Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Proc Natl Acad Sci U S A ; 121(28): e2320070121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968120

ABSTRACT

Hedgehog (Hh) signaling, an evolutionarily conserved pathway, plays an essential role in development and tumorigenesis, making it a promising drug target. Multiple negative regulators are known to govern Hh signaling; however, how activated Smoothened (SMO) participates in the activation of downstream GLI2 and GLI3 remains unclear. Herein, we identified the ciliary kinase DYRK2 as a positive regulator of the GLI2 and GLI3 transcription factors for Hh signaling. Transcriptome and interactome analyses demonstrated that DYRK2 phosphorylates GLI2 and GLI3 on evolutionarily conserved serine residues at the ciliary base, in response to activation of the Hh pathway. This phosphorylation induces the dissociation of GLI2/GLI3 from suppressor, SUFU, and their translocation into the nucleus. Loss of Dyrk2 in mice causes skeletal malformation, but neural tube development remains normal. Notably, DYRK2-mediated phosphorylation orchestrates limb development by controlling cell proliferation. Taken together, the ciliary kinase DYRK2 governs the activation of Hh signaling through the regulation of two processes: phosphorylation of GLI2 and GLI3 downstream of SMO and cilia formation. Thus, our findings of a unique regulatory mechanism of Hh signaling expand understanding of the control of Hh-associated diseases.


Subject(s)
Dyrk Kinases , Hedgehog Proteins , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Signal Transduction , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3 , Animals , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Mice , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Humans , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Cell Proliferation , Cilia/metabolism , Smoothened Receptor/metabolism , Smoothened Receptor/genetics , Nuclear Proteins , Repressor Proteins
2.
J Cell Sci ; 135(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35582972

ABSTRACT

Neural precursor cell-expressed developmentally down-regulated 8 (NEDD8), an ubiquitin-like protein, is an essential regulator of the DNA damage response. Numerous studies have shown that neddylation (conjugation of NEDD8 to target proteins) dysfunction causes several human diseases, such as cancer. Hence clarifying the regulatory mechanism of neddylation could provide insight into the mechanism of genome stability underlying the DNA damage response (DDR) and carcinogenesis. Here, we demonstrate that dual-specificity tyrosine-regulated kinase 2 (DYRK2) is a novel regulator of neddylation and maintains genome stability. Deletion of DYRK2 leads to persistent DNA double-strand breaks (DSBs) and subsequent genome instability. Mechanistically, DYRK2 promotes neddylation through forming a complex with NAE1, which is a component of NEDD8-activating enzyme E1, and maintaining its protein level by suppressing polyubiquitylation. The present study is the first to demonstrate that DYRK2 controls neddylation and is necessary for maintaining genome stability. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cullin Proteins , DNA Damage , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Cullin Proteins/metabolism , DNA Damage/genetics , Genomic Instability/genetics , Humans , NEDD8 Protein/genetics , NEDD8 Protein/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Dyrk Kinases
3.
Cancer Res ; 81(2): 414-425, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33318039

ABSTRACT

Expression of human protein kinase C delta (PKCδ) protein has been linked to many types of cancers. PKCδ is known to be a multifunctional PKC family member and has been rigorously studied as an intracellular signaling molecule. Here we show that PKCδ is a secretory protein that regulates cell growth of liver cancer. Full-length PKCδ was secreted to the extracellular space in living liver cancer cells under normal cell culture conditions and in xenograft mouse models. Patients with liver cancer showed higher levels of serum PKCδ than patients with chronic hepatitis or liver cirrhosis or healthy individuals. In liver cancer cells, PKCδ secretion was executed in an endoplasmic reticulum (ER)-Golgi-independent manner, and the inactivation status of cytosolic PKCδ was required for its secretion. Furthermore, colocalization studies showed that extracellular PKCδ was anchored on the cell surface of liver cancer cells via association with glypican 3, a liver cancer-related heparan sulfate proteoglycan. Addition of exogenous PKCδ activated IGF-1 receptor (IGF1R) activation and subsequently enhanced activation of ERK1/2, which led to accelerated cell growth in liver cancer cells. Conversely, treatment with anti-PKCδ antibody attenuated activation of both IGF1R and ERK1/2 and reduced cell proliferation and spheroid formation of liver cancer cells and tumor growth in xenograft mouse models. This study demonstrates the presence of PKCδ at the extracellular space and the function of PKCδ as a growth factor and provides a rationale for the extracellular PKCδ-targeting therapy of liver cancer. SIGNIFICANCE: PKCδ secretion from liver cancer cells behaves as a humoral growth factor that contributes to cell growth via activation of proliferative signaling molecules, which may be potential diagnostic or therapeutic targets.


Subject(s)
Biomarkers, Tumor/metabolism , Culture Media, Conditioned/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C-delta/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Movement , Cell Proliferation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation , Prognosis , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Elife ; 92020 08 06.
Article in English | MEDLINE | ID: mdl-32758357

ABSTRACT

Mammalian Hedgehog (Hh) signaling plays key roles in embryogenesis and uniquely requires primary cilia. Functional analyses of several ciliogenesis-related genes led to the discovery of the developmental diseases known as ciliopathies. Hence, identification of mammalian factors that regulate ciliogenesis can provide insight into the molecular mechanisms of embryogenesis and ciliopathy. Here, we demonstrate that DYRK2 acts as a novel mammalian ciliogenesis-related protein kinase. Loss of Dyrk2 in mice causes suppression of Hh signaling and results in skeletal abnormalities during in vivo embryogenesis. Deletion of Dyrk2 induces abnormal ciliary morphology and trafficking of Hh pathway components. Mechanistically, transcriptome analyses demonstrate down-regulation of Aurka and other disassembly genes following Dyrk2 deletion. Taken together, the present study demonstrates for the first time that DYRK2 controls ciliogenesis and is necessary for Hh signaling during mammalian development.


Subject(s)
Cilia/metabolism , Hedgehog Proteins/physiology , Organogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Signal Transduction , Animals , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Dyrk Kinases
5.
Int J Oncol ; 56(6): 1529-1539, 2020 06.
Article in English | MEDLINE | ID: mdl-32236621

ABSTRACT

Dual specificity tyrosine­phosphorylation­regulated kinase 2 (DYRK2) is a protein kinase that functions as a novel tumor suppressor. Previous studies have reported that DYRK2 expression is decreased in colorectal cancer compared with adjacent non­tumor tissues. However, the regulatory mechanisms by which the expression of DYRK2 is diminished remain unknown. The aim of the present study was to determine the regulatory mechanisms of DYRK2 expression. The present study identified the promoter regions of the DYRK2 gene and demonstrated that they contained CpG islands in human cancer cells. In addition, the DYRK2 promoter region exhibited a higher level of methylation in colorectal cancer tissues compared with healthy tissues from clinical samples. DYRK2 expression was increased at the mRNA and protein level in colorectal cancer cell lines by treatment with 5­Azacytidine, a demethylating agent. The results further demonstrated that knockdown of DNA methyltransferase (DNMT) 1 elevated DYRK2 expression in colorectal cancer cell lines. A colitis­related mouse carcinogenesis model also exhibited a lower DYRK2 level in colorectal cancer tissues compared with adjacent non­tumor tissues. In this model, nuclear staining of DNMT1 was detected in colorectal cancer cells, whereas a cytoplastic distribution pattern of DNMT1 staining was exhibited in healthy tissue. Overall, these findings suggested that DYRK2 expression was downregulated via transcriptional regulation by DNMT1 to elevate the proliferation of colorectal cancer cells.


Subject(s)
Colorectal Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , CpG Islands , DNA Methylation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Mice , Middle Aged , Neoplasm Transplantation , Promoter Regions, Genetic , Transcription, Genetic , Dyrk Kinases
6.
Anticancer Res ; 38(3): 1311-1316, 2018 03.
Article in English | MEDLINE | ID: mdl-29491054

ABSTRACT

BACKGROUND: CD147 (basigin/emmprin) is expressed on the surface of carcinoma cells. MATERIALS AND METHODS: For studying the efficacy of CD147-targeting medicine on CD147-expressing cells, we studied the effect of anti-CD147-labeled polymeric micelles (CD147ab micelles) that encapsulated a conjugate of doxorubicin with glutathione (GSH-DXR), with specific accumulation and cytotoxicity against CD147-expressing A431 human epidermoid carcinoma cells, Ishikawa human endometrial adenocarcinoma cells, and PC3 human prostate carcinoma cells. RESULTS: By treatment of each cell type with CD147ab micelles for 1 h, a specific accumulation of CD147ab micelles in CD147-expressing cells was observed. In addition, the cytotoxicity of GSH-DXR-encapsulated micelles against each cell type was measured by treatment of the micelles for 1 h. The cytotoxic effect of CD147ab micelles carrying GSH-DXR was 3- to 10-fold higher for these cells than that of micelles without GSH-DXR. CONCLUSION: These results suggest that GSH-DXR-encapsulated CD147ab micelles could serve as an effective drug delivery system to CD147-expressing carcinoma cells.


Subject(s)
Antibodies/chemistry , Basigin/metabolism , Doxorubicin/analogs & derivatives , Micelles , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Antibodies/immunology , Antibodies/pharmacology , Basigin/antagonists & inhibitors , Basigin/immunology , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/chemistry , Doxorubicin/pharmacology , Glutathione/analogs & derivatives , Glutathione/chemistry , Humans , Mice
7.
Cancer Sci ; 109(1): 112-120, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29095550

ABSTRACT

Suppression of bone metastasis can improve patient quality of life. Current drugs for bone metastasis have been shown to prolong progression-free survival but not overall survival; therefore, other potential therapeutic targets for bone metastasis should be investigated. Cell-surface antigens, such as CD24, have been recently shown to be involved in the metastasis of various cancers. However, whether CD24 plays a role in bone metastasis of lung cancer remains unknown. To observe metastasis of lung cancer cells by imaging technology, we introduced a near-infrared fluorescent protein, iRFP720, into a bone-seeking subclone established from lung cancer cells, HARA-B4 cells. The anchorage-independent growth of these cells was then evaluated by colony formation assays. We also compared cancer cell tropism to bone tissue with HARA-B4 cells in the presence or absence of CD24 by cell adhesion assays. To clarify the role of CD24 in bone metastasis, we intracardially injected CD24-knockdown HARA-B4 cells into mice and monitored metastasis through detection of iRFP720 using an in vivo imaging system. CD24-knockdown HARA-B4 cells in vitro showed reduced anchorage-independent growth and cancer cell tropism to bone. Bone metastasis was diminished in mice inoculated with CD24-knockdown HARA-B4 cells, which was rescued by add-back of CD24 in cells. Our findings indicate that iRFP720 is effective for in vivo imaging analysis of bone metastasis and that downregulation of CD24 suppresses bone metastasis of lung cancer cells. These findings collectively indicate that CD24 may be considered a promising new therapeutic candidate for the prevention of bone metastasis of lung cancer.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , CD24 Antigen/metabolism , Down-Regulation , Lung Neoplasms/metabolism , Animals , Bone Neoplasms/genetics , CD24 Antigen/genetics , Cell Adhesion , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Luminescent Proteins/metabolism , Lung Neoplasms/genetics , Mice , NIH 3T3 Cells , Neoplasm Transplantation
8.
FEBS Lett ; 591(6): 842-853, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28194753

ABSTRACT

The genome of eukaryotic cells is frequently exposed to damage by various genotoxins. Phosphorylation of histone H2AX at Serine 139 (γ-H2AX) is a hallmark of DNA damage. RNF8 monoubiquitinates γ-H2AX with the Lys63-linked ubiquitin chain to tether DNA repair molecules at DNA lesions. A high-throughput screening identified RNF8 as a binding partner of dual-specificity tyrosine phosphorylation-regulated kinase 2 (DYRK2). Notably, DNA damage-induced monoubiquitination of γ-H2AX is impaired in DYRK2-depleted cells. The foci formation of p53-binding protein 1 at DNA double-strand break sites is suppressed in DYRK2 knockdown cells, which fail to repair the DNA damage. A homologous recombination assay showed decreased repair efficiency in DYRK2-depleted cells. Our findings indicate direct interaction of DYRK2 with RNF8 in regulating response to DNA damage.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Cell Line, Tumor , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , HEK293 Cells , Histones/metabolism , Humans , Immunoblotting , Microscopy, Confocal , Mutation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Dyrk Kinases
9.
Hepatol Res ; 45(3): 315-25, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24802089

ABSTRACT

AIM: Despite an increasing demand, blood products are not always safe because most are derived from blood donations. One possible solution is the development and commercialization of recombinant fibrinogen, but this process remains poorly developed. This study aimed to develop an effective production system for producing risk-free fibrinogen using human hepatocellular cell lines and serum-free media. METHODS: Three human liver cancer cell lines (HepG2, FLC-4 and FLC-7) were cultivated in a serum-supplemented medium or two serum-free media (ASF104N and IS-RPMI) to compare their fibrinogen secretion abilities. Fibrinogen subunit gene expression was estimated by quantitative polymerase chain reaction. Massive fibrinogen production was induced using a 5-mL radial flow bioreactor (RFB) while monitoring glucose metabolism. Subsequently, fibrinogen's biochemical characteristics derived from these cells were analyzed. RESULTS: FLC-7 cell culture combined with IS-RPMI resulted in significantly better fibrinogen production (21.6 µg/10(7) cells per day). ASF104N had more positive effects on cell growth compared with IS-RPMI, whereas fibrinogen production was more efficient with IS-RPMI than with ASF104N. Changing the medium from ASF104N to IS-RPMI led to significantly increased fibrinogen gene expression and glucose consumption. In the RFB culture, the fibrinogen secretion rate of FLC-7 cells reached 0.73 µg/mL per day during a 42-day cultivation period. The subunit composition and clot formation activity of FLC-7 cell-derived fibrinogen corresponded to those of plasma fibrinogen. CONCLUSION: The FLC-7 cell culture system is suitable for large-scale fibrinogen preparation production.

10.
J Obstet Gynaecol Res ; 38(9): 1211-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22563698

ABSTRACT

AIM: CD147 is a membrane glycoprotein that is expressed in various cancer cells and is involved in tumor invasion and metastasis by inducing stromal fibroblastic cells to produce matrix metalloproteinases. This study was carried out to evaluate the correlation between CD147 expression and various clinicopathologic parameters, including histological grade and prognosis in a small sample set of human ovarian cancer patients. MATERIAL AND METHODS: Paraffin-embedded surgical tissue samples from 25 patients with ovarian serous and endometrioid adenocarcinoma were stained with anti-CD147 antibody (monoclonal antibody 12C3: MoAb 12C3) for immunohistochemical analysis. RESULTS: CD147 protein was expressed in 84.0% (21 of 25 cases) of cancerous lesions, but not in normal lesions. CD147 expression by ovarian cancer cells was inversely correlated with overall survival. There was no correlation between CD147 expression and histological grade. CONCLUSIONS: These results suggest that measurement of CD147 expression may enhance the understanding of the pathophysiology of epithelial ovarian cancer.


Subject(s)
Antibodies, Monoclonal , Basigin/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Endometrioid/metabolism , Ovarian Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/pathology , Female , Humans , Immunohistochemistry , Japan/epidemiology , Kaplan-Meier Estimate , Middle Aged , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovary/pathology , Pilot Projects
11.
Int J Oncol ; 40(6): 2122-30, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22447231

ABSTRACT

Ovarian clear cell carcinoma (OCCC) has several significant characteristics based on molecular features that are distinct from those of ovarian high-grade serous carcinoma. Cellular glycogen accumulation is the most conspicuous feature of OCCC and in the present study its metabolic mechanism was investigated. The amount of glycogen in cells cultured under hypoxia increased significantly and approximately doubled after 48 h (P<0.01) compared to that under normoxic conditions. Periodic acid-Schiff positive staining also demonstrated intracellular glycogen storage. Western blot analysis revealed that HIF1α, which was overexpressed and stabilized under hypoxic conditions, led to an increase in the levels of cellular glycogen synthase 1, muscle type (GYS1), and conversely to a decrease in inactive phosphorylated GYS1 at serine (Ser) 641. Additional increases were observed in both protein phosphatase 1, which dephosphorylates and thereby induces GYS1 enzyme activity, and glycogen synthase kinase 3 beta (GSK3ß) phosphorylated at Ser9, which is inactive on phosphorylation of GYS1 and subsequently induces its enzyme activity. By contrast, the level of PYGM-b decreased. These results indicated that the glycogen accumulation under a hypoxic environment resulted in the promotion of glycogen synthesis, but did not lead to inhibition of glycogen degradation and/or consumption. Under hypoxic conditions, HAC2 cells showed activation of the PI3K/AKT pathway caused by a mutation in exon 20 of PIK3CA, encoding the catalytic subunit p110α of PI3K. The resulting activation of AKT (phosphoSer473) also plays a role as a central enhancer in glycogen synthesis through suppression of GSK3ß via phosphorylation at Ser9. Hypoxia decreased the cytocidal activity of cisplatin and doxorubicin to various degrees. In conclusion, the hypoxic conditions together with HIF1 expression and stabilization increased the intracellular glycogen contents and resistance to the anticancer drugs.


Subject(s)
Carcinoma/metabolism , Glycogen/biosynthesis , Ovarian Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Base Sequence , Cell Hypoxia/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Class I Phosphatidylinositol 3-Kinases , DNA Mutational Analysis , Doxorubicin/pharmacology , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/genetics
12.
Mol Genet Metab ; 104(4): 566-73, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21982629

ABSTRACT

Pompe disease (glycogen storage disease type II) is an autosomal recessive myopathic disorder arising from the deficiency of lysosomal acid α-glucosidase (GAA). Activation of autophagy is a key pathophysiological feature in skeletal muscle fibers and fibroblasts from patients with Pompe disease. The accumulation of autophagic vacuoles has been shown to interfere with the efficacy of enzyme replacement therapy with recombinant human GAA. However, the induction mechanism of autophagy in Pompe disease is still unclear. In this study, we show that misfolded GAA-induced endoplasmic reticulum (ER) stress triggers autophagy in a manner regulated by p38 MAPK signaling pathways in fibroblasts from late-onset patients with Pompe disease. By studying normal fibroblasts and patient fibroblasts carrying a c.546G>T mutation, we uncovered that mutant GAA was rapidly degraded by proteasome. In addition, we found both activation of ER stress response and autophagy in these patient fibroblasts. Treatment with N-butyl-deoxynojirimycin (NB-DNJ), which acts as a pharmacological chaperone for certain mutant forms of GAA, led to attenuation of not only ER stress, but also autophagy in patient fibroblasts. Levels of phosphorylated p38 MAPK observed in patient fibroblasts were decreased after treatment with NB-DNJ. The autophagic response in patient fibroblasts was also negatively regulated by treatment with the p38 MAPK inhibitor SB203580. These findings define a critical role for ER stress in the activation of autophagy due to GAA mutation, and provide evidence that chaperone therapy may be a useful treatment for alleviation of autophagy in Pompe disease patients carrying a chaperon-responsive mutation.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Fibroblasts/metabolism , Glycogen Storage Disease Type II/metabolism , Point Mutation , alpha-Glucosidases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , Cells, Cultured , DNA Mutational Analysis , Enzyme Activation , Enzyme Activators/pharmacology , Fibroblasts/drug effects , Fibroblasts/enzymology , MAP Kinase Signaling System , Phosphoproteins/metabolism , Phosphorylation , Protein Folding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Unfolded Protein Response , alpha-Glucosidases/metabolism
13.
Int J Oncol ; 39(5): 1327-36, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21785823

ABSTRACT

In vitro tumor growth in a three-dimensional (3D) architecture has been demonstrated to play an important role in biology not only for developmental organogenesis and carcinogenesis, but also for analyses on reconstitution and maintenance in a variety of biological environments surrounding the cells. In addition to providing architectural similarity to living organisms, 3D culture with a radial flow bioreactor (RFB) can also closely mimic the living hypoxic microenvironment under which specific organogenesis or carcinogenesis occurs. The findings of the present study under the RFB culture conditions show that cancer cells underwent a shift from aerobic to hypoxic energy metabolism, in addition to protein expression to maintain the 3D structure. In RFB-cultured cells, protein stability of hypoxia-inducible factor 1 (HIF1) α, a subunit of HIF1, was increased without upregulation of its mRNA. Under these conditions, PHD2, HIF-prolyl-4-hydroxy-lase 2 and a HIF1 downstream enzyme, were stabilized without affecting the mRNA levels via downregulation of FK506-binding protein 8. PHD2 accumulation, which occurred concomitant with HIF1 stabilization, may have compensated for the lack of oxygen under hypoxic conditions to regulate the HIF levels. 3D-culture-induced overexpression of carbonic anhydrase (another representative HIF downstream enzyme) was found to occur independently of cell density in RFB--cultured cells, suggesting that the RFB provided an adequately hypoxic microenvironment for the cultured cells. From these results, it was hypothesized that the key factors are regulatory molecules, which stabilize and degrade HIF molecules, thereby activating the HIF1 pathway under a hypoxic milieu.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Microenvironment/drug effects , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Carcinoma, Squamous Cell/genetics , Cell Culture Techniques , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Nude , Spheroids, Cellular , Squamous Cell Carcinoma of Head and Neck , Transcriptional Activation/genetics , Tumor Cells, Cultured , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
14.
Eur J Haematol ; 85(1): 68-75, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20192985

ABSTRACT

OBJECTIVES: Bortezomib (PS-341; Velcade), a proteasome inhibitor, is used as a therapeutic agent for multiple myeloma. Bortezomib has been shown to strongly induce osteoblast differentiation and elevate the levels of osteoblast-related differentiation markers in the serum of patients with myeloma. Bortezomib also reportedly increases the activity of the transcription factor, Runx2. However, the mechanism of action by which bortezomib-elevated Runx2 activity mediates osteoblast differentiation remains unclear. On the other hand, fibroblast growth factor 2 (FGF-2) is found at high levels in patients with multiple myeloma. We previously reported that FGF-2 reduces the levels of the transcriptional coactivator with PDZ-binding motif (TAZ). We therefore investigated the effects of bortezomib on TAZ protein levels in the presence of FGF-2. METHODS: Osteoblastic MC3T3-E1 cells were treated with different concentrations of bortezomib in the presence or absence of FGF-2 and various biologic responses were investigated by immunoblotting, RT-PCR, quantitative PCR, and alizarin red staining. RESULTS: We found that bortezomib inhibited FGF-2-induced reduction of TAZ levels through a pathway other than that used for proteasome inhibition, while maintaining TAZ function, which in turn, enhanced the expression of Runx2-transcribed osteogenic differentiation markers. Bortezomib also suppressed the antimineralization effect of FGF-2. CONCLUSIONS: These findings suggest that bortezomib inhibited FGF-2-induced reduction of TAZ and consequently stimulated osteogenic differentiation independently of proteasome inhibition. These findings may contribute to elucidate the osteolytic mechanism in multiple myeloma, and to the development of new drugs for multiple myeloma and other osteolytic diseases.


Subject(s)
Boronic Acids/pharmacology , Fibroblast Growth Factor 2/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Proteasome Inhibitors , Pyrazines/pharmacology , Transcription Factors/metabolism , 3T3 Cells , Acyltransferases , Animals , Base Sequence , Boronic Acids/administration & dosage , Bortezomib , Cell Differentiation/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , DNA Primers/genetics , Humans , Mice , Models, Biological , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Osteoblasts/cytology , Osteogenesis/drug effects , Osteolysis/drug therapy , Osteolysis/etiology , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacology , Pyrazines/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Transcription Factors/genetics
15.
Int J Oncol ; 36(1): 77-83, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19956835

ABSTRACT

It was confirmed that CD147 (Emmprin) was expressed on the cell surface of carcinoma cells. For the purpose of studying the efficacy of a CD147-targeting agent on CD147-expressing carcinoma cells, we investigated the effect of a conjugate of glutathione-doxorubicin (GSH-DXR) encapsulated in an anti-CD147 antibody-labeled liposome (aCD147ab-liposome) in terms of specific accumulation and cytotoxicity in CD147-expressing human carcinoma cells. Expression of CD147 was not observed in many normal human tissues. However, slight expression of CD147 in kidney, prostate and breast tissues was observed. By contrast, high-level expression of CD147 in all carcinoma cells such as A431, PC3 and Ishikawa cell lines was confirmed by fluorescent microscopy and Western blot analysis. Specific accumulation of the aCD147ab-liposome in the above-described CD147-expressing cells was observed. GSH-DXR encapsulated in an aCD147ab-liposome expressed specific cytotoxicity against these carcinoma cells. These results suggested that target chemotherapy of GSH-DXR encapsulated in an aCD147ab-liposome on CD147-expressing carcinoma cells was effective.


Subject(s)
Antibodies, Monoclonal/chemistry , Basigin/chemistry , Carcinoma/metabolism , Doxorubicin/analogs & derivatives , Gene Expression Regulation, Neoplastic , Glutathione Transferase/chemistry , Glutathione/analogs & derivatives , Liposomes/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Doxorubicin/chemistry , Doxorubicin/pharmacology , Glutathione/chemistry , Glutathione/pharmacology , Humans , Mice , Time Factors , Tissue Distribution
16.
Int J Oncol ; 34(5): 1433-48, 2009 May.
Article in English | MEDLINE | ID: mdl-19360357

ABSTRACT

To confirm the usefulness of the radial flow type bioreactor (RFB) for a three-dimensional (3D) culture system, which provides a tissue architecture and molecular function mimicking the in vivo environment, molecular expression in the A431 human squamous carcinoma cell line during culture were analyzed under the physically different environments of 3D culture in the RFB, 2D culture in a monolayer as well as in nude mice. Time-dependent accumulation of autocrine transforming growth factor (TGF) beta1 was found in spent culture media obtained only from 3D cultured A431 cancer cells, which grew well with a stratified-sheet morphology. Cells in the RFB overexpressed matrix metalloproteinase 7 (MMP7) and showed an increased release of soluble 80-kDa fragments of E-cadherin into the media time-dependently, resulting in the reduction of E-cadherin protein at the cell surface without down-regulation of the mRNA. beta-Catenin and its nuclear partner, LEF1, were up-regulated and Wnt protein secretion was also accelerated. Additional up-regulation of the transcriptional factors, HMGA2 and down-stream Slug, was noted. TGFbeta1-dependent, MMP7-mediated up-regulation of beta-catenin/LEF1 signaling and TGFbeta1-activated HMGA2 pathways consequently converged with Slug overexpression, due to disassembly and further repression of E-cadherin expression, which was reproducible in the epithelial mesenchymal transition process without any manipulation. Other transcriptional factors, Notch/HEY1 and NF-kappaB, were also up-regulated in 3D-cultured cells. These signals recruited molecules related to extracellular matrix-cell remodeling and angiogenesis. Expression of several representative molecules in the 3D cultured cells was parallel with that in xenotransplanted A431 tumor tissues in nude mice. 3D culture of tumor cells in the RFB is a useful tool for cancer experimental biology and evaluation of cancer therapeutic-like systems in nude mice.


Subject(s)
Bioreactors , Cell Transdifferentiation/genetics , Matrix Metalloproteinase 7/physiology , Neoplasms/pathology , Smad Proteins/physiology , Transforming Growth Factor beta1/physiology , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Culture Techniques/methods , Cell Transdifferentiation/physiology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/physiology , Gene Expression Regulation, Neoplastic , Humans , Male , Matrix Metalloproteinase 7/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Xenograft Model Antitumor Assays
17.
Anal Biochem ; 377(1): 77-82, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18358228

ABSTRACT

Certain proteins insoluble in aqueous salt solutions are difficult to separate from impurities by immunoaffinity techniques, even when the proteins are solubilized with denaturants due to interference of the antigen-antibody reaction. Representative examples of such proteins are the ubiquitin-protein conjugates that accumulate in neuronal tissues of neurodegenerative diseases, the hallmark of such disorders. In this study, we developed a novel sample preparation method comprising two successive steps: Sodium dodecyl sulfate (SDS) removal from the SDS-containing extracts and renaturation of the denatured proteins. The application of this method was tested on ubiquitin-protein conjugates in the brains of Niemann-Pick type C disease mouse and in heat-shocked K562 erythroleukemia cells. The ubiquitin-protein conjugates in both cases are insoluble in Tris-buffered saline but soluble in 2% SDS. The SDS-solubilized fractions prepared from each of the samples were further pretreated by the method mentioned above, and the ubiquitin-protein conjugates were efficiently immunoprecipitated with the anti-ubiquitin antibody from them. This method was also applied successfully to the immunoprecipitation of flotillin-1, a lipid raft protein, from mouse brain extract prepared with 2% SDS. These results indicate that this simple protocol has potential applications for excellent immunoaffinity separation of the less-soluble proteins in diverse cells and tissues.


Subject(s)
Immunoprecipitation/methods , Proteins/chemistry , Proteins/isolation & purification , Sodium Dodecyl Sulfate/chemistry , Ubiquitin/chemistry , Ubiquitin/isolation & purification , Animals , Brain/cytology , Brain/immunology , Brain/metabolism , Detergents/chemistry , Heat-Shock Response , Humans , Intracellular Signaling Peptides and Proteins , K562 Cells , Membrane Proteins/chemistry , Membrane Proteins/immunology , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Mice , Niemann-Pick C1 Protein , Niemann-Pick Diseases , Protein Denaturation , Protein Renaturation , Proteins/immunology , Proteins/metabolism , Solubility , Time Factors , Ubiquitin/immunology , Ubiquitin/metabolism
18.
Biochem Biophys Res Commun ; 366(2): 471-5, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18067853

ABSTRACT

Transcriptional coactivator with PDZ-binding motif (TAZ) protein is a coactivator of Runx2 and corepressor of PPARgamma. It also induces differentiation of mesenchymal cells into osteoblasts. In this study, we found that FGF-2, which inhibits bone mineralization and stimulates cell proliferation, reduced the TAZ protein expression level in osteoblast-like cells, MC3T3-E1. This reduction was recovered by removing FGF-2 from the culture medium, which also restored the osteoblastic features of MC3T3-E1 cells. Furthermore, FGF-2-induced reduction of TAZ is blocked by a SAPK/JNK-specific inhibitor. These findings suggest that the expression of TAZ protein is involved in osteoblast proliferation and differentiation. This may help elucidate the discrepancies in the effect of FGF-2 and contribute to the understanding of FGF/FGFR-associated craniosynostosis syndrome etiology and treatment.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Proteins/metabolism , Transcription Factors/metabolism , 3T3 Cells , Acyltransferases , Animals , Cell Differentiation , Down-Regulation , Mice
19.
Int J Biochem Cell Biol ; 39(1): 171-80, 2007.
Article in English | MEDLINE | ID: mdl-16979371

ABSTRACT

Histones are key components of chromatin. We investigated histone H2A-immunoreactive proteins in acute monocytic leukemia THP-1 cells using three polyclonal antibodies raised against peptides corresponding to distinct regions of H2A. Two unknown immunoreactive proteins (9- and 12-kDa proteins), H2A (14kDa) and ubiquitinated H2A (23kDa) were found in the cell lysates prepared by immediate direct addition of SDS-PAGE sample buffer to the cells as well as in the nuclear and chromatin fractions. However, they were not found in the cytoplasmic fraction. The unknown proteins were successfully purified by immunoaffinity chromatography from the cell nucleus extract and identified as 9-kDa H2A(1-87) and 12-kDa H2A(1-114), suggesting that both were produced by limited proteolysis of intact H2A(1-129). The truncated forms of H2A probably persisted as chromatin constituents, since the stability of H2A(1-87) in the chromatin fraction was sensitive to treatment with micrococcal nuclease, and H2A(1-114) was solubilized with lower ionic strength from the chromatin fraction obtained by micrococcal nuclease treatment. Truncated H2A proteins in THP-1 cells were transiently increased in amount by short-term treatment with phorbol 12-myristate 13-acetate or all-trans-retinoic acid, both of which induce macrophage-like differentiation. Furthermore, these increases were suppressed by preceding treatment with carbobenzoxy-l-leucyl-l-leucyl-l-leucinal (MG132) but not with carbobenzoxy-l-isoleucyl-gamma-t-butyl-l-glutamyl-l-alanyl-l-leucinal (PSI), both of which are generally known as proteasome inhibitors. Our results suggest that histone H2A is cleaved at least at two sites by protease(s) that remain obscure, and might affect chromatins in the early stage of THP-1 cell differentiation.


Subject(s)
Antibodies/chemistry , Chromatin/chemistry , Histones/chemistry , Histones/isolation & purification , Monocytes/chemistry , Ubiquitins/chemistry , Ubiquitins/isolation & purification , Antibodies/immunology , Antineoplastic Agents/pharmacology , Carcinogens/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Chromatin/metabolism , Histones/biosynthesis , Histones/immunology , Humans , Leupeptins , Monocytes/metabolism , Protein Processing, Post-Translational/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology , Ubiquitins/biosynthesis , Ubiquitins/immunology
20.
Pediatr Int ; 48(6): 531-5, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17168969

ABSTRACT

BACKGROUND: In Japan, mass screening for neuroblastoma has been performed at 6 months of age to improve the prognosis of this condition for more than 20 years. In recent years, most neuroblastomas detected by mass screening were considered to have favorable biological features and sometimes tend to regress spontaneously. METHODS: The authors established non-treated observation criteria in 1997 and criteria for observation of residual tumor after first-line chemotherapy in 1999, and have made an effort to reduce the intensity of medical treatment for neuroblastoma. The authors examined outcomes of 79 patients who were found in the Shizuoka neuroblastoma mass screening at 6 months of age and who received medical treatment or underwent observation in Shizuoka Children's Hospital, Shizuoka, Japan, between December 1981 and December 2004. RESULTS: A total of 77 patients survived but the remaining two patients died from complications of medical treatment. None of the patients died due to progression of neuroblastoma. In the cases, non-treated observation was performed in 17. Of those, 12 patients are now under non-treated observation. Of their tumors, two have disappeared, nine have become smaller and another one has not change in size. Observation of residual tumor after first-line chemotherapy was performed in 15 cases, and three disappeared and the other 12 cases became smaller. Medical treatment-related complications were observed in 20 of 67 patients who received medical treatment, and 18 of the 20 patients were seen before establishing non-treated observation criteria. CONCLUSION: Non-treated observation and observation of residual tumor after first-line chemotherapy were useful to reduce medical treatment-related complications.


Subject(s)
Mass Screening , Neuroblastoma/diagnosis , Neuroblastoma/therapy , Adrenal Gland Neoplasms/diagnosis , Adrenal Gland Neoplasms/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/urine , Homovanillic Acid/urine , Humans , Infant , Japan/epidemiology , Mediastinal Neoplasms/diagnosis , Mediastinal Neoplasms/therapy , Neoplasm Regression, Spontaneous , Neoplasm Staging , Neuroblastoma/mortality , Neuroblastoma/urine , Prognosis , Retrospective Studies , Risk Factors , Survival Rate , Treatment Outcome , Vanilmandelic Acid/urine
SELECTION OF CITATIONS
SEARCH DETAIL