Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Mol Sci ; 23(16)2022 Aug 09.
Article in English | MEDLINE | ID: mdl-36012131

ABSTRACT

We examined proteomic profiles of rat liver extracellular vesicles (EVs) shed following treatment with a sub-toxic dose (500 mg/kg) of the pain reliever drug, acetaminophen (APAP). EVs representing the entire complement of hepatic cells were isolated after perfusion of the intact liver and analyzed with LC-MS/MS. The investigation was focused on revealing the function and cellular origin of identified EVs proteins shed by different parenchymal and non-parenchymal liver cells and their possible role in an early response of this organ to a toxic environment. Comparison of EV proteomic profiles from control and APAP-treated animals revealed significant differences. Alpha-1-macroglobulin and members of the cytochrome P450 superfamily were highly abundant proteins in EVs shed by the normal liver. In contrast, proteins like aminopeptidase N, metalloreductase STEAP4, different surface antigens like CD14 and CD45, and most members of the annexin family were detected only in EVs that were shed by livers of APAP-treated animals. In EVs from treated livers, there was almost a complete disappearance of members of the cytochrome P450 superfamily and a major decrease in other enzymes involved in the detoxification of xenobiotics. Additionally, there were proteins that predominated in non-parenchymal liver cells and in the extracellular matrix, like fibronectin, receptor-type tyrosine-protein phosphatase C, and endothelial type gp91. These differences indicate that even treatment with a sub-toxic concentration of APAP initiates dramatic perturbation in the function of this vital organ.


Subject(s)
Chemical and Drug Induced Liver Injury , Extracellular Vesicles , Acetaminophen/toxicity , Animals , Chemical and Drug Induced Liver Injury/metabolism , Chromatography, Liquid , Cytochrome P-450 Enzyme System/metabolism , Extracellular Vesicles/metabolism , Liver/metabolism , Proteomics , Rats , Tandem Mass Spectrometry
2.
Electrophoresis ; 42(12-13): 1388-1398, 2021 07.
Article in English | MEDLINE | ID: mdl-33837589

ABSTRACT

To identify changes in extracellular vesicles (EVs) secreted by the liver following drug-induced liver injury (DILI), rats were treated with a subtoxic dose (500 mg/kg) of the analgesic drug, acetaminophen (APAP). EVs were collected by liver perfusion of sham and APAP-treated rats. Changes in EVs morphology were examined by transmission electron microscopic analysis of negatively stained vesicles. Results from morphometric analysis of EVs revealed striking differences in their size and distribution. Proteome composition of EVs collected by liver perfusion was determined by mass spectrometry using methods of sample preparation that enabled better detection of both highly hydrophobic proteins and proteins with complex post-translational modifications. The collection of EVs after liver perfusion is an approach that enables the isolation of EVs shed not only by isolated hepatocytes, but also by the entire complement of hepatic cells. EVs derived after DILI had a lower content of alpha-1-macroglobulin, ferritin, and members of cytochrome 450 family. Fibronectin, aminopeptidase N, metalloreductase STEAP4, integrin beta, and members of the annexin family were detected only in APAP-treated samples of EVs. These results show that the present approach can provide valuable insights into the response of the liver following drug-induced liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Extracellular Vesicles , Acetaminophen/toxicity , Animals , Hepatocytes , Liver , Proteome , Rats
3.
Exp Mol Pathol ; 105(1): 89-97, 2018 08.
Article in English | MEDLINE | ID: mdl-29856983

ABSTRACT

The critical molecular and cellular mechanisms involved in the development and progression of prostate cancer remain elusive. In this report, we demonstrate that normal rat prostate epithelial cells (PEC) undergo spontaneous transformation at high passage (p > 85) evidenced by the acquisition of anchorage independent growth when plated on soft agar and tumorigenicity when injected into immunodeficient mice. In addition, we also report the discovery of a minor subpopulation of spontaneously transformed PEC derived from high passage PEC with the ability to migrate through a layer of 1% agar and form expanding colonies on the underlying plastic substratum. Comparison of these soft agar invasive (SAI) cells with low (p < 35), mid (p36-84) and high passage (p > 85) PEC identified marked differences in cell morphology, proliferation and motility. The SAI subpopulation was more tumorigenic than the high passage anchorage independent cultures from which they were isolated, as manifested by a decreased latency period and an increase in the size of tumors arising in immunodeficient mice. In contrast, low and mid passage cells were unable to grow on soft agar and failed to form tumors when injected into immunodeficient mice. Screening with antibody-based signaling arrays identified several differences in the altered expression levels of signaling proteins between SAI-derived cells and low or high passage PEC, including the up-regulation of EGFR and MAPK-related signaling pathways in SAI-selected cells. In summary, these studies suggest that the SAI assay selects for a novel, highly tumorigenic subpopulation of transformed cells that may represent an early step in the progression of slow growing prostatic carcinomas into more rapidly growing and aggressive tumors.


Subject(s)
Cell Separation/methods , Cell Transformation, Neoplastic , Epithelial Cells/cytology , Prostate/cytology , Animals , Cell Movement , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors/metabolism , MAP Kinase Signaling System , Male , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Primary Cell Culture/methods , Rats
4.
Carcinogenesis ; 37(4): 408-419, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26905589

ABSTRACT

Hepatocellular carcinoma (HCC) is a heterogeneous disease in which tumor subtypes can be identified based on the presence of adult liver progenitor cells. Having previously identified the mTOR pathway as critical to progenitor cell proliferation in a model of liver injury, we investigated the temporal activation of mTOR signaling in a rat model of hepatic carcinogenesis. The model employed chemical carcinogens and partial hepatectomy to induce progenitor marker-positive HCC. Immunohistochemical staining for phosphorylated ribosomal protein S6 indicated robust mTOR complex 1 (mTORC1) activity in early preneoplastic lesions that peaked during the first week and waned over the subsequent 10 days. Continuous administration of rapamycin by subcutaneous pellet for 70 days markedly reduced the development of focal lesions, but resulted in activation of the PI3K signaling pathway. To test the hypothesis that early mTORC1 activation was critical to the development and progression of preneoplastic foci, we limited rapamycin administration to the 3-week period at the start of the protocol. Focal lesion burden was reduced to a degree indistinguishable from that seen with continuous administration. Short-term rapamycin did not result in the activation of PI3K or mTORC2 pathways. Microarray analysis revealed a persistent effect of short-term mTORC1 inhibition on gene expression that resulted in a genetic signature reminiscent of normal liver. We conclude that mTORC1 activation during the early stages of hepatic carcinogenesis may be critical due to the development of preneoplastic focal lesions in progenitor marker-positive HCC. mTORC1 inhibition may represent an effective chemopreventive strategy for this form of liver cancer.


Subject(s)
Carcinoma, Hepatocellular/surgery , Gene Expression , Liver Neoplasms/surgery , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Disease Progression , Male , Rats , Rats, Inbred F344
5.
BMC Cancer ; 13: 463, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24098947

ABSTRACT

BACKGROUND: A major obstacle in treating colorectal cancer (CRC) is the acquired resistance to chemotherapeutic agents. An important protein in the regulation of cancer cell death and clinical outcome is Raf kinase inhibitor protein (RKIP). In contrast, activated signal transducer and activator of transcription 3 (STAT3) is a protein that promotes tumor cell survival by inhibiting apoptosis and has an important role in cancer progression in many of cancer types. The aim of this study was to evaluate the regulation of RKIP and STAT3 after treatment with clinically relevant chemotherapeutic agents (camptothecin (CPT) and oxaliplatin (OXP)) and the cytokine interleukin-6 (IL-6) in HCT116 colon cancer cells as well as evaluate the association between RKIP and STAT3 with clinical outcome of Stage II colon cancer patients. METHODS: HCT-116 colon cancer cells were treated with CPT, OXP, and IL-6 separately or in combination in a time and dose-dependent manner and examined for phosphorylated and non-phosphorylated RKIP and STAT3 via Western blot analysis. STAT3 transcriptional activity was measured via a luciferase reporter assay in HCT116 cells treated with CPT, IL-6 or transfected with JAK 1, 2 separately or in combination. We extended these observations and determined STAT3 and RKIP/ pRKIP in tumor microarrays (TMA) in stage II colon cancer patients. RESULTS: We demonstrate IL-6-mediated activation of STAT3 occurs in conjunction with the phosphorylation of RKIP in vitro in human colon cancer cells. OXP and CPT block IL-6 mediated STAT3 activation and RKIP phosphorylation via the inhibition of the interaction of STAT3 with gp130. We determined that STAT3 and nuclear pRKIP are significantly associated with poor patient prognosis in stage II colon cancer patients. CONCLUSIONS: In the analysis of tumor samples from stage II colon cancer patients and the human colon carcinoma cell line HCT116, pRKIP and STAT3, 2 proteins potentially involved in the resistance to conventional treatments were detected. The phosphorylation of pRKIP and STAT3 are induced by the cytokine IL-6 and suppressed by the chemotherapeutic drugs CPT and OXP. Therefore, these results suggest that STAT3 and pRKIP may serve as prognostic biomarkers in stage II colon cancer patients and may improve chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Organoplatinum Compounds/pharmacology , Phosphatidylethanolamine Binding Protein/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cytokine Receptor gp130/metabolism , Female , Gene Expression , Humans , Interleukin-6/metabolism , Interleukin-6/pharmacology , Janus Kinases/metabolism , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Oxaliplatin , Phosphorylation/drug effects , Prognosis , Protein Binding , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Transcription, Genetic , Tumor Burden
6.
Exp Mol Pathol ; 93(2): 250-60, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22613460

ABSTRACT

We have previously described the generation of a monoclonal antibody recognizing a novel cholangiocyte marker, designated BD.1, that is expressed by fetal and adult rat cholangiocytes but not hepatocytes or the hepatic progenitor cells known as oval cells. In the present report, we have undertaken a comprehensive examination of BD.1 expressed by long-term cultures of bile duct epithelial cells (BDEC) and prostate epithelial cells (PEC). We show that with continued passage, the levels of BD.1 expressed by BDEC and PEC drop significantly, a decrease that is temporally associated with transition from a diploid to an aneuploid karyotype. Cell cycle analysis revealed cell cycle dependent expression of BD.1 characterized by decreased BD.1 levels within the first 10 h after release from serum starvation followed by reacquisition as cells entered S phase. MAb BD.1 recognized a 170 kDa protein in Western blots and showed strong reactivity with a 170 kDa band in blots prepared from phosphoproteins isolated by metal affinity chromatography. Analysis by mass spectrometry of tryptic peptides generated from BD.1 purified by continuous elution electrophoresis identified the plus end microtubule-binding protein, CLIP170, in the fraction reactive with MAb BD.1. Double immunofluorescence with MAb BD.1 and a MAb specific for CLIP170 showed that both were reactive with intrahepatic bile ducts. However, overexpression or siRNA knockdown of CLIP170 in 293T cells did not significantly alter BD.1 levels, indicating that CLIP170 and BD.1 were distinct, co-migrating proteins. Immunoprecipitation analysis with MAb BD.1 and anti-CLIP170 antibodies showed that under microtubule depolymerizing conditions the two proteins could be co-precipitated with both antibodies, leading us to conclude they were capable of forming stable complexes. Two different protocols were devised to enrich for the CLIP170 binding protein recognized by MAb BD.1. Analysis of tryptic peptides by LC-ESI-MS/MS identified BD.1 as eIF3a, the largest subunit of the elongation initiation factor 3 (eIF3) complex. This identity was confirmed by the simultaneous knockdown of both BD.1 and eIF3a by eIF3a-specific siRNAs and by the strong reactivity of MAb BD.1 with the 170 kDa protein immunoprecipitated with the anti-eIF3a antibody, 5H10. Based on these findings, we concluded that the BD.1 antigen was identical to eIF3a, a multifunctional subunit of the eIf3 complex shown here to associate with microtubules through its interactions with CLIP170.


Subject(s)
Bile Ducts/metabolism , Biomarkers/metabolism , Epithelial Cells/metabolism , Eukaryotic Initiation Factor-3/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Antibodies, Monoclonal , Bile Ducts/cytology , Biomarkers/chemistry , Cell Cycle , Cell Separation/methods , Cells, Cultured , Epithelial Cells/cytology , Eukaryotic Initiation Factor-3/genetics , Flow Cytometry , Gene Knockdown Techniques , Male , Microtubules/metabolism , Peptide Mapping , Prostate/cytology , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Inbred F344 , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
7.
Exp Mol Pathol ; 93(1): 40-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22525806

ABSTRACT

Oval cell activation occurs under conditions of severe liver injury when normal hepatocyte proliferation is blocked. Recent studies have shown that a subset of hepatocellular carcinomas expresses oval cell markers, suggesting that these cells are targets of hepatocarcinogens. However, the signaling pathways that control oval cell activation and proliferation are not well characterized. Based on the role of the nutrient signaling kinase complex, mTORC1, in liver development, we investigated the role of this pathway in oval cell activation. Oval cell proliferation was induced in male Fisher rats by a modification of the traditional choline deficient plus ethionine model (CDE) or by 2-acetylaminoflourene treatment followed by 2/3 partial hepatectomy with or without initiation by diethylnitrosamine. To assess the role of mTOR in the oval cell response and development of preneoplastic foci, the effect of the mTORC1 inhibitor, rapamycin, was studied in all models. Rapamycin induced a significant suppression of the oval cell response in both models, an effect that coincided with a decrease in oval cell proliferation. Rapamycin administration did not affect the abundance of neutrophils or natural killer cells in CDE-treated liver or the expression of key cytokines. Gene expression studies revealed the fetal hepatocyte marker MKP-4 to be expressed in oval cells. In an experimental model of hepatic carcinogenesis, rapamycin decreased the size of preneoplastic foci and the rate of cell proliferation within the foci. mTORC1 signaling plays a key role in the oval cell response and in the development of preneoplastic foci. This pathway may be a target for the chemoprevention of hepatocellular carcinoma.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Hepatocytes/drug effects , Precancerous Conditions/drug therapy , Sirolimus/pharmacology , Animals , Carcinoma, Hepatocellular/prevention & control , Cell Proliferation/drug effects , Cell Shape , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Choline Deficiency/metabolism , Diethylnitrosamine/toxicity , Dual-Specificity Phosphatases/biosynthesis , Ethionine/toxicity , Fluorenes/toxicity , Gene Expression Profiling , Hepatectomy/methods , Liver Neoplasms, Experimental/prevention & control , Male , Mechanistic Target of Rapamycin Complex 1 , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Multiprotein Complexes , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Proteins/metabolism , Rats , TOR Serine-Threonine Kinases
8.
Exp Mol Pathol ; 89(3): 248-59, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20655306

ABSTRACT

Cholangiocarcinoma, a severe form of biliary cancer, has a high mortality rate resulting partially from the advanced stage of disease at earliest diagnosis. A better understanding of the progressive molecular and cellular changes occurring during spontaneous cholangiocarcinogenesis is needed to identify potential biomarkers for diagnosis/prognosis or targets for novel therapeutics. Here, we show that with continued passage (p) in vitro, rat bile duct epithelial cells (BDEC) accumulated neoplastic characteristics that by mid-passage (p31-85) included alterations in morphology, increased growth rate, growth factor independence, decreased cell adhesion, loss of cholangiocyte markers expressed at low passage (p<30), and onset of aneuploidy. At high passage (p>85), BDEC cultures showed increasing numbers of cells expressing activated, tyrosine phosphorylated ErbB-2/Neu, a receptor tyrosine kinase previously reported to be at elevated levels in cholangiocarcinomas. Enrichment for high passage ErbB-2/Neu-positive cells yielded several anchorage-independent sub-lines with elevated levels of activated ErbB-2/Neu and increased expression of cyclooxygenase-2 (COX-2). When injected into immunodeficient beige/nude/xid mice, these sub-lines formed poorly differentiated cystic tumors strongly positive for rat cholangiocyte markers, a finding consistent with a previous report showing the susceptibility of high passage, non-tumorigenic BDEC to transformation by activated ErbB-2/Neu. Mid passage BDEC, in contrast, were resistant to the transforming activity of activated ErbB-2/Neu and remained anchorage dependent in vitro and non-tumorigenic in vivo following stable transfection. Based on these findings, we concluded that during progression to high passage, cultured BDEC undergo preneoplastic changes that enhance their susceptibility to transformation by ErbB-2/Neu. The ability to generate cells at different points in the process of spontaneous neoplastic transformation offers a valuable model system for identifying molecular features that determine whether over-expression of activated ErbB-2/Neu is necessary and sufficient to induce neoplastic conversion.


Subject(s)
Cell Culture Techniques/methods , Cell Transformation, Neoplastic/genetics , Epithelial Cells/pathology , Receptor, ErbB-2/genetics , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Cell Separation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Epithelial Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Immunoblotting , Mice , Rats , Receptor, ErbB-2/metabolism , Transfection
9.
Cancer Res ; 65(23): 11010-7, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322250

ABSTRACT

Carcinoembryonic antigen (CEA)-related cell adhesion molecule 1 (CEACAM1) is a member of the CEA family of immunoglobulin-like adhesion molecules with two major splice variants, CEACAM1(a)-4L and CEACAM1(b)-4S, differing in the length of their COOH-terminal cytoplasmic tail. Both forms are down-regulated in prostate and liver carcinomas relative to normal tissues. We have previously shown in a nude mouse xenograft model that restoration of CEACAM1(a)-4L expression in human prostate carcinoma cells (PC-3) suppresses tumorigenicity, an effect observed with carcinomas from several other tissues but never established for hepatocellular carcinomas. In this report, we have examined the effect of CEACAM1(a)-4L on tumorigenicity of 1682A, a rat hepatocellular carcinoma that grows on the omentum when injected into the peritoneal cavity. Results show that restoration of CEACAM1(a)-4L expression at levels 13- and 0.45-fold compared with negative controls or normal hepatocytes, respectively, completely suppressed the formation of 1682A tumor nodules on the omentum at 3 weeks after injection. In contrast, 1682A cells infected with CEACAM1(b)-4S or an empty retroviral vector formed multiple clusters of tumor nodules. Although tumor nodules of 1682A cells positive and negative for CEACAM1(a)-4L did not display significant differences in histologic organization, aggregates formed in vitro by 1682A-L were smaller in size and displayed enlarged intercellular spaces relative to their 1682A-V counterparts. Restoration of CEACAM1(a)-4L expression did not elevate levels of apoptosis but seemed to cause an increase in the length of G1. This is the first demonstration of CEACAM1(a)-4L-induced tumor suppression in liver carcinomas using a quantifiable i.p. syngeneic transplantation model.


Subject(s)
Antigens, CD/physiology , Cell Adhesion Molecules/physiology , Liver Neoplasms, Experimental/pathology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Apoptosis/physiology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , G1 Phase/physiology , Gene Expression , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/physiology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Protein Isoforms , Rats , S Phase/physiology
10.
Oncotarget ; 8(16): 26041-26056, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28199961

ABSTRACT

We have shown previously that rapamycin, the canonical inhibitor of the mechanistic target of rapamycin (mTOR) complex 1, markedly inhibits the growth of focal lesions in the resistant hepatocyte (Solt-Farber) model of hepatocellular carcinoma (HCC) in the rat. In the present study, we characterized the proteome of persistent, pre-neoplastic focal lesions in this model. One group was administered rapamycin by subcutaneous pellet for 3 weeks following partial hepatectomy and euthanized 4 weeks after the cessation of rapamycin. A second group received placebo pellets. Results were compared to unmanipulated control animals and to animals that underwent an incomplete Solt-Farber protocol to activate hepatic progenitor cells. Regions of formalin-fixed, paraffin-embedded tissue were obtained by laser capture microdissection (LCM). Proteomic analysis yielded 11,070 unique peptides representing 2,227 proteins. Quantitation of the peptides showed increased abundance of known HCC markers (e.g., glutathione S-transferase-P, epoxide hydrolase, 6 others) and potential markers (e.g., aflatoxin aldehyde reductase, glucose 6-phosphate dehydrogenase, 10 others) in foci from placebo-treated and rapamycin-treated rats. Peptides derived from cytochrome P450 enzymes were generally reduced. Comparisons of the rapamycin samples to normal liver and to the progenitor cell model indicated that rapamycin attenuated a loss of differentiation relative to placebo. We conclude that early administration of rapamycin in the Solt-Farber model not only inhibits the growth of pre-neoplastic foci but also attenuates the loss of differentiated function. In addition, we have demonstrated that the combination of LCM and mass spectrometry-based proteomics is an effective approach to characterize focal liver lesions.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Proteome , Proteomics , Animals , Biomarkers , Chromatography, Liquid , Disease Models, Animal , Male , Peptides/metabolism , Proteomics/methods , Rats , Signal Transduction , Tandem Mass Spectrometry
11.
PLoS One ; 9(3): e92478, 2014.
Article in English | MEDLINE | ID: mdl-24658061

ABSTRACT

Raf kinase inhibitor protein (RKIP) is a member of the phosphatidylethanolamine-binding-protein (PEBP) family that modulates the action of many kinases involved in cellular growth, apoptosis, epithelial to mesenchymal transition, motility, invasion and metastasis. Previously, we described an inverse association between RKIP and signal transducers and activators of transcription 3 (STAT3) expression in gastric adenocarcinoma patients. In this study, we elucidated the mechanism by which RKIP regulates STAT3 activity in breast and prostate cancer cell lines. RKIP over expression inhibited c-Src auto-phosphorylation and activation, as well as IL-6-, JAK1 and 2-, and activated Raf-mediated STAT3 tyrosine and serine phosphorylation and subsequent activation. In MDA-231 breast cancer cells that stably over express RKIP, IL-6 treatment blocked STAT3 phosphorylation and transcriptional activation. Conversely, in RKIP knockdown MDA-231 cells: STAT3 phosphorylation and activation increased in comparison to parental MDA-231 cells. RKIP over expression resulted in constitutive physical interaction with STAT3 and blocked c-Src and STAT3 association. The treatment of DU145 prostate, but not PC3 prostate or MDA-231 breast, cancer cell lines with ENMD-1198 or MKC-1 dramatically increased expression of RKIP. Overexpression of RKIP sensitized PC3 and MDA-231 cells to MTI-induced apoptosis. Moreover, MTI treatment resulted in a decrease in Src-mediated STAT3 tyrosine phosphorylation and activation, an effect that was significantly enhanced by RKIP over expression. In stable RKIP over expressing MDA-231 cells, tumor xenograft growth induced by activated STAT3 is inhibited. RKIP synergizes with MTIs to induce apoptosis and inhibit STAT3 activation of breast and prostate cancer cells. RKIP plays a critical role in opposing the effects of pro-oncogenic STAT3 activation.


Subject(s)
Breast Neoplasms/physiopathology , Phosphatidylethanolamine Binding Protein/metabolism , Prostatic Neoplasms/physiopathology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Estrenes/pharmacology , Female , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/pharmacology , Janus Kinase 1/antagonists & inhibitors , Male , Mice , Neoplasm Transplantation , Phosphatidylethanolamine Binding Protein/biosynthesis , STAT3 Transcription Factor/metabolism , Transfection , Tubulin Modulators/pharmacology
12.
PLoS One ; 7(1): e29606, 2012.
Article in English | MEDLINE | ID: mdl-22235309

ABSTRACT

CEACAM1 is a multifunctional Ig-like cell adhesion molecule expressed by epithelial cells in many organs. CEACAM1-4L and CEACAM1-4S, two isoforms produced by differential splicing, are predominant in rat liver. Previous work has shown that downregulation of both isoforms occurs in rat hepatocellular carcinomas. Here, we have isolated an anchorage dependent clone, designated 253T-NT that does not express detectable levels of CEACAM1. Stable transfection of 253-NT cells with a wild type CEACAM1-4S expression vector induced an anchorage independent growth in vitro and a tumorigenic phenotype in vivo. These phenotypes were used as quantifiable end points to examine the functionality of the CEACAM1-4S transmembrane domain. Examination of the CEACAM1 transmembrane domain showed N-terminal GXXXG dimerization sequences and C-terminal tyrosine residues shown in related studies to stabilize transmembrane domain helix-helix interactions. To examine the effects of transmembrane domain mutations, 253-NT cells were transfected with transmembrane domain mutants carrying glycine to leucine or tyrosine to valine substitutions. Results showed that mutation of transmembrane tyrosine residues greatly enhanced growth in vitro and in vivo. Mutation of transmembrane dimerization motifs, in contrast, significantly reduced anchorage independent growth and tumorigenicity. 253-NT cells expressing CEACAM1-4S with both glycine to leucine and tyrosine to valine mutations displayed the growth-enhanced phenotype of tyrosine mutants. The dramatic effect of transmembrane domain mutations constitutes strong evidence that the transmembrane domain is an important determinant of CEACAM1-4S functionality and most likely by other proteins with transmembrane domains containing dimerization sequences and/or C-terminal tyrosine residues.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/metabolism , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Liver Neoplasms/pathology , Amino Acid Sequence , Animals , Antigens, CD/genetics , Carcinoma, Hepatocellular/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Proliferation , Cell Size , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Phenotype , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , Rats , Transfection , Tyrosine
13.
Article in English | MEDLINE | ID: mdl-24009878

ABSTRACT

BACKGROUND: Interest has been generated in the capacity of cellular-derived microvesicles to alter the fate of different target cells. Lung, liver, heart and brain-derived vesicles can alter the genetic phenotype of murine marrow cells; however, the stability of such changes and the mechanism of these changes remain unclear. In the present work, we show that lung-derived microvesicles (LDMV) alter the transcriptome and proteome of target marrow cells initially by mRNA and regulator(s) of transcription transfer, but that long term phenotype change is due solely to transfer of a transcriptional regulator with target cell. IN VIVO STUDIES: Whole bone marrow cells (WBM) were co-cultured with LDMV (both isolated from male C57BL/6 mice) or cultured alone (control). One week later, cultured WBM was transplanted into lethally-irradiated female C57BL/6 mice. Recipient mice were sacrificed 6 weeks later and WBM, spleens and livers were examined for the presence of lung-specific gene expression, including surfactants A, B, C and D, aquaporin-5, and clara cell specific protein, via real-time RT-PCR. Immunohistochemistry was also performed on lungs to determine the number of transplanted marrow-derived (Y chromosome+) type II pneumocytes (prosurfactant C+). Mice transplanted with LDMV co-cultured WBM expressed pulmonary epithelial cell genes in the cells of their bone marrow, livers and spleens and over fivefold more transplanted marrow-derived Y+/prosurfactant C+cells could be found in their lungs (vs. control mice). IN VITRO STUDIES: WBM (from mice or rats) was cultured with or without LDMV (from mice or rats) for 1 week then washed and cultured alone. WBM was harvested at 2-week intervals for real-time RT-PCR analysis, using species-specific surfactant primers, and for Western Blot analysis. Proteomic and microRNA microarray analyses were also performed on cells. LDMV co-cultured WBM maintained expression of pulmonary epithelial cell genes and proteins for up to 12 weeks in culture. Surfactant produced at later time points was specific only to the species of the marrow cell in culture indicating de novo mRNA transcription. These findings, in addition to the altered protein and microRNA profiles of LDMV co-cultured WBM, support a stable transcriptional mechanism for these changes. CONCLUSIONS: These data indicate that microvesicle alteration of cell fate is robust and long-term and represents an important new aspect of cellular biology.

14.
PLoS One ; 7(5): e37819, 2012.
Article in English | MEDLINE | ID: mdl-22662230

ABSTRACT

Helicobacter pylori (H. pylori) is a gram-negative, spiral-shaped bacterium that infects more than half of the world's population and is a major cause of gastric adenocarcinoma. The mechanisms that link H. pylori infection to gastric carcinogenesis are not well understood. In the present study, we report that the Raf-kinase inhibitor protein (RKIP) has a role in the induction of apoptosis by H. pylori in gastric epithelial cells. Western blot and luciferase transcription reporter assays demonstrate that the pathogenicity island of H. pylori rapidly phosphorylates RKIP, which then localizes to the nucleus where it activates its own transcription and induces apoptosis. Forced overexpression of RKIP enhances apoptosis in H. pylori-infected cells, whereas RKIP RNA inhibition suppresses the induction of apoptosis by H. pylori infection. While inducing the phosphorylation of RKIP, H. pylori simultaneously targets non-phosphorylated RKIP for proteasome-mediated degradation. The increase in RKIP transcription and phosphorylation is abrogated by mutating RKIP serine 153 to valine, demonstrating that regulation of RKIP activity by H. pylori is dependent upon RKIP's S153 residue. In addition, H. pylori infection increases the expression of Snail, a transcriptional repressor of RKIP. Our results suggest that H. pylori utilizes a tumor suppressor protein, RKIP, to promote apoptosis in gastric cancer cells.


Subject(s)
Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Stomach Neoplasms/metabolism , Antigens, Bacterial/metabolism , Apoptosis/genetics , Bacterial Proteins/metabolism , Cell Line, Tumor , Helicobacter pylori/pathogenicity , Humans , Interleukin-6/pharmacology , Phosphatidylethanolamine Binding Protein/genetics , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Stability , Protein Transport , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Snail Family Transcription Factors , Stomach Neoplasms/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects
15.
Cell Stress Chaperones ; 15(1): 39-53, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19415527

ABSTRACT

We previously described a cell surface reactive monoclonal antibody, MAb OC.10, which recognizes an epitope shared by rat fetal liver ductal cells, hepatic progenitor cells, mature cholangiocytes, and hepatocellular carcinomas (HCC). Here, intrasplenic injection of MAb OC.10 into newborn rats was shown by immunofluorescence microscopy to strongly label intrahepatic bile ducts. Furthermore, the in situ labeling of intrahepatic cholangiocytes by injecting MAb OC.10 increased the number of intraportal and intralobular bile ducts with well-defined lumens when compared to IgM-injected control animals. The antigen for MAb OC.10 was identified by mass spectrometry as Hsc70, a constitutively expressed heat shock protein belonging to the HSP70 family. Immunoblot analysis demonstrated that MAb OC.10 reacted with recombinant bovine Hsc70 protein, with protein immunoprecipitated from rat bile duct epithelial (BDE) cell lysates with monoclonal anti-Hsc70 antibody, and with Hsc70-FLAG protein over-expressed in human 293T cells. In addition, Hsc70-specific small interfering RNA reduced the amount of OC.10 antigen expressed in nucleofected BDE cells. Consistent with the specificity of MAb OC.10 for Hsc70, heat shock did not induce OC.10 expression in BDE cells, a characteristic of Hsp70. Immunofluorescence with BDE cells further suggested that MAb OC.10 binds a novel cell surface epitope of Hsc70. This was in contrast to a commercially available monoclonal anti-Hsc70 antibody that showed strong cytosolic reactivity. These findings demonstrate that presentation of the OC.10 epitope differs between cytosolic and surface forms of Hsc70 and may suggest distinct differences in protein conformation or epitope availability determined in part by protein-protein or protein-lipid interactions. Phage display and pepscan analysis mapped the epitope for MAb OC.10 to the N-terminal 340-384 amino acids of the ATPase domain of rat Hsc70. These findings suggest that MAb OC.10 recognizes an epitope on rat Hsc70 when presented on the cell surface that promotes morphogenic maturation of bile ducts in newborn rat liver. Furthermore, since we have shown previously that the OC.10 antigen is expressed on HCC subpopulations with oval cell characteristics, our current results indicate that Hsc70 has the potential to be expressed on the surface of certain tumor cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bile Ducts, Intrahepatic/growth & development , Epitopes/immunology , HSP70 Heat-Shock Proteins/immunology , Liver/growth & development , Amino Acid Sequence , Animals , Animals, Newborn , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Cells, Cultured , Epitope Mapping , Epitopes/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Molecular Sequence Data , Morphogenesis , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Rats
16.
J Proteomics ; 73(3): 678-88, 2010 Jan 03.
Article in English | MEDLINE | ID: mdl-19819359

ABSTRACT

The use of proteomic techniques in the monitoring of different production steps of plasma-derived clotting factor IX (pd F IX) was demonstrated. The first step, solid-phase extraction with a weak anion-exchange resin, fractionates the bulk of human serum albumin (HSA), immunoglobulin G, and other non-binding proteins from F IX. The proteins that strongly bind to the anion-exchange resin are eluted by higher salt concentrations. In the second step, anion-exchange chromatography, residual HSA, some proteases and other contaminating proteins are separated. In the last chromatographic step, affinity chromatography with immobilized heparin, the majority of the residual impurities are removed. However, some contaminating proteins still remain in the eluate from the affinity column. The next step in the production process, virus filtration, is also an efficient step for the removal of residual impurities, mainly high molecular weight proteins, such as vitronectin and inter-alpha inhibitor proteins. In each production step, the active component, pd F IX and contaminating proteins are monitored by biochemical and immunochemical methods and by LC-MS/MS and their removal documented. Our methodology is very helpful for further process optimization, rapid identification of target proteins with relatively low abundance, and for the design of subsequent steps for their removal or purification.


Subject(s)
Factor IX/isolation & purification , Plasma/chemistry , Proteomics/methods , Validation Studies as Topic , Anion Exchange Resins/chemistry , Anion Exchange Resins/pharmacology , Blood Coagulation Factors/analysis , Blood Coagulation Factors/isolation & purification , Chromatography, Affinity/methods , Chromatography, Agarose/methods , Clinical Laboratory Techniques , DEAE-Dextran/chemistry , DEAE-Dextran/pharmacology , Factor IX/analysis , Factor IX/metabolism , Hemofiltration/methods , Heparin/metabolism , Humans , Solid Phase Extraction/methods
17.
Exp Hematol ; 38(3): 233-45, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20079801

ABSTRACT

OBJECTIVE: Microvesicles have been shown to mediate intercellular communication. Previously, we have correlated entry of murine lung-derived microvesicles into murine bone marrow cells with expression of pulmonary epithelial cell-specific messenger RNA (mRNA) in these marrow cells. The present studies establish that entry of lung-derived microvesicles into marrow cells is a prerequisite for marrow expression of pulmonary epithelial cell-derived mRNA. MATERIALS AND METHODS: Murine bone marrow cells cocultured with rat lung, but separated from them using a cell-impermeable membrane (0.4-microm pore size), were analyzed using species-specific primers (for rat or mouse). RESULTS: These studies revealed that surfactant B and C mRNA produced by murine marrow cells were of both rat and mouse origin. Similar results were obtained using murine lung cocultured with rat bone marrow cells or when bone marrow cells were analyzed for the presence of species-specific albumin mRNA after coculture with rat or murine liver. These studies show that microvesicles both deliver mRNA to marrow cells and mediate marrow cell transcription of tissue-specific mRNA. The latter likely underlies the longer-term stable change in genetic phenotype that has been observed. We have also observed microRNA in lung-derived microvesicles, and studies with RNase-treated microvesicles indicate that microRNA negatively modulates pulmonary epithelial cell-specific mRNA levels in cocultured marrow cells. In addition, we have also observed tissue-specific expression of brain, heart, and liver mRNA in cocultured marrow cells, suggesting that microvesicle-mediated cellular phenotype change is a universal phenomena. CONCLUSION: These studies suggest that cellular systems are more phenotypically labile than previously considered.


Subject(s)
Bone Marrow Cells/metabolism , Exosomes/metabolism , RNA, Messenger/genetics , Transcription, Genetic , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Brain/cytology , Brain/metabolism , Cell Communication/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Cytoplasmic Granules , Liver/cytology , Liver/metabolism , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/cytology , Myocardium/metabolism , Peptides/genetics , Protein Precursors/genetics , Proteolipids/genetics , Rats , Rats, Inbred F344
18.
Transplantation ; 88(4): 486-95, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19696631

ABSTRACT

BACKGROUND: Pretreatment with retrorsine crosslinks host hepatocyte DNA and prevents proliferation after partial hepatectomy (PH), allowing selective expansion of transplanted progenitors. Shortcomings are length of protocol and carcinogenicity of retrorsine. METHODS: This report describes a rapid liver repopulation protocol using mitomycin C (MMC) to block proliferation of rat hepatocytes in response to PH. One week post-MMC treatment, dipeptidyl peptidase IV negative host rats were given a PH followed by injection of late gestation, newborn, or adult total liver isolates from dipeptidyl peptidase IV positive rats. For allogeneic transplantation, host rats received injections of anti-CD3 antibody before and after PH. RESULTS: Host liver staining 2 to 9 weeks posttransplantation revealed well-defined donor hepatocyte colonies with strong canalicular dipeptidyl peptidase IV activity. At the same cell dose, fetal and newborn isolates produced more colonies than adult liver isolates. Hepatocyte colonies also coexpressed marker proteins characteristic of adult hepatocytes and showed polarized localization of plasma membrane proteins. Host livers contained large clusters of sinusoids lined by dipeptidyl peptidase IV positive endothelial cells coexpressing the endothelial cell marker, RECA-1, but lacked the canalicular marker leucine aminopeptidase. Colonies containing donor hepatocytes, endothelial cells, and bile ducts were also observed. Similar levels of engraftment and expansion were achieved with allogeneic liver cell isolates by using anti-CD3 antibody treatment. CONCLUSIONS: The MMC transplantation model provides a rapid method for engraftment and expansion of hepatocytes, endothelial cells, and cholangiocytes and should be applicable to investigations centering on the role of endothelial cells in liver regeneration and the identification and characterization of putative endothelial, hepatocyte, and cholangiocyte progenitors.


Subject(s)
Bile Ducts/transplantation , Endothelial Cells/transplantation , Hepatocytes/transplantation , Animals , Animals, Newborn , Bile Ducts/cytology , Bile Ducts/enzymology , Cell Differentiation , Cell Proliferation/drug effects , Dipeptidyl Peptidase 4/deficiency , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Endothelial Cells/cytology , Endothelial Cells/enzymology , Female , Graft Enhancement, Immunologic , Hepatectomy , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/enzymology , Liver Regeneration , Mitomycin/pharmacology , Pregnancy , Pyrrolizidine Alkaloids/pharmacology , Rats , Rats, Inbred ACI , Rats, Inbred F344 , Rats, Mutant Strains , Transplantation, Homologous , Transplantation, Isogeneic
19.
Development ; 133(21): 4269-79, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17021035

ABSTRACT

We have used monoclonal antibodies against cell-surface developmental epitopes in combination with micromagnetic beads to isolate phenotypically defined subpopulations of cholangiocyte marker-positive fetal liver epithelial cells (CMP-FLEC). Differentiation potential was evaluated by injecting cell isolates from dipeptidyl peptidase IV (DPPIV) positive (DPPIV+) Fischer donor rats into the spleen of partially hepatectomized, DPPIV negative (DPPIV-) Fischer host rats exposed to retrorsine. At various time points, liver tissue was harvested and cells in DPPIV+ colonies were phenotyped by immunofluorescence and histochemical protocols. Functional differentiation and liver replacement were determined by comparing donor and host hepatocyte protein expression patterns and DPPIV enzyme activity in extracts from livers of host rats receiving CMP-FLEC. Our results showed that bipotentiality was retained during differentiation and maturation of CMP-FLEC, indicating that the acquisition of ductal morphology and phenotype were not indicative of lineage commitment. CMP-FLEC transplanted into the adult rat liver lost ductal and gained hepatocyte markers, and acquired protein expression patterns in 2D gels with a close similarity (>75% spot match) to host hepatocytes but differing significantly from the transplanted CMP-FLEC cell isolate (<25% spot match). The average size of donor hepatocyte colonies increased with time so that by 1 year, up to 70% of the host rat liver was replaced by CMP-FLEC derived DPPIV+ hepatocytes. Depletion of CMP-FLEC from fetal liver isolates resulted in a marked decrease in adult liver colonization, suggesting that a high percentage of the hepatocyte colonies in animals receiving total fetal liver isolates are derived from CMP-FLEC.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Biomarkers/metabolism , Liver Regeneration/physiology , Liver , Pyrrolizidine Alkaloids/toxicity , Animals , Antibodies, Monoclonal/metabolism , Cell Differentiation , Cell Lineage , Cell Transplantation , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Immunomagnetic Separation , Liver/cytology , Liver/drug effects , Liver/embryology , Liver/pathology , Phenotype , Rats , Rats, Inbred F344
20.
J Biol Chem ; 279(33): 35063-78, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15184366

ABSTRACT

In this paper, we have characterized the structure, evolutionary origin, and function of rat and human carcinoembryonic antigen-related cell adhesion molecule1 (CEACAM1) multifunctional Ig-like cell adhesion proteins that are expressed by many epithelial tissues. Restriction enzyme digestion reverse transcriptase-PCR analysis identified three cDNAs encoding novel CEACAM1 N-domains. Comparative sequence analysis showed that human and rat CEACAM1 N-domains segregated into two groups differing in similarity to rat CEACAM1(a)-4L and human CEACAM1. Sequence variability analysis indicated that both human and rat N-domains possessed two variable regions, and one contained a major adhesive epitope. Recombination analysis showed that the group of rat but not human N-domains with high sequence similarity was derived at least in part by recombination. Binding assays revealed that three monoclonal antibodies with strong reactivity for the CEACAM1(a)-4L N-domain showed no reactivity with CEACAM1(b)-4S, an allele with a different N-domain sequence. CEACAM1(b)-4S displayed adhesive activity efficiently blocked by a synthetic peptide corresponding to the adhesive epitope in CEACAM1(a)-4L. Blocking analysis also showed that the adhesive epitope for rat CEACAM1 was located downstream from the equivalent human and mouse epitopes. Glycosylation analysis demonstrated O-linked sugars on rat CEACAM1(b)-4S from COS-1 cells. However, this was not the alteration responsible for the lack of monoclonal antibody reactivity. When considered together with previous studies, our findings suggest an inverse relationship between functionality and amino acid sequence similarity to CEACAM1. Like IgG, the N-domain of CEACAM1 appears to tolerate 10-15% sequence diversification without loss of function but begins to show either altered specificity or diminished functionality at higher levels.


Subject(s)
Antigens, CD/chemistry , Antigens, Differentiation/chemistry , Carcinoembryonic Antigen/chemistry , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies, Monoclonal/chemistry , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Baculoviridae/genetics , Base Sequence , COS Cells , Cell Adhesion , Cell Adhesion Molecules , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Epitopes/chemistry , Fluorescent Antibody Technique, Indirect , Genetic Vectors , Glycosylation , Humans , Immunoglobulin G/chemistry , Immunoglobulin Variable Region , Insecta , Liver/metabolism , Male , Mice , Models, Genetic , Molecular Sequence Data , Peptide Biosynthesis , Phylogeny , Protein Isoforms , Protein Structure, Tertiary , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Recombination, Genetic , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL