Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Rinsho Ketsueki ; 65(3): 180-182, 2024.
Article in Japanese | MEDLINE | ID: mdl-38569863

ABSTRACT

Relapse or progressive disease after chimeric antigen receptor T-cell (CAR-T) treatment remains a major issue for poor-risk aggressive large B-cell lymphoma. However, limited data are available on post-CAR-T use of polatuzumab vedotin. Here we describe the case of a patient with diffuse large B-cell lymphoma (DLBCL) who experienced relapse three months after CD19-directed CAR-T therapy with tisagenlecleucel. However, the relapsed lesions rapidly disappeared following treatment with polatuzumab vedotin and rituximab. Notably, long-term remission was achieved without severe cytopenia, infections or peripheral neuropathy, showing the therapeutic benefit of polatuzumab vedotin for CAR-T failure.


Subject(s)
Immunoconjugates , Lymphoma, Large B-Cell, Diffuse , Receptors, Chimeric Antigen , Humans , Rituximab/therapeutic use , Antibodies, Monoclonal , Lymphoma, Large B-Cell, Diffuse/drug therapy , Chronic Disease , Antineoplastic Combined Chemotherapy Protocols
2.
Int J Hematol ; 117(1): 143-148, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36083572

ABSTRACT

Mogamulizumab (MOG), a humanized monoclonal anti-CCR4 antibody, exerts strong antibody-dependent cellular cytotoxic effects on CCR4-positive adult T-cell leukemia/lymphoma (ATLL) cells. As CCR4 is highly expressed on regulatory T cells as well as ATLL cells, pre-transplant MOG induces severe graft-versus-host disease (GvHD). However, limited data are available on post-transplant use of MOG for relapsed ATLL. Here we describe the case of a patient with ATLL who experienced post-transplant relapse with involvement of peripheral blood, skin, lungs, and lymph nodes. Neither tacrolimus dose reduction nor cytotoxic chemotherapy was effective, but a single dose of MOG (1 mg/kg) induced complete remission. After treatment with MOG, leukemic cells in the peripheral blood rapidly disappeared, and the skin, lymph node, and lung lesions gradually regressed. Most notably, the long-term remission was accompanied by recurrence of moderate acute GvHD (grade II, skin stage 2, gut stage 1, liver stage 0). Our findings indicate that MOG can augment allogeneic immune-mediated anti-tumor reactions through graft-versus-ATLL (GvATLL) even during post-transplant relapse involving the lymph nodes and lungs, along with inducing GvHD.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia-Lymphoma, Adult T-Cell , Lymphoma , Adult , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Recurrence
3.
Med Oncol ; 39(8): 118, 2022 Jun 08.
Article in English | MEDLINE | ID: mdl-35674939

ABSTRACT

We investigated the antitumor effects of oleanolic acid (OA) and ursolic acid (UA) on adult T-cell leukemia cells. OA and UA dose-dependently inhibited the proliferation of adult T-cell leukemia cells. UA-treated cells showed caspase 3/7 and caspase 9 activation. PARP cleavage was detected in UA-treated MT-4 cells. Activation of mTOR and PDK-1 was inhibited by UA. Autophagosomes were detected in MT-4 cells after UA treatment using electron microscopy. Consistently, mitophagy was observed in OA- and UA-treated MT-4 cells by confocal microscopy. The mitochondrial membrane potential in MT-4 cells considerably decreased, and mitochondrial respiration and aerobic glycolysis were significantly reduced following UA treatment. Furthermore, MT-1 and MT-4 cells were sorted into two regions based on their mitochondrial membrane potential. UA-treated MT-4 cells from both regions showed high activation of caspase 3/7, which were inhibited by Z-vad. Interestingly, MT-4 cells cocultured with sorted UA-treated cells showed enhanced proliferation. Finally, UA induced cell death and ex vivo PARP cleavage in peripheral blood mononuclear cells from patients with adult T-cell leukemia. Therefore, UA-treated MT-4 cells show caspase activation following mitochondrial dysfunction and may produce survival signals to the surrounding cells.


