Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Commun Biol ; 6(1): 1294, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38129572

ABSTRACT

Immunotherapy has attracted considerable attention as a therapeutic strategy for cancers including acute myeloid leukemia (AML). In this study, we found that the development of several aggressive subtypes of AML is slower in Rag2-/- mice despite the lack of B and T lymphocytes, even compared to the immunologically normal C57BL/6 mice. Furthermore, an orally active p53-activating drug shows stronger antileukemia effect on AML in Rag2-/- mice than C57BL/6 mice. Intriguingly, Natural Killer (NK) cells in Rag2-/- mice are increased in number, highly express activation markers, and show increased cytotoxicity to leukemia cells in a coculture assay. B2m depletion that triggers missing-self recognition of NK cells impairs the growth of AML cells in vivo. In contrast, NK cell depletion accelerates AML progression in Rag2-/- mice. Interestingly, immunogenicity of AML keeps changing during tumor evolution, showing a trend that the aggressive AMLs generate through serial transplantations are susceptible to NK cell-mediated tumor suppression in Rag2-/- mice. Thus, we show the critical role of NK cells in suppressing the development of certain subtypes of AML using Rag2-/- mice, which lack functional lymphocytes but have hyperactive NK cells.


Subject(s)
Killer Cells, Natural , Leukemia, Myeloid, Acute , Animals , Mice , Mice, Knockout , Mice, Inbred C57BL , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , T-Lymphocytes , DNA-Binding Proteins/genetics
2.
Cancer Discov ; 13(5): 1250-1273, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37067901

ABSTRACT

Cancer-relevant mutations in the oligomerization domain (OD) of the p53 tumor suppressor protein, unlike those in the DNA binding domain, have not been well elucidated. Here, we characterized the germline OD mutant p53(A347D), which occurs in cancer-prone Li-Fraumeni syndrome (LFS) patients. Unlike wild-type p53, mutant p53(A347D) cannot form tetramers and exists as a hyperstable dimeric protein. Further, p53(A347D) cannot bind or transactivate the majority of canonical p53 target genes. Isogenic cell lines harboring either p53(A347D) or no p53 yield comparable tumorigenic properties, yet p53(A347D) displays remarkable neomorphic activities. Cells bearing p53(A347D) possess a distinct transcriptional profile and undergo metabolic reprogramming. Further, p53(A347D) induces striking mitochondrial network aberration and associates with mitochondria to drive apoptotic cell death upon topoisomerase II inhibition in the absence of transcription. Thus, dimer-forming p53 demonstrates both loss-of-function (LOF) and gain-of-function (GOF) properties compared with the wild-type form of the protein. SIGNIFICANCE: A mutant p53 (A347D), which can only form dimers, is associated with increased cancer susceptibility in LFS individuals. We found that this mutant wields a double-edged sword, driving tumorigenesis through LOF while gaining enhanced apoptogenic activity as a new GOF, thereby yielding a potential vulnerability to select therapeutic approaches. See related commentary by Stieg et al., p. 1046. See related article by Gencel-Augusto et al., p. 1230. This article is highlighted in the In This Issue feature, p. 1027.


Subject(s)
Li-Fraumeni Syndrome , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Mitochondria/metabolism
3.
Oncotarget ; 13: 1359-1368, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36537913

ABSTRACT

Antibody drug conjugates (ADC) are one of the attractive modalities for the treatment of acute myeloid leukemia (AML). Previously, we have developed ASP1235, a novel ADC targeting Fms-like tyrosine kinase 3 (FLT3) which is widely expressed on the leukemic blasts of AML patients. In this study, we sought to evaluate the therapeutic effect of ASP1235 in combination with venetoclax plus azacitidine, a novel standard-of-care treatment for elderly AML patients, in ASP1235 poor sensitive AML cells. To identify the suitable preclinical model, we first evaluated the growth inhibitory effect of ASP1235 on several leukemia cell lines expressing FLT3 and found that THP-1 cells were partially sensitive to ASP1235 in vitro. Furthermore, ASP1235 showed marginal anti-tumor activity in a THP-1 xenograft model. Compared to the leukemic blasts in most of the relapsed or refractory (R/R) AML patients tested, THP-1 cells expressed equivalent protein levels of Bcl-2, suggesting that ASP1235 in combination with venetoclax plus azacitidine is a rational treatment in the THP-1 model. In vitro, ASP1235 showed a cytotoxic effect on THP-1 cells in combination with venetoclax, and the combination effect was greater than the additive effect. Furthermore, ASP1235 also showed a combination effect with venetoclax plus azacitidine treatment. Similarly, the combination of ASP1235, venetoclax and azacitidine showed a superior anti-tumor effect in a THP-1 xenograft model without obvious body weight loss. These findings provide supportive evidence that the triple combination of ASP1235, venetoclax and azacitidine would improve the clinical outcome of ASP1235 monotherapy and venetoclax plus azacitidine regimen in AML patients.


Subject(s)
Leukemia, Myeloid, Acute , fms-Like Tyrosine Kinase 3 , Humans , Animals , Mice , Heterografts , Azacitidine/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Leukemia, Myeloid, Acute/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
4.
Bone Marrow Transplant ; 57(5): 775-780, 2022 05.
Article in English | MEDLINE | ID: mdl-35228711

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-SCT) is a potentially curative therapy for FLT3 internal tandem duplication mutant (FLT3-ITD+) acute myeloid leukemia, but relapse rate is high. A recent study showed that sorafenib, a first generation FLT3 and multikinase inhibitor, enhanced graft-versus-leukemia (GVL) effects against FLT3-ITD+ leukemia via interleukin-15 (IL-15) production. However, it remains to be clarified whether this effect could be mediated by selective FLT3 inhibition. We investigated whether gilteritinib, a selective FLT3 inhibitor, could enhance GVL effects against FLT3-ITD transfected Ba/F3 leukemia (Ba/F3-FLT3-ITD) in mice. Oral administration of gilteritinib from day +5 to +14 after allo-SCT reduced expression of the co-inhibitory receptors PD-1 and TIGIT on donor CD8+ T cells and enhanced IL-15 expression in Ba/F3-FLT3-ITD. Bioluminescent imaging using luciferase-transfected Ba/F3-FLT3-ITD demonstrated that gilteritinib significantly suppressed leukemia expansion after allo-SCT, whereas it did not impact the morbidity or mortality of graft-versus-host disease (GVHD), resulting in significant improvement of overall survival. In conclusion, short-term administration of gilteritinib after allo-SCT enhanced GVL effects against FLT3-ITD+ leukemia without exacerbating GVHD.


Subject(s)
Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Aniline Compounds , Animals , CD8-Positive T-Lymphocytes , Interleukin-15 , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Mice , Mutation , Pyrazines , Transplantation, Homologous , fms-Like Tyrosine Kinase 3/genetics
5.
Sci Rep ; 11(1): 341, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431933

ABSTRACT

Single-cell level analysis is powerful tool to assess the heterogeneity of cellular components in tumor microenvironments (TME). In this study, we investigated immune-profiles using the single-cell analyses of endoscopically- or surgically-resected tumors, and peripheral blood mononuclear cells from gastric cancer patients. Furthermore, we technically characterized two distinct platforms of the single-cell analysis; RNA-seq-based analysis (scRNA-seq), and mass cytometry-based analysis (CyTOF), both of which are broadly embraced technologies. Our study revealed that the scRNA-seq analysis could cover a broader range of immune cells of TME in the biopsy-resected small samples of tumors, detecting even small subgroups of B cells or Treg cells in the tumors, although CyTOF could distinguish the specific populations in more depth. These findings demonstrate that scRNA-seq analysis is a highly-feasible platform for elucidating the complexity of TME in small biopsy tumors, which would provide a novel strategies to overcome a therapeutic difficulties against cancer heterogeneity in TME.


Subject(s)
Single-Cell Analysis , Stomach Neoplasms/pathology , Tumor Microenvironment , Adult , Biopsy , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , RNA-Seq , Stomach Neoplasms/genetics
6.
Sci Transl Med ; 12(526)2020 01 15.
Article in English | MEDLINE | ID: mdl-31941828

ABSTRACT

The immune status of the tumor microenvironment is a key indicator in determining the antitumor effectiveness of immunotherapies. Data support the role of activation and expansion of tumor-infiltrating lymphocytes (TILs) in increasing the benefit of immunotherapies in patients with solid tumors. We found that intratumoral injection of a tumor-selective oncolytic vaccinia virus encoding interleukin-7 (IL-7) and IL-12 into tumor-bearing immunocompetent mice activated the inflammatory immune status of previously poorly immunogenic tumors and resulted in complete tumor regression, even in distant tumor deposits. Mice achieving complete tumor regression resisted rechallenge with the same tumor cells, suggesting establishment of long-term tumor-specific immune memory. Combining this virotherapy with anti-programmed cell death-1 (PD-1) or anti-cytotoxic T lymphocyte antigen 4 (CTLA4) antibody further increased the antitumor activity as compared to virotherapy alone, in tumor models unresponsive to either of the checkpoint inhibitor monotherapies. These findings suggest that administration of an oncolytic vaccinia virus carrying genes encoding for IL-7 and IL-12 has antitumor activity in both directly injected and distant noninjected tumors through immune status changes rendering tumors sensitive to immune checkpoint blockade. The benefit of intratumoral IL-7 and IL-12 expression was also observed in humanized mice bearing human cancer cells. These data support further investigation in patients with non-inflamed solid tumors.


Subject(s)
Interleukin-12/metabolism , Interleukin-7/metabolism , Oncolytic Viruses/genetics , Animals , CTLA-4 Antigen/immunology , Female , Immune Checkpoint Inhibitors , Mice , Vaccinia virus/genetics
7.
Cancer Sci ; 111(2): 451-466, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31834974

ABSTRACT

The tumor suppressor gene p53 encodes a transcriptional activator that has two transactivation domains (TAD) located in its amino terminus. These two TAD can transactivate genes independently, and at least one TAD is required for p53 transactivation function. The 1st TAD (a.a. 1-40) is essential for the induction of numerous classical p53 target genes, while the second TAD (a.a. 41-61) suffices for tumor suppression, although its precise molecular function remains unclear. In this study, we comprehensively identified the sites to which p53 lacking the 1st TAD (Δ1stTAD-p53) binds, as well as its potential target genes. We found that the binding sequences for Δ1stTAD-p53 are divergent and include not only the canonical p53 consensus binding sequences but also sequences similar to those recognized by a number of other known transcription factors. We identified and analyzed the functions of three Δ1stTAD-p53 target genes, PTP4A1, PLK2 and RPS27L. All three genes were induced by both full-length p53 and Δ1stTAD-p53, and were dependent on the transactivation activity of the 2nd TAD. We also found that two of these, PTP4A1 and PLK2, are endoplasmic reticulum (ER) stress-inducible genes. We found that upon ER stress, PTP4A1 suppresses apoptosis while PLK2 induces apoptosis. These results reveal a novel Δ1stTAD-p53 downstream pathway that is dependent on the transcription activation activity of the 2nd TAD.


Subject(s)
Cell Cycle Proteins/genetics , Membrane Proteins/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Tyrosine Phosphatases/genetics , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Apoptosis , Binding Sites , Endoplasmic Reticulum Stress , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Oligonucleotide Array Sequence Analysis , Protein Binding , Protein Domains , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
8.
Oncotarget ; 10(58): 6111-6123, 2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31692922

ABSTRACT

Therapeutic effects of FLT3 inhibitors have been reported in acute myeloid leukemia (AML) with constitutively activating FLT3 mutations, including internal tandem duplication (ITD) and point mutation, which are found in approximately one-third of AML patients. One of the critical issues of treatment with FLT3 inhibitors in FLT3-mutated AML is drug resistance. FLT3 ligand (FL) represents a mechanism of resistance to FLT3 inhibitors, including quizartinib, midostaurin, and sorafenib, in AML cells harboring both wild-type and mutant FLT3 (FLT3 wt/FLT3 mut). Here, we investigated the effect of FL on the efficacy of gilteritinib, a FLT3 inhibitor, in AML-derived cells in vitro and in mice. In contrast to other FLT3 inhibitors, FL stimulation had little effect on growth inhibition or apoptosis induction by gilteritinib. The antitumor activity of gilteritinib was also comparable between xenograft mouse models injected with FL-expressing and mock MOLM-13 cells. In the FLT3 signaling analyses, gilteritinib inhibited FLT3wt and FLT3-ITD to a similar degree in HEK293 and Ba/F3 cells, and similarly suppressed FLT3 downstream signaling molecules (including ERK1/2 and STAT5) in both the presence and absence of FL in MOLM-13 cells. Co-crystal structure analysis showed that gilteritinib bound to the ATP-binding pocket of FLT3. These results suggest that gilteritinib has therapeutic potential in FLT3-mutated AML patients with FL overexpression.

9.
Sci Rep ; 9(1): 14627, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601997

ABSTRACT

Gastric cancer remains one of the leading causes of cancer death worldwide. Despite intensive investigations of treatments over the past three decades, the poor prognosis of patients with unresectable advanced or recurrent gastric cancer has not significantly changed, and improved therapies are required. Here, we report the identification of an oncogenic mutation in FGFR4 in a human gastric tumour that leads to constitutive activation of its product, FGFR4. The G636C-FGFR4 tyrosine kinase domain mutation was found in 1 of 83 primary human gastric tumours. The G636C mutation increased FGFR4 autophosphorylation, and activated FGFR4 downstream signalling molecules and enhanced anchorage-independent cell growth when expressed in NIH/3T3 cells. 3D-structural analysis and modelling of FGFR4 suggest that G636C destabilizes an auto-inhibitory conformation and stabilizes an active conformation, leading to increased kinase activation. Ba/F3 cell lines expressing the G636C-FGFR4 mutant were significantly more sensitive to ASP5878, a selective FGFR inhibitor, than the control. Oral administration of ASP5878 significantly inhibited the growth of tumours in mice engrafted with G636C-FGFR4/3T3 cells. Together, our results demonstrate that mutationally activated FGFR4 acts as an oncoprotein. These findings support the therapeutic targeting of FGFR4 in gastric cancer.


Subject(s)
Carcinogenesis/genetics , Proto-Oncogene Proteins/genetics , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Receptor, Fibroblast Growth Factor, Type 4/genetics , Stomach Neoplasms/genetics , 3T3 Cells , Animals , Carcinogenesis/drug effects , Humans , Male , Mice , Mutation , Phosphorylation/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Stomach/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Cancer Sci ; 109(11): 3532-3542, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30207029

ABSTRACT

The PHLDA family (pleckstrin homology-like domain family) of genes consists of 3 members: PHLDA1, 2, and 3. Both PHLDA3 and PHLDA2 are phosphatidylinositol (PIP) binding proteins and function as repressors of Akt. They have tumor suppressive functions, mainly through Akt inhibition. Several reports suggest that PHLDA1 also has a tumor suppressive function; however, the precise molecular functions of PHLDA1 remain to be elucidated. Through a comprehensive screen for p53 target genes, we identified PHLDA1 as a novel p53 target, and we show that PHLDA1 has the ability to repress Akt in a manner similar to that of PHLDA3 and PHLDA2. PHLDA1 has a so-called split PH domain in which the PH domain is divided into an N-terminal (ß sheets 1-3) and a C-terminal (ß sheets 4-7 and an α-helix) portions. We show that the PH domain of PHLDA1 is responsible for its localization to the plasma membrane and binding to phosphatidylinositol. We also show that the function of the PH domain is essential for Akt repression. In addition, PHLDA1 expression analysis suggests that PHLDA1 has a tumor suppressive function in breast and ovarian cancers.


Subject(s)
Breast Neoplasms/genetics , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Alternative Splicing , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Neoplasm Transplantation , Phosphatidylinositols/metabolism , Protein Binding , Transcription Factors/chemistry
11.
Mol Cancer Ther ; 16(1): 68-75, 2017 01.
Article in English | MEDLINE | ID: mdl-27837028

ABSTRACT

Hepatocellular carcinoma is an aggressive cancer with poor prognosis. Fibroblast growth factor 19, a member of the fibroblast growth factor family, is a ligand for fibroblast growth factor receptor 4. Moreover, it plays a crucial role in the progression of hepatocellular carcinoma. ASP5878 is a novel inhibitor of fibroblast growth factor receptors 1, 2, 3, and 4 that is under development. It inhibits fibroblast growth factor receptor 4 kinase activity with an IC50 of 3.5 nmol/L. ASP5878 potently suppressed the growth of the fibroblast growth factor 19-expressing hepatocellular carcinoma cell lines Hep3B2.1-7, HuH-7, and JHH-7. In the Hep3B2.1-7 cell line, ASP5878 inhibited the phosphorylation of fibroblast growth factor receptor 4 and its downstream signaling molecules as well as induced apoptosis. Oral administration of ASP5878 at 3 mg/kg induced sustained tumor regression in a subcutaneous xenograft mouse model using Hep3B2.1-7. In HuH-7, an orthotopic xenograft mouse model, ASP5878 induced complete tumor regression and dramatically extended the survival of the mice. These results suggest that ASP5878 is a potentially effective therapeutic agent for hepatocellular carcinoma patients with tumors expressing fibroblast growth factor 19. Mol Cancer Ther; 16(1); 68-75. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Fibroblast Growth Factors/genetics , Gene Expression , Liver Neoplasms/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrimidines/chemistry , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Cancer Sci ; 107(6): 734-45, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26998741

ABSTRACT

The tumor suppressor p53 functions by inducing the transcription of a collection of target genes. We previously attempted to identify p53 target genes by microarray expression and ChIP-sequencing analyses. In this study, we describe a novel p53 target gene, FUCA1, which encodes a fucosidase. Although fucosidase, α-l-1 (FUCA1) has been reported to be a lysosomal protein, we detected it outside of lysosomes and observed that its activity is highest at physiological pH. As there is a reported association between fucosylation and tumorigenesis, we investigated the potential role of FUCA1 in cancer. We found that overexpression of FUCA1, but not a mutant defective in enzyme activity, suppressed the growth of cancer cells and induced cell death. Furthermore, we showed that FUCA1 reduced fucosylation and activation of epidermal growth factor receptor, and concomitantly suppressed epidermal growth factor signaling pathways. FUCA1 loss-of-function mutations are found in several cancers, its expression is reduced in cancers of the large intestine, and low FUCA1 expression is associated with poorer prognosis in several cancers. These results show that protein defucosylation mediated by FUCA1 is involved in tumor suppression.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , alpha-L-Fucosidase/genetics , alpha-L-Fucosidase/metabolism , Cell Death , Cell Division , Cell Line , Cell Survival , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Fucose/metabolism , Humans , Mutant Proteins/biosynthesis , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Signal Transduction , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , alpha-L-Fucosidase/biosynthesis
13.
Sci Rep ; 6: 19174, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26754925

ABSTRACT

The transcription factors HSF1 and p53 both modulate the stress response, thereby protecting and facilitating the recovery of stressed cells, but both have the potential to promote tumor development. Here we show that a p53 target gene, IER5, encodes an activator of HSF1. IER5 forms a ternary complex with HSF1 and the phosphatase PP2A, and promotes the dephosphorylation of HSF1 at numbers of serine and threonine residues, generating a novel, hypo-phosphorylated active form of HSF1. IER5 is also transcriptionally upregulated in various cancers, although this upregulation is not always p53-dependent. The IER5 locus is associated with a so-called super enhancer, frequently associated with hyperactivated oncogenes in cancer cell lines. Enhanced expression of IER5 induces abnormal HSF1 activation in cancer cells and contributes to the proliferation of these cells under stressed conditions. These results reveal the existence of a novel IER5-mediated cancer regulation pathway that is responsible for the activation of HSF1 observed in various cancers.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/metabolism , Immediate-Early Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/metabolism , Cell Proliferation , DNA Damage , Enhancer Elements, Genetic , Gene Expression , Gene Expression Regulation , Heat Shock Transcription Factors , Humans , Immediate-Early Proteins/metabolism , Multigene Family , Multiprotein Complexes , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Nuclear Proteins/metabolism , Phosphorylation , Prognosis , Promoter Regions, Genetic , Protein Binding , Protein Phosphatase 2/metabolism , Signal Transduction , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
14.
Proc Natl Acad Sci U S A ; 111(23): E2404-13, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24912192

ABSTRACT

The molecular mechanisms underlying the development of pancreatic neuroendocrine tumors (PanNETs) have not been well defined. We report here that the genomic region of the PHLDA3 gene undergoes loss of heterozygosity (LOH) at a remarkably high frequency in human PanNETs, and this genetic change is correlated with disease progression and poor prognosis. We also show that the PHLDA3 locus undergoes methylation in addition to LOH, suggesting that a two-hit inactivation of the PHLDA3 gene is required for PanNET development. We demonstrate that PHLDA3 represses Akt activity and Akt-regulated biological processes in pancreatic endocrine tissues, and that PHLDA3-deficient mice develop islet hyperplasia. In addition, we show that the tumor-suppressing pathway mediated by MEN1, a well-known tumor suppressor of PanNETs, is dependent on the pathway mediated by PHLDA3, and inactivation of PHLDA3 and MEN1 cooperatively contribute to PanNET development. Collectively, these results indicate the existence of a novel PHLDA3-mediated pathway of tumor suppression that is important in the development of PanNETs.


Subject(s)
Genes, Tumor Suppressor , Loss of Heterozygosity , Neuroendocrine Tumors/genetics , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Animals , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , Cells, Cultured , DNA Methylation , Humans , Hyperplasia , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Knockout , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Nuclear Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred Lew , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
15.
J Biomed Opt ; 18(6): 061220, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23748701

ABSTRACT

We propose a method to visualize the arterial inflow, the vascular resistance, and the venous capacitance in the skin tissue from red, green, blue (RGB) digital color images. The arterial inflow and the venous capacitance in the skin tissue are visualized based on an increase in the rate of change in the total blood concentration and the change of the total blood concentration during upper limb occlusion at a pressure of 50 mmHg. The resultant arterial inflow with the measured mean arterial pressure also provides an image of the vascular resistance in human skin. The arterial inflow, the vascular resistance, and the venous capacitance acquired by the method are well correlated with those obtained from the conventional strain-gauge plethysmograph. The correlation coefficients R between the estimated values by the method and the measurements by the SPG are calculated to be 0.83 (P<0.001) for the arterial inflow, 0.77 (P<0.01) for the vascular resistance, and 0.77 (P<0.01) for the venous capacitance. The arterial inflow and the venous capacitance in the skin tissue are significantly higher in active subjects compared with the sedentary subjects, whereas the vascular resistance was significantly lower in the active subjects compared with the sedentary subjects. The results of the present study indicate the possibility of using the proposed method for evaluating the peripheral vascular functions in human skin.


Subject(s)
Skin/blood supply , Skin/pathology , Vasodilation , Algorithms , Arterial Pressure , Arteries/pathology , Color , Forearm/blood supply , Humans , Laser-Doppler Flowmetry , Light , Plethysmography , Pressure , Regional Blood Flow , Regression Analysis , Vascular Resistance
16.
Nanoscale Res Lett ; 8(1): 151, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23547763

ABSTRACT

We propose the metal-assisted chemical etching of Ge surfaces in water mediated by dissolved oxygen molecules (O2). First, we demonstrate that Ge surfaces around deposited metallic particles (Ag and Pt) are preferentially etched in water. When a Ge(100) surface is used, most etch pits are in the shape of inverted pyramids. The mechanism of this anisotropic etching is proposed to be the enhanced formation of soluble oxide (GeO2) around metals by the catalytic activity of metallic particles, reducing dissolved O2 in water to H2O molecules. Secondly, we apply this metal-assisted chemical etching to the nanoscale patterning of Ge in water using a cantilever probe in an atomic force microscopy setup. We investigate the dependences of probe material, dissolved oxygen concentration, and pressing force in water on the etched depth of Ge(100) surfaces. We find that the enhanced etching of Ge surfaces occurs only when both a metal-coated probe and saturated-dissolved-oxygen water are used. In this study, we present the possibility of a novel lithography method for Ge in which neither chemical solutions nor resist resins are needed.

17.
J Biomed Opt ; 18(6): 061220, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23264964

ABSTRACT

We propose a method to visualize the arterial inflow, the vascular resistance, and the venous capacitance in the skin tissue from red, green, blue (RGB) digital color images. The arterial inflow and the venous capacitance in the skin tissue are visualized based on an increase in the rate of change in the total blood concentration and the change of the total blood concentration during upper limb occlusion at a pressure of 50 mmHg. The resultant arterial inflow with the measured mean arterial pressure also provides an image of the vascular resistance in human skin. The arterial inflow, the vascular resistance, and the venous capacitance acquired by the method are well correlated with those obtained from the conventional strain-gauge plethysmograph. The correlation coefficients R between the estimated values by the method and the measurements by the SPG are calculated to be 0.83 (P < 0.001) for the arterial inflow, 0.77 (P < 0.01) for the vascular resistance, and 0.77 (P < 0.01) for the venous capacitance. The arterial inflow and the venous capacitance in the skin tissue are significantly higher in active subjects compared with the sedentary subjects, whereas the vascular resistance was significantly lower in the active subjects compared with the sedentary subjects. The results of the present study indicate the possibility of using the proposed method for evaluating the peripheral vascular functions in human skin.


Subject(s)
Image Processing, Computer-Assisted/methods , Skin/blood supply , Computer Simulation , Hand/blood supply , Humans , Monte Carlo Method , Photography , Regression Analysis , Skin/chemistry , Vascular Capacitance/physiology , Vascular Resistance/physiology
18.
J Biomed Opt ; 16(8): 086012, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21895324

ABSTRACT

In order to visualize human skin hemodynamics, we investigated a method that is specifically developed for the visualization of concentrations of oxygenated blood, deoxygenated blood, and melanin in skin tissue from digital RGB color images. Images of total blood concentration and oxygen saturation can also be reconstructed from the results of oxygenated and deoxygenated blood. Experiments using tissue-like agar gel phantoms demonstrated the ability of the developed method to quantitatively visualize the transition from an oxygenated blood to a deoxygenated blood in dermis. In vivo imaging of the chromophore concentrations and tissue oxygen saturation in the skin of the human hand are performed for 14 subjects during upper limb occlusion at 50 and 250 mm Hg. The response of the total blood concentration in the skin acquired by this method and forearm volume changes obtained from the conventional strain-gauge plethysmograph were comparable during the upper arm occlusion at pressures of both 50 and 250 mm Hg. The results presented in the present paper indicate the possibility of visualizing the hemodynamics of subsurface skin tissue.


Subject(s)
Image Processing, Computer-Assisted/methods , Photography/methods , Skin Pigmentation/physiology , Skin/blood supply , Adolescent , Adult , Arm , Blood Gas Monitoring, Transcutaneous , Female , Hemodynamics/physiology , Hemoglobins/analysis , Humans , Male , Melanins/analysis , Models, Biological , Monte Carlo Method , Oxyhemoglobins/analysis , Phantoms, Imaging , Plethysmography , Skin/chemistry , Therapeutic Occlusion
19.
J Nanosci Nanotechnol ; 11(4): 2968-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21776662

ABSTRACT

Germanium (Ge) is a promising substrate for semiconductor devices in the near future. However, wet-chemical preparations that enable the control of the structure of the Ge surface have not yet been developed. In this study, the surface structure of Ge(111) after HCl treatment is characterized by X-ray photoelectron spectroscopy (XPS), atomic force microscopy (AFM) and scanning tunneling microscopy (STM). XPS spectra revealed that purging with inert gas, such as nitrogen, is necessary to obtain a Ge surface free of oxide, probably because dissolved oxygen from air rapidly oxidizes the surface. Cl-terminated Ge surfaces are microscopically rough, but are composed of terraces and steps, as revealed by magnified STM images. Step edges run not along specific directions reflecting the crystallographic nature of the (111) surface but randomly. Many atomic-scale protrusions with the height of around 0.1 nm are dispersed on terraces. They are likely to be impurities such as carbon contaminants and water on Cl-terminated terraces attracted by Cl atoms with high electronegativity.


Subject(s)
Germanium/chemistry , Hydrochloric Acid/chemistry , Nanostructures/chemistry , Nanostructures/ultrastructure , Nitrogen/chemistry , Macromolecular Substances/chemistry , Materials Testing , Molecular Conformation , Particle Size , Surface Properties
20.
J Biol Chem ; 286(20): 18251-60, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21454683

ABSTRACT

The common polymorphism of p53 at codon 72, either encoding proline or arginine, has drawn attention as a genetic factor associated with clinical outcome or cancer risk for the last 2 decades. We now show that these two polymorphic variants differ in protein structure, especially within the N-terminal region and, as a consequence, differ in post-translational modification at the N terminus. The arginine form (p53-72R) shows significantly enhanced phosphorylation at Ser-6 and Ser-20 compared with the proline form (p53-72P). We also show diminished Mdm2-mediated degradation of p53-72R compared with p53-72P, which is at least partly brought about by higher levels of phosphorylation at Ser-20 in p53-72R. Furthermore, enhanced p21 expression in p53-72R-expressing cells, which is dependent on phosphorylation at Ser-6, was demonstrated. Differential p21 expression between the variants was also observed upon activation of TGF-ß signaling. Collectively, we demonstrate a novel molecular difference and simultaneously suggest a difference in the tumor-suppressing function of the variants.


Subject(s)
Codon , Genetic Predisposition to Disease , Neoplasms , Polymorphism, Genetic , Protein Processing, Post-Translational , Tumor Suppressor Protein p53 , Cell Line , Humans , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL