324 results on '"Rapp UR"'
Search Results
102. Generation dependent reduction of tTA expression in double transgenic NZL-2/tTA(CMV) mice.
- Author
-
Fedorov LM, Tyrsin OY, Sakk O, Ganscher A, and Rapp UR
- Subjects
- Aging genetics, Animals, Blotting, Southern, Crosses, Genetic, Embryo, Mammalian metabolism, Female, Gene Expression Profiling, Genes, Reporter genetics, Luciferases analysis, Luciferases metabolism, Male, Mice, Mice, Transgenic, RNA, Messenger genetics, RNA, Messenger metabolism, Time Factors, beta-Galactosidase analysis, beta-Galactosidase metabolism, Gene Expression Regulation, Luciferases genetics, Promoter Regions, Genetic genetics, Tetracycline metabolism, Transgenes genetics, beta-Galactosidase genetics
- Abstract
Despite the overall successful application of the tet-system to regulate gene expression in vitro and in vivo, nothing is known so far about the long-term stability of this system in transgenic mice. In this study, mice of generation F2, F3, F4, or F10 of two independent tTA(CMV) transgenic lines were bred with NZL-2 mice containing a tTA-responsive bidirectional promoter that allows the simultaneous expression of two reporter genes encoding luciferase and beta-galactosidase. Analysis of the expression of transgenes in double transgenic mice revealed a dramatic reduction of tTA transactivator mRNA over time. As a consequence, the expression of both reporter genes was gradually reduced from generation to generation in tissues of embryonic and adult NZL-2/tTA(CMV) mice. Luciferase activity in NZL-2/tTA(CMV)(F10) mice was reduced 8-10-fold compared to NZL-2/ tTA(CMV)(F2) mice, and beta-galactosidase expression was no longer detectable. In summary, we describe the long-term instability of the tet-system in our NZL-2/tTA(CMV) double transgenic mice. The molecular basis of this observation and experimental tools to overcome this limitation need to be addressed in future., (Copyright 2001 Wiley-Liss, Inc.)
- Published
- 2001
- Full Text
- View/download PDF
103. Selective expression of calcium-binding proteins S100a8 and S100a9 at distinct sites of hair follicles.
- Author
-
Schmidt M, Gillitzer R, Toksoy A, Bröcker EB, Rapp UR, Paus R, Roth J, Ludwig S, and Goebeler M
- Subjects
- Calgranulin A, Calgranulin B, Humans, Immunohistochemistry, In Situ Hybridization, Antigens, Differentiation biosynthesis, Calcium-Binding Proteins biosynthesis, Hair Follicle metabolism, S100 Proteins biosynthesis
- Published
- 2001
- Full Text
- View/download PDF
104. Extracellular signal regulated kinase 5 (ERK5) is required for the differentiation of muscle cells.
- Author
-
Dinev D, Jordan BW, Neufeld B, Lee JD, Lindemann D, Rapp UR, and Ludwig S
- Subjects
- Animals, Blotting, Western, Cell Differentiation, Cell Line, Enzyme Activation, Genes, Dominant, Genes, Reporter, Humans, MAP Kinase Kinase 5, MAP Kinase Signaling System, Mice, Mitogen-Activated Protein Kinase 7, Mitogen-Activated Protein Kinase Kinases metabolism, Muscle, Skeletal cytology, Oligonucleotides, Antisense metabolism, Promoter Regions, Genetic, RNA, Messenger metabolism, Signal Transduction, Time Factors, Transduction, Genetic, Transfection, Mitogen-Activated Protein Kinases metabolism, Muscles cytology
- Abstract
Extracellular signal regulated kinase 5 (ERK5) is a novel member of the mitogen-activated protein kinase (MAPK) family with a poorly defined physiological function. Since ERK5 and its upstream activator MEK5 are abundant in skeletal muscle we examined a function of the cascade during muscle differentiation. We show that ERK5 is activated upon induction of differentiation in mouse myoblasts and that selective activation of the pathway results in promoter activation of differentiation-specific genes. Moreover, myogenic differentiation is completely blocked when ERK5 expression is inhibited by antisense RNA. Thus, we conclude that the MEK5/ERK5 MAP kinase cascade is critical for early steps of muscle cell differentiation.
- Published
- 2001
- Full Text
- View/download PDF
105. Independent control of cell survival by Raf-1 and Bcl-2 at the mitochondria.
- Author
-
Zhong J, Troppmair J, and Rapp UR
- Subjects
- Animals, Caspases physiology, Cell Survival, Cells, Cultured, Doxorubicin pharmacology, Apoptosis, Mitochondria physiology, Proto-Oncogene Proteins c-bcl-2 physiology, Proto-Oncogene Proteins c-raf physiology
- Abstract
Bcl-2 family proteins play a critical role in the regulation of cell survival by controlling the activation of the cell death executing caspase machinery. Recent work demonstrated that they also provide a link between growth factor signaling and cell survival control. Raf-1 has been identified initially as an essential component of the mitogenic Ras-Raf-MEK-ERK cascade. However, expression of oncogenic Raf-1 also efficiently suppresses apoptotic cell death. This process requires mitochondrial translocation of Raf-1 which can be achieved either by co-expression of the anti-apoptotic protein Bcl-2 or by fusion with the transmembrane domain of the yeast outer mitochondrial membrane protein Mas 70p. It is currently unclear how mitochondrial Raf-1 prevents apoptosis. One possible mechanism involves the phosphorylation of the pro-apoptotic protein Bad resulting in the restoration of Bcl-2 function. Alternatively, the role of Bcl-2 could be limited to the mitochondrial translocation of Raf-1 and survival signaling by Raf-1 is Bcl-2 independent. To test for the mutual requirement of Raf-1 and Bcl-2 in apoptosis suppression the individual proteins were singly tested for survival activity in a genetic background which precludes the expression of the other. The results obtained in these studies demonstrate that ablation of Raf-1 or Bcl-2 expression in fibroblast cells significantly increases the sensitivity towards doxorubicin induced cell death. Reversion of the mutant phenotype could be achieved in either case by introducing a functional bcl-2 gene or a mitochondria targeted version of oncogenic Raf-1, demonstrating that each protein by itself is sufficient to confer protection. Our data thus suggest the existence of two separate pathways of survival signaling at the mitochondria controlled either by Bcl-2 or by Raf-1.
- Published
- 2001
- Full Text
- View/download PDF
106. Butyrate and propionate downregulate ERK phosphorylation in HT-29 colon carcinoma cells prior to differentiation.
- Author
-
Davido DJ, Richter F, Boxberger F, Stahl A, Menzel T, Lührs H, Löffler S, Dusel G, Rapp UR, and Scheppach W
- Subjects
- Alkaline Phosphatase analysis, Alkaline Phosphatase metabolism, Carcinoembryonic Antigen analysis, Carcinoembryonic Antigen metabolism, Down-Regulation, Humans, Mitogen-Activated Protein Kinase 3, Phosphorylation, Signal Transduction, Butyrates pharmacology, Cell Differentiation, HT29 Cells enzymology, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinases metabolism, Propionates pharmacology
- Abstract
We have characterized the effects of different short-chain fatty acids (SCFAs) on cell growth and differentiation as well as the phosphorylation state of ERK1 and 2 in the human colon adenocarcinoma cell line HT-29. Of the five SCFAs tested, only butyrate and propionate impaired cellular proliferation. Moreover, butyrate and propionate specifically resulted in a decrease in ERK1 and 2 phosphorylation at 3 and 6 hours post-treatment, suggesting a correlation between the ability of these SCFAs to inhibit cellular proliferation and decrease ERK phosphorylation. Notably, the decrease in ERK phosphorylation was observed prior to the induction of the differentiation markers alkaline phosphatase (AP) and carcinoembryonic antigen (CEA) by butyrate and propionate from days 6 to 18 post-treatment. In the case of butyrate- and propionate-induced differentiation, ERK phosphorylation is a marker and may play a role in the proliferation and/or differentiation states of this cell line.
- Published
- 2001
- Full Text
- View/download PDF
107. Influenza virus-induced AP-1-dependent gene expression requires activation of the JNK signaling pathway.
- Author
-
Ludwig S, Ehrhardt C, Neumeier ER, Kracht M, Rapp UR, and Pleschka S
- Subjects
- Activating Transcription Factor 2, Cell Line, Cyclic AMP Response Element-Binding Protein metabolism, DNA metabolism, DNA-Directed DNA Polymerase metabolism, Enzyme Activation, HeLa Cells, Humans, Interferon-beta genetics, MAP Kinase Kinase 7, Phosphorylation, Promoter Regions, Genetic, Protein Binding, RNA, Viral metabolism, Species Specificity, Transcription Factors metabolism, U937 Cells, Virus Replication, Gene Expression Regulation, Viral, Influenza A virus physiology, JNK Mitogen-Activated Protein Kinases, MAP Kinase Kinase 4, MAP Kinase Signaling System, Mitogen-Activated Protein Kinase Kinases metabolism, Transcription Factor AP-1 physiology
- Abstract
Influenza A virus infection of cells results in the induction of a variety of antiviral cytokines, including those that are regulated by transcription factors of the activating protein-1 (AP-1) family. Here we show that influenza virus infection induces AP-1-dependent gene expression in productively infected cells but not in cells that do not support viral replication. Among the AP-1 factors identified to bind to their cognate DNA element during viral infections of Madin-Darby canine kidney and U937 cells are those that are regulated via phosphorylation by JNKs. Accordingly, we observed that induction of AP-1-dependent gene expression correlates with a strong activation of JNK in permissive cells, which appears to be caused by viral RNA accumulation during replication. Blockade of JNK signaling at several levels of the cascade by transient expression of dominant negative kinase mutants and inhibitory proteins resulted in inhibition of virus-induced JNK activation, reduced AP-1 activity, and impaired transactivation of the IFN-beta promoter. Virus yields from transfected and infected cells in which JNK signaling was inhibited were higher compared with the levels from control cells. Therefore, we conclude that virus-induced activation of JNK and AP-1 is part of the innate antiviral response of the cell.
- Published
- 2001
108. Tet-system for the regulation of gene expression during embryonic development.
- Author
-
Fedorov LM, Tyrsin OY, Krenn V, Chernigovskaya EV, and Rapp UR
- Subjects
- Aging metabolism, Animals, Cytomegalovirus genetics, Doxycycline metabolism, Embryo, Mammalian cytology, Embryo, Mammalian drug effects, Embryo, Mammalian metabolism, Humans, Luciferases analysis, Luciferases biosynthesis, Luciferases genetics, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, Mice, Transgenic, Organ Specificity, Promoter Regions, Genetic genetics, beta-Galactosidase analysis, beta-Galactosidase biosynthesis, beta-Galactosidase genetics, Gene Expression Regulation, Developmental drug effects, Genes, Reporter genetics, Tetracycline metabolism
- Abstract
The ability to control gene expression in a temporal and spatial manner provides a new tool for the study of mammalian gene function particularly during development and oncogenesis. In this study the suitability of the tet-system for investigating embryogenesis was tested in detail. The tTACMV(M1) and rTACMV-3 (reverse Tc-controlled transactivator) transgenic mice were bred with NZL-2 bi-reporter mice containing the vector with a tTA/rTA responsive bidirectional promoter that allows simultaneous regulation of expression of two reporter genes encoding luciferase and beta-galactosidase. In both cases reporter genes were found to be expressed in a wide spectrum of tissues of double transgenic embryos and adult mice. The earliest expression was detected in tTACMV(M1)/NZL-2 embryos at embryonic day 10.5 (E10.5) and rTACMV-3/NZL-2 embryos at E13.5. Doxycycline abolished beta-gal expression in tTACMV(M1)/NZL-2 but induced it in rTACMV-3/NZL-2 embryos including late stages of embryo-genesis. The tTA and rtTA transactivators thus revealed a partially complementary mode of action during second half of embryonic development. These experiments demonstrated that both Tet regulatory systems function during embryonic development. We conclude that the Tet systems allows regulation of gene expression during embryonic development and that 'double reporter' animals like the NZL-2 mice are useful tools for the characterization of newly generated tet transactivator lines expressing tTA (or rtTA) in embryonic as well as in adult tissues.
- Published
- 2001
- Full Text
- View/download PDF
109. Active Ras induces heterodimerization of cRaf and BRaf.
- Author
-
Weber CK, Slupsky JR, Kalmes HA, and Rapp UR
- Subjects
- 14-3-3 Proteins, Binding Sites, Catalytic Domain, Cell Line, Dimerization, Humans, Peptide Fragments metabolism, Protein Isoforms, Tyrosine 3-Monooxygenase metabolism, Proto-Oncogene Proteins c-raf metabolism, ras Proteins physiology
- Abstract
Growth factor-induced signalling leads to activation of members of the Ras family and subsequent stimulation of different Raf isoforms. Within the mechanism of Raf activation, two isoforms of Raf, cRaf and BRaf, may cooperate. We investigated the relationship between cRaf and BRaf and found that active Ras induced heterodimerization of cRaf and BRaf, an effect that was dependent on the serine residue at position 621 of cRAF: Moreover, we also found that cRaf COOH-terminus constitutively associated with BRaf, whereas the NH(2) terminus did not, even in the presence of active RAS: These data suggest that Ras induces the cRaf-BRaf complex formation through the exposure of 14-3-3 binding sites in the COOH-terminus of cRAF: Thus, Ras-induced cRaf-Braf heterodimerization may explain the observed cooperativity of cRaf and BRaf in cells responding to growth factor signals.
- Published
- 2001
110. Apoptosis suppression by Raf-1 and MEK1 requires MEK- and phosphatidylinositol 3-kinase-dependent signals.
- Author
-
von Gise A, Lorenz P, Wellbrock C, Hemmings B, Berberich-Siebelt F, Rapp UR, and Troppmair J
- Subjects
- Enzyme Activation, Humans, MAP Kinase Kinase 1, Plasmids, Tumor Cells, Cultured, Apoptosis genetics, Mitogen-Activated Protein Kinase Kinases genetics, Phosphatidylinositol 3-Kinases genetics, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins c-raf genetics, Signal Transduction
- Abstract
Two Ras effector pathways leading to the activation of Raf-1 and phosphatidylinositol 3-kinase (PI3K) have been implicated in the survival signaling by the interleukin 3 (IL-3) receptor. Analysis of apoptosis suppression by Raf-1 demonstrated the requirement for mitochondrial translocation of the kinase in this process. This could be achieved either by overexpression of the antiapoptotic protein Bcl-2 or by targeting Raf-1 to the mitochondria via fusion to the mitochondrial protein Mas p70. Mitochondrially active Raf-1 is unable to activate extracellular signal-related kinase 1 (ERK1) and ERK2 but suppresses cell death by inactivating the proapoptotic Bcl-2 family member BAD. However, genetic and biochemical data also have suggested a role for the Raf-1 effector module MEK-ERK in apoptosis suppression. We thus tested for MEK requirement in cell survival signaling using the interleukin 3 (IL-3)-dependent cell line 32D. MEK is essential for survival and growth in the presence of IL-3. Upon growth factor withdrawal the expression of constitutively active MEK1 mutants significantly delays the onset of apoptosis, whereas the presence of a dominant negative mutant accelerates cell death. Survival signaling by MEK most likely results from the activation of ERKs since expression of a constitutively active form of ERK2 was as effective in protecting NIH 3T3 fibroblasts against doxorubicin-induced cell death as oncogenic MEK. The survival effect of activated MEK in 32D cells is achieved by both MEK- and PI3K-dependent mechanisms and results in the activation of PI3K and in the phosphorylation of AKT. MEK and PI3K dependence is also observed in 32D cells protected from apoptosis by oncogenic Raf-1. Additionally, we also could extend these findings to the IL-3-dependent pro-B-cell line BaF3, suggesting that recruitment of MEK is a common mechanism for survival signaling by activated Raf. Requirement for the PI3K effector AKT in this process is further demonstrated by the inhibitory effect of a dominant negative AKT mutant on Raf-1-induced cell survival. Moreover, a constitutively active form of AKT synergizes with Raf-1 in apoptosis suppression. In summary these data strongly suggest a Raf effector pathway for cell survival that is mediated by MEK and AKT.
- Published
- 2001
- Full Text
- View/download PDF
111. Influenza virus propagation is impaired by inhibition of the Raf/MEK/ERK signalling cascade.
- Author
-
Pleschka S, Wolff T, Ehrhardt C, Hobom G, Planz O, Rapp UR, and Ludwig S
- Subjects
- Active Transport, Cell Nucleus, Animals, Blotting, Western, Cell Line, Genes, Reporter, Humans, Immunohistochemistry, Influenza A virus genetics, Microscopy, Confocal, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Mitogen-Activated Protein Kinases antagonists & inhibitors, Phosphorylation, Proto-Oncogene Proteins c-raf antagonists & inhibitors, Proto-Oncogene Proteins c-raf metabolism, Recombinant Fusion Proteins metabolism, Ribonucleoproteins metabolism, Transfection, Viral Proteins metabolism, Virus Replication, Butadienes pharmacology, Enzyme Inhibitors pharmacology, Influenza A virus physiology, MAP Kinase Signaling System drug effects, Mitogen-Activated Protein Kinases metabolism, Nitriles pharmacology
- Abstract
Influenza A viruses are important worldwide pathogens in humans and different animal species. The functions of most of the ten different viral proteins of this negative-strand RNA virus have been well elucidated. However, little is known about the virus-induced intracellular signalling events that support viral replication. The Raf/MEK/ERK cascade is the prototype of mitogen-activated protein (MAP) kinase cascades and has an important role in cell growth, differentiation and survival. Investigation of the function of this pathway has been facilitated by the identification of specific inhibitors such as U0126, which blocks the cascade at the level of MAPK/ERK kinase (MEK). Here we show that infection of cells with influenza A virus leads to biphasic activation of the Raf/MEK/ERK cascade. Inhibition of Raf signalling results in nuclear retention of viral ribonucleoprotein complexes (RNPs), impaired function of the nuclear-export protein (NEP/NS2) and concomitant inhibition of virus production. Thus, signalling through the mitogenic cascade seems to be essential for virus production and RNP export from the nucleus during the viral life cycle.
- Published
- 2001
- Full Text
- View/download PDF
112. Activation of c-Jun NH2-terminal kinase/stress-activated protein kinase (JNK/SAPK) is critical for hypoxia-induced apoptosis of human malignant melanoma.
- Author
-
Kunz M, Ibrahim S, Koczan D, Thiesen HJ, Köhler HJ, Acker T, Plate KH, Ludwig S, Rapp UR, Bröcker EB, van Muijen GN, Flory E, and Gross G
- Subjects
- Gene Expression Regulation, Neoplastic physiology, Humans, Hypoxia physiopathology, Immunohistochemistry, JNK Mitogen-Activated Protein Kinases, Melanoma pathology, Melanoma physiopathology, Mitogen-Activated Protein Kinase Kinases metabolism, Necrosis, Neovascularization, Pathologic physiopathology, Proto-Oncogene Proteins c-jun metabolism, RNA, Messenger metabolism, Transcription, Genetic physiology, Transfection, Tumor Cells, Cultured enzymology, Tumor Cells, Cultured pathology, fas Receptor genetics, Apoptosis physiology, Hypoxia enzymology, MAP Kinase Kinase 4, Melanoma enzymology, Mitogen-Activated Protein Kinases metabolism, Neoplasm Invasiveness physiopathology, Neovascularization, Pathologic enzymology
- Abstract
Mitogen-activated protein kinase (MAPK) signaling was examined in malignant melanoma cells exposed to hypoxia. Here we demonstrate that hypoxia induced a strong activation of the c-Jun NH2-terminal kinase (JNK), also termed stress-activated protein kinase (SAPK), in the melanoma cell line 530 in vitro. Other members of the MAPK family, e.g., extracellular signal-regulated kinase and p38, remained unaffected by the hypoxic stimulus. Activated JNK/SAPK could also be observed in the vicinity of hypoxic tumor areas in melanoma metastases as detected by immunohistochemistry. Functional analysis of JNK/SAPK activation in the melanoma cell line 530 revealed that activation of JNK/SAPK is involved in hypoxia-mediated tumor cell apoptosis. Both a dominant negative mutant of JNK/SAPK (SAPKbeta K-->R) and a dominant negative mutant of the immediate upstream activator of JNK/SAPK, SEK1 (SEK1 K-->R), inhibited hypoxia-induced apoptosis in transient transfection studies. In contrast, overexpression of the wild-type kinases had a slight proapoptotic effect. Inhibition of extracellular signal-regulated kinase and p38 pathways by the chemical inhibitors PD98058 and SB203580, respectively, had no effect on hypoxiainduced apoptosis. Under normoxic conditions, no influence on apoptosis regulation was observed after inhibition of all three MAPK pathways. In contrast to recent findings, JNK/SAPK activation did not correlate with Fas or Fas ligand (FasL) expression, suggesting that the Fas/FasL system is not involved in hypoxia-induced apoptosis in melanoma cells. Taken together, our data demonstrate that hypoxia-induced JNK/SAPK activation appears to play a critical role in apoptosis regulation of melanoma cells in vitro and in vivo, independent of the Fas/FasL system.
- Published
- 2001
113. Specific function of B-Raf in mediating survival of embryonic motoneurons and sensory neurons.
- Author
-
Wiese S, Pei G, Karch C, Troppmair J, Holtmann B, Rapp UR, and Sendtner M
- Subjects
- Animals, Cell Survival physiology, Cells, Cultured, Embryo, Mammalian physiology, Embryonic and Fetal Development physiology, Ganglia, Spinal cytology, Mice, Spinal Cord cytology, Ganglia, Spinal embryology, Motor Neurons physiology, Neurons, Afferent physiology, Proto-Oncogene Proteins c-raf physiology, Spinal Cord embryology
- Abstract
Embryonic sensory and motoneurons depend on neurotrophic factors for survival. Here we show that their survival requires B-Raf, which, in this function, cannot be substituted by C-Raf. Sensory and motoneurons from b-raf-deficient mice do not respond to neurotrophic factors for their survival. However, these primary neurons can be rescued by transfection of a b-raf expression plasmid. In contrast, c-raf-deficient neurons survive in response to neurotrophic factors, similarly to neurons from wild-type mice. This points to an essential and specific function of B-Raf in mediating survival of sensory and motoneurons during development.
- Published
- 2001
- Full Text
- View/download PDF
114. The Ras-Raf relationship: an unfinished puzzle.
- Author
-
Kerkhoff E and Rapp UR
- Subjects
- Animals, Cell Division, Cell Membrane enzymology, Cytosol enzymology, Models, Biological, Protein Binding, Protein Kinase C chemistry, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism, Protein Structure, Tertiary, p21-Activated Kinases, Proto-Oncogene Proteins c-raf metabolism, ras Proteins metabolism
- Published
- 2001
- Full Text
- View/download PDF
115. Multiple signaling pathways regulate NF-kappaB-dependent transcription of the monocyte chemoattractant protein-1 gene in primary endothelial cells.
- Author
-
Goebeler M, Gillitzer R, Kilian K, Utzel K, Bröcker EB, Rapp UR, and Ludwig S
- Subjects
- Endothelium, Vascular cytology, Endothelium, Vascular physiology, Gene Expression Regulation drug effects, Haptens, Humans, Mitogen-Activated Protein Kinases physiology, Nickel pharmacology, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins drug effects, Nuclear Proteins metabolism, Promoter Regions, Genetic drug effects, RNA, Messenger drug effects, Signal Transduction physiology, Trans-Activators antagonists & inhibitors, Trans-Activators drug effects, Trans-Activators metabolism, Transcription, Genetic drug effects, Tumor Necrosis Factor-alpha pharmacology, Type C Phospholipases physiology, Umbilical Veins cytology, p38 Mitogen-Activated Protein Kinases, Chemokine CCL2 biosynthesis, Chemokine CCL2 genetics, Endothelium, Vascular metabolism, NF-kappa B pharmacology
- Abstract
The cytokine-induced C-C chemokine monocyte chemoattractant protein-1 (MCP-1) is an important regulator of leukocyte recruitment to sites of inflammatory challenge. Here, it is demonstrated that the widely distributed contact hapten NiCl(2), like tumor necrosis factor alpha (TNFalpha), induces monocyte-chemoattractant activity in primary human endothelial cells via induction of MCP-1. NiCl(2) rapidly activated mitogen-activated protein (MAP) kinase p38, and inhibition of p38 partially blocked NiCl(2)-induced MCP-1 messenger RNA and protein expression. Both NiCl(2)- and TNFalpha-induced MCP-1 synthesis was sensitive to D609, an inhibitor of phosphatidylcholine-dependent phospholipase C (PC-PLC). NiCl(2)-induced MCP-1 synthesis required activation of NF-kappaB since mutation of NF-kappaB-binding sites in the promoter resulted in complete loss of inducible promoter activity. Consistent with that finding, stimulation with NiCl(2) or TNFalpha activated IkappaB kinase-beta (IKKbeta), and transient transfection of dominant-negative IKKbeta strongly inhibited NiCl(2)- and TNFalpha-induced MCP-1 expression. However, D609 and the specific p38 inhibitor SB202190 did not affect NiCl(2)- and TNFalpha-induced IKKbeta activation, NF-kappaB DNA-binding activity, or transcriptional activity of a Gal4p65 fusion protein. This indicates that p38- and PC-PLC-dependent pathways directly regulate the transcriptional activity of NF-kappaB factors in the transcriptional complex. Consistent with that, inhibition of p38 blocked enhanced transcriptional activity induced by the transcriptional coactivator p300. Thus, it was concluded that at least 3 independent pathways regulate MCP-1 expression in endothelial cells. Its induction requires activation of the IKKbeta/IkappaBalpha/NF-kappaB signaling pathway, resulting in nuclear accumulation of p65 and subsequent recruitment of cofactors. Proper assembly and activity of this transcriptional complex is further modulated by the p38 MAP kinase cascade and a PC-PLC-dependent pathway.
- Published
- 2001
- Full Text
- View/download PDF
116. Ras-independent activation of the Raf/MEK/ERK pathway upon calcium-induced differentiation of keratinocytes.
- Author
-
Schmidt M, Goebeler M, Posern G, Feller SM, Seitz CS, Brocker EB, Rapp UR, and Ludwig S
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p21, Cyclins biosynthesis, Enzyme Activation, Epidermal Growth Factor pharmacology, Humans, Protein Kinase C physiology, Rabbits, Calcium physiology, Keratinocytes physiology, Mitogen-Activated Protein Kinases physiology, Proto-Oncogene Proteins c-raf physiology, ras Proteins physiology
- Abstract
MAPKs are crucially involved in the regulation of growth and differentiation of a variety of cells. To elucidate the role of MAPKs in keratinocyte differentiation, activation of ERK, JNK, and p38 in response to stimulation with extracellular calcium was analyzed. We provide evidence that calcium-induced differentiation of keratinocytes is associated with rapid and transient activation of the Raf/MEK/ERK pathway. Stimulation of keratinocytes with extracellular calcium resulted in activation of Raf isozymes and their downstream effector ERK within 10-15 min, but did not increase JNK or p38 activity. Calcium-induced ERK activation differed in kinetics from mitogenic ERK activation by epidermal growth factor and could be modulated by alterations of intracellular calcium levels. Interestingly, calcium stimulation led to down-regulation of Ras activity at the same time that ERK activation was initiated. Expression of a dominant-negative mutant of Ras also did not significantly impair calcium-induced ERK activation, indicating that calcium-mediated ERK activation does not require active Ras. Despite the transient nature of ERK activation, calcium-induced expression of the cyclin-dependent kinase inhibitor p21/Cip1 and the differentiation marker involucrin was sensitive to MEK inhibition, which suggests a role for the Raf/MEK/ERK pathway in early stages of keratinocyte differentiation.
- Published
- 2000
- Full Text
- View/download PDF
117. The Crk signaling pathway contributes to the bombesin-induced activation of the small GTPase Rap1 in Swiss 3T3 cells.
- Author
-
Posern G, Rapp UR, and Feller SM
- Subjects
- 3T3 Cells, Animals, COS Cells, Cells, Cultured, Fibroblasts drug effects, Fibroblasts metabolism, Guanine Nucleotide-Releasing Factor 2 genetics, Guanine Nucleotide-Releasing Factor 2 metabolism, MAP Kinase Kinase 1, MAP Kinase Signaling System, Mice, Mitogen-Activated Protein Kinase 3, Mitogen-Activated Protein Kinase Kinases metabolism, Mitogen-Activated Protein Kinases metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Oligonucleotides genetics, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-crk, Proto-Oncogene Proteins c-raf genetics, Signal Transduction, Transfection, src Homology Domains, Adaptor Proteins, Signal Transducing, Bombesin pharmacology, Proto-Oncogene Proteins physiology, rap1 GTP-Binding Proteins metabolism
- Abstract
Rap1 is a small GTPase implicated in cell proliferation and differentiation. The mechanisms how endogenous Rap1 is activated by many mitogenic stimuli including the neuropeptide bombesin remained unclear. Here we analyse which signaling pathways are necessary for Rap1 activation. Bombesin-mediated Rap1 activation in Swiss 3T3 and primary mouse embryo fibroblasts requires signaling components similar to those being essential for complex formation between p130Cas and Crk adapter proteins. The Crk/CRKL-binding region of the Rap1-specific exchange factor C3G (CBR) inhibits the bombesin-stimulated Rap1 activity in transfected Swiss 3T3 cells. Further characterization in COS cells showed that the CBR or a c-Crk I SH3 mutant specifically reduces both the basal as well as the stimulated Rap1 activity in a dose-dependent manner, whereas Ras is not affected. The CBR is complexed with endogenous c-Crk II and CRKL and blocks the protein association with catalytically active C3G. Such suppressors of Crk signaling do not affect Erk-phosphorylation induced by bombesin. Embryonic fibroblasts from b-raf knockout mice showed a bombesin-inducible Erk-phosphorylation, providing evidence that B-Raf does not link Rap1 to Erk-activation in bombesin-stimulated fibroblasts. We conclude that cellular Crk/CRKL complexes, recruited to upstream signaling components, contribute to basal and bombesin-induced Rap1 activity, which is independent from the Ras-Raf-Erk pathway under these circumstances.
- Published
- 2000
- Full Text
- View/download PDF
118. The neuronal apoptosis inhibitory protein suppresses neuronal differentiation and apoptosis in PC12 cells.
- Author
-
Götz R, Karch C, Digby MR, Troppmair J, Rapp UR, and Sendtner M
- Subjects
- Amino Acid Motifs, Animals, Apoptosis, Caspase 3, Caspases metabolism, Cell Differentiation, Humans, Mice, Nerve Growth Factor pharmacology, Nerve Tissue Proteins chemistry, Nerve Tissue Proteins genetics, Neurites physiology, Neuronal Apoptosis-Inhibitory Protein, Neurons enzymology, Neurons metabolism, PC12 Cells, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Receptors, Tumor Necrosis Factor, Tumor Necrosis Factor-alpha pharmacology, Nerve Tissue Proteins physiology, Neurons cytology
- Abstract
The human neuronal apoptosis inhibitory protein (NAIP) gene has been discovered as a candidate gene for spinal muscular atrophy, a genetic disorder characterized by motor neuron loss in the spinal cord. The telomeric NAIP gene on human chromosome 5 is deleted together with survival motor neurons (SMN) in many cases of the most severe forms of the disorder. NAIP, c-IAP1 (inhibitor of apoptosis-1), c-IAP2, X-IAP, survivin and Apollon comprise the mammalian inhibitors of the apoptosis family and contain an N-terminal domain with 1-3 imperfect repeats of an approximately 65 amino acids domain named the baculovirus IAP repeat (BIR) motif. We identified six NAIP genes in the mouse genome which were found to be expressed in a broad range of tissues. Furthermore, we have investigated the effects of NAIP in the rat pheochromocytoma PC12 cell line. These cells differentiate in the presence of nerve growth factor (NGF) into cells that resemble sympathetic neurons. We observed that NAIP overexpression impaired NGF-induced neurite outgrowth. The BIR motifs of NAIP (residues 1-345) were not required for this effect. However, the BIR domains of NAIP were essential to prevent apoptosis in PC12 cells after NGF deprivation or TNF-alpha receptor stimulation. Expression of full-length but not BIR-deleted-NAIP protects against cell death. This correlates with reduced activity of the cell death effector protease, caspase-3, in lysates of NAIP-PC12 cells, as measured by cleavage of the fluorogenic tetrapeptide substrate Asp-Glu-Val-Asp. Thus, unregulation of cellular differentiation and/or caspase suppression may contribute to motoneuron dysfunction and cell death in spinal muscular atrophy where NAIP is mutated.
- Published
- 2000
- Full Text
- View/download PDF
119. Serine/Threonine kinases 3pK and MAPK-activated protein kinase 2 interact with the basic helix-loop-helix transcription factor E47 and repress its transcriptional activity.
- Author
-
Neufeld B, Grosse-Wilde A, Hoffmeyer A, Jordan BW, Chen P, Dinev D, Ludwig S, and Rapp UR
- Subjects
- Animals, Base Sequence, Cell Line, DNA-Binding Proteins genetics, Gene Expression Regulation, Genes, Reporter, Humans, Intracellular Signaling Peptides and Proteins, MAP Kinase Kinase 2, Molecular Sequence Data, Phosphorylation, Precipitin Tests, Promoter Regions, Genetic, Protein Binding, Protein Serine-Threonine Kinases genetics, Recombinant Proteins, TCF Transcription Factors, Transcription Factor 7-Like 1 Protein, Transcriptional Activation genetics, Transfection, Yeasts, DNA-Binding Proteins metabolism, Mitogen-Activated Protein Kinase Kinases metabolism, Protein Serine-Threonine Kinases metabolism, Protein-Tyrosine Kinases metabolism, Repressor Proteins metabolism, Transcription Factors
- Abstract
In the search for physiological substrates of MAPK-activated protein (MAPKAP) kinases, we identified the basic helix-loop-helix (bHLH) transcription factor E47 as an interaction partner of chromosome 3p kinase (3pK) and MAPKAP-K2 (MK2). The E2A protein E47 is known to be involved in the regulation of tissue-specific gene expression and cell differentiation. E47 is a phosphoprotein, and we identified 3pK and MK2 as E47 kinases in vitro. Furthermore, the expression of either kinase results in a repression of the transcriptional activity of E47 on an E-box containing promoter. In summary, the MAPK-activated protein kinases 3pK and MK2 were identified to form an assembly with the bHLH protein E47 suggesting that these kinases are regulators of E47 activity and E47-dependent gene expression.
- Published
- 2000
- Full Text
- View/download PDF
120. Transactivation of naturally occurring HIV-1 long terminal repeats by the JNK signaling pathway. The most frequent naturally occurring length polymorphism sequence introduces a novel binding site for AP-1 factors.
- Author
-
Chen P, Flory E, Avots A, Jordan BW, Kirchhoff F, Ludwig S, and Rapp UR
- Subjects
- Binding Sites, Binding, Competitive, DNA-Binding Proteins genetics, Humans, JNK Mitogen-Activated Protein Kinases, MAP Kinase Kinase Kinases, Mutation, Oligodeoxyribonucleotides genetics, Polymorphism, Genetic, Protein Serine-Threonine Kinases metabolism, Signal Transduction genetics, T-Lymphocytes metabolism, Transcription Factor AP-1 genetics, Transcriptional Activation, Tumor Cells, Cultured, Mitogen-Activated Protein Kinase Kinase Kinase 11, HIV Long Terminal Repeat genetics, HIV-1 genetics, Mitogen-Activated Protein Kinases genetics
- Abstract
To study the role of MAPK cascades in the regulation of naturally occurring human immunodeficiency virus type 1 long terminal repeats (HIV-1 LTRs), we analyzed several HIV-1 LTRs from patients at different stages of disease progression. One of these naturally occurring HIV-1 LTRs contains an insertion termed the most frequent naturally occurring length polymorphism (MFNLP) and exhibited high inducibility upon T cell activation. We found that the protein kinase mixed lineage kinase 3/src-homology 3 domain-containing proline-rich kinase, a specific activator of the stress-activated protein kinase (SAPK)/JNK signaling pathway in T lymphocytes, induces high transcriptional activation of this promoter. Promoter inducibility is inhibited by the SAPK/JNK inhibitor, the JNK binding domain of the JNK interacting protein 1, and Tam-67 (N-terminal deletion mutant of c-Jun). In electrophoretic mobility shift assay, several protein complexes were found to bind to the MFNLP sequence in T cells. We identified AP-1 factors c-Fos and JunB as MFNLP-binding proteins, whose binding is abolished by introducing point mutations in the 3'-half of the MFNLP sequence. Introduction of these point mutations into the MFNLP containing HIV-1 LTR reduced src-homology 3 domain-containing proline-rich kinase -mediated transactivation. These data indicate that the AP-1-like binding site in the MFNLP sequence gives rise to a higher inducibility of natural HIV-LTRs by the SAPK/JNK signaling pathway.
- Published
- 2000
- Full Text
- View/download PDF
121. c-Raf regulates cell survival and retinal ganglion cell morphogenesis during neurogenesis.
- Author
-
Pimentel B, Sanz C, Varela-Nieto I, Rapp UR, De Pablo F, and de La Rosa EJ
- Subjects
- Animals, Apoptosis physiology, Cell Survival physiology, Chick Embryo, Gene Transfer Techniques, Retroviridae physiology, Cell Differentiation physiology, Proto-Oncogene Proteins c-raf physiology, Retina embryology, Retinal Ganglion Cells physiology
- Abstract
The signaling cascade Ras/Raf/mitogen-activated protein kinases modulates cell proliferation, differentiation, and survival, all key cellular processes during neural development. To better define the in vivo role of Raf during chick retinal neurogenesis, we interfered with Raf-dependent signaling during days 4.5 to 7.5 of embryonic development by expressing a dominant negative mutant of c-Raf (DeltaRaf), which blocks Ras-dependent Raf activation, and by overexpressing wild-type c-Raf. DeltaRaf expression induced an increase in cell death by apoptosis, whereas it did not affect overall cell proliferation and differentiation. In parallel, the number of Islet-1/2-positive and TUJ1-positive retinal ganglion cells were diminished in their definitive layer, whereas there was an increase in the number of mislocated Islet-1/2-positive cells. This disturbed morphogenesis correlated with a disruption of the optic fiber layer. Conversely, c-Raf overexpression caused moderate opposite effects on apoptosis. These results frame in vivo early neurogenesis processes in which c-Raf is essential.
- Published
- 2000
122. Raf induces NF-kappaB by membrane shuttle kinase MEKK1, a signaling pathway critical for transformation.
- Author
-
Baumann B, Weber CK, Troppmair J, Whiteside S, Israel A, Rapp UR, and Wirth T
- Subjects
- Animals, Cell Line, Genes, Reporter, I-kappa B Kinase, Recombinant Fusion Proteins metabolism, Recombinant Proteins metabolism, Spodoptera, Tetradecanoylphorbol Acetate pharmacology, Transfection, NF-kappa B metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-raf metabolism, Signal Transduction
- Abstract
NF-kappaB is regulated by inhibitor proteins (IkappaBs), which retain NF-kappaB in the cytoplasm. Signal-induced phosphorylation by the IkappaB-kinase complex containing the IkappaB-kinases 1 and 2 (IKK-1/2 or IKK-alpha/beta) and subsequent degradation of the IkappaB proteins are prerequisites for NF-kappaB activation. Many signals induce NF-kappaB, one of them being oncogenic Raf kinase. We investigated whether NF-kappaB induction is critical for Raf-mediated transformation. Here, we demonstrate that inhibition of NF-kappaB interferes with transformation by the Raf-oncogene, and we characterized the mechanism of NF-kappaB induction by activated Raf kinase and the tumor promoter phorbol 12-myristate 13-acetate (PMA). NF-kappaB activation by PMA and Raf critically depends on the IkappaB-kinase complex, most notably on IKK-2. A major signaling pathway induced by Raf is the mitogenic cytoplasmic kinase cascade. However, different inhibitors of this cascade do not affect PMA- and Raf-mediated NF-kappaB activation. Raf does not phosphorylate the IkappaB-kinase proteins directly. Raf rather synergizes with another membrane shuttle kinase MEKK1, and Raf-mediated activation of NF-kappaB is blocked by a dominant negative form of MEKK1. These results suggest that Raf induction of NF-kappaB is relayed by MEKK1, but not by the classical mitogenic cytoplasmic kinase cascade.
- Published
- 2000
- Full Text
- View/download PDF
123. Lung-targeted expression of the c-Raf-1 kinase in transgenic mice exposes a novel oncogenic character of the wild-type protein.
- Author
-
Kerkhoff E, Fedorov LM, Siefken R, Walter AO, Papadopoulos T, and Rapp UR
- Subjects
- Adenoma enzymology, Adenoma genetics, Adenoma pathology, Animals, Female, Gene Expression Regulation, Enzymologic, Humans, Immunohistochemistry, Lung enzymology, Lung pathology, Lung Neoplasms enzymology, Lung Neoplasms genetics, Lung Neoplasms pathology, Male, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, Mice, Transgenic, Promoter Regions, Genetic genetics, Proteolipids genetics, Proto-Oncogene Proteins c-raf analysis, Pulmonary Surfactants genetics, Recombinant Fusion Proteins genetics, Lung metabolism, Proto-Oncogene Proteins c-raf genetics
- Abstract
The c-Raf-1 kinase is a downstream effector of Ras signaling. Both proteins are highly oncogenic when they are mutationally activated, but only the Ras GTPase is frequently mutated in naturally occurring tumors. Although the c-Raf-1 protein was found to be amplified in different lung cancer cell lines, overexpression of the wild-type c-Raf-1 protein was shown to be insufficient to transform cultured cells. Here we have addressed the question of whether overexpression of the wild-type c-Raf-1 kinase can induce lung cancer in mice. We show that lung-targeted expression of oncogenically activated or wild-type c-Raf-1 proteins induces morphologically indistinguishable lung adenomas in transgenic mice. Compared with mice transgenic for the activated c-Raf-1-BxB, tumor development is delayed and occurs at a lower incidence in wild-type c-Raf-1 transgenic mice. Our studies show that the c-Raf-1 expression level is a critical parameter in tumor development and should be analyzed in more detail to evaluate its potential in the induction of cancer.
- Published
- 2000
124. Influenza virus-induced NF-kappaB-dependent gene expression is mediated by overexpression of viral proteins and involves oxidative radicals and activation of IkappaB kinase.
- Author
-
Flory E, Kunz M, Scheller C, Jassoy C, Stauber R, Rapp UR, and Ludwig S
- Subjects
- Antioxidants pharmacology, Base Sequence, Enzyme Activation, Free Radical Scavengers pharmacology, Free Radicals metabolism, Genes, Reporter, HIV Long Terminal Repeat, Hemagglutinin Glycoproteins, Influenza Virus genetics, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Humans, I-kappa B Kinase, Molecular Sequence Data, Nucleocapsid Proteins, Nucleoproteins genetics, Nucleoproteins metabolism, Protein Serine-Threonine Kinases genetics, Recombinant Proteins metabolism, T-Lymphocytes virology, Transcription, Genetic drug effects, Viral Core Proteins genetics, Viral Core Proteins metabolism, Viral Matrix Proteins genetics, Viral Matrix Proteins metabolism, Viral Proteins genetics, HIV-1 genetics, Influenza A virus growth & development, NF-kappa B metabolism, Oxidants metabolism, Protein Serine-Threonine Kinases metabolism, RNA-Binding Proteins, Transcriptional Activation, Viral Proteins metabolism
- Abstract
Influenza A viruses are capable of inducing the expression of a variety of cytokine and proapoptotic genes in infected cells. The promoter regions of most of these genes harbor binding sites for the transcription factor NF-kappaB which is an important mediator of immune and inflammatory responses. Our present study is based on an observation that influenza A virus infection of cells stimulates transcriptional activation of the HIV-1 long terminal repeat (LTR) which harbors two regulatory NF-kappaB elements, and is aimed at identifying the molecular mechanisms involved in this process. We found that the expression of influenza virus hemagglutinin (HA), matrix protein (M), and nucleoprotein (NP), as single factors is sufficient to transcriptionally activate the HIV-1 LTR. This process is mediated by oxidative radicals because treatment of cells with pyrrolidine dithiocarbamate, a scavenger of such radicals, abolished the transactivating ability. Expression of different influenza proteins induces activation of NF-kappaB-dependent gene expression but not transcriptional activation of an AP-1/Ets-dependent promoter, indicating a selectivity for NF-kappaB transactivation. Furthermore, influenza protein expression induces activation of IkappaB kinase (IKK). Accordingly coexpression of a catalytically inactive mutant of IKK abolishes influenza protein induced activation of NF-kappaB as well as HIV-1 LTR-dependent reporter gene expression, suggesting that IKK is an important intermediate within this signaling process. Taken together, our results show that various influenza virus proteins act as viral transactivators to modulate transcriptional activity of kappaB-element harboring promoters such as the HIV-LTR.
- Published
- 2000
- Full Text
- View/download PDF
125. The p150-Spir protein provides a link between c-Jun N-terminal kinase function and actin reorganization.
- Author
-
Otto IM, Raabe T, Rennefahrt UE, Bork P, Rapp UR, and Kerkhoff E
- Subjects
- 3T3 Cells, Amino Acid Sequence, Animals, Cells, Cultured, Insect Proteins genetics, JNK Mitogen-Activated Protein Kinases, Mice, Microfilament Proteins genetics, Mitogen-Activated Protein Kinase Kinases physiology, Mitogen-Activated Protein Kinases metabolism, Mitogen-Activated Protein Kinases physiology, Molecular Sequence Data, Phosphorylation, Actins metabolism, Drosophila Proteins, Insect Proteins metabolism, Microfilament Proteins metabolism, Mitogen-Activated Protein Kinase Kinases metabolism
- Abstract
The Jun N-terminal kinase (JNK) is a downstream effector of Rac and Cdc42 GTPases involved in actin reorganization [1-3]. A role of the Drosophila JNK homologue, Basket (DJNK/Bsk), in the regulation of cell shape changes and actin reorganization arises from its function in the process of dorsal closure [4-6]. One potential mechanism for induction of cytoskeletal changes by JNK is via transcriptional activation of the decapentaplegic gene (dpp, a member of the TGFbeta superfamily) [6]. A direct link between JNK signalling and actin organization has not yet been found, however. We have identified a novel DJNK-interacting protein, p150-Spir, that belongs to the Wiscott-Aldrich syndrome protein (WASP) homology domain 2 (WH2) family of proteins involved in actin reorganization [7] [8]. It is a multidomain protein with a cluster of four WH2 domains, a modified FYVE zinc-finger motif [9], and a DEJL motif, a docking site for JNK [10], at its carboxy-terminal end. In mouse fibroblasts, p150-Spir colocalized with F-actin and its overexpression induced clustering of filamentous actin around the nucleus. When coexpressed with p150-Spir in NIH 3T3 cells, JNK translocated to and colocalizes with p150-Spir at discrete spots around the nucleus. Carboxy-terminal sequences of p150-Spir were phosphorylated by JNK both in vitro and in vivo. We conclude that p150-Spir is a downstream target of JNK function and provides a direct link between JNK and actin organization.
- Published
- 2000
- Full Text
- View/download PDF
126. Overlapping and specific functions of Braf and Craf-1 proto-oncogenes during mouse embryogenesis.
- Author
-
Wojnowski L, Stancato LF, Larner AC, Rapp UR, and Zimmer A
- Subjects
- Animals, Cells, Cultured, Embryonic and Fetal Development, Gene Expression Regulation, Developmental, Isoenzymes genetics, Mice, Mutagenesis, Phenotype, Proto-Oncogenes, Proto-Oncogene Proteins c-raf genetics, Proto-Oncogene Proteins c-raf physiology
- Abstract
The three mammalian Raf serine/threonine protein kinases mediate the transduction of proliferative and differentiative signals from cell surface receptors to the nucleus. In vertebrates, Raf signaling has been implicated in the progression of mouse embryos through the two-cell stage and in the induction of posterior mesoderm. However, mouse embryos mutant for each of the Raf genes exhibit no developmental defects before mid-gestation. Here we describe the phenotype of mouse mutants with different combinations of mutant Craf-1 and Braf alleles. Our results show that Raf signaling is indeed indispensable for normal development beyond the blastocyst stage. However, due to a significant redundancy between Craf-1 and Braf, either gene is sufficient for normal development until mid-gestation. The molecular and developmental mechanisms for this redundancy were investigated by monitoring the expression of Raf genes throughout embryogenesis and by biochemical studies in mutant cell lines.
- Published
- 2000
- Full Text
- View/download PDF
127. Mitogenic signaling of Ras is regulated by differential interaction with Raf isozymes.
- Author
-
Weber CK, Slupsky JR, Herrmann C, Schuler M, Rapp UR, and Block C
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Enzyme Activation genetics, Humans, Isoenzymes genetics, Isoenzymes metabolism, Isoenzymes physiology, Molecular Sequence Data, Protein Binding genetics, Proto-Oncogene Proteins c-raf genetics, Proto-Oncogene Proteins c-raf metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Rabbits, Substrate Specificity genetics, Transfection, MAP Kinase Signaling System physiology, Mitogens physiology, Proto-Oncogene Proteins c-raf physiology, Proto-Oncogene Proteins p21(ras) physiology
- Abstract
In the mitogenic signaling cascade interaction of Ras with Raf represents a critical step for the regulation of cell growth and differentiation. The major effector of Ras, the serine/threonine kinase Raf exists as three isoforms with different tissue distributions. We demonstrate that transient transfection of oncogenic Ha-Ras leads to a preferential activation of endogenous c-Raf-1 in HEK 293 cells as opposed to A-Raf. In vitro binding studies using purified Ras binding domains of Raf as well as in vivo bindings tests with full length molecules reveals significantly lower binding affinities of A-Raf to Ha-Ras as compared to other Raf isoforms. The Ras-binding interface of c-Raf differs from A-Raf by a conservative Arg to Lys exchange at residue 59 or 22 respectively. Mutational analysis reveals that this residue represents a point of isozyme discrimination: c-Raf-R59K binds Ha-Ras weaker than the wildtype, likewise A-Raf-K22R increases its affinity to Ha-Ras in vivo and in vitro. Differential binding affinities are reflected in downstream signaling. Immunecomplex kinase assays reveal that Ha-Ras mediated Raf activation is decreased for c-Raf-R59K and increased for A-Raf-K22R when compared to the respective wildtype forms. Thus our observations introduce a new level of isoform discrimination in Ras/Raf signaling as a functional consequence of a conservative amino acid exchange in the Ras binding domains.
- Published
- 2000
- Full Text
- View/download PDF
128. [Deregulation of intracellular signal pathways in colorectal carcinoma].
- Author
-
Troppmair J and Rapp UR
- Subjects
- Apoptosis, Clinical Trials, Phase II as Topic, Colorectal Neoplasms pathology, Colorectal Neoplasms therapy, DNA Repair genetics, Humans, Mutation, Research, Tumor Cells, Cultured cytology, Tumor Cells, Cultured pathology, Colorectal Neoplasms genetics, Signal Transduction genetics
- Abstract
The analysis of the genetic causes of tumor development led to the identification of many genes affected in these diseases, which play a role in the control of cell proliferation and apoptosis. However, these studies also suggested that not all alterations are equal with regard to their effect on tumor growth, and they demonstrated that the deregulation of the classical cytoplasmic cascade made up by Ras-Raf-MEK and ERK plays a critical role in tumor development. These findings also raise the hope that via interference with a single pathway growth of a tumor carrying multiple genetic alterations will be affected.
- Published
- 2000
129. Transcriptional regulation of Fas gene expression by GA-binding protein and AP-1 in T cell antigen receptor.CD3 complex-stimulated T cells.
- Author
-
Li XR, Chong AS, Wu J, Roebuck KA, Kumar A, Parrillo JE, Rapp UR, Kimberly RP, Williams JW, and Xu X
- Subjects
- Base Sequence, Binding Sites, Binding, Competitive, CD3 Complex immunology, Cell Nucleus metabolism, Enhancer Elements, Genetic, Enzyme Inhibitors pharmacology, GA-Binding Protein Transcription Factor, Humans, Jurkat Cells, MAP Kinase Signaling System, Molecular Sequence Data, Neuropeptides metabolism, Promoter Regions, Genetic, Protein Binding, Signal Transduction drug effects, fas Receptor, CD3 Complex metabolism, DNA-Binding Proteins metabolism, Gene Expression Regulation, Neuropeptides genetics, Receptors, Antigen, T-Cell metabolism, Receptors, Tumor Necrosis Factor, T-Lymphocytes metabolism, Transcription Factor AP-1 metabolism, Transcription Factors metabolism
- Abstract
Fas (CD95 or APO-1), a transmembrane cell surface receptor of the tumor necrosis factor receptor family, is up-regulated in activated T lymphocytes. Our present study identified an upstream enhancer element (between nucleotide positions -862 and -682) containing a GA-binding protein (GABP) site and a low affinity activating protein-1 (AP-1)-binding site. T cell activation increased the DNA binding of GABP and AP-1 to this enhancer site. The specificity of GABP and AP-1 binding was demonstrated by competition electrophoretic mobility shift assay and supershift electrophoretic mobility shift assay with antibodies against GABP and AP-1, respectively. Mutational analysis of Fas promoter revealed that both GABP- and AP-1-binding sites were required for initiating Fas gene transcription. We further show that anti-CD3 mAb, phorbol 12-myristate 13-acetate, and phorbol 12-myristate 13-acetate/ionomycin strongly activated promoters carrying multiple copies of the Fas enhancer, and mutation of either the GABP or AP-1 binding site severely reduced transcriptional activity. Taken together, these results suggest that the transcription factors GABP and AP-1 play a critical role in the induction of Fas gene expression in T cell antigen receptor.CD3-stimulated Jurkat cells.
- Published
- 1999
- Full Text
- View/download PDF
130. Isotype-specific functions of Raf kinases.
- Author
-
Hagemann C and Rapp UR
- Subjects
- Animals, Autoantigens, Casein Kinase II, Ligands, MAP Kinase Signaling System, Mice, Mice, Knockout, Multiprotein Complexes, Proteasome Endopeptidase Complex, Protein Binding, Protein Serine-Threonine Kinases metabolism, Proteins metabolism, Pyruvate Kinase metabolism, Isoenzymes metabolism, Proto-Oncogene Proteins c-raf metabolism
- Abstract
The family of Raf-protein kinases consisting of A-Raf, B-Raf, and c-Raf-1 is involved in cellular processes which regulate proliferation, differentiation, and apoptosis. Cell-culture experiments and the knockout of individual Raf genes suggested that the three Raf isoforms have overlapping and unique regulatory functions. However, it is not known how these isotype-specific functions of Raf kinases occur in the cell. Published data suggest that Raf proteins might differ in the regulation of their activation as well as in their ability to connect to downstream signaling pathways. Since Raf is part of a multiprotein complex and protein-protein interactions are important for Raf signaling, we propose that isotype-specific functions can be achieved by isotype-restricted protein binding. Recently we were able to identify candidates for such Raf-isoform-specific interaction partners.
- Published
- 1999
- Full Text
- View/download PDF
131. The mitogen-activated protein (MAP) kinase p38 and its upstream activator MAP kinase kinase 6 are involved in the activation of signal transducer and activator of transcription by hyperosmolarity.
- Author
-
Bode JG, Gatsios P, Ludwig S, Rapp UR, Häussinger D, Heinrich PC, and Graeve L
- Subjects
- Animals, Base Sequence, COS Cells, DNA, Enzyme Activation, Intracellular Signaling Peptides and Proteins, MAP Kinase Kinase 6, Osmotic Pressure, Protein Tyrosine Phosphatase, Non-Receptor Type 11, Protein Tyrosine Phosphatase, Non-Receptor Type 6, Protein Tyrosine Phosphatases metabolism, Protein-Tyrosine Kinases metabolism, STAT1 Transcription Factor, Signal Transduction, p38 Mitogen-Activated Protein Kinases, Calcium-Calmodulin-Dependent Protein Kinases, DNA-Binding Proteins metabolism, Mitogen-Activated Protein Kinase Kinases metabolism, Mitogen-Activated Protein Kinases metabolism, Trans-Activators metabolism, Transcriptional Activation
- Abstract
Environmental stress (e.g. aniso-osmolarity and UV light), hypoxia/reoxygenation, and reactive oxygen species activate intracellular signaling cascades such as the "stress-responsive" mitogen-activated protein kinases and nuclear factor kappaB. We have recently shown that the Janus tyrosine kinase/signal transducer and activator of transcription (Jak/STAT) pathway is ligand-independently activated by hyperosmotic shock. In the present study, we show that besides STAT1 also the tyrosine phosphatase SHP2 became tyrosine-phosphorylated upon hyperosmolarity. SB 202190 and SB 203580 (specific inhibitors of p38) inhibited both STAT activation and tyrosine phosphorylation of SHP2 induced by hyperosmotic stress. Overexpression of wild-type p38 mitogen-activated protein kinase and its upstream activator mitogen-activated protein kinase kinase 6 (MKK6) resulted in an enhanced STAT1 tyrosine phosphorylation upon osmotic shock. Accordingly, overexpression of dominant negative mutants of p38 and MKK6 largely decreased hyperosmotic STAT1 activation and tyrosine phosphorylation of SHP2. Furthermore, we provide evidence that a genistein-sensitive tyrosine kinase different from Jak1 is involved in stress-activation of STAT1 and tyrosine phosphorylation of SHP2. These results strongly suggest that hyperosmotic shock activates STAT1 and SHP2 via p38 and its upstream activator MKK6.
- Published
- 1999
- Full Text
- View/download PDF
132. Binding of Gbetagamma subunits to cRaf1 downregulates G-protein-coupled receptor signalling.
- Author
-
Slupsky JR, Quitterer U, Weber CK, Gierschik P, Lohse MJ, and Rapp UR
- Subjects
- Animals, COS Cells, Cell Line, Chlorocebus aethiops, Humans, Kidney, Protein Binding, Proto-Oncogene Proteins p21(ras) physiology, Receptor, Bradykinin B2, Receptors, Bradykinin genetics, Receptors, Bradykinin physiology, Recombinant Fusion Proteins physiology, Transfection, Heterotrimeric GTP-Binding Proteins metabolism, MAP Kinase Signaling System physiology, Proto-Oncogene Proteins c-raf metabolism
- Abstract
Receptors of the seven transmembrane domain family are coupled to heterotrimeric G proteins [1]. Binding of ligand to these receptors induces dissociation of the heterotrimeric complex into free GTP-Galpha and Gbetagamma subunits, which then interact with their respective effector molecules to stimulate specific cellular responses. In some cases, these cellular responses involve mitogenic signalling [2]. The mitogen-activated protein (MAP) kinase cascade is initiated by the protein kinase cRaf1 and links growth factor receptor signalling to cell growth and differentiation [3]. The main activator of cRaf1 is the small GTP-binding protein Ras [4], and the binding of cRaf1 to GTP-Ras translocates cRaf1 to the plasma membrane, where it is activated [5]. It has been reported that cRaf1 associates directly with the beta subunit of heterotrimeric G proteins in vitro, and with the betagamma subunit complex in vivo [6], but the role of this association is not yet understood. Here, we show that cRaf1 associates with Gbeta1gamma2, and that this association in mammalian cells is significantly enhanced when active p21(Ras) is present or when cRaf1 is otherwise targeted to the membrane. Association with Gbeta1gamma2 has no effect on the kinase activity of cRaf1, but cRaf1 can affect Gbetagamma-mediated signalling events. Thus, membrane-localised cRaf1 inhibits G-protein-coupled receptor (GPCR)-stimulated activation of phospholipase Cbeta (PLCbeta) by sequestration of Gbetagamma subunits, an effect also observed with endogenous levels of cRaf1. Our data suggest that cRaf1 may be an important regulator of signalling by Gbetagamma, particularly in those GPCR systems that stimulate the MAP kinase cascade through the activation of p21(Ras).
- Published
- 1999
- Full Text
- View/download PDF
133. Anoxia-induced up-regulation of interleukin-8 in human malignant melanoma. A potential mechanism for high tumor aggressiveness.
- Author
-
Kunz M, Hartmann A, Flory E, Toksoy A, Koczan D, Thiesen HJ, Mukaida N, Neumann M, Rapp UR, Bröcker EB, and Gillitzer R
- Subjects
- Antigens, CD metabolism, Cell Hypoxia physiology, Gene Expression, Genes, Reporter, Humans, Immunohistochemistry, In Situ Hybridization, Melanoma genetics, Melanoma pathology, NF-kappa B metabolism, Necrosis, Neoplasm Invasiveness, Neoplasm Metastasis, Promoter Regions, Genetic, RNA, Messenger biosynthesis, Receptors, Chemokine metabolism, Receptors, Interleukin metabolism, Receptors, Interleukin-8A, Receptors, Interleukin-8B, Skin Neoplasms genetics, Skin Neoplasms pathology, Transcription Factor AP-1 metabolism, Transcriptional Activation genetics, Transcriptional Activation physiology, Tumor Cells, Cultured, Up-Regulation, Interleukin-8 metabolism, Melanoma metabolism, Skin Neoplasms metabolism
- Abstract
Besides its proinflammatory properties, interleukin-8 (IL-8) has been suggested as an important promoter for melanoma growth. To study the role of IL-8 in melanoma biology, we determined the in vivo expression of IL-8 mRNA by in situ hybridization in primary melanoma lesions and metastases. High levels of melanoma cell-associated IL-8-specific transcripts were exclusively detected in close vicinity of necrotic/hypoxic areas of melanoma metastases, whereas both in primary melanomas and in non-necrotic metastases IL-8 expression was low or absent. To analyze further the up-regulation of IL-8 mRNA expression in necrotic/hypoxic tumor areas, human melanoma cell lines of different aggressiveness exposed to severe hypoxic stress (anoxia) were used as an in vitro model. Anoxia induced IL-8 mRNA and protein expression in the highly aggressive/metastatic cell lines MV3 and BLM but not in the low aggressive cell lines IF6 and 530. As shown by IL-8 promoter-dependent reporter gene analysis and mRNA stability assays, elevated mRNA levels in melanoma cells were due to both enhanced transcriptional activation and enhanced IL-8 mRNA stability. Interestingly, transcriptional activation was abolished by mutations in the AP-1 and the NF-kappaB-like binding motifs, indicating that both sites are critical for IL-8 induction. Concomitantly, anoxia induced an enhanced binding activity of AP-1 and NF-kappaB transcription factors only in the highly aggressive cells. From our in vitro and in vivo data we suggest that anoxia-induced regulation of IL-8 might be a characteristic feature of aggressive tumor cells, thus indicating that IL-8 might play a critical role for tumor progression in human malignant melanoma.
- Published
- 1999
- Full Text
- View/download PDF
134. The JNK/SAPK activator mixed lineage kinase 3 (MLK3) transforms NIH 3T3 cells in a MEK-dependent fashion.
- Author
-
Hartkamp J, Troppmair J, and Rapp UR
- Subjects
- 3T3 Cells, Animals, Antigens, Polyomavirus Transforming genetics, Binding Sites, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Enhancer Elements, Genetic, Enzyme Activation, Enzyme Inhibitors pharmacology, Flavonoids pharmacology, Genes, Reporter, MAP Kinase Kinase 1, Mice, Mitogen-Activated Protein Kinase 1, Mitogen-Activated Protein Kinase 3, Phenotype, Phosphorylation drug effects, Protein Processing, Post-Translational drug effects, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Protein-Tyrosine Kinases antagonists & inhibitors, Recombinant Fusion Proteins physiology, Signal Transduction drug effects, Signal Transduction physiology, Transcription Factor AP-1 metabolism, Transfection, Mitogen-Activated Protein Kinase Kinase Kinase 11, Cell Transformation, Neoplastic metabolism, MAP Kinase Kinase Kinases, Mitogen-Activated Protein Kinase Kinases, Mitogen-Activated Protein Kinases, Protein Processing, Post-Translational physiology, Protein Serine-Threonine Kinases physiology, Protein-Tyrosine Kinases physiology
- Abstract
Mixed lineage kinases (MLKs) form a family of serin/threonine protein kinases with multiple protein/protein interaction domains (SH3, Cdc42 Rac interactive binding sequence, leucine zipper, and proline rich region), the physiological roles of which are largely unknown. We show that overexpression of wild type MLK3 leads to morphological transformation of NIH 3T3 fibroblasts and growth in soft agar. Consistent with this transforming potential, we demonstrate that MLK3 strongly induces transcription from a reporter construct that is driven by a composite AP-1-/Ets-1-enhancer element in HEK 293 cells. In the same cell system, MLK3 preferentially activates the c-Jun NH2-terminal kinase/stress-activated protein kinase (JNK/SAPK) mitogen-activated protein kinase cascade and to a lesser degree the extracellular signal-regulated kinase (ERK) pathway. Activation of the latter can be further enhanced by coexpression of wild type MEK1 and is blocked by the synthetic MEK inhibitor PD 098059 or a kinase-dead MEK1 mutant. Immunoprecipitated MLK3 catalyses the phosphorylation of MEK1 in vitro, but this phosphorylation leads only to a marginal activation. In support of these data, we also show that MEK1 is highly phosphorylated in vivo on Ser 217/221 in MLK3-transformed fibroblasts, whereas activating ERK phosphorylations are barely detectable. Nevertheless, MLK3-transformed NIH 3T3 fibroblasts are partially reverted when activation of MEK is specifically blocked with PD 098059. Our combined data show that although MLK3 is primarily an activator of the JNK/SAPK pathway, overexpression of the wild type protein leads to a transformed phenotype in NIH 3T3 cells that can be partially reversed by a synthetic MEK inhibitor. We conclude that the ERK pathway is necessary for MLK3-mediated transformation.
- Published
- 1999
135. CBP/p300 integrates Raf/Rac-signaling pathways in the transcriptional induction of NF-ATc during T cell activation.
- Author
-
Avots A, Buttmann M, Chuvpilo S, Escher C, Smola U, Bannister AJ, Rapp UR, Kouzarides T, and Serfling E
- Subjects
- Calcium-Calmodulin-Dependent Protein Kinases pharmacology, DNA-Binding Proteins physiology, Humans, Lymphocyte Activation drug effects, Signal Transduction, Transcription Factor AP-1 physiology, rac GTP-Binding Proteins, GTP-Binding Proteins physiology, Nuclear Proteins physiology, T-Lymphocytes immunology, Trans-Activators physiology
- Abstract
NF-ATc, an inducibly expressed transcription factor, controls gene expression in T lymphocytes and cardiomyocytes. We show here that the transcriptional co-activators CBP/p300 bind to and control the activity of the inducible N-terminal transactivation domain of NF-ATc, TAD-A. Similar to the N terminal transactivation domain of c-Jun, TAD-A is inducibly phosphorylated, but this phosphorylation is dispensable for the interaction with CBP/p300. Constitutive active versions of c-Raf and Rac synergistically enhance the CBP/p300-mediated increase of TAD-A activity, indicating the important role CBP/p300 plays in the integration of T cell activation signals. Since a mutation of CBP abolishing HAT activity is almost as active as wild-type CBP in T cells, functions of CBP/p300 other than histone acetylation appear to control the NF-AT-dependent transcription in T cells.
- Published
- 1999
- Full Text
- View/download PDF
136. Cot protooncoprotein activates the dual specificity kinases MEK-1 and SEK-1 and induces differentiation of PC12 cells.
- Author
-
Hagemann D, Troppmair J, and Rapp UR
- Subjects
- 3T3 Cells, Animals, Calcium-Calmodulin-Dependent Protein Kinases genetics, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cell Line, Transformed, Enzyme Activation, MAP Kinase Kinase 1, Mice, Mitogen-Activated Protein Kinase 1, Mitogen-Activated Protein Kinase 3, PC12 Cells, Phosphorylation, Protein Serine-Threonine Kinases biosynthesis, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins biosynthesis, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-jun metabolism, Rats, Receptor, EphA4, p38 Mitogen-Activated Protein Kinases, Cell Differentiation, Fetal Proteins metabolism, MAP Kinase Kinase Kinases, Mitogen-Activated Protein Kinase Kinases, Mitogen-Activated Protein Kinases, Protein Serine-Threonine Kinases metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins metabolism, Receptor Protein-Tyrosine Kinases metabolism
- Abstract
Mitogenic signals initiated at the plasma membrane are transmitted to the nucleus through an intricate signalling network. We identified the protooncoprotein Cot as a new component of mitogenic signalling cascades, which activates both the classic cytoplasmic cascade and the SAPK stress pathway. Wildtype and activated Cot phosphorylate and activate MEK-1 and SEK-1 in vitro. These findings are consistent with the sequence homology between Cot and the rat gene Tpl-2. Expression of oncogenic Cot in 293, NIH3T3 and PC12 cells leads to in vivo phosphorylation of endogenous c-Jun and Erk-1/2 suggesting that the serine/threonine kinase Cot functions beside c-Raf-1 and Mos as a direct activator of MEK-1. Furthermore, we have examined the biological effects of Cot on the phenotype of fibroblastic and neuronal cells. In order to test a potential c-Raf-1 dependency of Cot transformation, the effect of oncogenic Cot on Raf revertant CHP25 cells was determined. Cot could restore the transformed phenotype indicating that Cot transformation is not dependent on active c-Raf-1 and that Cot is not a target for the putative Raf inhibitor, which is presumably active in the revertant cell line. Expression of oncogenic versions of Raf as well as v-Mos leads to differentiation of PC12 cells. Cot also induces neurite outgrowth of PC12 cells. These data are consistent with the role of Cot in the classic mitogenic cascade and suggest that the simultaneously activated JNK/SAPK stress pathway has no antagonistic effects in this context.
- Published
- 1999
- Full Text
- View/download PDF
137. Ral and Rho-dependent activation of phospholipase D in v-Raf-transformed cells.
- Author
-
Frankel P, Ramos M, Flom J, Bychenok S, Joseph T, Kerkhoff E, Rapp UR, Feig LA, and Foster DA
- Subjects
- 3T3 Cells, Animals, Cell Line, Transformed, Enzyme Activation, GTP Phosphohydrolases physiology, Mice, Oncogene Proteins v-raf, Protein Kinase C metabolism, Signal Transduction physiology, ral GTP-Binding Proteins, Cell Transformation, Viral, GTP-Binding Proteins physiology, Phospholipase D metabolism, Retroviridae Proteins, Oncogenic physiology, Rho Factor physiology
- Abstract
Phospholipase D (PLD) activity is commonly elevated in response to mitogenic signals. We reported previously that although the transformed phenotype induced by v-Src was dependent upon Raf-1, the PLD activity induced by v-Src was independent of Raf-1. This observation suggested to us that Raf would not likely be an activator of PLD. However, upon examination of PLD activity in v-Raf-transformed cells, surprisingly, we found that PLD activity is elevated to levels that were even higher than that observed in v-Src-transformed cells. To characterize the mechanism of v-Raf-induced PLD activity, we examined the dependence of v-Raf-induced PLD activity upon protein kinase C (PKC) the small GTPases Ral and Rho, which have all been implicated in the activation of PLD. The v-Raf-induced PLD activity was inhibited by dominant negative mutants for both Ral and Rho. The dependence upon Ral was particularly surprising since Ral is a downstream target of Ras, which is an upstream activator of Raf. Depleting cells of PKC by long term phorbol ester treatment actually increased PLD activity in v-Raf-transformed cells, indicating that v-Raf-induced PLD activity is not dependent on PKC. These data describe a novel mechanism for PLD activation by v-Raf that is independent of PKC, but dependent upon both Ral and Rho GTPases., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
138. Different mitogen-activated protein kinase signaling pathways cooperate to regulate tumor necrosis factor alpha gene expression in T lymphocytes.
- Author
-
Hoffmeyer A, Grosse-Wilde A, Flory E, Neufeld B, Kunz M, Rapp UR, and Ludwig S
- Subjects
- Carrier Proteins pharmacology, Enzyme Activation, Enzyme Inhibitors pharmacology, Flavonoids pharmacology, Flow Cytometry, Humans, Imidazoles pharmacology, Ionomycin pharmacology, JNK Mitogen-Activated Protein Kinases, Pyridines pharmacology, Tetradecanoylphorbol Acetate pharmacology, Transcription, Genetic, Tumor Cells, Cultured, p38 Mitogen-Activated Protein Kinases, Adaptor Proteins, Signal Transducing, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Gene Expression Regulation drug effects, Mitogen-Activated Protein Kinases, Signal Transduction, T-Lymphocytes enzymology, Tumor Necrosis Factor-alpha genetics
- Abstract
Tumor necrosis factor a (TNF-alpha) is a potent proinflammatory cytokine and plays a crucial role in early events of inflammation. TNF-alpha is primarily produced by monocytes and T lymphocytes. In particular, T-cell-derived TNF-alpha plays a critical role in autoimmune inflammation and superantigen-induced septic shock. However, little is known about the intracellular signaling pathways that regulate TNF expression in T cells. Here we show that extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38-mitogen-activated protein kinase (MAPK) pathways control the transcription and synthesis of TNF-alpha in A3.01 T cells that produce the cytokine upon T cell activation by costimulation with 12-O-tetradecanoylphorbol-13-acetate (TPA) and ionomycin. Selective activation of each of the distinct MAPK pathways by expression of constitutively active kinases is sufficient for TNF-alpha promoter induction. Furthermore, blockage of all three pathways almost abolishes TPA/ionomycin-induced transcriptional activation of the TNF-alpha promoter. Selective inhibition of one or more MAPK pathways impairs TNF-alpha induction by TPA/ionomycin, indicating a cooperation between these signal transduction pathways. Our approach revealed that the MAPK kinase 6 (MKK6)/p38 pathway is involved in both transcriptional and posttranscriptional regulation of TNF expression. Moreover, analysis of the progressive 5' deletion mutants of the TNF-alpha promoter indicates that distinct promoter regions are targeted by either ERK-, JNK-, or p38-activating pathways. Thus, unlike what has been reported for other TNF-alpha-producing cells, all three MAPK pathways are critical and cooperate to regulate transcription of the TNF-alpha gene in T lymphocytes, suggesting a T-cell-specific regulation of the cytokine.
- Published
- 1999
- Full Text
- View/download PDF
139. Deamidation of Cdc42 and Rac by Escherichia coli cytotoxic necrotizing factor 1: activation of c-Jun N-terminal kinase in HeLa cells.
- Author
-
Lerm M, Selzer J, Hoffmeyer A, Rapp UR, Aktories K, and Schmidt G
- Subjects
- Bacterial Toxins genetics, Bacterial Toxins pharmacology, Base Sequence, Cell Cycle Proteins genetics, Cytotoxins genetics, Cytotoxins pharmacology, DNA, Complementary, Enzyme Activation, GTP-Binding Proteins genetics, HeLa Cells, Humans, JNK Mitogen-Activated Protein Kinases, Mass Spectrometry, Molecular Sequence Data, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins pharmacology, cdc42 GTP-Binding Protein, rac GTP-Binding Proteins, Bacterial Toxins metabolism, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cell Cycle Proteins metabolism, Cytotoxins metabolism, Escherichia coli metabolism, Escherichia coli Proteins, GTP-Binding Proteins metabolism, Mitogen-Activated Protein Kinases
- Abstract
Recently, Escherichia coli cytotoxic necrotizing factor 1 (CNF1) was shown to activate the low-molecular-mass GTPase RhoA by deamidation of Gln63, thereby inhibiting intrinsic and GTPase-activating protein (GAP)-stimulated GTPase activities (G. Schmidt, P. Sehr, M. Wilm, J. Selzer, M. Mann, and K. Aktories, Nature 387:725-729, 1997; G. Flatau, E. Lemichez, M. Gauthier, P. Chardin, S. Paris, C. Fiorentini, and P. Boquet, Nature 387:729-733, 1997). Here we report that in addition to RhoA, Cdc42 and Rac also are targets for CNF1 in vitro and in intact cells. Treatment of HeLa cells with CNF1 induced a transient formation of microspikes and formation of membrane ruffles. CNF1 caused a transient 10- to 50-fold increase in the activity of the c-Jun N-terminal kinase. Tryptic peptides of Cdc42 obtained from CNF1-treated cells by immunoprecipitation exhibited an increase in mass of 1 Da compared to control peptides, indicating the deamidation of glutamine 61 by the toxin. The same increase in mass was observed with the respective peptides obtained from CNF1-modified recombinant Cdc42 and Rac1. Modification of recombinant Cdc42 and Rac1 by CNF1 inhibited intrinsic and GAP-stimulated GTPase activities and retarded binding of 2'(3')-O-(N-methylanthraniloyl)GDP. The data suggest that recombinant as well as cellular Cdc42 and Rac are substrates for CNF1.
- Published
- 1999
- Full Text
- View/download PDF
140. The MKK6/p38 stress kinase cascade is critical for tumor necrosis factor-alpha-induced expression of monocyte-chemoattractant protein-1 in endothelial cells.
- Author
-
Goebeler M, Kilian K, Gillitzer R, Kunz M, Yoshimura T, Bröcker EB, Rapp UR, and Ludwig S
- Subjects
- Calcium-Calmodulin-Dependent Protein Kinases antagonists & inhibitors, Calcium-Calmodulin-Dependent Protein Kinases genetics, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cells, Cultured, Chemokine CCL2 genetics, Endothelium, Vascular cytology, Enzyme Activation, Enzyme Inhibitors pharmacology, Humans, Imidazoles pharmacology, JNK Mitogen-Activated Protein Kinases, MAP Kinase Kinase 6, Mitogen-Activated Protein Kinase 3, Multigene Family, Pyridines pharmacology, RNA, Messenger biosynthesis, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins physiology, p38 Mitogen-Activated Protein Kinases, Calcium-Calmodulin-Dependent Protein Kinases physiology, Chemokine CCL2 biosynthesis, Endothelium, Vascular metabolism, Gene Expression Regulation drug effects, Mitogen-Activated Protein Kinases, Signal Transduction physiology, Stress, Physiological physiopathology, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Monocyte chemoattractant protein-1 (MCP-1), a member of the C-C subfamily of chemokines, is important for the local recruitment of leukocytes to sites of inflammatory challenge. Here, we investigated endothelial signaling pathways involving members of the mitogen-activated protein (MAP) kinase superfamily and studied their role for MCP-1 expression in endothelium. We show that tumor necrosis factor-alpha (TNF-alpha), a potent inflammatory activator of endothelium, leads to activation of MAP kinases ERK, p38, and JNK in human umbilical vein endothelial cells (HUVEC). Contribution of MAP kinase pathways to TNF-alpha-induced synthesis of endothelial MCP-1 was then studied by pharmacologic inhibition and transient expression of dominant negative or constitutively active kinase mutants using flow cytometry, Northern blot, and luciferase reporter gene assays. Inhibition of Raf/MEK/ERK or SEK/JNK pathways had no significant effect on MCP-1 levels, whereas blocking the MKK6/p38 pathway by p38 inhibitors SB203580 or SB202190 or by a dominant negative mutant of MKK6, the upstream activator of p38, strongly inhibited TNF-alpha-induced expression of MCP-1. Consistent with that finding, expression of wild-type or constitutively active MKK6 significantly enhanced the effect of limiting TNF-alpha concentrations on MCP-1 synthesis. These data suggest a crucial role for the MKK6/p38 stress kinase cascade in TNF-alpha-mediated endothelial MCP-1 expression.
- Published
- 1999
141. Strict regulation of c-Raf kinase levels is required for early organogenesis of the vertebrate inner ear.
- Author
-
Sanz C, León Y, Troppmair J, Rapp UR, and Varela-Nieto I
- Subjects
- Animals, Apoptosis physiology, Chick Embryo, Ear, Inner cytology, Ear, Inner virology, Genetic Vectors, Morphogenesis, Nerve Growth Factors physiology, Oncogene Proteins v-raf, Organ Culture Techniques, Retroviridae genetics, Ear, Inner embryology, Protein-Tyrosine Kinases metabolism, Retroviridae Proteins, Oncogenic metabolism
- Abstract
Regulation of organogenesis involves a dynamic balance of the mechanisms regulating cell division, differentiation and death. Here we have investigated the pattern of expression of c-Raf kinase in the inner ear during early developmental stages and the consequences of manipulating c-Raf levels by misexpression of c-raf viral vectors in organotypic cultures of otic vesicle explants. We found that otic vesicles expressed c-Raf and its level remained constant during embryonic days 2 and 3 (E2-E3). c-Raf activity was increased in response to insulin like growth factor-I (IGF-I) and the activation by IGF-I of the c-Raf kinase pathway was a requirement to turn on cell proliferation in the otic vesicle. Overexpression of c-raf in E2.5 explants increased the proliferative response to low serum and IGF-I and blocked differentiation induced by retinoic acid. The increase in c-Raf levels also prevented nerve growth factor (NGF)-dependent induction of programmed cell death. Consistent with these results, the expression of a dominant negative c-Raf mutant potentiated retinoic acid action and decreased the rate of cell proliferation. We conclude that a strict control of c-Raf levels is essential for the co-ordination of the biological processes that operate simultaneously during early inner ear development.
- Published
- 1999
- Full Text
- View/download PDF
142. The RafC1 cysteine-rich domain contains multiple distinct regulatory epitopes which control Ras-dependent Raf activation.
- Author
-
Daub M, Jöckel J, Quack T, Weber CK, Schmitz F, Rapp UR, Wittinghofer A, and Block C
- Subjects
- Amino Acid Sequence, Animals, Binding Sites physiology, Cell Line, Enzyme Activation genetics, Epitopes genetics, Gene Expression Regulation genetics, Genes, Reporter genetics, Lipid Metabolism, Models, Molecular, Molecular Sequence Data, Mutagenesis genetics, Protein Binding physiology, Protein Kinase C chemistry, Rabbits, Signal Transduction physiology, Transcriptional Activation physiology, Cysteine chemistry, Proto-Oncogene Proteins c-raf chemistry, ras Proteins physiology
- Abstract
Activation of c-Raf-1 (referred to as Raf) by Ras is a pivotal step in mitogenic signaling. Raf activation is initiated by binding of Ras to the regulatory N terminus of Raf. While Ras binding to residues 51 to 131 is well understood, the role of the RafC1 cysteine-rich domain comprising residues 139 to 184 has remained elusive. To resolve the function of the RafC1 domain, we have performed an exhaustive surface scanning mutagenesis. In our study, we defined a high-resolution map of multiple distinct functional epitopes within RafC1 that are required for both negative control of the kinase and the positive function of the protein. Activating mutations in three different epitopes enhanced Ras-dependent Raf activation, while only some of these mutations markedly increased Raf basal activity. One contiguous inhibitory epitope consisting of S177, T182, and M183 clearly contributed to Ras-Raf binding energy and represents the putative Ras binding site of the RafC1 domain. The effects of all RafC1 mutations on Ras binding and Raf activation were independent of Ras lipid modification. The inhibitory mutation L160A is localized to a position analogous to the phorbol ester binding site in the protein kinase C C1 domain, suggesting a function in cofactor binding. Complete inhibition of Ras-dependent Raf activation was achieved by combining mutations K144A and L160A, which clearly demonstrates an absolute requirement for correct RafC1 function in Ras-dependent Raf activation.
- Published
- 1998
- Full Text
- View/download PDF
143. Activity of Rap1 is regulated by bombesin, cell adhesion, and cell density in NIH3T3 fibroblasts.
- Author
-
Posern G, Weber CK, Rapp UR, and Feller SM
- Subjects
- 3T3 Cells, Animals, Becaplermin, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cell Division drug effects, Cytochalasin D pharmacology, Guanosine Triphosphate metabolism, Kinetics, Mice, Phosphorylation, Platelet-Derived Growth Factor pharmacology, Proto-Oncogene Proteins c-raf metabolism, Proto-Oncogene Proteins c-sis, Tetradecanoylphorbol Acetate pharmacology, Transcription Factors metabolism, rap GTP-Binding Proteins, ras Proteins metabolism, Bombesin pharmacology, Cell Adhesion physiology, GTP-Binding Proteins metabolism
- Abstract
Rap1 and Ras are homologous GTPases that are implicated in cell proliferation and differentiation. At present, little is known about the regulation of Rap1 activity. Using a recently developed assay with activation-specific probes, we found increased activity of endogenous Rap1 in NIH3T3 cells after stimulation with the neuropeptide growth factor bombesin in a concentration- and time-dependent manner. The activity of endogenous Ras was unaffected. Analysis of putative effectors showed no activation of c-Raf-1 or B-Raf after bombesin stimulation. However, MAPK/Erk-phosphorylation and the proliferation rate was increased. In addition, Rap1 was activated during cell adhesion to coated and uncoated tissue culture plates, as well as in response to various mitogens. Surprisingly, the basal Rap1 activity was observed to be cell density-dependent, with low levels when cells were reaching confluency. The results suggest that Rap1 acts as an important mediator of mitogenic signals distinct to Ras activation.
- Published
- 1998
- Full Text
- View/download PDF
144. Cell cycle targets of Ras/Raf signalling.
- Author
-
Kerkhoff E and Rapp UR
- Subjects
- Animals, Cyclin D1 genetics, Cyclin D1 metabolism, Cyclin-Dependent Kinase Inhibitor p21, Cyclin-Dependent Kinase Inhibitor p27, Cyclins genetics, Cyclins metabolism, Genes, myc, Humans, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Protein Tyrosine Phosphatases metabolism, Cell Cycle genetics, Cell Cycle Proteins, Genes, ras, Proto-Oncogene Proteins c-raf genetics, Proto-Oncogene Proteins c-raf metabolism, Signal Transduction, Tumor Suppressor Proteins, cdc25 Phosphatases
- Abstract
The regulation of cell proliferation in multicellular organisms is a complex process, which is primarily regulated by external growth factors provided by surrounding cells. Once induced to proliferate, the passage through the mitotic cell cycle is directed by the components of the so called cell cycle machinery. Over the last years research on growth factor signal-transduction and the components of the cell cycle system has lead to a detailed knowledge of the mechanisms by which growth factors transmit their signals and of the relationship between the components of the cell cycle machinery. The remaining question how the growth factor mediated signal-transduction cascades couple with the cell cycle regulators has recently been a focus of interest.
- Published
- 1998
- Full Text
- View/download PDF
145. Activation of NF-kappa B by oncogenic Raf in HEK 293 cells occurs through autocrine recruitment of the stress kinase cascade.
- Author
-
Troppmair J, Hartkamp J, and Rapp UR
- Subjects
- 3T3 Cells, Animals, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Enzyme Activation, ErbB Receptors genetics, Gene Expression Regulation, Genes, Reporter, Humans, JNK Mitogen-Activated Protein Kinases, Mice, Mutation, Nitriles pharmacology, Protein Serine-Threonine Kinases metabolism, Quinazolines pharmacology, Suramin pharmacology, ras Proteins metabolism, Autocrine Communication, MAP Kinase Kinase Kinase 1, Mitogen-Activated Protein Kinases, NF-kappa B biosynthesis, Protein Kinases metabolism, Proto-Oncogene Proteins c-raf metabolism, Tyrphostins
- Abstract
Raf-1 kinase has been implicated in the induction of NF-kappa B activity by serum growth factors, phorbol ester and PTK oncogenes. Here we show that Raf activation of NF-kappa B, as measured in reporter gene assays, occurs indirectly and requires the stress kinase cascade. The stress pathway presumably becomes activated through induction of an autocrine loop by activated Raf (Raf-BXB) as suramin, the tyrphostin AG1478 and a dominant negative mutant of the EGF-R blocked NF-kappa B activation. Raf-BXB synergizes with SAPKs and a dominant negative mutant of SEK significantly reduces activation of NF-kappa B consistent with a role of this signaling pathway in the activation of NF-kappa B.
- Published
- 1998
- Full Text
- View/download PDF
146. Craf-1 protein kinase is essential for mouse development.
- Author
-
Wojnowski L, Stancato LF, Zimmer AM, Hahn H, Beck TW, Larner AC, Rapp UR, and Zimmer A
- Subjects
- Abnormalities, Multiple genetics, Animals, Base Sequence, Cell Division, Fetal Death genetics, Fibroblasts, Gene Expression Regulation, Developmental, Mice, Mice, Inbred Strains, Mice, Mutant Strains embryology, Molecular Sequence Data, Species Specificity, TNF Receptor-Associated Factor 3, Mice, Mutant Strains genetics, Mice, Mutant Strains growth & development, Mutation, Proteins genetics, Proteins metabolism
- Abstract
The three mammalian Raf serine/threonine protein kinases mediate the transduction of proliferative and differentiative signals from a variety of cell surface receptors to the nucleus. We report here that Craf-1 is essential for mouse development, as its mutation results in embryonic lethality. Developmental defects are found in mutant placentas as well as in the skin and in the lungs of mutant embryos. Craf-1 mutants also display a generalized growth retardation which is consistent with the ubiquitous expression of Craf-1 and which could be due to the reduced proliferation of mutant cells. Interestingly, the time-point of embryonal death varies depending on the genetic background. This suggests that Craf-1-mediated signaling is affected by genetic background-specific alleles of other genes., (Copyright 1998 Elsevier Science Ireland Ltd. All Rights Reserved.)
- Published
- 1998
- Full Text
- View/download PDF
147. Production and characterization of monoclonal antibodies against human BAD protein.
- Author
-
Afanassiev V, Troppmair J, Schuler M, Weber C, and Rapp UR
- Subjects
- Antibodies, Monoclonal, Antibody Specificity, Carrier Proteins immunology, Epitopes, Humans, Hybridomas, Immunoglobulin G immunology, Immunoglobulin Isotypes immunology, bcl-Associated Death Protein, Carrier Proteins analysis, Enzyme-Linked Immunosorbent Assay methods
- Abstract
More then 20 hybridoma cell lines secreting antibodies to recombinant human BAD protein were established. From five hybridomas monoclonal antibodies were purified and characterised. Four monoclonal antibodies belong to the IgG1 subclass. One monoclonal antibody (MAb) (designated as 6A11) belongs to the IgG2b subclass. All five MAbs have a specificity to different epitopes on the BAD protein. Four MAbs bound to the BAD + Bcl-2 complex, and one antibody (designated as 5E6) bound only BAD. A sandwich enzyme immunoassay specific to the human BAD molecule was developed using the different MAbs. The results show that the obtained antibodies are quite useful in studying the biological functions of BAD.
- Published
- 1998
- Full Text
- View/download PDF
148. Interaction between the protein kinase B-Raf and the alpha-subunit of the 11S proteasome regulator.
- Author
-
Kalmes A, Hagemann C, Weber CK, Wixler L, Schuster T, and Rapp UR
- Subjects
- Animals, Binding Sites, Cell Cycle Proteins, PC12 Cells, Phosphorylation, Rats, Yeasts, Proteins metabolism, Proto-Oncogene Proteins c-raf metabolism
- Abstract
Protein kinases of the Raf family act as signal-transducing elements downstream of activated cell surface receptors and are involved in the regulation of proliferation, differentiation, and cell survival. Whereas the role of c-Raf-1 as a mitogen-activated protein/extracellular signal-regulated kinase activator within the mitogenic cascade is well established, less is known about the mammalian Raf isoforms A-Raf and B-Raf. Here we report that B-Raf binds to PA28alpha, one of two subunits of the 11S regulator of proteasomes. PA28alpha was isolated as a B-Raf-binding protein in a yeast two-hybrid screen of a PC12 cDNA library. Both proteins can be coimmunoprecipitated after transient expression in 293 cells. No association could be found between PA28alpha and A-Raf or c-Raf-1. B-Raf binds to a region in PA28alpha that is important for its proteasome-activating function.
- Published
- 1998
149. The GABP-responsive element of the interleukin-2 enhancer is regulated by JNK/SAPK-activating pathways in T lymphocytes.
- Author
-
Hoffmeyer A, Avots A, Flory E, Weber CK, Serfling E, and Rapp UR
- Subjects
- 3T3 Cells, Animals, Enzyme Activation, GA-Binding Protein Transcription Factor, Humans, Ionomycin pharmacology, JNK Mitogen-Activated Protein Kinases, Mice, Phosphorylation, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Transcription, Genetic, Tumor Cells, Cultured, Mitogen-Activated Protein Kinase Kinase Kinase 11, Calcium-Calmodulin-Dependent Protein Kinases metabolism, DNA-Binding Proteins metabolism, Enhancer Elements, Genetic, Interleukin-2 genetics, MAP Kinase Kinase Kinases, Mitogen-Activated Protein Kinases, T-Lymphocytes enzymology, Transcription Factors metabolism
- Abstract
T cell activation leads via multiple intracellular signaling pathways to rapid induction of interleukin-2 (IL-2) expression, which can be mimicked by costimulation with 12-O-tetradecanoylphorbol-13-acetate (TPA) and ionomycin. We have identified a distal IL-2 enhancer regulated by the Raf-MEK-ERK signaling pathway, which can be induced by TPA/ionomycin treatment. It contains a dyad symmetry element (DSE) controlled by the Ets-like transcription factor GA-binding protein (GABP), a target of activated ERK. TPA/ionomycin treatment of T cells stimulates both mitogen-activated ERK, as well as the stress-activated mitogen-activated protein kinase family members JNK/SAPK and p38. In this study, we investigated the contribution of the stress-activated pathways to the induction of the distal IL-2 enhancer. We show that JNK- but not p38-activating pathways regulate the DSE activity. Furthermore, the JNK/SAPK signaling pathway cooperates with the Raf-MEK-ERK cascade in TPA/ionomycin-induced DSE activity. In T cells, overexpression of SPRK/MLK3, an activator of JNK/SAPK, strongly induces DSE-dependent transcription and dominant negative kinases of SEK and SAPK impair TPA/ionomycin-induced DSE activity. Blocking both ERK and JNK/SAPK pathways abolishes the DSE induction. The inducibility of the DSE is strongly dependent on the Ets-core motifs, which are bound by GABP. Both subunits of GABP are phosphorylated upon JNK activation in vivo and three different isoforms of JNK/SAPK, but not p38, in vitro. Our data suggest that GABP is targeted by signaling events from both ERK and JNK/SAPK pathways. GABP therefore is a candidate for signal integration and regulation of IL-2 transcription in T lymphocytes.
- Published
- 1998
- Full Text
- View/download PDF
150. High-intensity Raf signals convert mitotic cell cycling into cellular growth.
- Author
-
Kerkhoff E and Rapp UR
- Subjects
- 3T3 Cells, Animals, Cells, Cultured, Estrogen Antagonists pharmacology, Flow Cytometry, Immunoblotting, Mice, Receptors, Estrogen metabolism, Recombinant Fusion Proteins metabolism, Tamoxifen analogs & derivatives, Tamoxifen pharmacology, Cell Cycle physiology, Cell Division physiology, DNA Replication physiology, Mitosis physiology, Proto-Oncogene Proteins c-raf physiology
- Abstract
The selection of NIH 3T3 cells expressing a hydroxytamoxifen-inducible c-Raf-1-estrogen receptor fusion protein (c-Raf-1-BxB-ER) in the absence or presence of the inducer results in dramatic differences in the expression levels of the fusion protein. Hydroxytamoxifen-mediated constitutive activation of the Raf signal favors the selection of cells expressing low levels of c-Raf-1-BxB-ER. Cells selected in the absence of hydroxytamoxifen express up to 20 times higher levels of the inducible Raf kinase. Activation of the oncogenic Raf kinase in cells expressing low levels leads to a weak activation of the Raf/Mek/Erk cascade and the induction of S phase in confluent cells. The activation of cells expressing high levels of the kinase leads to a strong persistent signal and inhibits DNA synthesis and mitosis in proliferating cells. The inhibition of DNA synthesis and cell division is presumably due to the elevated expression of the cyclin-dependent kinase inhibitor p21cip1, similar to cells exposed to ionizing radiation. Despite the inhibition of DNA synthesis and mitosis, the constitutive activity of the Raf signaling pathway is still able to initiate cell growth. Activation of the high-intensity Raf signal in arrested serum-starved cells induces cell growth up to a size corresponding to that of M-phase cells in the absence of DNA synthesis. High-intensity Raf signals in proliferating cells consistently lead to an accumulation of cells with the size of M-phase cells and the DNA content of G1 cells or G2-M-phase cells. Therefore, the activation of Raf kinase is sufficient to drive cell growth, even in the presence of high levels of the cyclin-dependent kinase inhibitor p21cip1.
- Published
- 1998
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.