetylation/drug effects , Acute Disease , Animals , Apoptosis/drug effects , Cell Line , Colon/metabolism , Colon/pathology , Cytokines/biosynthesis , Enteritis/chemically induced , Enteritis/drug therapy , Enzyme Activation/drug effects , Epithelial Cells/metabolism , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Intestinal Mucosa/pathology , Mice , Tubulin/genetics , Uridine Diphosphate/analogs & derivatives , Uridine Diphosphate/pharmacology , rho GTP-Binding Proteins/metabolism
16.
Exp Mol Med ; 42(3): 216-27, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20164676

ABSTRACT

Growth factor stimulation induces Y783 phosphorylation of phosphoinositide-specific PLC-gamma1, and the subsequent activation of this enzyme in a cellular signaling cascade. Previously, we showed that a double point mutation, Y509A/F510A, of PLC-gamma1, abolished interactions with translational elongation factor 1-alpha. Here, we report that the Y509A/F510A mutant PLC-gamma1 displayed extremely high levels of Y783 phosphorylation and enhanced catalytic activity, compared to wild-type PLC-gamma1, upon treatment of COS7 cells with EGF. In quiescent COS7 cells, the Y509A/F510A mutant PLC-gamma1 exhibited a constitutive hydrolytic activity, whereas the wild-type counterpart displayed a basal level of activity. Upon treatment of COS7 cells with EGF, the Y783F mutation in Y509A/F510A PLC-gamma1 (Y509A/F510A/Y783F triple mutant) cells also led to an enhanced catalytic activity, whereas Y783F mutation alone displayed a basal level of activity. Our results collectively suggest that the Y509A/F510A mutant is more susceptible to receptor tyrosine kinase-induced Y783 phosphorylation than is wild-type PLC-gamma1, but no longer requires Y783 phosphorylation step for the Y509A/F510A mutant PLC-gamma1 activation in vivo.


Subject(s)
Amino Acid Substitution/genetics , Epidermal Growth Factor/pharmacology , Phosphatidylinositols/metabolism , Phospholipase C gamma/genetics , Phosphotyrosine/metabolism , Point Mutation/genetics , Amino Acid Substitution/drug effects , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation/drug effects , Hydrolysis/drug effects , Mutant Proteins/metabolism , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Rats
17.
Apoptosis ; 14(11): 1378-86, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19768546

ABSTRACT

Kahweol, the coffee-specific diterpene, has been reported for its tumor cell growth inhibitory activity and anti-carcinogenic activity. The mechanism by which kahweol initiates apoptosis remains poorly understood. In the present study, we investigated the effect of kahweol on the apoptotic pathway in U937 human promonocytic cells. We show that kahweol induces apoptosis in association with the activation of caspase 3 and cytochrome c release from the mitochondria to the cytosol, as well as down-regulation of anti-apoptotic proteins (Bcl-2, Bcl-xL, Mcl-1 and XIAP). Kahweol altered the phosphorylation state of members of the MAPKs and Akt. Ectopic expression of Bcl-2 or constitutive active Akt (myr-Akt) in U937 cells attenuates kahweol-induced apoptosis. In addition, we have also shown that JNK and Akt signal pathway plays a crucial role in kahweol-induced apoptosis in U937 cells. Taken together, our results show the activity of kahweol to modulate multiple components in apoptotic response of human leukemia cells and raise the possibility a novel therapeutic strategy in hematological malignancies.


Subject(s)
Apoptosis/drug effects , Diterpenes/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis Regulatory Proteins/metabolism , Caspase Inhibitors , Cell Proliferation/drug effects , Down-Regulation , Humans , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Signal Transduction/drug effects , U937 Cells
18.
BMB Rep ; 41(12): 868-74, 2008 Dec 31.
Article in English | MEDLINE | ID: mdl-19123978

ABSTRACT

Neurofilaments (NFs) are neuronal intermediate filaments composed of light (NF-L), middle (NF-M), and heavy (NF-H) subunits. NF-L self-assembles into a "core" filament with which NF-M or NF-H co-assembles to form the neuronal intermediate filament. Recent reports show that point mutations of the NF-L gene result in Charcot-Marie-Tooth disease (CMT). However, the most recently described rod domain mutant of human NF-L (A148V) has not been characterized in cellular level. We cloned human NF-L and used it to engineer the A148V. In phenotypic analysis using SW13 cells, A148V mutation completely abolished filament formation despite of presence of NF-M. Moreover, A148V mutation reduced the levels of in vitro self-assembly using GST-NF-L (H/R) fusion protein whereas control (A296T) mutant did not affect the filament formation. These results suggest that alanine at position 148 is essentially required for NF-L self-assembly leading to subsequent filament formation in neuronal cells.


Subject(s)
Neurofilament Proteins/chemistry , Neurofilament Proteins/genetics , Alanine/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Cell Line , DNA Primers/genetics , DNA, Complementary/genetics , Humans , Intermediate Filaments/chemistry , Intermediate Filaments/metabolism , Multiprotein Complexes , Mutagenesis, Site-Directed , Neurofilament Proteins/metabolism , Neurons/metabolism , PC12 Cells , Phenotype , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
19.
J Neurosci Methods ; 161(2): 199-204, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17157386

ABSTRACT

Neurofilaments (NFs) are heteropolymers composed of light (NF-L), middle (NF-M), and heavy (NF-H) subunits, present in most neurons. NF-L polymerizes on its own to provide a scaffold on which regular NFs form via the cross-bridging of NF-M or NF-H. To clarify the mechanism of regulation of NF-L self-assembly, we developed an assay using truncated mutant NF-L fused to glutathione-S transferase (GST). Western immunoblotting data show that the GST-fused head-rod domains of NF-L are necessary and sufficient for detecting assembled NF-L. The levels of self-assembled NF-L subunits detected using GST fusion proteins were consistent with those detected by electron microscopy and turbidity assay. Our results collectively imply that GST-fused head-rod domains of NF-L are critical tools for analyzing NF-L self-assembly in vitro.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/ultrastructure , Blotting, Western/methods , Neurofilament Proteins/metabolism , Neurofilament Proteins/ultrastructure , Animals , Cell Line, Tumor , Humans , Mutagenesis, Site-Directed , PC12 Cells , Rats , Structure-Activity Relationship
20.
Nat Cell Biol ; 8(12): 1389-97, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17128263

ABSTRACT

Growth hormone binds to its membrane receptor (GHR), whereby it regulates many cellular functions, including proliferation, differentiation and chemotaxis. However, although the activation of growth hormone-mediated signalling is well understood, the precise mechanism responsible for its regulation has not been elucidated. Here, we demonstrate that phospholipase Cgamma1 (PLCgamma1) modulates the action of growth hormone-mediated signalling by interacting with tyrosine kinase Jak2 (janus kinase 2) in a growth hormone-dependent manner. In the absence of PLCgamma1 (PLCgamma1(-/-)), growth hormone-induced JAK2 and STAT5 phosphorylation significantly increased in mouse embryonic fibroblasts (MEFs). Furthermore, the re-expression of PLCgamma1 reduced growth hormone-induced Jak2 activation. Growth hormone-induced Jak2 phosphorylation was enhanced by siRNA-specific knockdown of PLCgamma1. Interestingly, PLCgamma1 physically linked Jak2 and protein tyrosine phosphatase-1B (PTP-1B) by binding to both using different domains, and this process was implicated in the modulation of cytokine signalling through Jak2. In addition, in PLCgamma1(-/-) MEFs, growth hormone-dependent c-Fos activation was upregulated and growth hormone-induced proliferation was potentiated. These results suggest that PLCgamma1 has a key function in the regulation of growth hormone-mediated signalling by negatively regulating Jak2 activation.


Subject(s)
Down-Regulation/drug effects , Growth Hormone/pharmacology , Janus Kinase 2/metabolism , Phospholipase C gamma/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction/drug effects , Animals , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Humans , Mice , NIH 3T3 Cells , Phospholipase C gamma/deficiency , Protein Binding/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Recombinant Fusion Proteins/metabolism , STAT5 Transcription Factor/metabolism , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...