Subject(s)
Antineoplastic Agents, Phytogenic , Leukemia-Lymphoma, Adult T-Cell , Oleanolic Acid , Triterpenes , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukocytes, Mononuclear/metabolism , Mitochondria/metabolism , Oleanolic Acid/metabolism , Oleanolic Acid/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triterpenes/metabolism , Triterpenes/pharmacology , Ursolic Acid
4.
Invest New Drugs ; 39(1): 122-130, 2021 02.
Article in English | MEDLINE | ID: mdl-32914311

ABSTRACT

Adult T cell leukemia (ATL) is an aggressive and malignant blood disease. We previously reported that steroid-structured cucurbitacin D (CuD) induces apoptosis in ATL cells. In this study, we investigated the effects of mitogen-activated protein kinase (MAPK) signaling inhibitors on CuD-induced cell death in peripheral blood lymphocytes (PBLs) isolated from ATL/acute lymphoblastic leukemia (ALL) patients and two human leukemia cell lines (MT-1 and MT-4). PBLs were isolated from an ATL/ALL patient as well as from a healthy donor. Cell surface markers were examined using flow cytometry. Serum cytokine levels were estimated using LEGENDplex or analyzed at the Center for Clinical and Translational Research of Kyushu University Hospital. Cell proliferation was assessed using the Cell Titer-Glo luminescent cell viability assay. Protein expression was determined by western blotting. PBLs from patients highly expressed CD4 and CD5. Serum from the patient contained high levels of interleukin (IL)-8, IL-10, IL-18, and interferon-γ compared to the healthy donor. CuD-induced cell death was enhanced by the mitogen-activated protein kinase kinase (MEK)1/2 inhibitor U0126. However, a c-Jun N-terminal kinase (JNK) inhibitor prevented CuD-induced cell death. Immunoblot analyses revealed that CuD reduced the phosphorylation of extracellular signal-regulated kinase (ERK), p38, and JNK, and co-treatment with CuD and U0126 did not affect the phosphorylation of ERK. MEK1/2 and p38 inhibitors enhanced CuD-induced cell death, and U0126 enhanced the CuD-induced de-phosphorylation of ERK in MT-1 and MT-4 cells. We conclude that CuD reduces ERK activation, resulting in enhanced antitumor effects on leukemic cells.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell/pathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Triterpenes/pharmacology , Animals , CD4 Antigens/biosynthesis , CD5 Antigens/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Humans , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice
6.
Rinsho Ketsueki ; 58(12): 2411-2413, 2017.
Article in Japanese | MEDLINE | ID: mdl-29332876

ABSTRACT

An 85-year-old male presented with 1-year history of a right breast mass. Needle biopsy of the mass revealed diffuse proliferation of large lymphoid cells that were positive for CD20, BCL2, BCL6, and MUM1 and negative for CD5, CD10, MYC, and EBER. The patient was diagnosed as having diffuse large B-cell lymphoma, a type of primary breast lymphoma (PBL). Sex hormone imbalance, which causes conditions such as gynecomastia, is associated with PBL development in males. Estramustine is a nitrogen mustard moiety linked to estradiol. For 5 years, the patient underwent estramustine therapy for treating prostate cancer. Our case suggests an important role of estrogen in PBL development.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/diagnostic imaging , Estramustine/therapeutic use , Lymphoma, Large B-Cell, Diffuse/diagnostic imaging , Prostatic Neoplasms/drug therapy , Aged, 80 and over , Breast Neoplasms/pathology , Humans , Male
7.
PLoS One ; 9(7): e103240, 2014.
Article in English | MEDLINE | ID: mdl-25054240

ABSTRACT

WNT signaling mediates various physiological and pathological processes. We previously showed that WNT10A is a novel angio/stromagenic factor involved in such processes as tumor growth, wound healing and tissue fibrosis. In this study, we investigated the role of WNT10A in promoting the fibrosis that is central to the pathology of acute interstitial nephritis (AIN). We initially asked whether there is an association between kidney function (estimated glomerular filtration rate; eGFR) and WNT10A expression using kidney biopsies from 20 patients with AIN. Interestingly, patients with WNT10A expression had significantly lower eGFR than WNT10A-negative patients. However, changes in kidney function were not related to the level of expression of other WNT family members. Furthermore, there was positive correlation between WNT10A and α-SMA expression. We next investigated the involvement of WNT10A in kidney fibrosis processes using COS1 cells, a kidney fibroblast cell line. WNT10A overexpression increased the level of expression of fibronectin and peroxiredoxin 5. Furthermore, WNT10A overexpression renders cells resistant to apoptosis induced by hydrogen peroxide and high glucose. Collectively, WNT10A may induce kidney fibrosis and associate with kidney dysfunction in AIN.


Subject(s)
Fibroblasts/pathology , Kidney/pathology , Nephritis, Interstitial/genetics , Nephritis, Interstitial/pathology , Wnt Proteins/genetics , Actins/analysis , Actins/genetics , Acute Disease , Aged , Apoptosis , Fibroblasts/metabolism , Fibronectins/metabolism , Fibrosis , Gene Expression Regulation , Humans , Kidney/metabolism , Kidney/physiopathology , Male , Nephritis, Interstitial/physiopathology , Oxidative Stress , Wnt Proteins/analysis , Wnt Proteins/metabolism
8.
Anticancer Agents Med Chem ; 14(7): 1042-50, 2014.
Article in English | MEDLINE | ID: mdl-24521151

ABSTRACT

The aurora kinases are serine/threonine kinases that are essential for mitosis and contribute to tumorigenesis. Therefore, aurora kinases hold promise for molecularly targeted therapy. In the present study, we demonstrated that aurora B kinase (AURKB) is overexpressed in both cisplatin- and oxaliplatin-resistant cells. Downregulation of AURKB sensitized cells to both cisplatin and oxaliplatin, but not to paclitaxel, 5-FU or hydrogen peroxide. Interestingly, we found that both cisplatin- and oxaliplatin-resistant cells were hypersensitive to the AURKB specific inhibitors, AZD1152 HQPA and ZM447439, suggesting that both cisplatin- and oxaliplatinresistant cells develop an addiction to AURKB. These data provide evidence that aurora kinase inhibitors can overcome both cisplatin and oxaliplatin resistance. Therefore, AURKB inhibitors could offer potential benefits if used after first-line platinum-based chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinases/antagonists & inhibitors , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Organoplatinum Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Aurora Kinases/metabolism , Benzamides/pharmacology , Cell Line, Tumor , Humans , Organophosphates/pharmacology , Oxaliplatin , Quinazolines/pharmacology
9.
Sci Rep ; 2: 379, 2012.
Article in English | MEDLINE | ID: mdl-22532927

ABSTRACT

Mieap, a p53-inducible protein, controls mitochondrial integrity by inducing the accumulation of lysosomal proteins within mitochondria. This phenomenon is designated MALM, for Mieap-induced accumulation of lysosome-like organelles within mitochondria. To identify this novel Mieap-interacting protein(s), we performed a two-dimensional image-converted analysis of liquid chromatography and mass spectrometry (2DICAL) on the proteins immunoprecipitated by an anti-Mieap antibody. We indentified 14-3-3γ as one of the proteins that was included in the Mieap-binding protein complex when MALM was induced. The interaction between Mieap and 14-3-3γ was confirmed on the exogenous and endogenous proteins. Interestingly, 14-3-3γ was localized within mitochondria when MALM occurred. A 14-3-3γ deficiency did not affect the accumulation of Mieap and lysosomal proteins within mitochondria, but dramatically inhibited the elimination of oxidized mitochondrial proteins. These results suggest that 14-3-3γ plays a critical role in eliminating oxidized mitochondrial proteins during the MALM process by interacting with Mieap within mitochondria.

10.
PLoS One ; 7(1): e30767, 2012.
Article in English | MEDLINE | ID: mdl-22292033

ABSTRACT

Mieap, a p53-inducible protein, controls mitochondrial quality by repairing unhealthy mitochondria. During repair, Mieap induces the accumulation of intramitochondrial lysosomal proteins (designated MALM for Mieap-induced accumulation of lysosome-like organelles within mitochondria) by interacting with NIX, leading to the elimination of oxidized mitochondrial proteins. Here, we report that an additional mitochondrial outer membrane protein, BNIP3, is also involved in MALM. BNIP3 interacts with Mieap in a reactive oxygen species (ROS)-dependent manner via the BH3 domain of BNIP3 and the coiled-coil domains of Mieap. The knockdown of endogenous BNIP3 expression severely inhibited MALM. Although the overexpression of either BNIP3 or NIX did not cause a remarkable change in the mitochondrial membrane potential (MMP), the co-expression of all three exogenous proteins, Mieap, BNIP3 and NIX, caused a dramatic reduction in MMP, implying that the physical interaction of Mieap, BNIP3 and NIX at the mitochondrial outer membrane may regulate the opening of a pore in the mitochondrial double membrane. This effect was not related to cell death. These results suggest that two mitochondrial outer membrane proteins, BNIP3 and NIX, mediate MALM in order to maintain mitochondrial integrity. The physical interaction of Mieap, BNIP3 and NIX at the mitochondrial outer membrane may play a critical role in the translocation of lysosomal proteins from the cytoplasm to the mitochondrial matrix.


Subject(s)
Membrane Proteins/physiology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/physiology , Tumor Suppressor Proteins/physiology , Cell Line, Tumor , HCT116 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Models, Biological , Organelles/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Protein Transport/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
PLoS One ; 6(1): e16054, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21264221

ABSTRACT

The accumulation of unhealthy mitochondria results in mitochondrial dysfunction, which has been implicated in aging, cancer, and a variety of degenerative diseases. However, the mechanism by which mitochondrial quality is regulated remains unclear. Here, we show that Mieap, a novel p53-inducible protein, induces intramitochondrial lysosome-like organella that plays a critical role in mitochondrial quality control. Mieap expression is directly regulated by p53 and is frequently lost in human cancer as result of DNA methylation. Mieap dramatically induces the accumulation of lysosomal proteins within mitochondria and mitochondrial acidic condition without destroying the mitochondrial structure (designated MALM, for Mieap-induced accumulation of lysosome-like organelles within mitochondria) in response to mitochondrial damage. MALM was not related to canonical autophagy. MALM is involved in the degradation of oxidized mitochondrial proteins, leading to increased ATP synthesis and decreased reactive oxygen species generation. These results suggest that Mieap induces intramitochondrial lysosome-like organella that plays a critical role in mitochondrial quality control by eliminating oxidized mitochondrial proteins. Cancer cells might accumulate unhealthy mitochondria due to p53 mutations and/or Mieap methylation, representing a potential cause of the Warburg effect.


Subject(s)
Lysosomes , Mitochondria/ultrastructure , Mitochondrial Proteins/physiology , Organelles/chemistry , Tumor Suppressor Protein p53/physiology , Humans , Lysosomes/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Protein Processing, Post-Translational , Protein Stability , Transcriptional Activation
12.
PLoS One ; 6(1): e16060, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21264228

ABSTRACT

Maintenance of healthy mitochondria prevents aging, cancer, and a variety of degenerative diseases that are due to the result of defective mitochondrial quality control (MQC). Recently, we discovered a novel mechanism for MQC, in which Mieap induces intramitochondrial lysosome-like organella that plays a critical role in the elimination of oxidized mitochondrial proteins (designated MALM for Mieap-induced accumulation of lysosome-like organelles within mitochondria). However, a large part of the mechanisms for MQC remains unknown. Here, we report additional mechanisms for Mieap-regulated MQC. Reactive oxygen species (ROS) scavengers completely inhibited MALM. A mitochondrial outer membrane protein NIX interacted with Mieap in a ROS-dependent manner via the BH3 domain of NIX and the coiled-coil domain of Mieap. Deficiency of NIX also completely impaired MALM. When MALM was inhibited, Mieap induced vacuole-like structures (designated as MIV for Mieap-induced vacuole), which engulfed and degraded the unhealthy mitochondria by accumulating lysosomes. The inactivation of p53 severely impaired both MALM and MIV generation, leading to accumulation of unhealthy mitochondria. These results suggest that (1) mitochondrial ROS and NIX are essential factors for MALM, (2) MIV is a novel mechanism for lysosomal degradation of mitochondria, and (3) the p53-Mieap pathway plays a pivotal role in MQC by repairing or eliminating unhealthy mitochondria via MALM or MIV generation, respectively.


Subject(s)
Lysosomes , Membrane Proteins/physiology , Mitochondria/pathology , Mitochondrial Proteins/physiology , Organelles/chemistry , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Cell Line , Humans , Lysosomes/metabolism , Membrane Proteins/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/pharmacology , Transcriptional Activation , Tumor Suppressor Proteins/metabolism
13.
Cancers (Basel) ; 3(4): 3909-20, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-24213117

ABSTRACT

We previously reported that the transcription factor Zinc Finger Protein 143 (ZNF143) regulates the expression of genes associated with cell cycle and cell division, and that downregulation of ZNF143 induces cell cycle arrest at G2/M. To assess the function of ZNF143 expression in the cell cycle, we established two cells with forced expression of ZNF143 derived from PC3 prostate cancer cell lines. These cell lines overexpress genes associated with cell cycle and cell division, such as polo-like kinase 1 (PLK1), aurora kinase B (AURKB) and some minichromosome maintenance complex components (MCM). However, the doubling time of cells with forced expression of ZNF143 was approximately twice as long as its control counterpart cell line. Analysis following serum starvation and re-seeding showed that PC3 cells were synchronized at G1 in the cell cycle. Also, ZNF143 expression fluctuated, and was at its lowest level in G2/M. However, PC3 cells with forced expression of ZNF143 synchronized at G2/M, and showed lack of cell cycle-dependent fluctuation of nuclear expression of MCM proteins. Furthermore, G2/M population of both cisplatin-resistant PCDP6 cells over-expressing ZNF143 (derived from PC3 cells) and cells with forced expression of ZNF143 was significantly higher than that of each counterpart, and the doubling time of PCDP6 cells is about 2.5 times longer than that of PC3 cells. These data suggested that fluctuations in ZNF143 expression are required both for gene expression associated with cell cycle and for cell division.

14.
Int J Oncol ; 37(5): 1133-41, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20878061

ABSTRACT

NEEP21, also designated D4S234E or NSG1, is an endosomal protein expressed in neuronal cells under normal conditions. Here, we report that NEEP21 is a direct transcriptional target gene of the tumor suppressor p53. NEEP21 expression is inducible in non-neuronal human cancer cell lines by exposure to adriamycin, hydrogen peroxide, UV and γ-ray in a p53-dependent manner. Chromatin immunoprecipitation assay indicated that a potential p53-binding site (p53BS) is located in intron 1 of the NEEP21 gene. A reporter assay confirmed that p53BS has p53-responsive activity. The heterologous luciferase gene containing p53BS is also transactivated by p73-ß and p63-γ. The introduction of the NEEP21 gene into various cancer cell lines suppressed cell growth. Infection with an adenovirus vector containing NEEP21 induced apoptotic cell death via caspase-3 activation in many cancer cell lines. The expression of NEEP21 mRNA was remarkably induced by γ-ray irradiation in the spleen of p53+/+ mice but not in that of p53-/- mice. These results suggest that NEEP21 may play a critical role in apoptosis as a mediator of p53.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Nerve Tissue Proteins/genetics , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Cell Separation , Chromatin Immunoprecipitation , Flow Cytometry , Gene Expression , Humans , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
15.
Oncol Rep ; 24(5): 1193-200, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20878110

ABSTRACT

The tumor suppressor p53 is a transcription factor that induces the transcription of various target genes in response to DNA damage and it protects the cells from malignant transformation. In this study, we performed cDNA microarray analysis and found that the transmembrane protein containing sushi domain (TMPS) gene, which encodes a putative type I transmembrane protein, is a novel p53-target gene. TMPS contains a sushi domain in the extracellular region, which is associated with protein-protein interaction. TMPS expression is induced by endogenous p53 under genotoxic stress in several cancer cell lines. Reporter assay revealed p53-dependent transactivation of the p53 binding-sites (BSs) located in the intron 1 of TMPS. Chromatin immunoprecipitation (ChIP) assay showed that p53 binds to these BSs in vivo. Overexpression of TMPS induced apoptosis through the activation of caspase-3, 8, and 9 in various cancer cell lines. Moreover, γ-irradiation induced the expression of TMPS mRNA in the spleen and colon of p53+/+ mice but not in those of p53-/- mice. These data indicate that TMPS may play a role in p53-dependent apoptosis under DNA damage condition.


Subject(s)
Apoptosis/physiology , Membrane Proteins/physiology , Tumor Suppressor Protein p53/physiology , Amino Acid Sequence , Animals , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , Hep G2 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Protein Array Analysis , Protein Structure, Tertiary , Rats , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Int J Oncol ; 36(5): 1253-60, 2010 May.
Article in English | MEDLINE | ID: mdl-20372800

ABSTRACT

UNC5A is an axon-guidance molecule, and plays a critical role in neuronal development and differentiation as a netrin-1 receptor. Emerging evidence suggests that axon guidance molecules including UNC5A regulate apoptosis in non-neuronal cells. Here, we report that UNC5A regulates apoptosis as a downstream target of p53. UNC5A expression was strongly induced by exogenous and endogenous p53. Chromatin immunoprecipitation (ChIP) revealed that p53 binds to a sequence in the promoter region of the UNC5A gene. Reporter assays showed that this sequence exhibits p53-dependent transcriptional activity. Overexpression of UNC5A significantly suppressed colony formation of two glioblastoma cell lines-U373MG and T98G. UNC5A dramatically induced apoptosis through the activation of caspase-3 in various cancer cell lines, including LS174T (colon cancer), U373MG (glioblastoma), SH-SY5Y (neuroblastoma), and SKNAS (neuroblastoma). Finally, gamma irradiation strongly induced the expression of UNC5A mRNA in the spleen and colon of p53+/+ mice, but not in those of p53-/- mice, implying that the transcription of UNC5A in vivo is regulated by p53. These results suggest that UNC5A is a novel transcriptional target of p53 and plays a role in p53-dependent apoptosis.


Subject(s)
Apoptosis , Genes, p53 , Receptors, Cell Surface/biosynthesis , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Mice , Models, Genetic , Netrin Receptors , Promoter Regions, Genetic , RNA Interference
17.
Cancer Sci ; 99(12): 2444-54, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19018766

ABSTRACT

Aneuploidy is a hallmark of human cancers. Although the maintenance of genomic integrity by p53 is important in preventing aneuploidy, its mechanism remains to be elucidated. Here we report evidence that B-cell linker protein (BLNK) mediates the inhibition of cytokinesis, which generates tetraploidy but prevents aneuploidy. We identified BLNK as a transcriptional target of p53. Surprisingly, ectopic expression of exogenous BLNK inhibited cytokinesis, resulting in the formation of tetraploid cells. Indeed, BLNK was involved in the generation of spontaneously arising binucleate tetraploid cells. Interestingly, cytokinesis after DNA damage was inhibited in p21(-/-) and p53+/+ cells, but not in p53(-/-) cells. BLNK knockdown in p53+/+ and p21(-/-) cells enhanced cytokinesis after DNA damage, leading to the generation of aneuploid cells. In addition, a BLNK-downregulated human pre-B leukemia cell line showed increased cytokinesis and aneuploidy after DNA damage compared with two other pre-B leukemia cell lines expressing higher levels of BLNK. These results suggest that BLNK acts as a mediator of p53 in the inhibition of cytokinesis, which prevents aneuploidy. We propose that the inhibition of cytokinesis is crucial for the maintenance of genomic integrity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Aneuploidy , Cytokinesis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , HCT116 Cells , Humans , In Situ Hybridization, Fluorescence , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Models, Genetic , Tumor Suppressor Protein p53/genetics
18.
Cancer Res ; 67(4): 1451-60, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17308083

ABSTRACT

Although the regulation of tumor angiogenesis is believed to be one of the core functions of p53, the mechanism still remains to be elucidated. Here, we report that semaphorin 3F (SEMA3F), an axon guidance molecule, is involved in p53-regulated antiangiogenesis. The expression level of SEMA3F mRNA was increased by both exogenous and endogenous p53. Chromatin immunoprecipitation assay indicated that a potent p53-binding sequence in intron 1 of SEMA3F interacts with p53 and that it has a p53-responsive transcriptional activity. Overexpression of SEMA3F inhibited in vitro cell growth of the lung cancer cell line H1299. In nude mice assay, the size of the H1299 tumors expressing SEMA3F was much smaller, and they showed lesser number of blood vessels as compared with the control tumors. Moreover, tumors derived from the p53-knockdown colorectal cancer cell line LS174T displayed a remarkable enhancement of tumor vessel formation as compared with control tumors containing normal levels of p53. The expression levels of SEMA3F and neuropilin-2 (NRP2), the functional receptor for SEMA3F, in p53-knockdown LS174T tumors were lower than those in the control tumors. Adenovirus-mediated SEMA3F gene transfer induced the remarkable in vitro growth suppression of the stable transformant of H1299 cells, which express high levels of NRP2. These results suggest that p53 negatively regulates tumor vessel formation and cell growth via the SEMA3F-NRP2 pathway.


Subject(s)
Genes, Tumor Suppressor , Membrane Proteins/genetics , Neoplasms/blood supply , Neoplasms/genetics , Nerve Tissue Proteins/genetics , Cell Growth Processes/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 3 , Gene Expression Regulation, Neoplastic , Genes, p53 , HCT116 Cells , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/metabolism , Neuropilin-2/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction , Transcription, Genetic
19.
J Hum Genet ; 51(8): 652-664, 2006.
Article in English | MEDLINE | ID: mdl-16897187

ABSTRACT

The tumor suppressor p53 plays a crucial role in the cellular response to DNA damage by transcriptional activation of numerous downstream genes. Although a considerable number of p53 target genes have been reported, the precise mechanism of p53-regulated tumor suppression still remains to be elucidated. Here, we report a novel role of the DFNA5 gene in p53-mediated etoposide-induced cell death. The DFNA5 gene has been previously reported to be responsible for autosomal-dominant, nonsyndromic hearing impairment. The expression of the DFNA5 gene was strongly induced by exogenous and endogenous p53. The chromatin immunoprecipitation assay indicated that a potential p53-binding sequence is located in intron 1 of the DFNA5 gene. Furthermore, the reporter gene assay revealed that the sequence displays p53-dependent transcriptional activity. The ectopic expression of DFNA5 enhanced etoposide-induced cell death in the presence of p53; however, it was inhibited in the absence of p53. Finally, the expression of DFNA5 mRNA was remarkably induced by gamma-ray irradiation in the colon of p53(+/+) mice but not in that of p53(-/-) mice. These results suggest that DFNA5 plays a role in the p53-regulated cellular response to genotoxic stress probably by cooperating with p53.


Subject(s)
DNA Damage , Hearing Loss/genetics , Receptors, Estrogen/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Base Sequence , Binding Sites/drug effects , COS Cells , Chlorocebus aethiops , Etoposide/pharmacology , Exons/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human/genetics , Humans , Mice , Microarray Analysis , Molecular Sequence Data , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Transcription, Genetic/drug effects , Tumor Cells, Cultured
20.
Proc Natl Acad Sci U S A ; 102(8): 2796-801, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15705719

ABSTRACT

A 48-kDa beta-N-acetylglucosamine (GlcNAc)-binding protein was isolated from mouse brain by GlcNAc-agarose column chromatography. The N-terminal amino acid residues showed the protein to be a mouse Na(+)/K(+)-ATPase beta1-subunit. When the recombinant FLAG-beta1-subunit expressed in Sf-9 cells was applied to a GlcNAc-agarose column, only the glycosylated 38- and 40-kDa proteins bound to the column. In the absence of KCl, little of the proteins bound to a GlcNAc-agarose column, but the 38- and 40-kDa proteins bound in the presence of KCl at concentrations above 1 mM. Immunohistochemical study showed that the beta1-subunit and GlcNAc-terminating oligosaccharides are at the cell contact sites. Inclusion of anti-beta1-subunit antibody or chitobiose in cell aggregation assays using mouse neural cells resulted in inhibition of cell aggregation. These results indicate that the Na(+)/K(+)-ATPase beta1-subunit is a potassium-dependent lectin that binds to GlcNAc-terminating oligosaccharides: it may be involved in neural cell interactions.


Subject(s)
Acetylglucosamine/metabolism , Polysaccharides/metabolism , Potassium/pharmacology , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Brain/metabolism , Cell Adhesion , Disaccharides/pharmacology , Glycosylation , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Protein Subunits , Sepharose/metabolism , Sodium-Potassium-Exchanging ATPase/analysis , Sodium-Potassium-Exchanging ATPase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...