Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38906678

ABSTRACT

H3.1 histone is predominantly synthesized and enters the nucleus during the G1/S phase of the cell cycle, as a new component of duplicating nucleosomes. Here, we found that p53 is necessary to secure the normal behavior and modification of H3.1 in the nucleus during the G1/S phase, in which p53 increases C-terminal domain nuclear envelope phosphatase 1 (CTDNEP1) levels and decreases enhancer of zeste homolog 2 (EZH2) levels in the H3.1 interactome. In the absence of p53, H3.1 molecules tended to be tethered at or near the nuclear envelope (NE), where they were predominantly trimethylated at lysine 27 (H3K27me3) by EZH2, without forming nucleosomes. This accumulation was likely caused by the high affinity of H3.1 toward phosphatidic acid (PA). p53 reduced nuclear PA levels by increasing levels of CTDNEP1, which activates lipin to convert PA into diacylglycerol. We moreover found that the cytosolic H3 chaperone HSC70 attenuates the H3.1-PA interaction, and our molecular imaging analyses suggested that H3.1 may be anchored around the NE after their nuclear entry. Our results expand our knowledge of p53 function in regulation of the nuclear behavior of H3.1 during the G1/S phase, in which p53 may primarily target nuclear PA and EZH2.


Subject(s)
Cell Nucleus , Enhancer of Zeste Homolog 2 Protein , Histones , Tumor Suppressor Protein p53 , Histones/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Nucleus/metabolism , Humans , Enhancer of Zeste Homolog 2 Protein/metabolism , G1 Phase , S Phase , Nuclear Envelope/metabolism , Methylation , Animals , Nucleosomes/metabolism
2.
Commun Biol ; 5(1): 403, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35501367

ABSTRACT

Estrogen receptor (ER) positive breast cancer represents 75% of all breast cancers in women. Although patients with ER+ cancers receive endocrine therapies, more than 30% develop resistance and succumb to the disease, highlighting the need to understand endocrine resistance. Here we show an unexpected role for the cell polarity protein SCRIB as a tumor-promoter and a regulator of endocrine resistance in ER-positive breast cancer cells. SCRIB expression is induced by estrogen signaling in a MYC-dependent manner. SCRIB interacts with SLC3A2, a heteromeric component of leucine amino acid transporter SLC7A5. SLC3A2 binds to the N-terminus of SCRIB to facilitate the formation of SCRIB/SLC3A2/LLGL2/SLC7A5 quaternary complex required for membrane localization of the amino acid transporter complex. Both SCRIB and SLC3A2 are required for cell proliferation and tamoxifen resistance in ER+ cells identifying a new role for the SCRIB/SLC3A2 complex in ER+ breast cancer.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Fusion Regulatory Protein 1, Heavy Chain , Membrane Proteins , Tamoxifen , Tumor Suppressor Proteins , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Proliferation , Cytoskeletal Proteins , Estrogens , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Humans , Large Neutral Amino Acid-Transporter 1 , Membrane Proteins/genetics , Receptors, Estrogen , Tamoxifen/pharmacology , Tumor Suppressor Proteins/genetics
3.
Cell Host Microbe ; 29(6): 941-958.e10, 2021 06 09.
Article in English | MEDLINE | ID: mdl-33989515

ABSTRACT

Infection with CagA-producing Helicobacter pylori plays a causative role in the development of gastric cancer. Upon delivery into gastric epithelial cells, CagA deregulates prooncogenic phosphatase SHP2 while inhibiting polarity-regulating kinase PAR1b through complex formation. Here, we show that CagA/PAR1b interaction subverts nuclear translocation of BRCA1 by inhibiting PAR1b-mediated BRCA1 phosphorylation. It hereby induces BRCAness that promotes DNA double-strand breaks (DSBs) while disabling error-free homologous recombination-mediated DNA repair. The CagA/PAR1b interaction also stimulates Hippo signaling that circumvents apoptosis of DNA-damaged cells, giving cells time to repair DSBs through error-prone mechanisms. The DSB-activated p53-p21Cip1 axis inhibits proliferation of CagA-delivered cells, but the inhibition can be overcome by p53 inactivation. Indeed, sequential pulses of CagA in TP53-mutant cells drove somatic mutation with BRCAness-associated genetic signatures. Expansion of CagA-delivered cells with BRCAness-mediated genome instability, from which CagA-independent cancer-predisposing cells arise, provides a plausible "hit-and-run mechanism" of H. pylori CagA for gastric carcinogenesis.


