106 results on '"Ralph A. Bradshaw"'
Search Results
2. Proliferation of Human Corneal Endothelia in Organ Culture Stimulated by Wounding and the Engineered Human Fibroblast Growth Factor 1 Derivative TTHX1114
- Author
-
Ralph A. Bradshaw, Jennifer Jenkins-Eveleth, Jessica Weant, Sarah Pizzuto, and David Eveleth
- Subjects
Adult ,Male ,0301 basic medicine ,endothelium ,Endothelium ,Population ,FGF1 ,Protein Engineering ,Organ culture ,Fibroblast growth factor ,Cell junction ,03 medical and health sciences ,Organ Culture Techniques ,0302 clinical medicine ,cornea ,growth factors ,medicine ,Humans ,Pharmacology (medical) ,Progenitor cell ,education ,Aged ,Cell Proliferation ,Pharmacology ,Wound Healing ,education.field_of_study ,Chemistry ,Endothelium, Corneal ,Fuchs endothelial corneal dystrophy ,Contact inhibition ,Original Articles ,Middle Aged ,Cell biology ,Endothelial stem cell ,Ophthalmology ,030104 developmental biology ,medicine.anatomical_structure ,cardiovascular system ,030221 ophthalmology & optometry ,Fibroblast Growth Factor 1 ,Female - Abstract
Purpose: Corneal endothelial dystrophies are characterized by endothelial cell loss and dysfunction. Recent evidence suggests that corneal endothelial cells (CECs) can regenerate although they do not do so under normal conditions. This work sought to test whether CECs can be stimulated to proliferate in organ culture by wounding and/or by treatment with the engineered human fibroblast growth factor 1 (FGF1) derivative TTHX1114. Methods: Human donor corneas obtained from eye banks were maintained in organ culture in the presence or absence of TTHX1114. Wounds in the corneas were created by quartering the corneas. The CEC monolayer was identified as a regular layer by Hoechst staining of the nuclear DNA with cell outlines delineated by immunohistochemical identification of ZO-1. Nuclei and nuclei incorporating 5-ethynyl-2′-deoxyuridine (EdU) were counted using ImageJ. Results: CECs in normal corneas in undisturbed monolayers had low, but measurable, rates of proliferation. CECs at the edge of a wound had higher rates of proliferation, probably due to the release of contact inhibition. TTHX1114 increased proliferation at wound edges. After 7 days of culture, proliferating CECs formed contiguous groups of labeled cells that did not migrate away from one another. TTHX1114-treated cells, including the EdU labeled proliferating cells, retained normal morphology, including cell/cell junction ZO-1 staining. Conclusions: Proliferation of CECs in organ-cultured corneas is low, but can be stimulated by wounding or by the administration of TTHX1114 with the effects of each being additive. The CEC monolayer appears to have a population of progenitor cells that are susceptible to stimulation.
- Published
- 2020
3. Characterization of a Novel Caveolin Modulator That Reduces Vascular Permeability and Ocular Inflammation
- Author
-
William C. Sessa, Ralph A. Bradshaw, David Eveleth, Bo Tao, and Pascal Bernatchez
- Subjects
Vascular Endothelial Growth Factor A ,0301 basic medicine ,Caveolin 1 ,Biomedical Engineering ,Inflammation ,Vascular permeability ,vascular endothelial growth factor (VEGF) ,Article ,Nitric oxide ,Capillary Permeability ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,caveolin (Cav) ,Caveolin ,medicine ,Humans ,vascular permeability ,Endothelial Cells ,In vitro ,Cell biology ,Endothelial stem cell ,Ophthalmology ,Vascular endothelial growth factor A ,030104 developmental biology ,chemistry ,peptides ,cardiovascular system ,030221 ophthalmology & optometry ,medicine.symptom - Abstract
Purpose Caveolin (Cav) regulates various aspect of endothelial cell signaling and cell-permeable peptides (CPPs) fused to domains of Cav can reduce retinal damage and inflammation in vivo. Thus, the goal of the present study was to identify a novel CPP that improves delivery of a truncated Cav modulator in vitro and in vivo. Methods Phage display technology was used to identify a small peptide (RRPPR) that was internalized into endothelial cells. Fusions of Cav with the peptide were compared to existing molecules in three distinct assays, vascular endothelial growth factor-A (VEGF) induced nitric oxide (NO) release, VEGF induced vascular leakage, and in a model of immune mediated uveitis. Results RRPPR was internalized efficiently and was potent in blocking NO release. Fusing RRPPR with a minimal Cav inhibitory domain (CVX51401) dose-dependently blocked NO release, VEGF induced permeability, and retinal damage in a model of uveitis. Conclusions CVX51401 is a novel Cav modulator that reduces VEGF and immune mediated inflammation. Translational relevance CVX51401 is an optimized Cav modulator that reduces vascular permeability and ocular inflammation that is poised for clinical development.
- Published
- 2021
4. NGF and ProNGF: Regulation of neuronal and neoplastic responses through receptor signaling
- Author
-
Jordane Biarc, Hubert Hondermarck, Ralph A. Bradshaw, Jay Pundavela, Robert J. Chalkley, and Alma L. Burlingame
- Subjects
Male ,Cancer Research ,Tropomyosin receptor kinase A ,PC12 Cells ,Receptor tyrosine kinase ,Neurotrophic factors ,Receptors ,Nerve Growth Factor ,Low-affinity nerve growth factor receptor ,Epidermal growth factor receptor ,Cancer ,NGF ,Neurons ,biology ,phosphorylation ,Adaptor Proteins ,growth factor ,Cell biology ,Gene Expression Regulation, Neoplastic ,trkA ,Neurological ,Molecular Medicine ,Female ,Signal transduction ,signaling ,Platelet-derived growth factor receptor ,Receptor ,Signal Transduction ,Urologic Diseases ,medicine.medical_specialty ,Breast Neoplasms ,Nerve Tissue Proteins ,Receptors, Nerve Growth Factor ,Article ,Internal medicine ,Breast Cancer ,Genetics ,medicine ,Animals ,Humans ,Protein Precursors ,Receptor, trkA ,Molecular Biology ,Neoplastic ,Neurosciences ,Prostatic Neoplasms ,Rats ,Vesicular Transport ,Adaptor Proteins, Vesicular Transport ,Endocrinology ,Nerve growth factor ,Gene Expression Regulation ,nervous system ,receptor tyrosine kinase ,biology.protein - Abstract
Nerve growth factor (NGF) and its precursor (proNGF) are primarily considered as regulators of neuronal function that induce their responses via the tyrosine kinase receptor TrkA and the pan-neurotrophin receptor p75NTR. It has been generally held that NGF exerts its effects primarily through TrkA, inducing a cascade of tyrosine kinase-initiated responses, while proNGF binds more strongly to p75NTR. When this latter entity interacts with a third receptor, sortilin, apoptotic responses are induced in contrast to the survival/differentiation associated with the other two. Recent studies have outlined portions of the downstream phosphoproteome of TrkA in the neuronal PC12 cells and have clarified the contribution of individual docking sites in the TrkA endodomain. The patterns observed showed a similarity with the profile induced by the epidermal growth factor receptor, which is extensively associated with oncogenesis. Indeed, as with other neurotrophic factors, the distribution of TrkA and p75NTR is not limited to neuronal tissue, thus providing an array of targets outside the nervous systems. One such source is breast cancer cells, in which NGF and proNGF stimulate breast cancer cell survival/growth and enhance cell invasion, respectively. This latter activity is exerted via TrkA (as opposed to p75NTR) in conjunction with sortilin. Another tissue overexpressing proNGF is prostate cancer and here the ability of cancer cells to induce neuritogenesis has been implicated in cancer progression. These studies show that the non-neuronal functions of proNGF/NGF are likely integrated with their neuronal activities and point to the clinical utility of these growth factors and their receptors as biomarkers and therapeutic targets for metastasis and cancer pain.
- Published
- 2015
5. Phosphoproteomics of Tyrosine Kinases in the Nervous System
- Author
-
Robert J. Chalkley and Ralph A. Bradshaw
- Subjects
Nervous system ,medicine.anatomical_structure ,Chemistry ,medicine ,Phosphoproteomics ,Tyrosine kinase ,Cell biology - Published
- 2016
6. Receptor tyrosine kinase signaling – a proteomic perspective
- Author
-
Jordane Biarc, Ralph A. Bradshaw, Alma L. Burlingame, and Robert J. Chalkley
- Subjects
Cancer Research ,Proteome ,biology ,Protein Conformation ,Receptor Protein-Tyrosine Kinases ,Protein tyrosine phosphatase ,Phosphoproteins ,SH2 domain ,Article ,Receptor tyrosine kinase ,Cell biology ,Biochemistry ,ROR1 ,Genetics ,biology.protein ,Animals ,Humans ,Molecular Medicine ,Phosphorylation ,NCK1 ,Signal transduction ,Molecular Biology ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The RTKs are one of the most important families mediating transmembrane signaling and they participate and are instrumental in regulating a broad range of physiological activities. Indeed, tyrosine kinases in general, and the processes that they control and/or stimulate, provide a rich source of drug targets, particularly in growth related disorders such as cancer (Zwick et al., 2002; Krause and Van Etten, 2005). However, there remain many questions regarding their activation and downstream signaling and the application of proteomic analyses promises to answer many of them. There have been relatively few detailed studies of this type to date and it will require considerably more of them to better define the pathways with respect to both the major and minor PTMs that, along with the protein-protein interactions, are the means to direct the flow of the signals generated. It will take such approaches to define the specificity that characterize the individual families, even appreciating that to some degree all are capable of activating many, if not all, of the principal pathways. It will also be necessary to understand, in the highly complex networks of intracellular phosphorylation (that contain thousands of sites of modification and clearly have not yet been fully determined in any paradigm), exactly which kinases modify which substrates, and to work out the inter-relationships with other modifications such as O-GlcNAcylation and acetylation. Only then will it be possible to determine which modifications are physiologically significant and which are simply background. Along theway, these studies should continue to provide potential drug targets and perhaps improve the current lackluster biomarker discovery track record.
- Published
- 2011
7. The TrK Receptor Family
- Author
-
Hubert Hondermarck, Yohann Demont, and Ralph A. Bradshaw
- Subjects
biology ,Neurodegeneration ,medicine.disease ,Receptor tyrosine kinase ,Cell biology ,nervous system ,Tumor progression ,Apoptosis ,Trk receptor ,medicine ,biology.protein ,Receptor ,PI3K/AKT/mTOR pathway ,Neurotrophin - Abstract
The Trk family is composed of three principal members (A, B and C), also termed neurotrophin tyrosine kinase receptors (NTRK1, NTRK2, NTRK3), with several additional splice variants, which in humans are located on three different chromosomes (1, 9, 15). The Trk proteins are receptors for the neurotrophin family of growth factors (NGF, BDNF, NT-3, and NT4/5); they also bind and respond more weakly to the proforms of these neurotrophins. Both the neurotrophins and pro-neurotrophins utilize other receptors (p75NTR, sortilin) that can interact with the Trks and affect their activity. The Trks are predominantly expressed in nervous tissues, where they are essential for the growth, development, and maintenance of select types of both peripheral and central neurons and are involved in neurodegenerative diseases. The Trks are also found in some nonneuronal tissues and in a wide variety of tumors, where they can play an active role in tumor progression and metastasis. The activated Trk receptors primarily bind and signal through Shc, FRS2, and PLCγ, which in turn activate PI3K, the Erks, and the hydrolysis of inositol phospholipids, among other events, leading to the modulation of gene transcription.
- Published
- 2015
8. Characterization of Symmetric Complexes of Nerve Growth Factor and the Ectodomain of the Pan-neurotrophin Receptor, p75NTR
- Author
-
Mart Saarma, Veli-Matti Leppänen, Elaine Stephens, J. Gunther Grossmann, Martin C. Moncrieffe, Brandon T. Ruotolo, Tom L. Blundell, Carol V. Robinson, Jukka P. Aurikko, and Ralph A. Bradshaw
- Subjects
Models, Molecular ,Light ,Stimulation ,Tropomyosin receptor kinase A ,Receptor, Nerve Growth Factor ,Biochemistry ,Mass Spectrometry ,Article ,03 medical and health sciences ,0302 clinical medicine ,Nerve Growth Factor ,Humans ,Scattering, Radiation ,Computer Simulation ,Cysteine ,Receptor, trkA ,Receptor ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,biology ,Kinase ,Chemistry ,Cell Biology ,Ligand (biochemistry) ,Molecular biology ,Recombinant Proteins ,Protein Structure, Tertiary ,3. Good health ,Cell biology ,Molecular Weight ,Nerve growth factor ,Solubility ,nervous system ,Ectodomain ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Chromatography, Gel ,biology.protein ,sense organs ,Ultracentrifugation ,030217 neurology & neurosurgery ,Neurotrophin - Abstract
Nerve growth factor (NGF) is the ligand for two unrelated cellular receptors, TrkA and p75(NTR), and acts as a mediator in the development and maintenance of the mammalian nervous system. Signaling through TrkA kinase domains promotes neuronal survival, whereas activation of the p75(NTR) "death domains" induces apoptosis under correct physiological conditions. However, co-expression of these receptors leads to enhanced neuronal survival upon NGF stimulation, possibly through a ternary p75(NTR) x NGF x TrkA complex. We have expressed human p75(NTR) ligand binding domain as a secreted glycosylated protein in Trichoplusia ni cells. Following assembly and purification of soluble p75(NTR) x NGF complexes, mass spectrometry, analytical ultracentrifugation, and solution x-ray scattering measurements are indicative of 2:2 stoichiometry, which implies a symmetric complex. Molecular models of the 2:2 p75(NTR) x NGF complex based on these data are not consistent with the further assembly of either symmetric (2:2:2) or asymmetric (2:2:1) ternary p75(NTR) x NGF x TrkA complexes.
- Published
- 2005
9. Human Embryonic Kidney Cells (HEK-293 Cells): