Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Cell Death Differ ; 22(6): 1035-46, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25476776

ABSTRACT

Mitochondrial outer membrane permeabilization (MOMP) is a critical control point during apoptosis that results in the release of pro-apoptotic mitochondrial contents such as cytochrome c. MOMP is largely controlled by Bcl-2 family proteins such as Bax, which under various apoptotic stresses becomes activated and oligomerizes on the outer mitochondrial membrane. Bax oligomerization helps promote the diffusion of the mitochondrial contents into the cytoplasm activating the caspase cascade. In turn, Bax is regulated primarily by anti-apoptotic Bcl-2 proteins including Bcl-xL, which was recently shown to prevent Bax from accumulating at the mitochondria. However, the exact mechanisms by which Bcl-xL regulates Bax and thereby MOMP remain partially understood. In this study, we show that the small CHCH-domain-containing protein CHCHD2 binds to Bcl-xL and inhibits the mitochondrial accumulation and oligomerization of Bax. Our data show that in response to apoptotic stimuli, mitochondrial CHCHD2 decreases prior to MOMP. Furthermore, when CHCHD2 is absent from the mitochondria, the ability of Bcl-xL to inhibit Bax activation and to prevent apoptosis is attenuated, which results in increases in Bax oligomerization, MOMP and apoptosis. Collectively, our findings establish CHCHD2, a previously uncharacterized small mitochondrial protein with no known homology to the Bcl-2 family, as one of the negative regulators of mitochondria-mediated apoptosis.


Subject(s)
Mitochondrial Proteins/metabolism , Transcription Factors/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Survival/physiology , DNA-Binding Proteins , HeLa Cells , Humans , Immunoprecipitation , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Protein Binding , Transcription Factors/genetics , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
2.
Oncogene ; 33(39): 4767-77, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-24141779

ABSTRACT

Basal-like breast tumors are aggressive cancers associated with high proliferation and metastasis. Chemotherapy is currently the only treatment option; however, resistance often occurs resulting in recurrence and patient death. Some extremely aggressive cancers are also associated with hypoxia, inflammation and high leukocyte infiltration. Herein, we discovered that the neural-specific transcription factor, Engrailed 1 (EN1), is exclusively overexpressed in these tumors. Short hairpin RNA (shRNA)-mediated knockdown of EN1 triggered potent and selective cell death. In contrast, ectopic overexpression of EN1 in normal cells activated survival pathways and conferred resistance to chemotherapeutic agents. Exogenous expression of EN1 cDNA reprogrammed the breast epithelial cells toward a long-lived, neural-like phenotype displaying dopaminergic markers. Gene expression microarrays demonstrated that the EN1 cDNA altered transcription of a high number of inflammatory molecules, notably chemokines and chemokine receptors, which could mediate prosurvival pathways. To block EN1 function, we engineered synthetic interference peptides (iPeps) comprising the EN1-specific sequences that mediate essential protein-protein interactions necessary for EN1 function and an N-terminal cell-penetrating peptide/nuclear localization sequence. These EN1-iPeps rapidly mediated a strong apoptotic response in tumor cells overexpressing EN1, with no toxicity to normal or non EN1-expressing cells. Delivery of EN1-iPeps into basal-like cancer cells significantly decreased the fifty percent inhibitory concentrations (IC50) of chemotherapeutic drugs routinely used to treat breast cancer. Lastly, matrix-assisted laser desorption/ionization-time of flight mass spectrometry and immunoprecipitation assays demonstrated that EN1-iPeps captured targets involved in transcriptional and post-transcriptional regulation. Importantly, the EN1-iPeps bound the glutamyl-prolyl tRNA synthetase (EPRS) target, which has been associated with the transcript-specific translational control of inflammatory proteins and activation of amino-acid stress pathways. This work unveils EN1 as an activator of intrinsic inflammatory pathways associated with prosurvival in basal-like breast cancer. We further build upon these results and describe the engineering of iPeps targeting EN1 (EN1-iPeps) as a novel and selective therapeutic strategy to combat these lethal forms of breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Homeodomain Proteins/physiology , Neoplasm Recurrence, Local/metabolism , Neoplasms, Basal Cell/metabolism , Peptides/pharmacology , Amino Acid Sequence , Antineoplastic Agents/metabolism , Apoptosis , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/pharmacology , Drug Screening Assays, Antitumor , Female , Humans , Inhibitory Concentration 50 , Kaplan-Meier Estimate , Molecular Sequence Data , Molecular Targeted Therapy , Neoplasms, Basal Cell/mortality , Neoplasms, Basal Cell/pathology , Peptides/metabolism
3.
Appl Environ Microbiol ; 79(7): 2471-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23377929

ABSTRACT

In Listeria monocytogenes serotype 4b isolates from sporadic listeriosis, heavy metal resistance was primarily encountered in certain clonal groups (ECI, ECII, and ECIa). All arsenic-resistant isolates harbored the arsenic resistance cassette previously identified in pLI100; ECIa harbored additional arsenic resistance genes and a novel cadmium resistance determinant in a conserved chromosomal locus.


Subject(s)
Arsenic/toxicity , Cadmium/toxicity , Drug Resistance, Bacterial , Genes, Bacterial , Listeria monocytogenes/drug effects , Listeria monocytogenes/genetics , Listeriosis/microbiology , Humans , Listeria monocytogenes/isolation & purification
4.
Nucleosides Nucleotides Nucleic Acids ; 27(6): 850-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18600551

ABSTRACT

We investigated the interacting proteins and intracellular localization of CTP synthetase 1 (CTPS1) in mammalian cells. CTPS1 interacted with a GST- peptidyl prolyl isomerase, Pin1 fusion (GST-Pin1) in a Ser 575 (S575) phosphorylation-dependent manner. Immunoprecipitation experiments demonstrated that CTPS1 also bound tubulin, and thirteen additional coimmunoprecipitating proteins were identified by mass spectrometry. Immunolocalization experiments showed that tubulin and CTPS1 colocalized subcellularly. Taxol treatment enhanced this but cotreatment of cells with the CTPS inhibitor, cyclopentenyl cytosine (CPEC), and taxol failed to disrupt the colocalization. Thus, these studies provide novel information on the potential interacting proteins that may regulate CTPS1 function or intracellular localization.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Carbon-Nitrogen Ligases/immunology , Cell Line , Humans , Immunoprecipitation , Mass Spectrometry , Peptidylprolyl Isomerase/metabolism , Protein Binding
5.
Toxicol Appl Pharmacol ; 214(1): 16-23, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16410015

ABSTRACT

Epidemiological studies have implicated zinc (Zn2+) in the toxicity of ambient particulate matter (PM) inhalation. We previously showed that exposure to metal-laden PM inhibits protein tyrosine phosphatase (PTP) activity in human primary bronchial epithelial cells (HAEC) and leads to Src-dependent activation of EGFR signaling in B82 and A431 cells. In order to elucidate the mechanism of Zn2+-induced EGFR activation in HAEC, we treated HAEC with 500 microM ZnSO4 for 5-20 min and measured the state of activation of EGFR, c-Src and PTPs. Western blots revealed that exposure to Zn2+ results in increased phosphorylation at both trans- and autophosphorylation sites in the EGFR. Zn2+-mediated EGFR phosphorylation did not require ligand binding and was ablated by the EGFR kinase inhibitor PD153035, but not by the Src kinase inhibitor PP2. Src activity was inhibited by Zn2+ treatment of HAEC, consistent with Src-independent EGFR transactivation in HAEC exposed to Zn2+. The rate of exogenous EGFR dephosphorylation in lysates of HAEC exposed to Zn2+ or V4+ was significantly diminished. Moreover, exposure of HAEC to Zn2+ also resulted in a significant impairment of dephosphorylation of endogenous EGFR. These data show that Zn2+-induced activation of EGFR in HAEC involves a loss of PTP activities whose function is to dephosphorylate EGFR in opposition to baseline EGFR kinase activity. These findings also suggest that there are marked cell-type-specific differences in the mechanism of EGFR activation induced by Zn2+ exposure.


Subject(s)
Bronchi/metabolism , Epithelial Cells/metabolism , ErbB Receptors/physiology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Respiratory Mucosa/metabolism , Zinc/toxicity , Blotting, Western , Cations, Divalent , Cells, Cultured , Humans , Phosphorylation , Respiratory Mucosa/cytology , Signal Transduction , Tissue Culture Techniques , src-Family Kinases/metabolism
6.
Leukemia ; 18(11): 1857-63, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15385935

ABSTRACT

Cyclopentenyl cytosine (CPEC) is a carbocyclic cytidine analog inhibitor of CTP synthetase and experimental drug for combination chemotherapy. CPEC treatment (50 nM) depleted intracellular CTP and induced a specific S-phase arrest and erythroid differentiation of human erythroleukemia K562 cells. The equilibrative nucleoside transporters (ENT1, 2) facilitated uptake of CPEC into K562 cells as evidenced by both NBMPR and dipyridamole inhibition of CPEC-mediated CTP depletion and erythroid differentiation. Incubation with the pyridinylimidazole p38 MAPK inhibitors, SB203580 or SB220025, suppressed both the CPEC-induced cell cycle arrest and differentiation of K562 cells. SB203580 also prevented the cell cycle arrest and erythroid differentiation of K562 cells induced by Leflunomide (LEF), a non-nucleoside inhibitor of the de novo pyrimidine pathway, without affecting LEF-induced depletion of pyrimidine pools. Finally, selective knockdown of p38 MAPK by using Smart Pooltrade mark siRNA to p38 MAPK significantly decreased the CPEC-induced differentiation of K562 cells. These results suggest that endogenous activity of p38 MAP kinases may be required for committing K562 cells to cell cycle arrest and erythroid differentiation under conditions of CTP depletion.


Subject(s)
Cell Differentiation/drug effects , Cytidine Triphosphate/metabolism , Cytidine/analogs & derivatives , Cytidine/pharmacology , Erythroid Precursor Cells/drug effects , Thioinosine/analogs & derivatives , p38 Mitogen-Activated Protein Kinases/metabolism , Affinity Labels/pharmacology , Carbon-Nitrogen Ligases/antagonists & inhibitors , Dipyridamole/pharmacology , Enzyme Inhibitors/pharmacology , Equilibrative Nucleoside Transporter 1/metabolism , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Humans , Isoxazoles/pharmacology , K562 Cells , Leflunomide , Leukemia, Myeloid , Phosphodiesterase Inhibitors/pharmacology , Pyrimidines/metabolism , RNA, Small Interfering/pharmacology , S Phase/drug effects , Thioinosine/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
7.
Cell Mol Life Sci ; 60(2): 321-36, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12678497

ABSTRACT

The de novo biosynthesis of pyrimidine nucleotides provides essential precursors for multiple growth-related events in higher eukaryotes. Assembled from ATP, bicarbonate and glutamine, the uracil and cytosine nucleotides are fuel for the synthesis of RNA, DNA, phospholipids, UDP sugars and glycogen. Over the past 2 decades considerable progress has been made in elucidating the mechanisms by which cellular pyrimidines are modulated to meet the needs of the cell. Recent studies demonstrate that CAD, a rate-limiting enzyme in the de novo synthesis of pyrimidines, is regulated through reversible phosphorylation, Myc-dependent transcriptional changes and caspase-mediated degradation. These studies point to increasing evidence for cooperation between key cell signaling pathways and basic elements of cellular metabolism, and suggest that these events have the potential to determine distinct cellular fates, including growth, differentiation and death. This review highlights some of the recent advances in the regulation of pyrimidine synthesis by growth-factor-stimulated signaling pathways.


Subject(s)
Pyrimidine Nucleotides/biosynthesis , Signal Transduction , Allosteric Regulation , Animals , Apoptosis , Aspartate Carbamoyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Carbon-Nitrogen Ligases/metabolism , Caspases/metabolism , Cell Differentiation , Cell Division , Cyclic AMP-Dependent Protein Kinases , Dihydroorotase/metabolism , Genes, myc , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Chemical , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism
8.
Int J Food Microbiol ; 65(1-2): 55-62, 2001 Apr 11.
Article in English | MEDLINE | ID: mdl-11322701

ABSTRACT

PulseNet is a national network of pubic health and food regulatory laboratories established in the US to detect clusters of foodborne disease and respond quickly to foodborne outbreak investigations. PulseNet laboratories currently subtype Escherichia coli O157:H7, non-typhoidal Salmonella, and Shigella isolates by a highly standardized 1-day pulsed-field gel electrophoresis (PFGE), and exchange normalized DNA "fingerprint" patterns via the Internet. We describe a standardized molecular subtyping protocol for subtyping Listeria monocytogenes that was recently added to PulseNet. The subtyping can be completed within 30 h from the time a pure culture of the bacteria is obtained.


Subject(s)
DNA, Bacterial/analysis , Electrophoresis, Gel, Pulsed-Field/methods , Listeria monocytogenes/classification , Bacterial Typing Techniques , DNA Restriction Enzymes , DNA, Bacterial/isolation & purification , Databases, Factual , Food Microbiology , Internet , Laboratories , Listeria monocytogenes/genetics , Public Health , United States
9.
J Biol Chem ; 275(37): 28688-94, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10862777

ABSTRACT

The activity of the serine/threonine kinase c-Raf (Raf) is inhibited by increased intracellular cAMP. This is believed to require phosphorylation with the cAMP-dependent protein kinase (PKA), although the mechanism by which PKA inhibits Raf is controversial. We investigated the requirement for PKA phosphorylation using Raf mutants expressed in HEK293 or NIH 3T3 cells. Phosphopeptide mapping of (32)P-labeled Raf (WT) or a mutant lacking a putative PKA phosphorylation site (serine to alanine, S43A) confirmed that serine 43 (Ser(43)) was the major cAMP (forskolin)-stimulated phosphorylation site in vivo. Interestingly, the EGF-stimulated Raf kinase activity of the S43A mutant was inhibited by forskolin equivalently to that of the WT Raf. Forskolin also inhibited the activation of an N-terminal deletion mutant Delta5-50 Raf completely lacking this phosphorylation site. Although WT Raf was phosphorylated by PKA, phosphorylation did not inhibit Raf catalytic activity in vitro, nor did forskolin treatment inhibit the activity of an N-terminally truncated Raf protein (Raf 22W) or a full-length Raf protein (Raf-CAAX) expressed in NIH 3T3 cells. In contrast, forskolin inhibited the EGF-dependent activation of a Raf isoform (B-Raf), lacking an analogous phosphorylation site to Ser(43). Thus, these results demonstrate that PKA exerts its inhibitory effects independently of direct Raf phosphorylation and suggests instead that PKA prevents an event required for the EGF-dependent activation of Raf.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Serine/metabolism , Catalytic Domain , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/physiology , Epidermal Growth Factor/pharmacology , Humans , Liver/metabolism , MAP Kinase Signaling System , Phosphorylation
10.
Nature ; 403(6767): 328-32, 2000 Jan 20.
Article in English | MEDLINE | ID: mdl-10659854

ABSTRACT

The de novo synthesis of pyrimidine nucleotides is required for mammalian cells to proliferate. The rate-limiting step in this pathway is catalysed by carbamoyl phosphate synthetase (CPS II), part of the multifunctional enzyme CAD. Here we describe the regulation of CAD by the mitogen-activated protein (MAP) kinase cascade. When phosphorylated by MAP kinase in vitro or activated by epidermal growth factor in vivo, CAD lost its feedback inhibition (which is dependent on uridine triphosphate) and became more sensitive to activation (which depends upon phosphoribosyl pyrophosphate). Both these allosteric regulatory changes favour biosynthesis of pyrimidines for growth. They were accompanied by increased epidermal growth factor-dependent phosphorylation of CAD in vivo and were prevented by inhibition of MAP kinase. Mutation of a consensus MAP kinase phosphorylation site abolished the changes in CAD allosteric regulation that were stimulated by growth factors. Finally, consistent with an effect of MAP kinase signalling on CPS II activity, epidermal growth factor increased cellular uridine triphosphate and this increase was reversed by inhibition of MAP kinase. Hence these studies may indicate a direct link between activation of the MAP kinase cascade and de novo biosynthesis of pyrimidine nucleotides.


Subject(s)
Aspartate Carbamoyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Dihydroorotase/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Multienzyme Complexes/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Aspartate Carbamoyltransferase/antagonists & inhibitors , Aspartate Carbamoyltransferase/chemistry , Aspartate Carbamoyltransferase/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/antagonists & inhibitors , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/chemistry , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , Cell Line , Cricetinae , Dihydroorotase/antagonists & inhibitors , Dihydroorotase/chemistry , Dihydroorotase/genetics , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Mesocricetus , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Molecular Sequence Data , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutagenesis, Site-Directed , Phosphoribosyl Pyrophosphate/metabolism , Phosphorylation , Pyrimidine Nucleotides/biosynthesis , Rats , Uridine Triphosphate/metabolism
11.
Transplantation ; 69(5): 773-80, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10755525

ABSTRACT

BACKGROUND: Chronic rejection is influenced by a variety of risk factors, including histoincompatibility and ischemia. Glycine, a cytoprotective agent, has been shown to protect against ischemia-reperfusion injury in the liver, inactivate hepatic resident macrophages, minimize cyclosporin A-induced nephrotoxicity, and exhibit immunosuppressive properties in vitro. The aim of this study was to investigate whether dietary glycine could reduce development of chronic rejection. METHODS: Lewis recipients of Fisher-344 abdominal aortic allografts received diets that contained either 5% glycine plus 15% casein or 20% casein as control for 10 weeks. Vascular lesions of aortic isografts and allografts were evaluated quantitatively with image analysis and cell counting. RESULTS: No significant vascular changes were observed in isografts (mean medial areas of 3.3 +/- 0.3x0(5) microm2). However, dramatic intimal thickening (neointimal area 2.1+/-0.3) and medial thinning (1.5+/-0.3) were observed in allografts from rats fed control diet. In contrast, glycine significantly reduced the neointima by 45% (1.2+/-0.3) and protected the media (3.5+/-0.2). This led to intima to media area ratios almost twice as large in the control group as in glycine-fed rats (2.2+/-0.4 vs. 1.1+/-0.3, P<0.05). Moreover, infiltrating leukocytes, especially macrophages, were reduced significantly in the adventitia by glycine. In addition, glycine inhibited proliferation and migration of rat aortic smooth muscle cells in culture by 45 and 60%, respectively. CONCLUSION: These results indicate that dietary glycine minimizes histopathological changes of chronic rejection by reducing the immune response and, in part, by minimizing proliferation and migration of smooth muscle cells.


Subject(s)
Aorta, Abdominal/transplantation , Glycine/administration & dosage , Graft Rejection/prevention & control , Animals , Aorta/surgery , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Cell Count/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Chronic Disease , Diet , Female , Glycine/blood , Glycine/therapeutic use , Leukocytes/pathology , Male , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Transplantation, Homologous , Transplantation, Isogeneic , Tunica Intima/metabolism
12.
Gastroenterology ; 118(1): 90-100, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10611157

ABSTRACT

BACKGROUND & AIMS: Infectious diarrhea caused by viruses plus enterotoxigenic bacteria is often more severe than diarrhea induced by either pathogen alone. We postulated that the increased cell adenosine 3',5'-cyclic monophosphate (cAMP) concentration observed during infection by enterotoxigenic organisms retards the intestinal repair process by blocking activation of mitogen-activated protein kinases (MAPKs) in proliferating intestinal cells. METHODS: We evaluated the effects of glutamine on MAPK activity, thymidine incorporation, and cell number in glutamine-starved and -sufficient rat intestinal crypt cells (IEC-6). RESULTS: In glutamine-starved cells, 10 mmol/L glutamine in the absence of serum stimulated [(3)H]thymidine incorporation 8-fold. This effect was inhibited by 60% with 8-(4-chlorophenylthio) (8-CPT)-cAMP (100 micromol/L) + isobutyl methylxanthine (100 micromol/L). In cells not starved of glutamine, glutamine stimulated thymidine incorporation by 3-fold, and 8-CPT-cAMP completely blocked the mitogenic effect. Inhibition of proliferation by cAMP persisted for at least 68 hours after cAMP removal. In vitro kinase assays showed that glutamine signaling requires an intact ERK (extracellular signal-related kinase) pathway in unstarved cells. In starved cells, at least one other pathway (JNK) was activated by glutamine, and the mitogenic inhibition by 8-CPT-cAMP was incomplete. Other intestinal fuels (glucose and acetate) were not mitogenic. CONCLUSIONS: Increased levels of intracellular cAMP inhibit ERKs but only partially reduce glutamine-stimulated proliferation in enterocytes adapted to low glutamine.


Subject(s)
Cyclic AMP/metabolism , Glutamine/metabolism , Intestinal Mucosa/metabolism , Mitogen-Activated Protein Kinases/metabolism , Cell Division/drug effects , Cell Line , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , DNA/biosynthesis , DNA/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Flavonoids/pharmacology , Glutamine/pharmacology , Intestines/cytology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Oxygen Consumption/drug effects , Thionucleotides/pharmacology , Thymidine/metabolism
13.
J Immunol ; 164(1): 176-82, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10605009

ABSTRACT

Previously, it was shown that glycine prevented increases in intracellular calcium ([Ca2+]i) in Kupffer cells. Since Kupffer cells and T lymphocytes are derived from the same pluripotent stem cell, it was hypothesized that glycine would prevent increases in [Ca2+]i in lymphocytes and inhibit cell proliferation. Lymphocyte proliferation was measured in one-way MLC with spleen cells from DA and Lewis rats and in enriched T lymphocyte preparations stimulated by immobilized anti-CD3 Ab. Glycine caused a dose-dependent decrease in cell proliferation to about 40% of control. Con A caused a dose-dependent increase in [Ca2+]i in Jurkat cells which was blunted maximally with 0.6 mM glycine. The effect of glycine was dependent on extracellular chloride and reversed by strychnine, an antagonist of the glycine-gated chloride channel. Similar results were obtained with rat T lymphocytes stimulated by anti-CD3 Ab. Surprisingly, glycine had no effect on IL-2 production in the mixed lymphocyte culture; therefore, the effect of glycine on IL-2-dependent proliferation was tested. Glycine and rapamycin caused dose-dependent decreases in IL-2-stimulated growth of Ctll-2 cells to about 60% and 40%, respectively, of control. Moreover, glycine also inhibited the IL-2-stimulated growth of rat splenic lymphocytes. It is concluded that glycine blunts proliferation in an IL-2-independent manner. This is consistent with the hypothesis that glycine activates a glycine-gated chloride channel and hyperpolarizes the cell membrane-blunting increases in [Ca2+]i that are required for transcription of factors necessary for cell proliferation.


Subject(s)
Glycine/physiology , Growth Inhibitors/physiology , Interleukin-2/physiology , T-Lymphocytes/cytology , Animals , CD3 Complex/immunology , Calcium/metabolism , Cell Division/drug effects , Cell Division/immunology , Chloride Channel Agonists , Concanavalin A/pharmacology , Cyclosporine/pharmacology , Female , Glycine/agonists , Growth Inhibitors/agonists , Growth Inhibitors/pharmacology , Humans , Immune Sera/pharmacology , Intracellular Fluid/metabolism , Jurkat Cells , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Rats , Rats, Inbred Lew , Strychnine/agonists , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
Am J Physiol ; 277(5): G1048-54, 1999 11.
Article in English | MEDLINE | ID: mdl-10564111

ABSTRACT

The aim of this study was to determine which PGE(2) receptors and signal transduction pathways are responsible for the stimulation of oxygen uptake in liver. Hepatic parenchymal cells isolated from female Sprague-Dawley rats were incubated either with PGE(2), 17-phenyl-omega-trinor PGE(2) (an EP(1)-specific agonist), or 11-deoxy PGE(1) (an EP(2)/EP(4)-specific agonist), and oxygen consumption was measured. Both PGE(2) and 11-deoxy PGE(1) stimulated oxygen consumption. However, an EP(1) agonist was without effect. Although PGE(2) elevated intracellular calcium, this occurred at concentrations approximately 500-fold lower than that required to stimulate oxygen uptake. PGE(2)-stimulated increases in cAMP formation correlated well with the increase in oxygen consumption. Dibutyryl cAMP also increased oxygen consumption. Furthermore, N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide, a cell-permeable inhibitor of protein kinase A (PKA), reduced the stimulation of oxygen uptake by PGE(2). Incubation of isolated parenchymal cell mitochondria with the purified catalytic subunit of PKA and ATP increased both state 3 rates of oxygen uptake and the respiratory control ratio by approximately 50%. Activation of these events was prevented by incubation with the PKA inhibitory peptide, PKI. These findings are consistent with the hypothesis that PGE(2) stimulates oxygen consumption via an EP(2) and/or EP(4) subclass of receptors through the actions of cAMP on a cAMP-dependent protein kinase.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Liver/enzymology , Oxygen/pharmacokinetics , Receptors, Prostaglandin E/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenosine Diphosphate/pharmacology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Anti-Ulcer Agents/pharmacology , Bucladesine/pharmacology , Cells, Cultured , Electron Transport/physiology , Female , Liver/chemistry , Liver/cytology , Mitochondria/drug effects , Mitochondria/enzymology , Oxygen Consumption/drug effects , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Succinic Acid/pharmacology
15.
Am J Physiol ; 277(5): L924-31, 1999 11.
Article in English | MEDLINE | ID: mdl-10564177

ABSTRACT

We have previously shown that exposure to combustion-derived metals rapidly (within 20 min) activated mitogen-activated protein kinases (MAPK), including extracellular signal-regulated kinase (ERK), in the human bronchial epithelial cell line BEAS. To study the mechanisms responsible for metal-induced activation of ERK, we examined the effect of noncytotoxic exposures to As, Cu, V, or Zn on the kinases upstream of ERK in the epidermal growth factor (EGF) receptor signaling pathway. Western blotting using phospho-specific ERK1/2 antibody demonstrated the selective MEK1/2 inhibitor PD-98059 blocked metal-induced phosphorylation of ERK1/2. Meanwhile, Western blotting using a phospho-specific MEK1/2 antibody showed that these metals induce a rapid phosphorylation of MEK1/2. Kinase activity assays confirmed the activation of MEK1/2 by metal treatment. Immunoprecipitation studies demonstrated that As, Cu, V, or Zn induces EGF receptor phosphorylation. Furthermore, the EGF receptor-specific tyrosine kinase inhibitor (PD-153035) significantly blocked the phosphorylation of MEK1/2 initiated by metals. Interestingly, we observed low levels of Raf-1 activity that were not increased by metal exposure in these cells through kinase activity assay. Finally, transfection assays showed that MEK1/2 inhibition could inhibit trans-activation of Elk1, a transcription factor in the ERK pathway, in BEAS cells exposed to metals. Together, these data demonstrate that As, Cu, V, and Zn can activate the EGF receptor signaling pathway in BEAS cells and suggest that this mechanism may be involved in pulmonary responses to metal inhalation.


Subject(s)
DNA-Binding Proteins , Epithelial Cells/enzymology , ErbB Receptors/physiology , MAP Kinase Signaling System/drug effects , Metals, Heavy/pharmacology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Pulmonary Alveoli/cytology , Transcription Factors , Air Pollutants/pharmacology , Arsenic/pharmacology , Cell Line, Transformed , Copper/pharmacology , Epithelial Cells/chemistry , Epithelial Cells/cytology , Humans , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase Kinases/metabolism , Potassium Channels/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Pulmonary Alveoli/chemistry , Pulmonary Alveoli/drug effects , Vanadium/pharmacology , Zinc/pharmacology , ets-Domain Protein Elk-1
16.
Am J Respir Cell Mol Biol ; 21(3): 357-64, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10460753

ABSTRACT

We previously showed that exposure to metal-laden combustion particles disregulates protein tyrosine phosphate homeostasis in human airway epithelial cells (HAEC). More recently, we reported that exposure to certain metal ions activates mitogen-activated protein kinases in HAEC. To study the mechanism responsible, we examined the effects of arsenic (As), vanadium (V), and zinc (Zn) on tyrosine phosphate catabolism in BEAS S6 cells or cultured human bronchial epithelial cells. Western blots and immunocytochemical analyses showed that exposure to noncytotoxic levels of As, V, or Zn resulted in increased levels of protein phosphotyrosines in HAEC. Tyrosine phosphatase activity, measured against [(32)P]-labeled PolyGlu:Tyr, was markedly inhibited in cells treated with V or Zn but was unaffected by exposure to As. Fast performance liquid chromatography fractionation and subsequent in-gel phosphatase activity assay of HAEC protein extracts revealed the presence of numerous tyrosine phosphatases, of varying molecular weights, that were effectively inhibited by exposure to V or Zn ions. As had no discernible effect on these enzymes. The protein tyrosine phosphatase PTP1B, immunoprecipitated from HAEC, was similarly inhibited by V and Zn but not by As ions. These data show that V and Zn may induce tyrosine phosphate accumulation by inhibiting dephosphorylation and implicate kinase activation as the mechanism in HAEC exposed to As. These findings suggest that metal exposure can activate signaling pathways through multiple mechanisms.


Subject(s)
Arsenic/pharmacology , Bronchi/enzymology , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Vanadium/pharmacology , Zinc/pharmacology , Bronchi/cytology , Bronchi/drug effects , Carrier Proteins/metabolism , Cell Line, Transformed , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Humans , Immunohistochemistry , Membrane Proteins/metabolism , Phosphotyrosine/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Time Factors
17.
J Biol Chem ; 274(13): 8917-24, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10085136

ABSTRACT

The calcium-dependent tyrosine kinase (CADTK), also known as Pyk2/RAFTK/CAKbeta/FAK2, is a cytoskeleton-associated tyrosine kinase. We compared CADTK regulation with that of the highly homologous focal adhesion tyrosine kinase (FAK). First, we generated site-specific CADTK mutants. Mutation of Tyr402 eliminated autophosphorylation and significantly decreased kinase activity. Mutation of Tyr881, a putative Src kinase phosphorylation site predicted to bind Grb2, had little effect on CADTK regulation. Src family tyrosine kinases resulted in CADTK tyrosine phosphorylation even when co-expressed with the Tyr402/Tyr881 double mutant, suggesting that Src/Fyn etc. phosphorylate additional tyrosine residues. Interestingly, CADTK tyrosine-phosphorylated FAK when both were transiently expressed, but FAK did not phosphorylate CADTK. Biochemical experiments confirmed direct CADTK phosphorylation of FAK. This phosphorylation utilized tyrosine residues other than Tyr397, Tyr925, or Tyr576/Tyr577, suggesting that new SH2-binding sites might be created by CADTK-dependent FAK phosphorylation. Last, expression of the CADTK carboxyl terminus (CRNK) abolished CADTK but not FAK autophosphorylation. In contrast, FAK carboxyl terminus overexpression inhibited both FAK and CADTK autophosphorylation, suggesting that a FAK-dependent cytoskeletal function may be necessary for CADTK activation. Thus, CADTK and FAK, which both bind to some, but not necessarily the same, cytoskeletal elements, may be involved in coordinate regulation of cytoskeletal structure and signaling.


Subject(s)
Calcium/pharmacology , Cell Adhesion Molecules/metabolism , Cytoskeleton/enzymology , Protein-Tyrosine Kinases/metabolism , Animals , Binding Sites/genetics , Cell Line , Enzyme Activation/drug effects , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Gene Expression Regulation, Enzymologic/genetics , Humans , Mutagenesis, Site-Directed , Mutation/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Rats , Vanadates/pharmacology , src Homology Domains/genetics , src-Family Kinases/metabolism
18.
Carcinogenesis ; 20(1): 27-33, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9934846

ABSTRACT

Increased cell proliferation most likely plays a key role in peroxisome proliferator-induced liver cancer. Recently, Kupffer cells were shown to be responsible for Wy-14,643-induced cell proliferation. However, the mechanism by which peroxisome proliferators activate Kupffer cells is unknown. Since gut-derived endotoxin is a known activator of Kupffer cells, the hypothesis that it is involved was evaluated. Increased cell proliferation and peroxisome induction were unaffected by gut sterilization. Moreover, endotoxin was not detectable in portal blood following treatment with Wy-14,643. Therefore, it is concluded that gut-derived endotoxin is not responsible for Kupffer cell activation. To test the hypothesis that Kupffer cells are activated by Wy-14,643 directly, Kupffer cell superoxide production was measured following treatment in vitro. Wy-14,643 increased superoxide production in a dose-dependent manner (0.1 and 50 microM) with half-maximal stimulation at 2.5 microM. Diethylhexylphthalate (DEHP) and ethylhexanol did not increase superoxide production even at doses 50 times higher than Wy-14,643; however, monoethylhexylphthalate (MEHP) activated superoxide production as effectively as Wy-14,643 with half-maximal stimulation at 5 microM. Treatment with Wy-14,643 for 21 days caused a 2-fold increase in Kupffer cell superoxide production while DEHP did not. Pretreatment of Kupffer cells with staurosporine (0.01-10 pM) completely blocked generation of superoxide demonstrating that protein kinase C is required. Moreover, Wy-14,643 increased Kupffer cell protein kinase C activity 3-fold. Pretreatment of Kupffer cells with the amino acid glycine (0.01-3 mM), which blunts calcium signaling, inhibited Wy-14,643-stimulated superoxide production and increased protein kinase C activity completely. These data are consistent with the hypothesis that potent peroxisome proliferators (Wy-14,643 and MEHP) directly activate Kupffer cell production of oxidants via mechanisms involving protein kinase C. Further, peroxisome proliferator treatments that sustain elevated rates of cell proliferation (e.g. Wy-14,643) activate Kupffer cell superoxide production following long-term dietary treatment supporting the hypothesis that Kupffer cell-derived oxidants are involved in peroxisome proliferator-induced neoplasia.


Subject(s)
Diethylhexyl Phthalate/analogs & derivatives , Kupffer Cells/metabolism , Oxidants/biosynthesis , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , Superoxides/metabolism , Animals , Calcium Signaling/drug effects , Cell Division/drug effects , Cell Transformation, Neoplastic/chemically induced , Diet , Diethylhexyl Phthalate/pharmacology , Diethylhexyl Phthalate/toxicity , Dose-Response Relationship, Drug , Endotoxins/blood , Endotoxins/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glycine/pharmacology , Intestines/microbiology , Kupffer Cells/drug effects , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/virology , Male , Oxidative Stress , Peroxisome Proliferators/administration & dosage , Peroxisome Proliferators/toxicity , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Pyrimidines/administration & dosage , Pyrimidines/toxicity , Rats , Rats, Sprague-Dawley , Staurosporine/pharmacology
19.
Am J Physiol ; 275(3): L551-8, 1998 09.
Article in English | MEDLINE | ID: mdl-9728050

ABSTRACT

We have previously shown that in vitro exposure to metallic compounds enhances expression of interleukin (IL)-6, IL-8, and tumor necrosis factor-alpha in human bronchial epithelial cells. To characterize signaling pathways involved in metal-induced expression of inflammatory mediators and to identify metals that activate them, we studied the effects of As, Cr, Cu, Fe, Ni, V, and Zn on the mitogen-activated protein kinases (MAPK) extracellular receptor kinase (ERK), c-Jun NH2-terminal kinase (JNK), and P38 in BEAS cells. Noncytotoxic concentrations of As, V, and Zn induced a rapid phosphorylation of MAPK in BEAS cells. Activity assays confirmed marked activation of ERK, JNK, and P38 in BEAS cells exposed to As, V, and Zn. Cr and Cu exposure resulted in a relatively small activation of MAPK, whereas Fe and Ni did not activate MAPK under these conditions. Similarly, the transcription factors c-Jun and ATF-2, substrates of JNK and P38, respectively, were markedly phosphorylated in BEAS cells treated with As, Cr, Cu, V, and Zn. The same acute exposure to As, V, or Zn that activated MAPK was sufficient to induce a subsequent increase in IL-8 protein expression in BEAS cells. These data suggest that MAPK may mediate metal-induced expression of inflammatory proteins in human bronchial epithelial cells.


Subject(s)
Bronchi/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Epithelial Cells/physiology , Metals/pharmacology , Mitogen-Activated Protein Kinases , Activating Transcription Factor 2 , Arsenic/pharmacology , Bronchi/drug effects , Bronchi/enzymology , Chromium/pharmacology , Clone Cells , Copper/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Humans , Iron/pharmacology , JNK Mitogen-Activated Protein Kinases , Nickel/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factors/metabolism , Vanadium/pharmacology , Zinc/pharmacology , p38 Mitogen-Activated Protein Kinases
20.
EMBO J ; 17(9): 2574-83, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9564040

ABSTRACT

In GN4 rat liver epithelial cells, angiotensin II (Ang II) produces intracellular calcium and protein kinase C (PKC) signals and stimulates ERK and JNK activity. JNK activation appears to be mediated by a calcium-dependent tyrosine kinase (CADTK). To define the ERK pathway, we established GN4 cells expressing an inhibitory Ras(N17). Induction of Ras(N17) blocked EGF- but not Ang II- or phorbol ester (TPA)-dependent ERK activation. In control cells, Ang II and TPA produced minimal increases in Ras-GTP level and Raf kinase activity. PKC depletion by chronic TPA exposure abolished TPA-dependent ERK activation but failed to diminish the effect of Ang II. In PKC-depleted cells, Ang II increased Ras-GTP level and activated Raf and ERK in a Ras-dependent manner. In PKC depleted cells, Ang II stimulated Shc and Cbl tyrosine phosphorylation, suggesting that without PKC, Ang II activates another tyrosine kinase. PKC-depletion did not alter Ang II-dependent tyrosine phosphorylation or activity of p125(FAK), CADTK, Fyn or Src, but PKC depletion or incubation with GF109203X resulted in Ang II-dependent EGF receptor tyrosine phosphorylation. In PKC-depleted cells, EGF receptor-specific tyrosine kinase inhibitors blocked Ang II-dependent EGF receptor and Cbl tyrosine phosphorylation, and ERK activation. In summary, Ang II can activate ERK via two pathways; the latent EGF receptor, Ras-dependent pathway is equipotent to the Ras-independent pathway, but is masked by PKC action. The prominence of this G-protein coupled receptor to EGF receptor pathway may vary between cell types depending upon modifiers such as PKC.


Subject(s)
Angiotensin II/pharmacology , Epidermal Growth Factor/pharmacology , Epithelial Cells/metabolism , ErbB Receptors/metabolism , ErbB Receptors/physiology , GTP-Binding Proteins/metabolism , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Cell Line , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , ErbB Receptors/antagonists & inhibitors , Humans , Liver/drug effects , Liver/metabolism , Quinazolines/pharmacology , Rats , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...