Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Int J Immunopathol Pharmacol ; 19(3): 517-24, 2006.
Article in English | MEDLINE | ID: mdl-17026836

ABSTRACT

Peroxiredoxins (Prx) comprise an extended family of small antioxidant proteins which conserve a thioredoxin-dependent catalytic function that can contribute to cell protection from reactive oxygen species (ROS). ROS generation is one of the deleterious intracellular effects of ionizing radiation, but the role of Prx during radiation treatment has not been extensively explored. Present experiments measure effects of ionizing radiation on expression of human Prx types I (PAGA), II (NKEF-B) and IV (AOE372) in human myeloid leukemia cells (K562). Prx gene transcription was analyzed by amplifying with RT-PCR cDNAs complementary to each Prx-specific coding sequence and by identifying the derived products with Southern blotting procedure. Transcripts of GAPDH were used as the endogenous standard for semi-quantitative comparisons. No consistent increase in Prx gene expression was detected at time intervals up to 72 h after gamma radiation doses that caused cell cycle arrest and nuclear damage (maximum 20 Gy). Immunoblots also were consistent with a prolonged expression or stability of the Prx I/II proteins. Similarly, a cytotoxic concentration of the oxidant hemin, which stimulates rapid hemoglobinization of K562 cells, caused no induction of Prx gene expression. Our results indicate a high Prx stability in human radio-resistant leukemia cells.


Subject(s)
Gene Expression Regulation, Leukemic/radiation effects , Leukemia, Myeloid/genetics , Peroxidases/genetics , Humans , K562 Cells , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Oxidative Stress , Peroxiredoxins , Radiation Tolerance
2.
Int J Immunopathol Pharmacol ; 17(1): 5-14, 2004.
Article in English | MEDLINE | ID: mdl-15000861

ABSTRACT

Previous results demonstrated that the occurrence of death in human peripheral B lymphocytes by TNF-alpha was paralleled by the activation of the cytoplasmic Jak1 and Tyk2 protein kinases, along with the recruitment of transcription factors Stat3 and Stat5b. In this study we demonstrate that the balance of survival signals in the presence of TNF-alpha was altered by the addition of a salicylate compound, the endonuclease inhibitor aurintricarboxylic acid (ATA). Apoptosis effected by TNF-alpha alone was suppressed by ATA and this event was paralleled by phosphorylation and nuclear translocation of Jak2, Stat2, Stat4 and NF-kB, along with inhibition of caspase activation. These results confirm that among the different cellular responses evoked by TNF-alpha in human B cells, recruitment of Jak/Stat proteins and possible related gene modulation represent contributing factors and address the issue of the development of potential therapeutic strategies aimed at the control of systemic or local effects produced by TNF-alpha.


Subject(s)
Aurintricarboxylic Acid/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , DNA-Binding Proteins/metabolism , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/toxicity , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Janus Kinase 1 , NF-kappa B/physiology , STAT3 Transcription Factor , Trans-Activators/genetics
3.
Clin Lab Med ; 21(3): 495-512, viii, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11572139

ABSTRACT

This article provides an overview of arbovirus diseases from the perspective of laboratory diagnosis and related responsibilities of health personnel. Although the disease manifestations are very diverse, general lessons relevant to optimal use of laboratory resources can be drawn from medically important examples. This approach is warranted, because under conditions of bioterrorism or emergence of a novel pathogen, ecologic clues that ordinarily facilitate identification of an arbovirus infection most likely will be absent.


Subject(s)
Arthropod Vectors , Virus Diseases/diagnosis , Virus Diseases/transmission , Animals , Biological Warfare , Humans
4.
Lab Invest ; 80(7): 993-1006, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10908145

ABSTRACT

The transcription factor Stat5a critically mediates prolactin (PRL)-induced mammary gland development and lactogenesis. PRL also stimulates growth and differentiation of prostate tissue. Specifically, hyperprolactinemia gives rise to prostate hyperplasia, and prostate size is reduced in PRL-deficient mice. We therefore investigated the importance of Stat5a for prostate development and function by examining Stat5a-null mice. The absence of Stat5a in mice was associated with a distinct prostate morphology characterized by an increased prevalence of local disorganization within acinar epithelium of ventral prostates. Affected acini were typically filled with desquamated, granular epithelial cells that had become embedded in dense, coagulated secretory material. These features were reminiscent of acinar cyst formation and degeneration frequently observed in human benign prostate hyperplasia, however, cystic changes in prostate acini of Stat5a-deficient mice were not associated with increased prostate size or morphologic hallmarks of epithelial hyperplasia. Instead, immunohistochemistry of the prostate-specific secretory marker, probasin, suggested that hypersecretory function of the epithelium could underlie local congestion and cyst formation in prostates of Stat5a-null mice. Serum testosterone and PRL levels were normal in Stat5a knockout mice, but prostate PRL receptor expression was reduced as determined by immunohistochemistry. Expression levels or activation states of other PRL signal transduction proteins, including Stat5b, Stat3, Stat1, ERK1, and ERK2 were not altered. The present study offers the first evidence for a direct role of Stat5a in the maintenance of normal tissue architecture and function of the mouse prostate.


Subject(s)
DNA-Binding Proteins/deficiency , Milk Proteins , Prostate/pathology , Prostatic Diseases/etiology , Prostatic Diseases/pathology , Trans-Activators/deficiency , Animals , Apoptosis , Cell Division , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/pathology , Epithelial Cells/physiology , Epithelium/metabolism , Epithelium/pathology , Male , Mice , Mice, Knockout/genetics , Prolactin/blood , Prolactin/physiology , Prostate/metabolism , Prostate/physiopathology , Prostatic Diseases/genetics , Prostatic Diseases/metabolism , Receptors, Prolactin/metabolism , Reference Values , STAT5 Transcription Factor , Signal Transduction , Testosterone/blood , Trans-Activators/genetics , Trans-Activators/metabolism
5.
Cytokine Growth Factor Rev ; 10(2): 131-57, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10743504

ABSTRACT

Stat5a and Stat5b are discretely encoded transcription factors that mediate signals for a broad spectrum of cytokines. Their activation is often an integral component of redundant cytokine signal cascades involving complex cross-talk and pleiotropic gene regulation by Stat5 has been implicated in cellular functions of proliferation, differentiation and apoptosis with relevance to processes of hematopoiesis and immunoregulation, reproduction, and lipid metabolism. Although Stat5a and Stat5b show peptide sequence similarities of > 90%, targeted gene disruptions in mice yield distinctive phenotypes. Prolactin-directed mammary gland maturation fails without functional Stat5a, while disruption of Stat5b in males mitigates growth hormone effects on hepatic function and body mass. The molecular basis for this biologic dichotomy is probably multifaceted. Limited structural dissimilarities between the Stat5a and Stat5b transactivation domains, or subtle differences in the DNA-binding affinities of Stat5 dimer pairs undoubtedly influence gene regulation, but cell-dependent asymmetries in availability of phosphorylated Stat5 can be an underlying factor. Differences in serine phosphorylation(s) of Stat5a and Stat5b, or Stat5 associations with adaptor proteins or co-transcription factors are other potential sources of functional disparity and the signal amplitude, frequency or duration also can be significant. In addition to Stat5 signal attenuation by phosphatase actions or classical feedback inhibition, truncated forms of Stat5 lacking in transactivation capacity may compete upstream for activation and diminish access of full length molecules to DNA binding sites.


Subject(s)
DNA-Binding Proteins/metabolism , Milk Proteins , Receptors, Cytokine/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcriptional Activation , Amino Acid Sequence , Animals , Cytokines/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Cytokine/genetics , STAT5 Transcription Factor , Serine/metabolism , Trans-Activators/genetics , Tumor Suppressor Proteins
6.
J Immunol ; 161(12): 6503-9, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9862674

ABSTRACT

In a manner similar to many other cytokines, treatment of cells with granulocyte CSF (G-CSF) has been shown to induce the tyrosine phosphorylation of the STAT proteins. Activation of Stat1 and Stat5 by G-CSF requires the membrane-proximal cytoplasmic domain of the receptor, including box1 and box2, while G-CSF-stimulated tyrosine phosphorylation of Stat3 also requires a region distal to box 2. In this study, we show that although the membrane-proximal 55 amino acids of the G-CSF receptor are sufficient for activation of Stat5, the maximal rate of Stat5 activation requires an additional 30 amino acids of the cytoplasmic domain. In contrast, the distal carboxyl-terminal region of the receptor appears to down-regulate Stat5 activation in that deletion of this carboxyl terminus results in increased amplitude and prolonged duration of Stat5 activation by G-CSF. Significantly, expression of a truncated dominant-negative Stat5 protein in hemopoietic cells not only inhibits G-CSF-dependent cell proliferation, but also suppresses cell survival upon G-CSF withdrawal. We further show that a potential protein tyrosine phosphatase may play a critical role in the down-regulation of G-CSF-stimulated Stat5 activation. These results demonstrate that two distinct cytoplasmic regions of the G-CSF receptor are involved in the regulation of the intensity and duration of Stat5 activation, and that Stat5 may be an important player in G-CSF-mediated cell proliferation and survival.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/physiology , Milk Proteins , Protein Structure, Tertiary , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Trans-Activators/physiology , Animals , COS Cells , Cell Division , Cell Survival , DNA, Complementary/genetics , Hematopoietic Stem Cells/metabolism , Mice , Receptors, Granulocyte Colony-Stimulating Factor/chemistry , Receptors, Granulocyte Colony-Stimulating Factor/genetics , STAT5 Transcription Factor , Signal Transduction , Structure-Activity Relationship , Transcription, Genetic , Transfection
7.
Blood ; 91(8): 3017-27, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9531615

ABSTRACT

Multiple biologic effects of interferon-alpha (IFN-alpha), including cell growth inhibition and antiviral protection, are initiated by tyrosine phosphorylation of STAT proteins. Although this signal pathway has been intensively investigated, the relevance of STAT signal persistence has received scant attention. Using paired isogenic lymphoma cells (Daudi), which either are sensitive or resistant to growth inhibition by IFN-alpha, we found comparable initial tyrosine phosphorylation of multiple STAT proteins; however, the phosphorylation durations and associated DNA-binding activities diverged. Phosphorylation and DNA-binding capacity of STAT1 decreased after 4 to 8 hours in resistant cells, as compared with 24 to 32 hours in sensitive cells, whereas phosphorylation of STAT3 and STAT5b was briefer in both lines. Functional significance of the prolonged STAT1 signal, therefore, was explored by experimental interruption of tyrosine phosphorylation, either by premature withdrawal of the IFN-alpha or deferred addition of pharmacologically diverse antagonists: staurosporine (protein kinase inhibitor), phorbol 12-myristate 13-acetate (growth promoter), or aurintricarboxylic acid (ligand competitor). Results indicated that an approximately 18-hour period of continued STAT1 phosphorylation was associated with growth arrest, but that antiviral protection developed earlier. These differences provide novel evidence of a temporal dimension to IFN-alpha signal specificity and show that duration of STAT1 activation may be a critical variable in malignant cell responsiveness to antiproliferative therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Interferon-gamma/pharmacology , Lymphoma/genetics , Lymphoma/pathology , Milk Proteins , Signal Transduction/genetics , Trans-Activators/genetics , Cell Division/drug effects , Cell Division/genetics , Humans , Recombinant Proteins , STAT1 Transcription Factor , STAT5 Transcription Factor , Signal Transduction/drug effects , Tumor Cells, Cultured
8.
Cancer Res ; 58(9): 1914-9, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9581833

ABSTRACT

Type I interferons (IFN alpha and IFN beta) are presently used in the adjuvant treatment of several human cancers. However, these cytokines have demonstrated only modest success in breast cancer therapy, and research efforts have focused on improving their efficacy. Recent progress in understanding the molecular mechanisms underlying the antiproliferative effects of IFNs has identified the cytoplasmic transcription factor Stat1 as a critical mediator. It is, therefore, possible that IFN-induced growth inhibition of mammary epithelial cells is counteracted by other cytokines that also use Stat1. One such candidate IFN-antagonist with particular relevance to breast cancer is the mammotropic hormone prolactin (PRL). The main goal of this study was to examine whether PRL would interfere with type I IFN (IFN alpha/beta) signal transduction by competing for limited cytoplasmic Stat factors. A second aim was to test whether pretreatment of mammary tumor cell lines with IFN gamma could enhance the effect of IFN alpha/beta. By analyzing the effect of PRL on IFN alpha/beta-induced tyrosine phosphorylation of Stat proteins and their binding to IFN-regulated genes, we now report that costimulation of PRL receptors did not interfere with IFN alpha/beta signals in several human breast cancer cell lines, including T47D, MCF-7, and BT-20. Specifically, PRL did not affect IFN alpha/beta-induced tyrosine phosphorylation or heterodimerization of Stat1 and Stat2 in any cell line. Instead, IFN alpha/beta- and PRL-induced tyrosine phosphorylation of Stat1 was additive and occurred without evidence of competition for limited concentrations of cytoplasmic Stat1. A similar additive relationship was observed on IFN alpha/beta- and PRL-induced Stat3 tyrosine phosphorylation. Furthermore, electrophoretic mobility shift assays showed that type I IFNs induced predominantly Stat1-Stat2 or Stat1-Stat3 heteromeric complexes with various IFN-response elements of IFN-stimulated genes, whereas PRL induced Stat1 homodimers. Despite significant mutual use of Stats by IFNs and PRL, these results indicated a high degree of signaling specificity in the two receptor systems, and that cytoplasmic levels of Stat proteins were not limiting. Similarly, PRL did not interfere with the growth-inhibitory effect of IFN beta. On the other hand, the study indicated that pretreatment of human breast cancer cell lines with IFN gamma enhanced the growth-inhibitory action of type I IFNs, suggesting a possible avenue for improving the effect of type I IFNs in the treatment of breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Interferon Type I/pharmacology , Prolactin/pharmacology , Signal Transduction , Trans-Activators/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Drug Therapy, Combination , Female , Humans , Immunoblotting , Phosphorylation , Rabbits , STAT1 Transcription Factor , STAT2 Transcription Factor , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology , Tyrosine/metabolism
9.
J Biol Chem ; 273(1): 28-32, 1998 Jan 02.
Article in English | MEDLINE | ID: mdl-9417042

ABSTRACT

Biological effects of many hormones and cytokines are mediated through receptor-associated Jak tyrosine kinases and cytoplasmic Stat transcription factors, including critical physiological processes such as immunity, reproduction, and cell growth and differentiation. Pharmaceuticals that control Jak-Stat pathways are therefore of considerable interest. Here we demonstrate that a single Jak-Stat pathway can be activated by aurintricarboxylic acid (ATA), a negatively charged triphenylmethane derivative (475 Da) with anti-apoptotic properties. In prolactin (PRL)-dependent Nb2 lymphocytes, ATA sustained cell growth in the absence of hormone and mimicked rapid PRL-induced tyrosine phosphorylation of Jak2 and activation of Stat5a and Stat5b with tyrosine phosphorylation, heterodimerization, DNA binding, and induction of the Stat5-regulated pim-1 protooncogene. ATA also mimicked PRL activation of serine kinases ERK1 and ERK2. However, unlike PRL, ATA did not regulate Stat1 or Stat3. ATA also did not affect Jak3, which is activated in these cells by interleukin-2 family cytokines. Although the mechanism and specificity by which ATA activates Jak2, Stat5, and ERKs in Nb2 cells are still unclear, the present study demonstrates that certain hormone or cytokine effects on Jak-Stat pathways can be discretely imitated by a low molecular weight, non-peptide pharmaceutical. The results are also consistent with Stat5 involvement in lymphocyte growth and survival.


Subject(s)
Apoptosis/drug effects , Aurintricarboxylic Acid/pharmacology , DNA-Binding Proteins/metabolism , Lymphoma/metabolism , Milk Proteins , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Trans-Activators/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA/metabolism , Enzyme Activation , Gene Expression Regulation , Janus Kinase 2 , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphoma/pathology , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-pim-1 , Receptors, Prolactin/metabolism , STAT5 Transcription Factor , Tumor Cells, Cultured , Tyrosine/metabolism
10.
Nat Med ; 3(8): 908-12, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9256284

ABSTRACT

We found that simian virus 40 (SV40) induces mesotheliomas in hamsters and that 60% of human mesotheliomas contain and express SV40 sequences, results now confirmed by others [ref. 3-5, and presentations by D. Griffiths & R. Weiss, F. Galateau-SallE, and H.I.P. at "Simian virus 40: A possible human polyoma virus," NIH workshop, 27-28 January 1997, Bethesda, MD (transcript available through SAG Corp., Washington, DC 20008)]. Mesothelioma, an aggressive malignancy resistant to therapy, originates from the serosal lining of the pleural, pericardial and peritoneal cavities. The incidence of mesothelioma continues to increase worldwide because of exposure to crocidolite asbestos. However, at least 20% of mesotheliomas in the United States are not associated with asbestos exposure, and only a minority of people exposed to high concentrations of asbestos develop mesothelioma. Thus, other carcinogens may induce mesothelioma in individuals not exposed to asbestos, and/or may render particular individuals more susceptible to the carcinogenic effect of asbestos. We investigated whether the expression of the SV40 large T-antigen (Tag) interferes with the normal expression of the tumor suppressor gene p53 in human mesotheliomas. We found that SV40 Tag retains its ability to bind and to inactivate p53, a cellular protein that when normally expressed plays an important role in suppressing tumor growth and in inducing sensitivity to therapy. Our findings do not establish a cause-and-effect relation, but indicate that the possibility that SV40 contributes to the development of human mesotheliomas should be carefully investigated.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , Simian virus 40/immunology , Tumor Suppressor Protein p53/metabolism , Gene Expression Regulation, Neoplastic , Genes, p53 , Humans , Immunohistochemistry , Mesothelioma/genetics , Mesothelioma/pathology , Mutation , Pleural Neoplasms/pathology , Protein Binding , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
11.
J Biol Chem ; 272(22): 14098-103, 1997 May 30.
Article in English | MEDLINE | ID: mdl-9162035

ABSTRACT

Transcription factors of the Stat gene family are selectively activated by many hormones and cytokines. Stat5 originally was cloned as a prolactin-stimulated DNA-binding protein, but is also activated by non-lactogenic cytokines in many cell types. The recent identification of two distinct Stat5 genes, which encode a 94-kDa Stat5a and a 92-kDa Stat5b as well as several lower molecular weight isoforms, suggests additional complexity and combinatorial possibilities for transcriptional regulation. We now report a biochemical analysis of prolactin activation of Stat proteins in Nb2 lymphocytes, which was associated with: 1) rapid tyrosine phosphorylation of Stat5a, Stat5b, a COOH-terminally truncated 80-kDa Stat5 form, Stat1alpha, and Stat3; 2) rapid and selective formation of Stat5a/b heterodimers, without involvement of Stat1alpha or Stat3; 3) marked serine, but not threonine phosphorylation of Stat5a and Stat5b; and 4) the appearance of two qualitatively distinct Stat5 protein complexes, which discriminated between oligonucleotides corresponding to the prolactin response elements of the beta-casein and interferon regulatory factor-1 gene promoters. Collectively, our analyses showed that Stat5a and Stat5b respond similarly to prolactin receptor activation, but also suggested that the two genes have evolved unique properties that may contribute to the specificity of receptors that utilize Stat5 signaling proteins.


Subject(s)
DNA-Binding Proteins/metabolism , Lymphocytes/metabolism , Milk Proteins , Prolactin/pharmacology , Signal Transduction/drug effects , Trans-Activators/metabolism , Cell Line , Humans , Phosphorylation , Protein Binding/drug effects , STAT5 Transcription Factor , Serine/metabolism , Tumor Suppressor Proteins , Tyrosine/metabolism
12.
Mol Cell Biol ; 13(12): 7515-21, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8246969

ABSTRACT

Alpha and gamma interferons rapidly induce several early response genes in primary human diploid fibroblasts. The transcription rates of these genes are maximal after 1 h of interferon treatment and return to basal levels within 8 h. Three different interferon-activated DNA-binding complexes (ISGF3, GAF, and FcRF gamma) that are responsible for transcriptional activation of cellular genes have been characterized. Assembly of these complexes requires tyrosine phosphorylation of one or more of the protein components. In this report, we demonstrate that a nuclear tyrosine phosphatase is responsible for the deactivation of these interferon-regulated transcription factors and the subsequent transcriptional downregulation of the corresponding genes. Furthermore, tyrosine phosphorylation is required for nuclear localization of the 91-kDa protein that is part of all three interferon-induced transcription complexes. These results provide the first evidence for a nuclear tyrosine phosphatase activity as a mechanism of transcriptional regulation.


Subject(s)
Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Protein Tyrosine Phosphatases/metabolism , Base Sequence , Cell Nucleus/metabolism , Cells, Cultured , DNA/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Humans , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Molecular Sequence Data , Recombinant Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Vanadates/pharmacology
13.
Cancer Lett ; 67(1): 13-9, 1992 Oct 30.
Article in English | MEDLINE | ID: mdl-1423240

ABSTRACT

Several clinical protocols are attempting to utilize the combined anti-proliferative signal of interferon-alpha (INF-alpha) and tamoxifen on cancer cells. We demonstrated here that the effect of these two agents on the growth of the premacrophage U937 cells is antagonistic. This antagonistic effect is paralleled by the ability of tamoxifen to modulate the INF-alpha-induced hyperpolarization in these cells. INF-alpha-induced hyperpolarization was shown before to be an integral part of the anti-proliferative signal of this agent. Tamoxifen liberates Ca2+ from intracellular stores of U937 cells but this effect of the drug is not the cause of its antagonistic effect with the anti-proliferative signal of IFN-alpha. We suggest therefore, that the combined effect of these two anti-cancer drugs could also be advantageous for macrophage proliferation.


Subject(s)
Interferon-alpha/pharmacology , Macrophages/drug effects , Tamoxifen/pharmacology , Calcium/metabolism , Cell Division/drug effects , Cell Line , Chlorides/metabolism , Humans , Membrane Potentials/drug effects
14.
Virology ; 190(1): 210-20, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1529530

ABSTRACT

Binding of interferons (IFNs) to their cell surface receptors stimulates rapid translocation of cytoplasmic proteins to the nucleus and the expression of a variety of cellular genes within minutes. Translocated proteins subsequently bind to the interferon-stimulated response element (ISRE) located in the promoters of all IFN-activated cellular genes. We report here that ouabain, a specific inhibitor of the Na/K ATPase, selectively inhibited transcription of several IFN-alpha-induced cellular RNAs under conditions in which some other well-described signal transduction pathways remained intact. The latter included induction of human metallothionein 2A (HMT2A) by phorbol ester and induction of IP-10 RNA by IFN-gamma. Ouabain itself induced RNA of the protooncogene c-fos which conversely was inhibited by IFN-alpha. Specificity of the ouabain effects on IFN alpha-induced RNAs with respect to a direct action on the Na/K ATPase was shown with a transfected monkey CV-1 cell line which expresses the ouabain-insensitive rat alpha 1 subunit. Electrophoretic mobility shift assays (EMSAs) using nuclear extracts from ouabain-treated cells demonstrated that ouabain decreased IFN alpha-induced binding of the ISGF3 complex to the ISRE. Reconstitution experiments showed that this effect of ouabain is not due to the inhibition of IFN alpha activation of the ISGF3 alpha subcomponent, which occurs in the cytoplasm, but a selective depletion of the ISGF3 gamma factor which in concert with activated ISGF3 alpha induces interferon-stimulated gene (54 kDa) transcription. These findings imply that intracellular ion balance can selectively regulate the half-life of the ISGF3 gamma protein or the ability of this protein to complex with ISGF3 alpha to activate IFN alpha-regulated cellular genes.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Interferon-alpha/physiology , Ouabain/pharmacology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Base Sequence , Cells, Cultured , HeLa Cells , Humans , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, alpha Subunit , Interferon-Stimulated Gene Factor 3, gamma Subunit , Melanoma , Molecular Sequence Data , Oligodeoxyribonucleotides , Proto-Oncogene Proteins c-fos/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured
15.
J Interferon Res ; 12(4): 249-55, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1331257

ABSTRACT

Cytochalasin B, a drug that alters microfilament structure, was found to modulate interferon-alpha (IFN-alpha)-induced changes in ion fluxes, in motional freedom of spin probes, and lateral diffusion of surface antigens. These changes occur in Daudi cells inherently sensitive to the antiproliferative signal of IFN-alpha, but not in insensitive cells, and were associated with the antiproliferative signal previously. The biophysical effects of cytochalasin B were detected by flow cytometric quantitation of membrane potential using an oxonol dye, by electron spin resonance (ESR) spectrometry, and by measurements of fluorescence recovery after photobleaching (FRAP) of surface antigens using a laser-interactive cell imaging system. Cytochalasin B treatment increased an IFN-alpha-induced membrane potential shift by -5 mV. The motional freedom of 5-doxyl-stearic acid changed from 0.67 to 0.63, as expressed by the order parameter, S, with IFN-alpha treatment and was prevented by cytochalasin B. Changes in the lateral diffusion of surface antigens induced by IFN-alpha treatment, D = 5.3 x 10(-10) without treatment and D = 7.8 x 10(-10) cm2/s with treatment, was blocked by cytochalasin B. In contrast, the microtubule stabilizers taxol and D2O and the microtubule depolymerizing colcemid were ineffective at dose levels sufficient to cause the characteristic cell physiological alterations of these agents. These results implicate microfilaments but not the microtubule system in transduction of the antiproliferative signal by IFN-alpha in Daudi cells.


Subject(s)
Actin Cytoskeleton/drug effects , Cytochalasin B/pharmacology , Cytoskeleton/physiology , Interferon-alpha/pharmacology , Signal Transduction/drug effects , Cell Membrane/physiology , Demecolcine/pharmacology , Electron Spin Resonance Spectroscopy , Flow Cytometry , Humans , Membrane Potentials/drug effects , Microscopy, Fluorescence , Microtubules/drug effects , Spin Labels , Tumor Cells, Cultured
16.
Carcinogenesis ; 12(11): 1993-9, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1934283

ABSTRACT

The effects of transforming growth factor beta (TGF-beta) on cell-to-cell communication were investigated in the log phase of growth in normal BE and in adenovirus-12 SV40 hybrid virus transformed BE cells (strain BEAS-2B). Gap junctions in these cells were identified immunocytochemically. Exposure of BE cells to exogenous TGF-beta (0.04-4.0 pM) in serum-free keratinocyte growth medium (KGM) for 1 or 24 h reduced the rate of fluorescent dye transfer (i.e. cell-to-cell communication) by 30-50% in BE cells. Inversely, in BEAS-2B cells, TGF-beta after 1 h induced a 2- to 10-fold increase in the rate of dye transfer. After 24 h of TGF-beta, communication among BEAS-2B cells was not significantly different from controls (no exogenous TGF-beta). The protein kinase C (PKC) inhibitor H-7 induced a dose-dependent enhancement in communication, which was even higher in the presence of TGF-beta (4 pM X 24 h). The calmodulin antagonist W-7 enhanced communication in BEAS-2B cells independently of the presence of TGF-beta. In keratinocyte basal medium (KBM) supplemented with EGF (5 ng/ml) or with TGF-beta (4.0 pM) dye transfer was reduced or enhanced respectively. The combination of EGF and TGF-beta in KBM antagonized the stimulatory effect of the latter on communication in BEAS-2B cells. In BE cells, continuous exposure (4 days) to TGF-beta in KGM induced a dose-dependent inhibition of proliferation and an increased expression of a keratinized, epidermoid phenotype. This correlated with a reduction in the expression of a mucous secretory phenotype. Increased exposure to TGF-beta (0.04-4.0 pM) decreased the labeling index in BEAS-2B cells, but the cells retained a growth advantage over normal BE cells, and did not express a keratinized epidermoid morphology. With respect to dye transfer as an index of cell-to-cell communication, we conclude (i) that an inhibition or enhancement of communication is involved in the response of bronchial epithelial cells to mitogens (e.g. epidermal growth factor) or growth inhibitors (e.g. TGF-beta), (ii) that PKC and Ca(2+)-calmodulin-dependent processes regulate dye transfer, and (iii) the effects of TGF-beta are mediated by PKC.


Subject(s)
Bronchi/physiology , Bronchial Neoplasms/physiopathology , Cell Communication/drug effects , Transforming Growth Factor beta/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Calmodulin/antagonists & inhibitors , Cells, Cultured , Culture Media/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Epidermal Growth Factor/pharmacology , Epithelium , Humans , Isoquinolines/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors , Sulfonamides/pharmacology , Time Factors
17.
J Med ; 22(4-5): 255-71, 1991.
Article in English | MEDLINE | ID: mdl-1664849

ABSTRACT

In earlier studies, we showed that when interferon alpha (IFN alpha) binds to cell receptors, it alters membrane potentials. In IFN-sensitive Daudi cells derived from Burkitt's lymphoma patients, IFNs produce dose-dependent hyperpolarization, decreased plasma membrane viscosity, modulation of the microfilament system, and altered synthesis of cAMP, cGMP and prostaglandins. No such changes were seen in IFN alpha-resistant Daudi cells. In this study, we found that indomethacin, pentoxifylline, or the cGMP inducer sodium azide (NaN3) had no significant effect on the IFN alpha induced membrane potential changes, or on membrane viscosity changes as measured by flow cytometry and electron spin resonance spectrometry. In tissue culture, indomethacin did not alter the anti-proliferative effect of IFN alpha on Daudi cells. Indomethacin may influence IFN synthesis, but not it's antiproliferative actions. Relatively high doses of pentoxifylline slightly inhibited proliferation of Daudi cells and synergized with IFN alpha. Measurement of early biophysical changes induced by IFNs may represent a new screening method to rapidly explore certain types of IFN alpha potentiating agents.


Subject(s)
Azides/pharmacology , Indomethacin/pharmacology , Interferon-alpha/physiology , Pentoxifylline/pharmacology , Burkitt Lymphoma/pathology , Cyclic GMP/metabolism , Drug Interactions , Electron Spin Resonance Spectroscopy , Humans , In Vitro Techniques , Interferon-alpha/metabolism , Membrane Potentials/drug effects , Sodium Azide , Tumor Cells, Cultured/drug effects
18.
AJR Am J Roentgenol ; 155(6): 1247-50, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2173385

ABSTRACT

Paragangliomas of the retroperitoneum arise from specialized neural crest cells distributed along the aorta in association with the sympathetic chain. In order to ascertain characteristic CT features of extraadrenal retroperitoneal paragangliomas to differentiate them from other retroperitoneal tumors, 31 discrete tumors and two cases of paragangliomatosis in 28 patients were reviewed retrospectively, and the CT features were correlated with clinical and pathologic findings. There were 16 men and 12 women. Average age was 37 years (range, 11-70 years). Twenty-four patients (86%) had hypertension. Of these, catecholamine levels were elevated in all 18 patients who had biochemical studies. Four patients (14%) had malignant paragangliomas. The discrete tumors were classified by location as suprarenal (26%), renal hilar (32%), or infrarenal (42%). Suprarenal paragangliomas could not be distinguished from the ipsilateral adrenal gland on CT. The average size of functional tumors was smaller (7.0 cm) than that of nonfunctional tumors (12.0 cm), but the sizes of the two groups overlapped. Smaller tumors were more likely to be homogeneous and have well-defined margins than were larger tumors. Our findings indicate that extraadrenal retroperitoneal paragangliomas are functionally active more often than previously reported and that they are readily detected by CT as soft-tissue masses closely associated with the entire length of the abdominal aorta. However, no CT feature was found that was unique for paraganglioma.


Subject(s)
Neoplasms, Multiple Primary/epidemiology , Paraganglioma, Extra-Adrenal/epidemiology , Retroperitoneal Neoplasms/epidemiology , Tomography, X-Ray Computed , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged , Neoplasms, Multiple Primary/diagnostic imaging , Neoplasms, Multiple Primary/pathology , Paraganglioma, Extra-Adrenal/diagnostic imaging , Paraganglioma, Extra-Adrenal/pathology , Retroperitoneal Neoplasms/diagnostic imaging , Retroperitoneal Neoplasms/pathology , Retrospective Studies
19.
J Interferon Res ; 10(6): 605-11, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2128303

ABSTRACT

Increasing interest is being focused on the role of protein kinase C (PKC) in the mode of action of interferons (IFNs). Here we report that IFN-alpha induced a transient translocation of PKC from the cytosol to the particulate fraction of U937 cells. In contrast, after IFN-gamma treatment, no significant change in PKC activity could be observed. IFN-induced changes in membrane potential were also examined by means of a potential sensitive oxonal dye and flow cytometry. Hyperpolarization was induced by both IFN-alpha and IFN-gamma. The protein kinase inhibitor H-7 blocked the hyperpolarization induced by IFN-alpha but not by IFN-gamma. These concordant results suggest that PKC is involved in the early signals of IFN-alpha but not of IFN-gamma in U937 cells.


Subject(s)
Interferon Type I/physiology , Protein Kinase C/physiology , Signal Transduction/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Cell Line , Humans , Interferon-gamma/physiology , Isoquinolines/pharmacology , Membrane Potentials/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors , Signal Transduction/drug effects
20.
Exp Cell Res ; 187(2): 197-202, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2318263

ABSTRACT

The effect of human recombinant DNA interferon-alpha type A (rIFN alpha A) on nuclear lipid biosynthesis and on in vitro nucleic acid synthesis was investigated in Daudi lymphoma cells sensitive (Daudi Is) or resistant (Daudi Ir) to the antiproliferative activity of this glycoprotein. In the Daudi Is studied at 90 min and up to 8 h, relative proportions of 3H-labeled nuclear lipids were reproducibly altered as compared to controls: phosphatidylcholine increased while phosphatidylserine and total neutral lipids decreased. These changes were not detected in parallel studies of the whole cell extracts. No significant changes in the profiles of nuclear lipids were observed in Daudi Ir. Decreased rates of alpha DNA polymerase and of RNA transcription were evident within 90 min in the Daudi Is nuclei but not in untreated controls or nuclei from rIFN alpha A-treated Daudi Ir cells, thus suggesting a possible relationship of the rapid alterations of nuclear lipid biosynthesis in Daudi Is cells to the rIFN alpha A antiproliferative activity.


Subject(s)
Cell Division/drug effects , Cell Nucleus/metabolism , DNA Replication/drug effects , Interferon Type I/pharmacology , Lipids/biosynthesis , RNA, Neoplasm/biosynthesis , Transcription, Genetic/drug effects , Tumor Cells, Cultured/metabolism , Burkitt Lymphoma , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , DNA Repair , DNA, Neoplasm/biosynthesis , Glycerol/metabolism , Humans , Lipids/isolation & purification , Phospholipids/biosynthesis , Recombinant Proteins , Thymine Nucleotides/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Uridine Triphosphate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...