Subject(s)
Antigens, Bacterial/metabolism , BRCA1 Protein/metabolism , Bacterial Proteins/metabolism , Epithelial Cells/metabolism , Genomic Instability , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Stomach Neoplasms/microbiology , Adult , Aged , Aged, 80 and over , Animals , Carcinogenesis/metabolism , Cell Line , DNA Breaks, Double-Stranded , Epithelial Cells/microbiology , Female , Gene Expression Regulation, Neoplastic , Helicobacter pylori/pathogenicity , Host-Pathogen Interactions , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Serine-Threonine Kinase 3 , Signal Transduction , Stomach/microbiology , Tumor Suppressor Protein p53/metabolism
4.
Proc Natl Acad Sci U S A ; 116(45): 22673-22682, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31636183

ABSTRACT

Despite the omnipresence of specific host-symbiont associations with acquisition of the microbial symbiont from the environment, little is known about how the specificity of the interaction evolved and is maintained. The bean bug Riptortus pedestris acquires a specific bacterial symbiont of the genus Burkholderia from environmental soil and harbors it in midgut crypts. The genus Burkholderia consists of over 100 species, showing ecologically diverse lifestyles, and including serious human pathogens, plant pathogens, and nodule-forming plant mutualists, as well as insect mutualists. Through infection tests of 34 Burkholderia species and 18 taxonomically diverse bacterial species, we demonstrate here that nonsymbiotic Burkholderia and even its outgroup Pandoraea could stably colonize the gut symbiotic organ and provide beneficial effects to the bean bug when inoculated on aposymbiotic hosts. However, coinoculation revealed that the native symbiont always outcompeted the nonnative bacteria inside the gut symbiotic organ, explaining the predominance of the native Burkholderia symbiont in natural bean bug populations. Hence, the abilities for colonization and cooperation, usually thought of as specific traits of mutualists, are not unique to the native Burkholderia symbiont but, to the contrary, competitiveness inside the gut is a derived trait of the native symbiont lineage only and was thus critical in the evolution of the insect gut symbiont.


Subject(s)
Burkholderia/physiology , Heteroptera/microbiology , Host-Pathogen Interactions , Intestines/microbiology , Symbiosis , Animals , Models, Biological
5.
Emerg Infect Dis ; 25(5): 883-890, 2019 05.
Article in English | MEDLINE | ID: mdl-31002057

ABSTRACT

Bacillus cereus is associated with foodborne illnesses characterized by vomiting and diarrhea. Although some B. cereus strains that cause severe extraintestinal infections and nosocomial infections are recognized as serious public health threats in healthcare settings, the genetic backgrounds of B. cereus strains causing such infections remain unknown. By conducting pulsed-field gel electrophoresis and multilocus sequence typing, we found that a novel sequence type (ST), newly registered as ST1420, was the dominant ST isolated from the cases of nosocomial infections that occurred in 3 locations in Japan in 2006, 2013, and 2016. Phylogenetic analysis showed that ST1420 strains belonged to the Cereus III lineage, which is much closer to the Anthracis lineage than to other Cereus lineages. Our results suggest that ST1420 is a prevalent ST in B. cereus strains that have caused recent nosocomial infections in Japan.


Subject(s)
Bacillus cereus/classification , Bacillus cereus/genetics , Bacteremia , Cross Infection/microbiology , Gram-Positive Bacterial Infections/microbiology , Alleles , Cross Infection/epidemiology , DNA, Bacterial , Genes, Bacterial , Genotype , Gram-Positive Bacterial Infections/epidemiology , Humans , Japan/epidemiology , Molecular Typing , Phylogeny
6.
Vet Pathol ; 56(5): 703-710, 2019 09.
Article in English | MEDLINE | ID: mdl-30866742

ABSTRACT

Equine herpesvirus 1 (EHV-1) uses equine major histocompatibility complex class I (MHC class I) as an entry receptor. Exogenous expression of equine MHC class I genes in murine cell lines confers susceptibility to EHV-1 infection. To examine the in vivo role of equine MHC class I as an entry receptor for EHV-1, we generated transgenic (Tg) mice expressing equine MHC class I under the control of the CAG promoter. Equine MHC class I protein was expressed in the liver, spleen, lung, and brain of Tg mice, which was confirmed by Western blot. However, equine MHC class I antigen was only detected in bronchiolar epithelium and not in other tissues, using the immunofluorescence method employed in this study. Both Tg and wild-type (WT) mice developed pneumonia 3 days after intranasal infection with EHV-1. The bronchiolar epithelial cells of Tg mice showed more severe necrosis, compared with those in WT mice. In addition, the number of virus antigen-positive cells in the lungs was higher in Tg mice than in WT mice. These results suggest that exogenous expression of equine MHC class I renders mice more susceptible to EHV-1 infection.


Subject(s)
Herpesviridae Infections/immunology , Herpesvirus 1, Equid , Histocompatibility Antigens Class I/metabolism , Horses , Lung Diseases/virology , Animals , Histocompatibility Antigens Class I/genetics , Lung Diseases/immunology , Mice , Mice, Transgenic
7.
mSystems ; 3(5)2018.
Article in English | MEDLINE | ID: mdl-30417107

ABSTRACT

Bacillus anthracis is a Gram-positive endospore-forming bacterial species that causes anthrax in both humans and animals. In Zambia, anthrax cases are frequently reported in both livestock and wildlife, with occasional transmission to humans, causing serious public health problems in the country. To understand the genetic diversity of B. anthracis strains in Zambia, we sequenced and compared the genomic DNA of B. anthracis strains isolated across the country. Single nucleotide polymorphisms clustered these strains into three groups. Genome sequence comparisons revealed a large deletion in strains belonging to one of the groups, possibly due to unequal crossing over between a pair of rRNA operons. The deleted genomic region included genes conferring resistance to bacitracin, and the strains with the deletion were confirmed with loss of bacitracin resistance. Similar deletions between rRNA operons were also observed in a few B. anthracis strains phylogenetically distant from Zambian strains. The structure of bacitracin resistance genes flanked by rRNA operons was conserved only in members of the Bacillus cereus group. The diversity and genomic characteristics of B. anthracis strains determined in this study would help in the development of genetic markers and treatment of anthrax in Zambia. IMPORTANCE Anthrax is caused by Bacillus anthracis, an endospore-forming soil bacterium. The genetic diversity of B. anthracis is known to be low compared with that of Bacillus species. In this study, we performed whole-genome sequencing of Zambian isolates of B. anthracis to understand the genetic diversity between closely related strains. Comparison of genomic sequences revealed that closely related strains were separated into three groups based on single nucleotide polymorphisms distributed throughout the genome. A large genomic deletion was detected in the region containing a bacitracin resistance gene cluster flanked by rRNA operons, resulting in the loss of bacitracin resistance. The structure of the deleted region, which was also conserved among species of the Bacillus cereus group, has the potential for both deletion and amplification and thus might be enabling the species to flexibly control the level of bacitracin resistance for adaptive evolution.

8.
Int J Cancer ; 142(3): 607-617, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28975613

ABSTRACT

Cancer-associated extracellular vesicles (EVs) are intimately involved in establishment of tumor microenvironment and occurrence of metastasis. However, previous studies have mainly relied on experiments with cultured cell lines or mouse models, making it difficult to gain a full understanding of EV functions in human body. Hence, we extracted EVs directly from surgically resected viable clear cell renal cell carcinoma (ccRCC) tissues and adjacent normal renal tissues (n = 20). Quantitative LC/MS analysis identified 3,871 tissue-exudative EV (Te-EV) proteins, among which azurocidin (AZU1) was highly enriched in tumor Te-EVs (p = 2.85 × 10-3 , fold-change = 31.59). Importantly, AZU1 content was also significantly higher in serum EVs from ccRCC patients compared to those from healthy donors. We further found that ccRCC-derived EVs had AZU1-dependent membrane permeabilizing activity for the vascular endothelial cell layer. Thus Te-EVs should be ideal resource for investigation of physiological EV functions.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Carcinoma, Renal Cell/pathology , Carrier Proteins/metabolism , Endothelial Cells/pathology , Kidney Neoplasms/pathology , Animals , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Female , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Kidney Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Proteome/metabolism
9.
Neoplasia ; 19(3): 185-195, 2017 03.
Article in English | MEDLINE | ID: mdl-28152424

ABSTRACT

BACKGROUND & AIM: p53 activation by cellular stresses induces the transcription of hundreds of its target genes. To elucidate the entire picture of its downstream pathway, we screened a cDNA microarray dataset of adriamycin-treated HCT116 p53-/- or p53+/+ cells and identified EPSIN 3 as a novel p53 target. METHODS: Potential p53 binding sequences in the EPSIN 3 locus were evaluated by reporter and CHIP assays. To investigate the role of EPSIN 3 in the p53 downstream pathway, we assessed DNA damage-induced apoptosis in EPSIN 3-knockdown HCT116 cells or Epsin 3-deficient mice. In addition, we evaluated EPSIN 3 expression levels in various tissues, including gastric adenocarcinoma, human gastric mucosa with or without Helicobacter pylori infection, and mouse acute gastritis tissues induced by indomethacin. RESULTS: In response to DNA damage, p53 induced the expression of EPSIN 3 through the p53 binding elements in the EPSIN 3 promoter and the first intron. Knockdown of EPSIN 3 resulted in resistance to DNA damage-induced apoptosis both in vitro and in vivo. EPSIN 3 expression was down-regulated in gastric cancer tissues compared with normal tissues. In addition, Helicobacter pylori infection and indomethacin-induced acute gastritis repressed EPSIN 3 expression in gastric mucosa. CONCLUSIONS: EPSIN 3 is a novel p53 target and a key mediator of apoptosis. Chronic or acute mucosal inflammation as well as p53 inactivation induced down-regulation of EPSIN 3 and subsequently caused apoptosis resistance, which is a hallmark of cancer cells.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Apoptosis/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Cell Proliferation , Gastritis/genetics , Gastritis/metabolism , Gastritis/microbiology , Gastritis/pathology , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , HCT116 Cells , Humans , Protein Binding , Proteolysis , Stomach Neoplasms/pathology , Stress, Physiological , Tumor Suppressor Protein p53/genetics
10.
Biomed Res ; 37(1): 21-7, 2016.
Article in English | MEDLINE | ID: mdl-26912137

ABSTRACT

More than 50% of people in the world are infected with Helicobacter pylori (H. pylori), which induces various gastric diseases. Especially, epidemiological studies have shown that H. pylori infection is a major risk factor for gastric cancer. It has been reported that the levels of interleukin (IL)-1ß are upregulated in gastric tissues of patients with H. pylori infection. In this study, we investigated the induction mechanism of IL-1ß during H. pylori infection. We found that IL-1ßmRNA and protein were induced in phorbol-12-myristate-13-acetate (PMA)-differentiated THP-1 cells after H. pylori infection. This IL-1ß production was inhibited by a caspase-1 inhibitor and a ROS inhibitor. Furthermore, K(+) efflux and Ca(2+) signaling were also involved in this process. These data suggest that NOD-like receptor (NLR) family, pyrin domain containing 3 (NLRP3) and its complex, known as NLRP3 inflammasome, are involved in IL-1ß production during H. pylori infection because it is reported that NLRP3 inflammasome is activated by ROS, K(+) efflux and/or Ca(2+) signaling. These findings may provide therapeutic strategy for the control of gastric cancer in H. pylori-infected patients.


Subject(s)
Helicobacter Infections/metabolism , Helicobacter pylori , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Adenosine Triphosphate/metabolism , Calcium Signaling , Caspase 1/metabolism , Cell Line , Extracellular Space/metabolism , Helicobacter Infections/genetics , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Humans , Interleukin-1beta/genetics , Intracellular Space/metabolism , Macrophages/immunology , Potassium/metabolism , Reactive Oxygen Species/metabolism
11.
J Vet Med Sci ; 77(8): 993-5, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25797134

ABSTRACT

To follow-up anthrax in Zambia since the outbreak in 2011, we have collected samples from the environment and the carcasses of anthrax-suspected animals, and have tried to isolate Bacillus anthracis. In the process of identification of B. anthracis, we collected two isolates, of which colonies were similar to B. anthracis; however, from the results of identification using the molecular-based methods, two isolates were genetically related to the highly pathogenic B. cereus, of which clinical manifestation is severe and fatal (e.g., pneumonia). In this study, we showed the existence of bacteria suspected to be highly pathogenic B. cereus in Zambia, indicating the possibility of an outbreak caused by highly pathogenic B. cereus.


Subject(s)
Bacillus cereus/genetics , Animals , Anthrax/epidemiology , Anthrax/microbiology , Bacillus anthracis/genetics , Bacillus anthracis/isolation & purification , Bacillus anthracis/pathogenicity , Bacillus cereus/isolation & purification , Bacillus cereus/pathogenicity , Disease Outbreaks , Environmental Microbiology , Humans , Phylogeny , Zambia/epidemiology
12.
PLoS One ; 10(3): e0122004, 2015.
Article in English | MEDLINE | ID: mdl-25774512

ABSTRACT

Anthrax is an important zoonotic disease worldwide that is caused by Bacillus anthracis, a spore-forming pathogenic bacterium. A rapid and sensitive method to detect B. anthracis is important for anthrax risk management and control in animal cases to address public health issues. However, it has recently become difficult to identify B. anthracis by using previously reported molecular-based methods because of the emergence of B. cereus, which causes severe extra-intestinal infection, as well as the human pathogenic B. thuringiensis, both of which are genetically related to B. anthracis. The close genetic relation of chromosomal backgrounds has led to complexity of molecular-based diagnosis. In this study, we established a B. anthracis multiplex PCR that can screen for the presence of B. anthracis virulent plasmids and differentiate B. anthracis and its genetically related strains from other B. cereus group species. Six sets of primers targeting a chromosome of B. anthracis and B. anthracis-like strains, two virulent plasmids, pXO1 and pXO2, a bacterial gene, 16S rRNA gene, and a mammalian gene, actin-beta gene, were designed. The multiplex PCR detected approximately 3.0 CFU of B. anthracis DNA per PCR reaction and was sensitive to B. anthracis. The internal control primers also detected all bacterial and mammalian DNAs examined, indicating the practical applicability of this assay as it enables monitoring of appropriate amplification. The assay was also applied for detection of clinical strains genetically related to B. anthracis, which were B. cereus strains isolated from outbreaks of hospital infections in Japan, and field strains isolated in Zambia, and the assay differentiated B. anthracis and its genetically related strains from other B. cereus group strains. Taken together, the results indicate that the newly developed multiplex PCR is a sensitive and practical method for detecting B. anthracis.


Subject(s)
Bacillus anthracis/genetics , Bacillus cereus/genetics , Multiplex Polymerase Chain Reaction , Animal Diseases/diagnosis , Animal Diseases/microbiology , Animals , Anthrax/diagnosis , Anthrax/microbiology , Bacillus anthracis/classification , Bacillus cereus/classification , Chromosomes, Bacterial , Gram-Positive Bacterial Infections/diagnosis , Gram-Positive Bacterial Infections/microbiology , Humans , Plasmids/genetics , RNA, Bacterial , RNA, Ribosomal, 16S/genetics , Sensitivity and Specificity
13.
Springerplus ; 3: 288, 2014.
Article in English | MEDLINE | ID: mdl-25019040

ABSTRACT

The abnormal expression of microRNAs (miRNAs) is implicated in various human diseases, including cancers. Accordingly, miRNA expressions have been examined in many cancer tissues and blood, but there have been few studies examining smear samples from bone marrow (BM) or peripheral blood. Here we successfully isolated small RNAs from BM smears using a mirVana miRNA Isolation Kit with our original modifications. The isolated small RNAs were then used to measure the levels of representative miRNAs such as miR-155, let-7a, and U6 via real-time PCR with a specific TaqMan probe, although peaks for the ribosomal RNAs (18S, and 28S) were not identified. The PCR curves of the miRNAs were indistinguishable from those from BM living cells from the same donor. Finally, our method for BM smears identified numerous abnormally altered miRNAs (significantly decreased, 39 miRNAs; significantly increased, 27 miRNAs) in follicular lymphomas (FL) compared with normal donors via TaqMan real-time PCR miRNA array. The array indicated that miR-451 showed the greatest decrease in FL (a 345-fold decrease), while miR-338-5p showed the greatest increase in FL (172-fold) relative to normal donors. The miRNAs identified by our study might serve as markers to predict the invasion of FL cells into BM without biopsy. Furthermore, our method will provide a new avenue for the analysis of miRNAs in BM smear samples from various hematologic diseases.

14.
Genome Announc ; 2(2)2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24604644

ABSTRACT

In August 2011, an anthrax outbreak occurred among Hippopotamus amphibius hippopotamuses and humans in Zambia. Here, we report the draft genome sequence of the Bacillus anthracis outbreak strain CZC5, isolated from tissues of H. amphibius hippopotamuses that had died in the outbreak area.

15.
Mol Cell ; 43(1): 45-56, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21726809

ABSTRACT

Deregulation of SHP2 is associated with malignant diseases as well as developmental disorders. Although SHP2 is required for full activation of RAS signaling, other potential roles in cell physiology have not been elucidated. Here we show that SHP2 dephosphorylates parafibromin/Cdc73, a core component of the RNA polymerase II-associated factor (PAF) complex. Parafibromin is known to act as a tumor suppressor that inhibits cyclin D1 and c-myc by recruiting SUV39H1 histone methyltransferase. However, parafibromin can also act in the opposing direction by binding ß-catenin, thereby activating promitogenic/oncogenic Wnt signaling. We found that, on tyrosine dephosphorylation by SHP2, parafibromin acquires the ability to stably bind ß-catenin. The parafibromin/ß-catenin interaction overrides parafibromin/SUV39H1-mediated transrepression and induces expression of Wnt target genes, including cyclin D1 and c-myc. Hence, SHP2 governs the opposing functions of parafibromin, deregulation of which may cause the development of tumors or developmental malformations.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Tumor Suppressor Proteins/metabolism , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Mass Spectrometry , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/analysis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Tyrosine/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
16.
Arch Biochem Biophys ; 498(1): 35-42, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20363211

ABSTRACT

CagA protein is the most assessed effecter molecule of Helicobacter pylori. In this report, we demonstrate how CagA protein regulates the functions of dendritic cells (DC) against H. pylori infection. In addition, we found that CagA protein was tyrosine-phosphorylated in DC. The responses to cagA-positive H. pylori in DC were reduced in comparison to those induced by cagA-negative H. pylori. CagA-overexpressing DC also exhibited a decline in the responses against LPS stimulation and the differentiation of CD4(+) T cells toward Th1 type cells compared to wild type DC. In addition, the level of phosphorylated IRF3 decreased in CagA-overexpressing DC stimulated with LPS, indicating that activated SHP-2 suppressed the enzymatic activity of TBK1 and consequently IRF3 phosphorylation. These data suggest that CagA protein negatively regulates the functions of DC via CagA phosphorylation and that cagA-positive H. pylori strains suppress host immune responses resulting in their chronic colonization of the stomach.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Helicobacter pylori/physiology , Immunosuppressive Agents/immunology , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/metabolism , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Interferon Regulatory Factor-3/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/immunology , Stomach/microbiology , Stomach/pathology , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Tyrosine/metabolism
17.
Int J Cancer ; 125(11): 2497-504, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19588494

ABSTRACT

Infection with cagA-positive Helicobacter pylori is associated with gastric carcinoma. The cagA-encoded CagA protein is delivered into gastric epithelial cells and, upon tyrosine phosphorylation at the C-terminal EPIYA segments, binds and deregulates SHP-2 oncoprotein. On the basis of the differential alignment of the EPIYA segments, CagA can be subdivided into Western CagA, which is produced by H. pylori isolated in Western countries, and East Asian CagA, which is produced by H. pylori circulating in East Asian countries. Western CagA contains EPIYA-A, EPIYA-B and variable numbers of EPIYA-C segments, whereas East Asian CagA contains EPIYA-A, EPIYA-B and variable numbers of EPIYA-D segments. Upon tyrosine phosphorylation, EPIYA-C and EPIYA-D, respectively, serve as low-affinity and high-affinity SHP-2-binding sites. We previously reported that systemic expression of East Asian CagA (CagA-ABDD) induces gastrointestinal and hematopoietic malignancies in mice. In this study, we generated transgenic mice that systemically express Western CagA (CagA-ABCCC), the levels of which are comparable to those in mice expressing East Asian CagA. The mice developed gastric epithelial hypertrophy and gastrointestinal tumors and also showed lymphoid abnormality but not myeloid abnormalities such as granulocytosis and myeloid leukemia found in mice carrying East Asian CagA. The incidence of tumors in mice expressing Western CagA was significantly lower than that in mice expressing East Asian CagA. Our results indicate that Western CagA is qualitatively less oncogenic than East Asian CagA. Differential oncogenic potential of geographically distinct CagA isoforms may contribute to the differential prevalence of gastric carcinoma between East Asian countries and Western countries.


Subject(s)
Adenocarcinoma/microbiology , Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Stomach Neoplasms/microbiology , Animals , Asia, Eastern , Female , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Western World
18.
Proc Natl Acad Sci U S A ; 105(3): 1003-8, 2008 Jan 22.
Article in English | MEDLINE | ID: mdl-18192401

ABSTRACT

Infection with cagA-positive Helicobacter pylori is associated with gastric adenocarcinoma and gastric mucosa-associated lymphoid tissue (MALT) lymphoma of B cell origin. The cagA-encoded CagA protein is delivered into gastric epithelial cells via the bacterial type IV secretion system and, upon tyrosine phosphorylation by Src family kinases, specifically binds to and aberrantly activates SHP-2 tyrosine phosphatase, a bona fide oncoprotein in human malignancies. CagA also elicits junctional and polarity defects in epithelial cells by interacting with and inhibiting partitioning-defective 1 (PAR1)/microtubule affinity-regulating kinase (MARK) independently of CagA tyrosine phosphorylation. Despite these CagA activities that contribute to neoplastic transformation, a causal link between CagA and in vivo oncogenesis remains unknown. Here, we generated transgenic mice expressing wild-type or phosphorylation-resistant CagA throughout the body or predominantly in the stomach. Wild-type CagA transgenic mice showed gastric epithelial hyperplasia and some of the mice developed gastric polyps and adenocarcinomas of the stomach and small intestine. Systemic expression of wild-type CagA further induced leukocytosis with IL-3/GM-CSF hypersensitivity and some mice developed myeloid leukemias and B cell lymphomas, the hematological malignancies also caused by gain-of-function SHP-2 mutations. Such pathological abnormalities were not observed in transgenic mice expressing phosphorylation-resistant CagA. These results provide first direct evidence for the role of CagA as a bacterium-derived oncoprotein (bacterial oncoprotein) that acts in mammals and further indicate the importance of CagA tyrosine phosphorylation, which enables CagA to deregulate SHP-2, in the development of H. pylori-associated neoplasms.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gastrointestinal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Helicobacter pylori/metabolism , Hematologic Neoplasms/metabolism , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cell Transformation, Neoplastic/genetics , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Mice , Mice, Transgenic , Phosphotyrosine/metabolism
19.
Nature ; 447(7142): 330-3, 2007 May 17.
Article in English | MEDLINE | ID: mdl-17507984

ABSTRACT

Helicobacter pylori cagA-positive strains are associated with gastritis, ulcerations and gastric adenocarcinoma. CagA is delivered into gastric epithelial cells and, on tyrosine phosphorylation, specifically binds and activates the SHP2 oncoprotein, thereby inducing the formation of an elongated cell shape known as the 'hummingbird' phenotype. In polarized epithelial cells, CagA also disrupts the tight junction and causes loss of apical-basolateral polarity. We show here that H. pylori CagA specifically interacts with PAR1/MARK kinase, which has an essential role in epithelial cell polarity. Association of CagA inhibits PAR1 kinase activity and prevents atypical protein kinase C (aPKC)-mediated PAR1 phosphorylation, which dissociates PAR1 from the membrane, collectively causing junctional and polarity defects. Because of the multimeric nature of PAR1 (ref. 14), PAR1 also promotes CagA multimerization, which stabilizes the CagA-SHP2 interaction. Furthermore, induction of the hummingbird phenotype by CagA-activated SHP2 requires simultaneous inhibition of PAR1 kinase activity by CagA. Thus, the CagA-PAR1 interaction not only elicits the junctional and polarity defects but also promotes the morphogenetic activity of CagA. Our findings revealed that PAR1 is a key target of H. pylori CagA in the disorganization of gastric epithelial architecture underlying mucosal damage, inflammation and carcinogenesis.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Polarity , Epithelial Cells/cytology , Epithelial Cells/enzymology , Helicobacter pylori , Protein Serine-Threonine Kinases/metabolism , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cell Line , Epithelial Cells/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Structure, Quaternary , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Tight Junctions/metabolism
20.
Oncogene ; 23(39): 6590-602, 2004 Aug 26.
Article in English | MEDLINE | ID: mdl-15235584

ABSTRACT

The eukaryotic cell cycle is regulated by sequential activation and inactivation of cyclin-cyclin-dependent kinase (Cdk) complexes. In this work, we screened human cDNAs that can rescue yeast Saccharomyces cerevisiae from lethality caused by ectopic expression of human cyclin E and isolated a cDNA encoding ESXR1, a paired-like homeodomain-containing protein with a unique C-terminal proline-rich repeat region. In adult tissues, ESXR1 is primarily expressed in the testis. We demonstrate that ESXR1 prevents degradation of ubiquitinated cyclins in human cells. Accordingly, elevation of ESXR1 level results in accumulation of cyclin A and cyclin B1 and thereby provokes M-phase arrest. In human cells, the 65-kDa full-length ESXR1 protein is capable of proteolytically processing into N-terminal 45-kDa and C-terminal 20-kDa fragments. The C-terminal fragment, containing a proline-rich repeat region, is localized to the cytoplasm and displays the ability to inhibit cyclin degradation. In contrast, the N-terminal fragment, containing a paired-like homeodomain, is localized exclusively in the nucleus, suggesting that it plays a role in transcription. Our results indicate that proteolytic processing of ESXR1 plays a role in concerted regulation of the cell cycle and transcription in human cells.


Subject(s)
Cyclins/metabolism , Homeodomain Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , DNA, Complementary , Down-Regulation , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Hydrolysis , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL