Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Appl Environ Microbiol ; 82(18): 5603-11, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27401975

ABSTRACT

UNLABELLED: Spinosad, a highly effective insecticide, has an excellent environmental and mammalian toxicological profile. Global market demand for spinosad is huge and growing. However, after much effort, there has been almost no improvement in the spinosad yield from the original producer, Saccharopolyspora spinosa Here, we report the heterologous expression of spinosad using Saccharopolyspora erythraea as a host. The native erythromycin polyketide synthase (PKS) genes in S. erythraea were replaced by the assembled spinosad gene cluster through iterative recombination. The production of spinosad could be detected in the recombinant strains containing the whole biosynthesis gene cluster. Both metabolic engineering and UV mutagenesis were applied to further improve the yield of spinosad. The final strain, AT-ES04PS-3007, which could produce spinosad with a titer of 830 mg/liter, has significant potential in industrial applications. IMPORTANCE: This work provides an innovative and promising way to improve the industrial production of spinosad. At the same time, it also describes a successful method of heterologous expression for target metabolites of interest by replacing large gene clusters.


Subject(s)
Insecticides/metabolism , Macrolides/metabolism , Saccharopolyspora/metabolism , Cloning, Molecular , Drug Combinations , Gene Expression , Metabolic Engineering , Metabolic Networks and Pathways/genetics , Multigene Family
2.
Biotechniques ; 59(4): 193-4, 196, 198-200, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26458547

ABSTRACT

We report a novel method for the seamless cloning of large DNA fragments (SCLF) of up to 44 kb or larger from Streptomyces chromosomal DNA. SCLF is based on homologous recombination in Streptomyces and is easy to perform. The strategy of SCLF is to flank the target sequence in the chromosomal DNA with two identical restriction sites by the insertion of plasmids containing that site at either end of the fragment, which is then isolated by plasmid rescue through the self-ligation of restriction digested genomic DNA. The method involves three steps: (i) placing a certain restriction site (CRS) at the 3'-end of the target sequence by insertion through homologous recombination of a plasmid containing the CRS; (ii) inserting through homologous recombination at the 5'-end of the target sequence a linearized self-suicide vector with the identical CRS; (iii) digesting the genomic DNA with the certain restriction enzyme followed by self-ligation in order to plasmid rescue the target fragment. SCLF can be applied to other Actinomycetales, and further optimizations may reduce the amount of time required to perform this technique.


Subject(s)
Cloning, Molecular/methods , DNA/genetics , Homologous Recombination/genetics , Streptomyces/genetics , Genetic Vectors , Genome, Fungal
3.
J Pharmacol Exp Ther ; 354(3): 261-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26105954

ABSTRACT

The bioactive lipid sphingosine-1-phosphate (S1P) and its receptors (S1P1-5) play critical roles in many pathologic processes, including cancer. The S1P axis has become a bona fide therapeutic target in cancer. JTE-013 [N-​(2,​6-​dichloro-​4-​pyridinyl)-​2-​[1,​3-​dimethyl-​4-​(1-​methylethyl)-​1H-​pyrazolo[3,​4-​b]pyridin-​6-​yl]-​hydrazinecarboxamide], a known S1P2 antagonist, suffers from instability in vivo. Structurally modified, more potent, and stable S1P2 inhibitors would be desirable pharmacological tools. One of the JTE-013 derivatives, AB1 [N-(1H-4-isopropyl-1-allyl-3-methylpyrazolo[3,4-b]pyridine-6-yl)-amino-N'-(2,6-dichloropyridine-4-yl) urea], exhibited improved S1P2 antagonism compared with JTE-013. Intravenous pharmacokinetics indicated enhanced stability or slower clearance of AB1 in vivo. Migration assays in glioblastoma showed that AB1 was slightly more effective than JTE-013 in blocking S1P2-mediated inhibition of cell migration. Functional studies in the neuroblastoma (NB) cell line SK-N-AS showed that AB1 displayed potency at least equivalent to JTE-013 in affecting signaling molecules downstream of S1P2. Similarly, AB1 inhibition of the growth of SK-N-AS tumor xenografts was improved compared with JTE-013. Cell viability assays excluded that this enhanced AB1 effect is caused by inhibition of cancer cell survival. Both JTE-013 and AB1 trended to inhibit (C-C motif) ligand 2 expression and were able to significantly inhibit subsequent tumor-associated macrophage infiltration in NB xenografts. Interestingly, AB1 was more effective than JTE-013 in inhibiting the expression of the profibrotic mediator connective tissue growth factor. The terminal deoxynucleotidyl transferase-mediated digoxigenin-deoxyuridine nick-end labeling assay and cleaved caspase-3 detection further demonstrated that apoptosis was increased in AB1-treated NB xenografts compared with JTE-013. Overall, the modification of JTE-013 to produce the AB1 compound improved potency, intravenous pharmacokinetics, cellular activity, and antitumor activity in NB and may have enhanced clinical and experimental applicability.


Subject(s)
Antineoplastic Agents/pharmacology , Chlorambucil/analogs & derivatives , Neuroblastoma/drug therapy , Receptors, Lysosphingolipid/antagonists & inhibitors , Animals , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chlorambucil/pharmacology , Connective Tissue Growth Factor/metabolism , Female , Heterografts , Humans , Mice , Mice, Nude , Neuroblastoma/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors
4.
Appl Environ Microbiol ; 81(16): 5326-34, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26025902

ABSTRACT

Avermectin (AVM) and ivermectin (IVM) are potent pesticides and acaricides which have been widely used during the past 30 years. As insect resistance to AVM and IVM is greatly increasing, alternatives are urgently needed. Here, we report two novel AVM derivatives, tenvermectin A (TVM A) and TVM B, which are considered a potential new generation of agricultural and veterinary drugs. The molecules of the TVMs were designed based on structure and pharmacological property comparisons among AVM, IVM, and milbemycin (MBM). To produce TVMs, a genetically engineered strain, MHJ1011, was constructed from Streptomyces avermitilis G8-17, an AVM industrial strain. In MHJ1011, the native aveA1 gene was seamlessly replaced with milA1 from Streptomyces hygroscopicus. The total titer of the two TVMs produced by MHJ1011 reached 3,400 mg/liter. Insecticidal tests proved that TVM had enhanced activities against Tetranychus cinnabarinus and Bursaphelenchus xylophilus, as desired. This study provides a typical example of exploration for novel active compounds through a new method of polyketide synthase (PKS) reassembly for gene replacement. The results of the insecticidal tests may be of use in elucidating the structure-activity relationship of AVMs and MBMs.


Subject(s)
Acaricides/metabolism , Anthelmintics/metabolism , Ivermectin/analogs & derivatives , Metabolic Engineering , Streptomyces/genetics , Streptomyces/metabolism , Acaricides/chemistry , Animals , Anthelmintics/chemistry , Ivermectin/chemistry , Ivermectin/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Survival Analysis , Tetranychidae/drug effects , Tylenchida/drug effects
5.
Cell Biol Int ; 39(10): 1131-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25952685

ABSTRACT

Hepatocyte growth factor (HGF) was identified as an endogenous tissue protective agent against apoptosis in many cell types. The mechanism by which HGF protects primary endothelial cells (ECs) has not yet been completely elucidated. FOXO1 and FOXO3a, two members of the FOXO family, are the most abundant FOXO isoforms in mature endothelial cells. In this study, we aimed to explore whether FOXO1 and FOXO3a play similar roles in HGF-mediated protection against apoptosis in mature endothelial cells. Our result showed that HGF prevented ECs from oxidative-stress induced apoptosis in part by inducing the phosphorylation of FOXO proteins. FOXO1 and FOXO3a are equally important in this process by regulating the expression of Bim, PUMA, FasL, and TRAIL.


Subject(s)
Apoptosis/physiology , Endothelial Cells/physiology , Forkhead Transcription Factors/physiology , Hepatocyte Growth Factor/physiology , Cells, Cultured , Fas Ligand Protein/genetics , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Humans , Oxidative Stress/physiology , TNF-Related Apoptosis-Inducing Ligand/genetics
6.
J Asian Nat Prod Res ; 17(4): 384-90, 2015.
Article in English | MEDLINE | ID: mdl-25633191

ABSTRACT

This work presents the metabolites of isocorynoxeine (ICOR), which is one of four bioactive tetracyclic oxindole alkaloids isolated from Uncaria hooks used commonly in the traditional Chinese medicines and Kampo medicines. After oral administration of 40 mg kg(-1) ICOR to rats, bile was drained and analyzed by LC-MS. Two phase I metabolites, namely 11-hydroxyisocorynoxeine (M1) and 10-hydroxyisocorynoxeine (M2), and two phase II metabolites, namely 11-hydroxyisocorynoxeine 11-O-ß-D-glucuronide (M3) and 10-hydroxyisocorynoxeine 10-O-ß-D-glucuronide (M4), were isolated from rat excreta and bile, respectively, whose structures were elucidated on the basis of CD, NMR, and MS.


Subject(s)
Indole Alkaloids/pharmacokinetics , Uncaria/chemistry , Administration, Oral , Animals , Bile/chemistry , Bile/metabolism , Chromatography, Liquid , Glucuronides/chemistry , Indole Alkaloids/chemistry , Male , Medicine, Chinese Traditional , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Rats
7.
J Pediatr Surg ; 49(8): 1286-91, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25092091

ABSTRACT

BACKGROUND/PURPOSE: Neuroblastoma (NB) is the most common extracranial solid tumor of childhood. Preliminary data derived from a human angiogenesis array in NB showed that the bioactive lipid sphingosine-1-phosphate (S1P) induced the secretion of several angiogenesis-related proteins including the important inflammatory factor chemokine (C-C motif) ligand 2 (CCL2). In the present study, we investigated the mechanism of S1P-induced CCL2 expression in NB. METHODS: Quantitative real-time PCR and CCL2 ELISA were conducted to detect the mRNA expression and protein secretion of CCL2 in NB cells. Gain and loss of function studies were performed by using specific S1PR antagonists, adenoviral transduction and siRNA transfection. Macrophage F4/80 receptor in NB xenografts was detected by quantitative real-time PCR and immunohistochemistry staining. RESULTS: S1P induced CCL2 mRNA expression and protein secretion in a time- and concentration-dependent manner in NB cells. Blockade of S1P2 signaling using the selective S1P2 antagonist JTE-013 inhibited S1P-induced CCL2 expression. Overexpression of S1P2 by adenoviral transduction increased CCL2 secretion while knockdown of S1P2 by siRNA transfection decreased S1P-induced CCL2 secretion in NB cells. Macrophage infiltration, as detected by F4/80 staining, was significantly decreased in JTE-013-treated NB xenografts. CONCLUSIONS: Taken together, our data for the first time demonstrate that S1P induced the macrophage-recruiting factor CCL2 expression in NB cells via S1P2, providing new insights into the complicated functions of S1P2 in cancer.


Subject(s)
Chemokines, C/genetics , Gene Expression Regulation, Neoplastic , Lysophospholipids/pharmacology , Neuroblastoma/genetics , RNA, Neoplasm/genetics , Sphingosine/analogs & derivatives , Chemokines, C/biosynthesis , Chemokines, C/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Neuroblastoma/metabolism , Neuroblastoma/pathology , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Sphingosine/pharmacology , Tumor Cells, Cultured
8.
Korean J Physiol Pharmacol ; 17(4): 315-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23946691

ABSTRACT

Here, we show that radicicol, a fungal antibiotic, resulted in marked inhibition of inducible nitric oxide synthase (iNOS) transcription by the pancreatic beta cell line MIN6N8a in response to cytokine mixture (CM: TNF-α, IFN-γ, and IL-1ß). Treatment of MIN6N8a cells with radicicol inhibited CM-stimulated activation of NF-κB/Rel, which plays a critical role in iNOS transcription, in a dose-related manner. Nitrite production in the presence of PD98059, a specific inhibitor of the extracellular signal-regulated protein kinase-1 and 2 (ERK1/2) pathway, was dramatically diminished, suggesting that the ERK1/2 pathway is involved in CM-induced iNOS expression. In contrast, SB203580, a specific inhibitor of p38, had no effect on nitrite generation. Collectively, this series of experiments indicates that radicicol inhibits iNOS gene expression by blocking ERK1/2 signaling. Due to the critical role that NO release plays in mediating destruction of pancreatic beta cells, the inhibitory effects of radicicol on iNOS expression suggest that radicicol may represent a useful anti-diabetic activity.

9.
Pediatr Blood Cancer ; 60(9): 1418-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23704073

ABSTRACT

BACKGROUND: Neuroblastoma (NB) is the most common extra-cranial solid tumor in childhood. Poor outcomes for children with advanced disease underscore the need for novel therapeutic strategies. FTY720, an immunomodulating drug approved for multiple sclerosis, has been investigated in oncology with promising preclinical activities. To date, its effect in NB has not been explored. Herein we describe our preclinical experience with FTY720, alone or in combination with topotecan, and its putative mechanism of action in NB. PROCEDURE: MTT assay was performed to assess the effect of FTY720 on cell viability. A NB xenograft model was employed to assess the efficacy of FTY720 on tumor growth. Quantitative real-time PCR and Western blot were employed to determine changes of mRNA and protein expression, respectively. Liquid chromatography/tandem mass spectrometry was used to measure sphingolipid levels. RESULTS: FTY720, but not FTY720-P induced NB cell death. FTY720 inhibited the growth of NB xenografts and enhanced the tumor-suppressive effect of topotecan both in vitro and in vivo. FTY720 significantly inhibited sphingosine kinase 2 (SphK2) mRNA and protein expression in NB cells. Pro-apoptotic sphingosine levels were increased in NB cells and NB xenografts treated with FTY720. FTY720-induced cell death was caspase-independent and involved the dephosphorylation of Akt and BAD at Ser136. CONCLUSIONS: Our data demonstrate that FTY720 has potent preclinical anti-cancer activity in NB. Its unique death signaling mechanism, interference with the sphingolipid pathway, acts cooperatively with that of topotecan, suggesting that FTY720 related molecules may be useful in NB treatment.


Subject(s)
Immunosuppressive Agents , Neuroblastoma/drug therapy , Propylene Glycols , Signal Transduction/drug effects , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Topoisomerase I Inhibitors , Topotecan , Animals , Cell Death , Child , Child, Preschool , Drug Synergism , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/agonists , Immunosuppressive Agents/pharmacology , Infant , Male , Mice , Mice, Nude , Neuroblastoma/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Propylene Glycols/agonists , Propylene Glycols/pharmacology , Sphingosine/agonists , Sphingosine/pharmacology , Topoisomerase I Inhibitors/agonists , Topoisomerase I Inhibitors/pharmacology , Topotecan/agonists , Topotecan/pharmacology , Xenograft Model Antitumor Assays
10.
J Mol Med (Berl) ; 90(8): 971-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22406864

ABSTRACT

We have recently discovered that c-Jun executes a non-transcriptional function to stabilize hypoxia inducible factor 1α (HIF-1α) and that pseudolaric acid B (PAB) accelerates HIF-1α degradation and phosphorylates c-Jun at Ser63/73. In this study, PAB was used as a probe to investigate whether and how the Ser63/73 phosphorylation of c-Jun regulates its functions. The PAB-induced reduction of HIF-1α protein was rescued through supplying additional non-phosphorylated c-Jun. However, c-Jun siRNA, which reduced both the PAB-driven phosphorylated c-Jun and the total c-Jun protein, did not prevent the PAB-induced decrease in HIF-1α. HIF-1α was revealed to be co-immunoprecipitated only with the non-phosphorylated c-Jun. PAB increased the phosphorylated c-Jun while reducing the non-phosphorylated c-Jun at Ser63/73, which impaired its function in stabilizing HIF-1α. Consequently, PAB led to the degradation of HIF-1α, thus resulting in the decreased HIF-1α-dependent expression of mdr-1 and VEGF. We accordingly propose a function-converter model of c-Jun: the Ser63/73 phosphorylation serves as a function converter to convert c-Jun from its non-transcriptional function to its transcriptional function.


Subject(s)
Diterpenes/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , HeLa Cells , Hep G2 Cells , Humans , Immunoprecipitation , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Stability/drug effects , Reverse Transcriptase Polymerase Chain Reaction
11.
Korean J Physiol Pharmacol ; 15(2): 101-6, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21660150

ABSTRACT

We demonstrate that glycoprotein isolated from Dioscorea batatas (GDB) has immunostimulatory effects including macrophage activation. Analysis of infiltration of inflammatory cells into peritoneal cavity showed GDB treatment significantly increased the recruitment of macrophages, lymphocytes, neutrophils, and monocytes into the peritoneal cavity. Treatment of spleen cells isolated from C57BL/6 mice with GDB significantly increased the proliferation of B cells and T cells induced by LPS and ConA, respectively. Treatment with GDB significantly increased the cytolytic capacity of NK cells and macrophages against YAC-1 and B16 cells, respectively. In order to further confirm and investigate the mechanism of GDB on macrophage activation, we analyzed the effects of GDB on the cytokine expression including iNOS, IL-1ß, and TNF-α in mouse macrophage cell line, RAW 264.7 cells. RT-PCR and ELISA showed that GDB increased the expression of IL-1ß, and TNF-α, whereas iNOS was not induced by GDB. Collectively, this series of experiments indicates that GDB stimulates immune system including macrophage activation.

12.
Cancer Prev Res (Phila) ; 4(8): 1325-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21576349

ABSTRACT

Metabolism of sphingolipids into downstream lipid mediators followed by signaling modulates tumor microenvironment and the cancer cells to influence tumor progression. As such, sphingolipid signaling represents a novel way to modulate tumor biology. Neuroblastoma (NB), the most common extracranial solid tumor of childhood, is highly angiogenic and often displays poor prognosis. However, the role of sphingolipid mediators is not known in NB. We found that NB expresses high levels of sphingosine kinase-2, which is essential for the formation of sphingosine-1-phosphate (S1P). S1P induced VEGF expression in SK-N-AS NB cells. The effect occurred at the transcriptional level. Hypoxia in combination with S1P had a synergistic effect on VEGF expression. Strong correlation was detected between S1P receptor-2 (S1P(2)) and VEGF mRNAs in 11 different cell lines and 17 NB tissues. Blockade of S1P(2) with the selective antagonist JTE-013 significantly inhibited S1P-induced VEGF expression. Overexpression and knockdown of S1P(2) in SK-N-AS cells increased or inhibited S1P-induced VEGF secretion, respectively. Interestingly, JTE-013 significantly inhibited tumor growth, VEGF mRNA expression, and induced apoptosis in the NB tumor xenografts. Taken together, our data suggest that enhanced formation of sphingolipid mediator S1P in NB profoundly influences tumor microenvironment by inducing VEGF expression via S1P(2). Modulation of sphingolipid signaling by inhibiting S1P(2) may constitute a novel strategy to control NB.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Neuroblastoma/metabolism , Sphingolipids/metabolism , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay/methods , Humans , Hypoxia/metabolism , In Situ Nick-End Labeling , Lysophospholipids/metabolism , Male , Mice , Mice, Nude , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Vascular Endothelial Growth Factor A/metabolism
13.
Korean J Physiol Pharmacol ; 14(6): 353-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21311674

ABSTRACT

This study demonstrates the ability of magnolol, a hydroxylated biphenyl compound isolated from Magnolia officinalis, to inhibit LPS-induced expression of iNOS gene and activation of NF-κB/Rel in RAW 264.7 cells. Immunohisto-chemical staining of iNOS and Western blot analysis showed magnolol to inhibit iNOS gene expression. Reporter gene assay and electrophoretic mobility shift assay showed that magnolol inhibited NF-κB/Rel transcriptional activation and DNA binding, respectively. Since p38 is important in the regulation of iNOS gene expression, we investigated the possibility that magnolol to target p38 for its anti-inflammatory effects. A molecular modeling study proposed a binding position for magnolol that targets the ATP binding site of p38 kinase (3GC7). Direct interaction of magnolol and p38 was further confirmed by pull down assay using magnolol conjugated to Sepharose 4B beads. The specific p38 inhibitor SB203580 abrogated the LPS-induced NF-κB/Rel activation, whereas the selective MEK-1 inhibitor PD98059 did not affect the NF-κB/Rel. Collectively, the results of the series of experiments indicate that magnolol inhibits iNOS gene expression by blocking NF-κB/Rel and p38 kinase signaling.

14.
Pediatr Blood Cancer ; 54(2): 316-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19824073

ABSTRACT

Wilms tumor is the most common malignant renal tumor in children. However, to date no Wilms tumor mouse model is available due to the lack of Wilms tumor cell lines. Herein for the first time we report an orthotopic xenograft mouse model utilizing the recently described Wilms tumor cell line WiT49. It has a high tumor occurrence rate (85%) without metastasis. Hematoxylin and eosin staining showed it is subcapsular in location and mainly biphasic with stromal and epithelial components while blastemal component is unappreciable. This model provides the prerequisite for the screening and development of new anti-tumor agents for Wilms tumor.


Subject(s)
Cell Line, Tumor , Disease Models, Animal , Wilms Tumor , Animals , Male , Mice , Mice, Nude , Wilms Tumor/pathology
15.
Cancer Res ; 69(19): 7704-12, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19738058

ABSTRACT

Although hypoxia-inducible factor-1alpha (HIF-1alpha) has long been intensively investigated as a drug target by interfering with its expression or transcriptional function, the regulatory mechanisms of HIF-1alpha remain to be further clarified. We report here that c-Jun associates with HIF-1alpha via its oxygen-dependent degradation domain, masks the sites for ubiquitination, and thus protects HIF-1alpha from proteasome-executing degradation. All of these together resulted in the stabilization and accumulation of HIF-1alpha, consequently promoting the transcription of its target gene and driving angiogenesis-related events. The stabilization of HIF-1alpha was dependent on the domains of c-Jun for DNA binding and heterodimerization but independent of the Ser(63/73) phosphorylation that is critical for transcriptional function. These findings highlight a previously unrecognized nontranscriptional function of c-Jun on the one hand and a distinct regulatory mechanism of HIF-1alpha activity on the other, consequently offering profound mechanistic insights into multiple events simultaneously involving both c-Jun and HIF-1alpha in tumor progression.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Endothelial Cells/metabolism , Female , HeLa Cells , Humans , Neovascularization, Pathologic/metabolism , Oxygen/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-jun/genetics , RNA, Small Interfering/genetics , Transcription, Genetic , Ubiquitination , Uterine Cervical Neoplasms/metabolism
16.
Cancer Lett ; 276(2): 171-9, 2009 Apr 18.
Article in English | MEDLINE | ID: mdl-19131156

ABSTRACT

Sphingosine-1-phosphate (S1P) is an important regulator of cellular functions via interaction with its receptors S1P(1-5). To date, nothing is known about the S1P receptor expression and the effects of S1P signaling in Wilms tumor. In this study, we found ubiquitous expression of S1P receptors in Wilms tumor specimens and cell lines. We demonstrated that S1P(1) acted as a promigratory modulator by employing S1P(1) antagonist VPC44116, S1P(1) siRNA and adenoviral transduction in Wilms tumor cells. Further, we clarified that S1P(1)-mediated migration occurred via Gi coupling and activation of PI3K and Rac1. In addition, S1P stimulated WiT49 cell invasion through S1P(1)/Gi signaling pathway. We consider that targeting S1P(1) may be a point of therapeutic intervention in Wilms tumor.


Subject(s)
Kidney Neoplasms/pathology , Lysophospholipids/physiology , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Wilms Tumor/pathology , Cell Line, Tumor , Cell Movement , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Humans , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/physiology , RNA, Small Interfering/pharmacology , Receptors, Lysosphingolipid/analysis , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/physiology , Sphingosine/physiology , rac1 GTP-Binding Protein/physiology
17.
J Urol ; 181(3): 1347-52, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19157443

ABSTRACT

PURPOSE: Cyclooxygenase-2 has been reported to be ubiquitously expressed in Wilms tumor, the most common malignant renal tumor in children. However, to our knowledge the regulation mechanism of cyclooxygenase-2 expression remains unexplored. MATERIALS AND METHODS: Quantitative real-time polymerase chain reaction and Western blot were performed to detect cyclooxygenase-2 mRNA and protein expression in WiT49 cells upon stimulation by S1P (Biomol(R)), and S1P(2) and cyclooxygenase-2 mRNA expression in 10 freshly frozen Wilms tumor tissues and matched normal tissues. Over expression, blockade and down-regulation of S1P(2) were determined using adenoviral transduction, the S1P(2) antagonist JTE-013 (Tocris Bioscience, Ellisville, Missouri) and small interfering RNA (Dharmacon, Lafayette, Colorado) transfection, respectively. The prostaglandin E(2) level in WiT49 cells was determined by gas chromatography/mass spectrometry. RESULTS: S1P induced cyclooxygenase-2 mRNA and protein expression in WiT49 cells in a concentration dependent manner. Over expression of S1P(2) in WiT49 cells led to a significant increase in cyclooxygenase-2 mRNA and protein expression as well as subsequent prostaglandin E(2) synthesis. In addition, pretreatment of those cells that over expressed S1P(2) with the S1P(2) selective antagonist JTE-013 completely blocked S1P induced cyclooxygenase-2 protein expression. In accordance with these results silencing S1P(2) in WiT49 cells down-regulated S1P induced cyclooxygenase-2 expression. Further research in 10 Wilms tumor specimens showed that S1P(2) mRNA is greatly increased in Wilms tumor. CONCLUSIONS: S1P induced cyclooxygenase-2 expression in Wilms tumor and this effect was mediated by S1P(2). This finding extends the biological function of S1P(2) and provides the biochemical basis for developing inhibitors targeting the S1P/cyclooxygenase-2 signaling pathway.


Subject(s)
Cyclooxygenase 2/biosynthesis , Kidney Neoplasms/enzymology , Receptors, Lysosphingolipid/physiology , Wilms Tumor/enzymology , Humans , Signal Transduction , Sphingosine-1-Phosphate Receptors , Tumor Cells, Cultured
18.
Mol Cancer Res ; 6(10): 1649-56, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18922980

ABSTRACT

Connective tissue growth factor (CTGF), a member of the CCN family of secreted matricellular proteins, regulates fibrosis, angiogenesis, cell proliferation, apoptosis, tumor growth, and metastasis. However, the role of CTGF and its regulation mechanism in Wilms' tumor remains largely unknown. We found that the bioactive lipid sphingosine-1-phosphate (S1P) induced CTGF expression in a concentration- and time-dependent manner in a Wilms' tumor cell line (WiT49), whereas FTY720-phosphate, an S1P analogue that binds all S1P receptors except S1P2, did not. Further, the specific S1P2 antagonist JTE-013 completely inhibited S1P-induced CTGF expression, whereas the S1P1 antagonist VPC44116 did not, indicating that this effect was mediated by S1P2. This was confirmed by adenoviral transduction of S1P2 in WiT49 cells, which showed that overexpression of S1P2 increased the expression of CTGF. Induction of CTGF by S1P was sensitive to ROCK inhibitor Y-27632 and c-Jun NH2-terminal kinase inhibitor SP600125, suggesting the requirement of RhoA/ROCK and c-Jun NH2-terminal kinase pathways for S1P-induced CTGF expression. Interestingly, the expression levels of CTGF were decreased in 8 of 10 Wilms' tumor tissues compared with matched normal tissues by quantitative real-time PCR and Western blot analysis. In vitro, human recombinant CTGF significantly inhibited the proliferation of WiT49 cells. In addition, overexpression of CTGF resulted in significant inhibition of WiT49 cell growth. Taken together, these data suggest that CTGF protein induced by S1P2 might act as a growth inhibitor in Wilms' tumor.


Subject(s)
Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Neoplasms/pathology , Receptors, Lysosphingolipid/metabolism , Wilms Tumor/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Connective Tissue Growth Factor , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Lysophospholipids/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Suppression, Genetic/drug effects , Wilms Tumor/enzymology , Wilms Tumor/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
19.
Biosci Biotechnol Biochem ; 71(9): 2214-22, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17827681

ABSTRACT

In this study we purified a fibrinolytic enzyme from the culture supernatant of Flammulina velutipes mycelia by ion exchange and gel filtration chromatographies, it was designated as F. velutipes protease (FVP-I). This purification protocol resulted in 18.52-fold purification of the enzyme at a final yield of 0.69%. The molecular mass of the purified enzyme was estimated to be 37 kDa by SDS-PAGE, fibrin-zymography and size exclusion by FPLC. This protease effectively hydrolyzed fibrin, preferentially digesting alpha-chain over beta-and gamma-gamma chain. Optimal protease activity was found to occur at a pH of 6.0 and a temperature of 20 to 30 degrees C. The protease activity was inhibited by Cu2+, Fe2+ and Fe3+ ions, but was found to be enhanced by Mn2+ and Mg2+ ions. Furthermore, FVP-I activity was potently inhibited by EDTA and EGTA, and it was found to exhibit a higher specificity for chromogenic substrate S-2586 for chymotrypsin, indicating that the enzyme is a chymotrypsin-like metalloprotease. The first 20 amino acid residues of the N-terminal sequence of FVP-I were LTYRVIPITKQAVTEGTELL. They had a high degree of homology with hypothetical protein CC1G_11771, GeneBank Accession no. EAU86463.


Subject(s)
Agaricales/enzymology , Endopeptidases/isolation & purification , Endopeptidases/metabolism , Fibrinolysis , Amino Acid Sequence , Chromatography, Gel , Endopeptidases/chemistry , Enzyme Activation/drug effects , Hydrogen-Ion Concentration , Metals/pharmacology , Molecular Sequence Data , Molecular Weight , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Temperature
20.
J Ethnopharmacol ; 108(1): 38-45, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16806764

ABSTRACT

We demonstrate that KIOM-79, combined extracts obtained from Magnolia officinalis, Pueraria lobata, Glycyrrhiza uralensis, and Euphorbia pekinensis, inhibits LPS-induced expression of iNOS gene in RAW 264.7 cells. Treatment of RAW 264.7 cells with KIOM-79 inhibited LPS-stimulated nitric oxide production in a dose-related manner. Immunohisto-chemical staining of iNOS and RT-PCR analysis showed that the decrease of NO was due to the inhibition of iNOS gene expression. Immunostaining of p65, EMSA, and reporter gene assay showed that KIOM-79 inhibited NF-kappa/Rel nuclear translocation, DNA binding, and transcriptional activation, respectively. Western immunoblot analysis of p38 kinase showed KIOM-79 significantly inhibited the phosphoylation of p38 kinase which is important in the regulation of iNOS gene expression. Collectively, this series of experiments indicates that KIOM inhibits iNOS gene expression by blocking NF-kappa/Rel and p38 kinase signaling. Due to the critical role that NO release plays in mediating inflammatory responses, the inhibitory effects of KIOM-79 on iNOS suggest that KIOM-79 may represent a useful anti-inflammatory agent.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/drug effects , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-rel/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Enzyme Activation/drug effects , Euphorbia/chemistry , Female , Gene Expression Regulation, Enzymologic/drug effects , Glycyrrhiza uralensis/chemistry , Macrophages/enzymology , Macrophages/metabolism , Magnolia/chemistry , Mice , NF-kappa B/metabolism , Phytotherapy , Plant Extracts/chemistry , Proto-Oncogene Proteins c-rel/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...