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










Publication year range
1.
Int J Biochem Cell Biol ; 57: 123-34, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25449262

ABSTRACT

Members of the casitas B-lineage lymphoma (Cbl) family (Cbl, Cbl-b and Cbl-c) of ubiquitin ligases serve as negative regulators of receptor tyrosine kinases (RTKs). An essential role of Cbl-family protein-dependent ubiquitination for efficient ligand-induced lysosomal targeting and degradation is now well-accepted. However, a more proximal role of Cbl and Cbl-b as adapters for CIN85-endophilin recruitment to mediate ligand-induced initial internalization of RTKs is supported by some studies but refuted by others. Overexpression and/or incomplete depletion of Cbl proteins in these studies is likely to have contributed to this dichotomy. To address the role of endogenous Cbl and Cbl-b in the internalization step of RTK endocytic traffic, we established Cbl/Cbl-b double-knockout (DKO) mouse embryonic fibroblasts (MEFs) and demonstrated that these cells lack the expression of both Cbl-family members as well as endophilin A, while they express CIN85. We show that ligand-induced ubiquitination of EGFR, as a prototype RTK, was abolished in DKO MEFs, and EGFR degradation was delayed. These traits were reversed by ectopic human Cbl expression. EGFR endocytosis, assessed using the internalization of (125)I-labeled or fluorescent EGF, or of EGFR itself, was largely retained in Cbl/Cbl-b DKO compared to wild type MEFs. EGFR internalization was also largely intact in Cbl/Cbl-b depleted MCF-10A human mammary epithelial cell line. Inducible shRNA-mediated knockdown of CIN85 in wild type or Cbl/Cbl-b DKO MEFs had no impact on EGFR internalization. Our findings, establish that, at physiological expression levels, Cbl, Cbl-b and CIN85 are largely dispensable for EGFR internalization. Our results support the model that Cbl-CIN85-endophilin complex is not required for efficient internalization of EGFR, a prototype RTK.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , ErbB Receptors/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Animals , Cell Line , Endocytosis , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Transfection
2.
J Carcinog ; 10: 29, 2011.
Article in English | MEDLINE | ID: mdl-22190871

ABSTRACT

BACKGROUND: Well over a quarter of human breast cancers are ErbB2-driven and constitute a distinct subtype with substantially poorer prognosis. Yet, there are substantial gaps in our understanding of how ErbB2 tyrosine kinase activity unleashes a coordinated program of cellular and extracellular alterations that culminate in aggressive breast cancers. Cellular models that exhibit ErbB2 kinase dependency and can induce metastatic breast cancer in immune competent hosts are likely to help bridge this gap. MATERIALS AND METHODS: Here, we derived and characterized a cell line model obtained from a transgenic ErbB2/Neu-driven mouse mammary adenocarcinoma. RESULTS: The MPPS1 cell line produces metastatic breast cancers when implanted in the mammary fat pads of immune-compromised as well as syngeneic immune-competent hosts. MPPS1 cells maintain high ErbB2 overexpression when propagated in DFCI-1 or related media, and their growth is ErbB2-dependent, as demonstrated by concentration-dependent inhibition of proliferation with the ErbB kinase inhibitor Lapatinib. When grown in 3-dimensional (3-D) culture on Matrigel, MPPS1 cells predominantly form large irregular cystic and solid structures. Remarkably, low concentrations of Lapatinib led to a switch to regular acinar growth on Matrigel. Immunofluorescence staining of control vs. Lapatinib-treated acini for markers of epithelial polarity revealed that inhibition of ErbB2 signaling led to rapid resumption of normal mammary epithelium-like cell polarity. CONCLUSIONS: The strict dependence of the MPPS1 cell system on ErbB2 signals for proliferation and alterations in cell polarity should allow its use to dissect ErbB2 kinase-dependent signaling pathways that promote loss of cell polarity, a key component of the epithelial mesenchymal transition and aggressiveness of ErbB2-driven breast cancers.

3.
Bioarchitecture ; 1(1): 45-48, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21866262

ABSTRACT

ESCRT pathway proteins play a key role in sorting ubiquitinated membrane receptors towards lysosomes providing an important mechanism for attenuating cell surface receptor signaling. However, recent studies point to a positive role of ESCRT proteins in signal transduction in multiple species studied under physiological and pathological conditions. ESCRT components such as Tsg101 and Hrs are overexpressed in human cancers and Tsg101 depletion is detrimental for cell proliferation, survival and transformed phenotype of tumor cells. However, the mechanisms underlying the positive contributions of ESCRT pathway to surface receptor signaling have remained unclear. In a recent study, we showed that Tsg101 and Vps4 are essential for translocation of active Src from endosomes to focal adhesion and invadopodia, thereby revealing a role of ESCRT pathway in promoting Src-mediated migration and invasion. We discuss the implications of these and other recent studies which together suggest a role for the ESCRT pathway in recycling of endocytic cargo proteins, aside from its role in lysosomal targeting, potentially explaining the positive roles of ESCRT proteins in signal transduction.

4.
Cancer Biol Ther ; 11(2): 263-76, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21088503

ABSTRACT

The receptor tyrosine kinase ErbB2 is overexpressed in up to a third of breast cancers, allowing targeted therapy with ErbB2-directed humanized antibodies such as Trastuzumab. Concurrent targeting of ErbB2 stability with HSP90 inhibitors is synergistic with Trastuzumab, suggesting that pharmacological agents that can inhibit HSP90 as well as signaling pathways activated by ErbB2 could be useful against ErbB2-overexpressing breast cancers. The triterpene natural product Celastrol inhibits HSP90 and several pathways relevant to ErbB2-dependent oncogenesis including the NFκB pathway and the proteasome, and has shown promising activity in other cancer models. Here, we demonstrate that Celastrol exhibits in vitro antitumor activity against a panel of human breast cancer cell lines with selectivity towards those overexpressing ErbB2. Celastrol strongly synergized with ErbB2-targeted therapeutics Trastuzumab and Lapatinib, producing higher cytotoxicity with substantially lower doses of Celastrol. Celastrol significantly retarded the rate of growth of ErbB2-overexpressing human breast cancer cells in a mouse xenograft model with only minor systemic toxicity. Mechanistically, Celastrol not only induced the expected ubiquitinylation and degradation of ErbB2 and other HSP90 client proteins, but it also increased the levels of reactive oxygen species (ROS). Our studies show that the Michael Acceptor functionality in Celastrol is important for its ability to destabilize ErbB2 and exert its bioactivity against ErbB2-overexpressing breast cancer cells. These studies suggest the potential use of Michael acceptor-containing molecules as novel therapeutic modalities against ErbB2-driven breast cancer by targeting multiple biological attributes of the driver oncogene.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Triterpenes/administration & dosage , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Synergism , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/therapeutic use , Humans , Inhibitory Concentration 50 , Lapatinib , Mice , Mice, SCID , Pentacyclic Triterpenes , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/therapeutic use , Signal Transduction , Trastuzumab , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
5.
Proc Natl Acad Sci U S A ; 107(37): 16107-12, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805499

ABSTRACT

Active Src localization at focal adhesions (FAs) is essential for cell migration. How this pool is linked mechanistically to the large pool of Src at late endosomes (LEs)/lysosomes (LY) is not well understood. Here, we used inducible Tsg101 gene deletion, TSG101 knockdown, and dominant-negative VPS4 expression to demonstrate that the localization of activated cellular Src and viral Src at FAs requires the endosomal-sorting complexes required for transport (ESCRT) pathway. Tsg101 deletion also led to impaired Src-dependent activation of STAT3 and focal adhesion kinase and reduced cell migration. Impairment of the ESCRT pathway or Rab7 function led to the accumulation of active Src at aberrant LE/LY compartments followed by its loss. Analyses using fluorescence recovery after photo-bleaching show that dynamic mobility of Src in endosomes is ESCRT pathway-dependent. These results reveal a critical role for an ESCRT pathway-dependent LE/LY trafficking step in Src function by promoting localization of active Src to FAs.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , src-Family Kinases/metabolism , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , DNA-Binding Proteins/metabolism , Humans , Lysosomes/metabolism , Mice , Protein Transport , Transcription Factors/metabolism , src-Family Kinases/genetics
6.
J Biol Chem ; 285(2): 1555-68, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19826000

ABSTRACT

Non-malignant mammary epithelial cells (MECs) undergo acinar morphogenesis in three-dimensional Matrigel culture, a trait that is lost upon oncogenic transformation. Rho GTPases are thought to play important roles in regulating epithelial cell-cell junctions, but their contributions to acinar morphogenesis remain unclear. Here we report that the activity of Rho GTPases is down-regulated in non-malignant MECs in three-dimensional culture with particular suppression of Rac1 and Cdc42. Inducible expression of a constitutively active form of Vav2, a Rho GTPase guanine nucleotide exchange factor activated by receptor tyrosine kinases, in three-dimensional MEC culture activated Rac1 and Cdc42; Vav2 induction from early stages of culture impaired acinar morphogenesis, and induction in preformed acini disrupted the pre-established acinar architecture and led to cellular outgrowths. Knockdown studies demonstrated that Rac1 and Cdc42 mediate the constitutively active Vav2 phenotype, whereas in contrast, RhoA knockdown intensified the Vav2-induced disruption of acini, leading to more aggressive cell outgrowth and branching morphogenesis. These results indicate that RhoA plays an antagonistic role to Rac1/Cdc42 in the control of mammary epithelial acinar morphogenesis.


Subject(s)
Mammary Glands, Human/growth & development , Morphogenesis/physiology , Proto-Oncogene Proteins c-vav/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Line, Transformed , Female , Humans , Mammary Glands, Human/cytology , Proto-Oncogene Proteins c-vav/genetics , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
7.
Cancer Res ; 68(22): 9147-56, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010886

ABSTRACT

Podosomes mediate cell migration and invasion by coordinating the reorganization of actin cytoskeleton and focal matrix degradation. MMP and serine proteases have been found to function at podosomes. The lysosomal cysteine cathepsins, a third major class of matrix-degrading enzymes involved in tumor invasion and tissue remodeling, have yet to be linked to podosomes with the exception of cathepsin K in osteoclasts. Using inhibitors and shRNA-mediated depletion, we show that cathepsin B participates in podosomes-mediated focal matrix degradation and invasion in v-Src-transformed fibroblasts. We observed that lysosomal marker LAMP-1 localized at the center of podosome rosettes protruding into extracellular matrix using confocal microscopy. Time-lapse live-cell imaging revealed that lysosomal vesicles moved to and fused with podosomes. Disruption of lysosomal pH gradient with Bafilomycin A1, chloroquine, or ammonium chloride greatly enhanced the formation of podosomes and increased the matrix degradation. Live-cell imaging showed that actin structures, induced shortly after Bafilomycin A1 treatment, were closely associated with lysosomes. Overall, our results suggest that cathepsin B, delivered by lysosomal vesicles, is involved in the matrix degradtion of podosomes.


Subject(s)
Actins/physiology , Cathepsin B/physiology , Cell Transformation, Neoplastic , Cellular Structures/physiology , Extracellular Matrix/metabolism , Genes, src , Lysosomes/physiology , Animals , Cathepsin B/antagonists & inhibitors , Fibroblasts , Gelatin/metabolism , Lysosomal Membrane Proteins/analysis , Lysosomal Membrane Proteins/physiology , Macrolides/pharmacology , Mice , NIH 3T3 Cells , Neoplasm Invasiveness
8.
Cancer Biol Ther ; 7(10): 1630-40, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18769124

ABSTRACT

ErbB2 (or Her2/Neu) overexpression in breast cancer signifies poorer prognosis, yet it has provided an avenue for targeted therapy as demonstrated by the success of the humanized monoclonal antibody Trastuzumab (Herceptin). Resistance to Trastuzumab and eventual failure in most cases, however, necessitate alternate ErbB2-targeted therapies. HSP90 inhibitors such as 17-allylaminodemethoxygeldanamycin (17-AAG), potently downregulate the cell surface ErbB2. While the precise mechanisms of Trastuzumab or 17-AAG action remain unclear, ubiquitinylation-dependent proteasomal or lysosomal degradation of ErbB2 appears to play a substantial role. As Trastuzumab and 17-AAG induce the recruitment of distinct E3 ubiquitin ligases, Cbl and CHIP respectively, to ErbB2, we hypothesized that 17-AAG and Trastuzumab combination could induce a higher level of ubiquitinylation and downregulation of ErbB2 as compared to single drug treatments. We present biochemical and cell biological evidence that combined 17-AAG and Trastuzumab treatment of ErbB2-overexpressing breast cancer cell lines leads to enhanced ubiquitinylation, downregulation from the cell surface and lysosomal degradation of ErbB2. Importantly, combined 17-AAG and Trastuzumab treatment induced synergistic growth arrest and cell death specifically in ErbB2-overexpressing but not in ErbB2-low breast cancer cells. Our results suggest the 17-AAG and Trastuzumab combination as a mechanism-based combinatorial targeted therapy for ErbB2-overexpressing breast cancer patients.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Benzoquinones/administration & dosage , Breast Neoplasms/metabolism , Lactams, Macrocyclic/administration & dosage , Lysosomes/metabolism , Receptor, ErbB-2/metabolism , Ubiquitin/chemistry , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Death , Cell Line, Tumor , Flow Cytometry , HSP90 Heat-Shock Proteins/metabolism , Humans , Microscopy, Fluorescence , Models, Biological , Protein Structure, Tertiary , Trastuzumab
9.
Dev Biol ; 316(2): 191-9, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18334253

ABSTRACT

The lin-12/Notch signaling pathway is conserved from worms to humans and is a master regulator of metazoan development. Here, we demonstrate that lin-12/Notch gain-of-function (gf) animals display precocious alae at the L4 larval stage with a significant increase in let-7 expression levels. Furthermore, lin-12(gf) animals display a precocious and higher level of let-7 gfp transgene expression in seam cells at L3 stage. Interestingly, lin-12(gf) mutant rescued the lethal phenotype of let-7 mutants similar to other known heterochronic mutants. We propose that lin-12/Notch signaling pathway functions in late developmental timing, upstream of or in parallel to the let-7 heterochronic pathway. Importantly, the human microRNA let-7a was also upregulated in various human cell lines in response to Notch 1 activation, suggesting an evolutionarily conserved cross-talk between let-7 and the canonical lin-12/Notch signaling pathway.


Subject(s)
Caenorhabditis elegans/genetics , MicroRNAs/genetics , Mutation , Animals , Animals, Genetically Modified , Blotting, Northern , Caenorhabditis elegans/growth & development , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Larva , Phenotype , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Cancer Res ; 67(9): 4164-72, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483327

ABSTRACT

Epidermal growth factor receptor (EGFR), a member of the ErbB family of receptor tyrosine kinases, is overexpressed in as many as 60% cases of breast and other cancers. EGFR overexpression is a characteristic of highly aggressive molecular subtypes of breast cancer with basal-like and BRCA1 mutant phenotypes distinct from ErbB2-overexpressing breast cancers. Yet, EGFR is substantially weaker compared with ErbB2 in promoting the oncogenic transformation of nontumorigenic human mammary epithelial cells (human MEC), suggesting a role for cooperating oncogenes. Here, we have modeled the co-overexpression of EGFR and a biologically and clinically relevant potential modifier c-Src in two distinct immortal but nontumorigenic human MECs. Using a combination of morphologic analysis and confocal imaging of polarity markers in three-dimensional Matrigel culture together with functional analyses of early oncogenic traits, we show for the first time that EGFR and c-Src co-overexpression but not EGFR or c-Src overexpression alone unleashes an oncogenic signaling program that leads to hyperproliferation and loss of polarity in three-dimensional acinar cultures, marked enhancement of migratory and invasive behavior, and anchorage-independent growth. Our results establish that EGFR overexpression in an appropriate context (modeled here using c-Src overexpression) can initiate oncogenic transformation of nontumorigenic human MECs and provide a suitable in vitro model to interrogate human breast cancer-relevant oncogenic signaling pathways initiated by overexpressed EGFR and to identify modifiers of EGFR-mediated breast oncogenesis.


Subject(s)
Breast Neoplasms/enzymology , Cell Transformation, Neoplastic/metabolism , ErbB Receptors/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , Animals , Breast Neoplasms/pathology , CSK Tyrosine-Protein Kinase , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/pathology , Collagen , Drug Combinations , Epithelial Cells/enzymology , Epithelial Cells/pathology , Female , Humans , Laminin , Mice , Mice, Nude , Proteoglycans , Tumor Cells, Cultured , src-Family Kinases
11.
Curr Pharm Des ; 12(32): 4215-28, 2006.
Article in English | MEDLINE | ID: mdl-17100624

ABSTRACT

Toll-like receptors (TLRs) are sensors of microbial products that initiate host defense responses in multicellular organisms. They are mainly linked to innate immunity and bridging to adaptive immunity, signaling through different TLRs responsible for a wide range of biological responses. The intracellular signaling pathways through Toll/interleukin-1 receptor (IL-1R) domains result in recruitment of the cytoplasmic adaptor molecules, with subsequent activation of a signaling cascade leading to nuclear factor-kappa B (NF-kappaB). TLR-signaling induces host inflammatory response and the inflammation becomes more severe in the absence of several extra and intra cellular negative regulators of TLR-signaling. In the intestine, TLR-dependent activation of NF-kappaB plays a vital role in maintaining epithelial homeostasis as well as regulating infections and inflammation, while dysregulation of TLR-signaling is associated with the pathogenesis of inflammatory bowel diseases (IBD). Recent findings regarding innate immunity-mediated regulation of intestinal pathophysiology prove that development of new drugs targeting TLRs including antagonists of TLR-signaling and agonists of their negative regulators has a potential impact on therapeutic strategies for intestinal inflammatory diseases.


Subject(s)
Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/immunology , Inflammation/drug therapy , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/immunology , Animals , Gastrointestinal Diseases/pathology , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Signal Transduction
12.
Am J Physiol Gastrointest Liver Physiol ; 291(4): G735-43, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16959957

ABSTRACT

Midkine (MK) is a unique growth and differentiation factor that modulates the proliferation and migration of various cells; however, little is known regarding its relationship to intestinal diseases. The aim of this study was to investigate MK expression and its role in dextran sulfate sodium (DSS)-induced colitis in rats. The expressions of MK, receptor-like protein-tyrosine phosphatase (RPTP)-beta, and proinflammatory cytokines were examined in rat colonic tissues after the development of DSS-induced colitis using Northern blotting, immunohistochemistry, and laser-capture microdissection (LCM) coupled with RT-PCR. The effects of MK on the migration of intestinal epithelial cells (IEC-6) were also evaluated in vitro using an intestinal wound repair model. MK expression was significantly increased in damaged colonic mucosa, mainly from day 3 to day 5 after the end of DSS administration, with abundant MK immunoreactive signals detected in submucosal fibroblasts. Expressions of proinflammatory cytokines were most strongly induced on day 1, which preceded the augmentation of MK expression. Results of LCM coupled with RT-PCR clearly indicated RPTP-beta expression in colonic epithelial cells. The migration assay showed that wound repair in the MK-treated groups was accelerated dose dependently. The present results showed for the first time that intestinal inflammation upregulates the MK-RPTP-beta system, which may stimulate mucosal regeneration during the process of healing of colitis. Additional investigations regarding the role of MK may contribute to the development of new options for the treatment of inflammatory bowel diseases.


Subject(s)
Colitis/metabolism , Colon/metabolism , Cytokines/metabolism , Gene Expression Regulation , Wound Healing , Animals , Cell Line , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Colitis/chemically induced , Colitis/pathology , Cytokines/genetics , Dextran Sulfate/pharmacology , Fibroblasts/metabolism , Interleukin-1/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Midkine , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Clin Vaccine Immunol ; 13(1): 132-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16426010

ABSTRACT

We recently demonstrated that the pattern recognition receptors (PRRs) toll-like receptor 2 (TLR2), TLR4, and CD14 are expressed in mouse colonic epithelium in a compartmentalized manner. Here we report the localization of TLR5, the receptor for bacterial flagellin, and its distinctive down-regulation during experimental colitis. Guts from normal BALB/c mice and those with dextran sodium sulfate (DSS)-induced colitis were compared. Each gut was divided into seven segments (stomach, small intestine [three parts], and colon [three parts]), and epithelial cells and crypt units were collected by scraping and EDTA treatment, respectively. Northern blotting showed that TLR5 mRNA was preferentially expressed in the epithelium of the proximal colon in normal mice. Laser capture microdissection coupled to reverse transcriptase PCR confirmed this localization. TLR5 protein expression reflected mRNA expression, as evidenced by Western blotting. In mice with acute colitis, inflammation occurred mainly in the distal colon. Interestingly, while TLR2, TLR4, and CD14 were up-regulated in the inflamed colon, TLR5 was down-regulated at both the mRNA and protein levels. Decreased TLR5 expression was more evident during chronic colitis. Additional in vitro studies using a mouse cell line, Colon-26, showed that gamma interferon (IFN-gamma) time- and dose-dependently down-regulates TLR5. In conclusion, epithelial cells, mainly in the proximal colon, constitutively express TLR5. TLR5 expression is down-regulated in vivo during acute and chronic DSS-induced colitis, in contrast to the expression of TLR2, TLR4, and CD14. The mechanism governing TLR5 regulation may therefore differ from that controlling other PRRs. Finally, IFN-gamma may be involved in down-regulating TLR5 expression.


Subject(s)
Cecum/chemistry , Colitis/metabolism , Down-Regulation , Toll-Like Receptor 5/metabolism , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/immunology , Disease Models, Animal , Disease Progression , Epithelium , Humans , Interferon-gamma/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Mice , RNA, Messenger/biosynthesis , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/immunology , Up-Regulation
14.
J Immunol ; 173(2): 1406-16, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15240737

ABSTRACT

TLR4, a member of pattern recognition receptors, is the main receptor of LPS. MD-2 physically associates with TLR4 on the cell surface and confers LPS responsiveness. Helicobacter pylori LPS is one of the major virulence factors for induction of gastritis. We demonstrated in this study the role of MD-2 in TLR4-dependent signaling in H. pylori-associated gastritis. Gastric biopsy samples collected from patients with and without H. pylori infection and four gastric cancer cell lines were used for this study. TLR-4 and MD-2 expression in biopsy specimens and the cell lines was examined by using RT-PCR. Localization of TLR-4 in histological sections was evaluated by immunohistochemistry. For in vitro functional assays, we established stable transfectants of AGS cells expressing TLR4 and MD-2. Cellular distribution of TLR4 was examined by flow cytometry. NF-kappaB activation and activation of IL-8 and MD-2 promoters were assessed by reporter gene assay. H. pylori infection up-regulated the TLR4 and MD-2 expression in gastric mucosa. TLR4 staining was observed predominantly in epithelial cells, located in both the cytoplasm and at the apical surface. MD-2 transfection in AGS cells markedly increased cell surface expression of TLR4 and augmented the activation of NF-kappaB and IL-8 promoter upon stimulation with H. pylori LPS. Live H. pylori also stimulated transcriptional activation of MD-2. This study revealed that MD-2 expression is elevated in gastric epithelial cells during H. pylori infection, suggesting that the TLR4/MD-2 system is a potent receptor complex involved in the response to H. pylori LPS in the stomach.


Subject(s)
Antigens, Surface/metabolism , Gastritis/metabolism , Helicobacter Infections/immunology , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Adult , Antigens, Surface/genetics , Female , Gastritis/immunology , Gastritis/microbiology , Genes, Reporter , Helicobacter Infections/metabolism , Helicobacter pylori/immunology , Helicobacter pylori/metabolism , Humans , Interleukin-8/genetics , Lipopolysaccharides/metabolism , Lymphocyte Antigen 96 , Male , Membrane Glycoproteins/genetics , Middle Aged , NF-kappa B/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Toll-Like Receptor 4 , Toll-Like Receptors
15.
Am J Physiol Gastrointest Liver Physiol ; 286(3): G508-14, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14563668

ABSTRACT

For the production and vesicle storage of histamine, Enterochromaffin-like (ECL) cells express histidine decarboxylase (HDC) and vesicular monoamine transporter 2 (VMAT2). Although HDC and VMAT2 show dynamic changes during gastric ulcer healing, the control system of their expression has not been fully investigated. In the present study, we investigated the effect of transforming growth factor-alpha (TGF-alpha) and proinflammatory cytokines on HDC and VMAT2 expression in rat ECL cells. Time course changes in the expression of TGF-alpha during the healing of acetic acid-induced ulcers were studied. EGF receptor (EGFR) expression was also examined in ECL cells, whereas the direct effects of TGF-alpha and proinflammatory cytokines on HDC and VMAT2 expression in ECL cells were investigated using in vivo and in vitro models. During the process of ulcer healing, expression of TGF-alpha mRNA was markedly augmented. Furthermore, EGFR was identified in isolated ECL cells. TGF-alpha stimulated HDC and VMAT2 mRNA expression and protein production and also increased histamine release from ECL cells. Selective EGFR tyrosine kinase inhibitor tyrphostin AG1478 almost completely inhibited HDC and VMAT2 gene expression induced by TGF-alpha in vivo and in vitro. During gastric mucosal injury, TGF-alpha was found to stimulate ECL cell functions by increasing HDC and VMAT2 expression.


Subject(s)
Enterochromaffin Cells/metabolism , Histidine Decarboxylase/biosynthesis , Membrane Glycoproteins/biosynthesis , Membrane Transport Proteins , Neuropeptides , Transforming Growth Factor alpha/pharmacology , Acetic Acid , Animals , Blotting, Northern , Cell Separation , Cells, Cultured , Enterochromaffin Cells/drug effects , Gastric Mucosa/cytology , Gastric Mucosa/pathology , Gene Expression Regulation, Enzymologic/drug effects , Histidine Decarboxylase/metabolism , Immunohistochemistry , Kinetics , Male , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
16.
Aliment Pharmacol Ther ; 18 Suppl 1: 106-12, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12925147

ABSTRACT

BACKGROUND: Midkine has been reported to bind to receptor-like protein tyrosine phosphatase (RPTP)-beta and to play important roles in growth and differentiation of various cells. Midkine is expressed in rat stomach during experimental ulcer healing, suggesting that the midkine-RPTP-beta system has some physiological functions in the stomach. Rebamipide is a mucoprotective drug used for the treatment of gastric ulcers. We have tested the hypothesis that the ulcer healing mechanism stimulated by rebamipide is linked physiologically to the gastric midkine-RPTP-beta system. MATERIALS AND METHODS: Seven-week-old-male Wistar rats were used. Midkine and RPTP-beta gene expression in rat stomach was investigated by laser capture microdissection coupled with the reverse transcription-polymerase chain reaction (RT-PCR). The effects of rebamipide on midkine and RPTP-beta expression in rat stomach and the gastric epithelial cell line RGM1 were evaluated by RT-PCR and Northern blot analyses. RESULTS: Midkine and RPTP-beta expression was detected in the gastric mucosal, submucosal and muscle layers. Rebamipide stimulated both midkine and RPTP-beta expression in rat stomach and RGM1 cells. CONCLUSION: Rebamipide may protect the gastric mucosa by regulating midkine and RPTP-beta expression.


Subject(s)
Alanine/analogs & derivatives , Alanine/pharmacology , Anti-Ulcer Agents/pharmacology , Carrier Proteins/metabolism , Cytokines , Gastric Mucosa/metabolism , Nerve Tissue Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Quinolones/pharmacology , Animals , Blotting, Northern , Cells, Cultured , Male , Midkine , RNA/metabolism , Rats , Rats, Wistar , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Reverse Transcriptase Polymerase Chain Reaction/methods
17.
J Immunol ; 170(8): 3977-85, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682225

ABSTRACT

Pattern recognition receptors (PRRs), which include the Toll-like receptors (TLRs), are involved in the innate immune response to infection. TLR4 is a model for the TLR family and is the main LPS receptor. We wanted to determine the expression of TLR4 and compare it with that of TLR2 and CD14 along the gastrointestinal mucosa of normal and colitic BALB/c mice. Colitis was induced with 2.5% dextran sodium sulfate (DSS). Mucosa from seven segments of the digestive tract (stomach, small intestine in three parts, and colon in three parts) was isolated by two different methods. Mucosal TLR4, CD14, TLR2, MyD88, and IL-1beta mRNA were semiquantified by Northern blotting. TLR4 protein was determined by Western blotting. TLR4/MD-2 complex and CD14 were evaluated by immunohistochemistry. PRR genes were constitutively expressed and were especially stronger in colon. TLR4 and CD14 mRNA were increased in the distal colon, but TLR2 mRNA was expressed more strongly in the proximal colon, and MyD88 had a uniform expression throughout the gut. Accordingly, TLR4 and CD14 protein levels were higher in the distal colon. TLR4/MD-2 and CD14 were localized at crypt bottom epithelial cells. TLR4/MD2, but not CD14, was found in mucosal mononuclear cells. Finally, DSS-induced inflammation was localized in the distal colon. All genes studied were up-regulated during DSS-induced inflammation, but the normal colon-stressed gut distribution was preserved. Our findings demonstrate that TLR4, CD14, and TLR2 are expressed in a compartmentalized manner in the mouse gut and provide novel information about the in vivo localization of PRRs.


Subject(s)
Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Antigens, Ly/biosynthesis , Antigens, Ly/genetics , Antigens, Ly/metabolism , Blotting, Northern , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Gene Expression Regulation/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Lymphocyte Antigen 96 , Macromolecular Substances , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Models, Animal , Myeloid Differentiation Factor 88 , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Up-Regulation/genetics , Up-Regulation/immunology
18.
J Lab Clin Med ; 141(2): 102-5, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12577045

ABSTRACT

Lafutidine is a novel histamine H(2)-receptor antagonist with a potent and long-lasting anti-acid secretory effect that has also been found to have a potent gastroprotective effect. We investigated the effect of lafutidine on gastric mucosal injury induced in rats with the use of water-immersion restraint stress (WRS) by examining serum calcitonin gene-related peptide (CGRP) concentrations, which we measured with the use of an enzyme immunometric assay. WRS-induced mucosal erosive injury in the stomach was reduced significantly by both lafutidine and famotidine pretreatment (from 7.79 +/- 2.02 mm(2) to 3.09 +/- 0.74 mm(2) and 4.05 +/- 1.18 mm(2), respectively). A single administration of lafutidine or famotidine did not change the serum CGRP concentration from the control value when these drugs were administered without WRS. Lafutidine pretreatment before WRS caused a significant increase in serum CGRP concentration compared with famotidine (lafutidine, 86.64 +/- 9.52 pg/mL; famotidine, 47.55 +/- 4.35 pg/mL; control, 58.43 +/- 6.07 pg/mL). Our results suggest that lafutidine augments CGRP release from the rat stomach when administered before the induction of WRS.


Subject(s)
Acetamides/pharmacology , Calcitonin Gene-Related Peptide/blood , Histamine H2 Antagonists/pharmacology , Immersion , Piperidines/pharmacology , Pyridines/pharmacology , Restraint, Physical , Stress, Physiological/blood , Animals , Famotidine/pharmacology , Male , Rats , Rats, Sprague-Dawley , Stomach Ulcer/etiology , Stomach Ulcer/prevention & control , Stress, Physiological/complications , Water
19.
J Lab Clin Med ; 142(6): 364-71, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14713888

ABSTRACT

Hypergastrinemia is known to cause hyperplasia of the gastric mucosa, especially in gastric enterochromaffinlike (ECL) cells. In some clinical conditions causing hypergastrinemia, such as long-term gastric-acid inhibition and gastric-mucosa atrophy, hyperplastic ECL cells may develop into gastric carcinoid tumors. A newly developed gastrin-receptor antagonist, S-0509, has been reported to block gastrin-induced stimulation of gastric-acid secretion. We therefore investigated whether S-0509 inhibits the omeprazole- and gastrin-stimulated hyperproliferation of gastric mucosa, especially of ECL cells. Daily administration of omeprazole and gastrin in male Sprague-Dawley rats induced marked hypergastrinemia and increased proliferation of gastric-mucosa cells. The numbers of ECL cells and of ECL cells producing messenger RNA for regenerating gene, a potent growth factor for gastric-mucosa cells, were also augmented by long-term administration of omeprazole and gastrin. Coadministration of S-0509 with omeprazole or gastrin almost completely inhibited the omeprazole- and gastrin-induced changes in gastric mucosa, including mucosal thickening and ECL hyperplasia. S-0509 did not induce gastric-mucosa atrophy, even when administered for as long as 4 weeks. In summary, we have found that a newly developed gastrin receptor antagonist, S-0509, inhibits omeprazole- and gastrin-induced mucosal hyperplasia, especially ECL-cell hyperplasia, in rats.


Subject(s)
Benzophenones/pharmacology , Gastric Mucosa/drug effects , Membrane Transport Proteins , Neuropeptides , Omeprazole/pharmacology , Phenylurea Compounds/pharmacology , Receptors, Cholecystokinin/antagonists & inhibitors , Animals , Cell Division/drug effects , Enterochromaffin Cells/drug effects , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Gastrins/blood , Histidine Decarboxylase/genetics , Male , Membrane Glycoproteins/genetics , Proliferating Cell Nuclear Antigen/analysis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Vesicular Biogenic Amine Transport Proteins
20.
Dig Dis Sci ; 47(10): 2275-85, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12395901

ABSTRACT

Hepatocyte growth factor (HGF), a potent inducer of cell migration with morphogenic and mitogenic actions was reported to have key roles in the repair of various tissues. In order to evaluate the role of HGF in the repair process of inflammatory bowel disease, we have investigated the HGF expression in a dextran sodium sulfate (DSS) colitis model. We randomly assigned rats to a colitis group or to a placebo group; the former received a 7-day course of 5% DSS (mw 5 kDa) in drinking water. DSS-induced severe colitis in rats manifested with weight loss, diarrhea, and intestinal bleeding. Animals were killed from day 1 through 7 and on days 9 and 14 after the end of DSS administration. After DSS was withdrawn, disease activity subsided gradually and HGF expression was significantly enhanced along with the augmented expression of IL-1beta, TNF-alpha, and cyclooxygenase-2, accompanied by an increased number of proliferating epithelial cells in colon. These findings suggest that proinflammatory cytokines and cyclooxygenase-2 may have an important role in the mucosal repair in inflammatory bowel disease through increased production of HGF.


Subject(s)
Colitis/immunology , Cytokines/physiology , Hepatocyte Growth Factor/metabolism , Isoenzymes/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Animals , Colitis/chemically induced , Colitis/pathology , Colon/immunology , Colon/pathology , Cyclooxygenase 2 , Dextran Sulfate , Interleukin-1/physiology , Male , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...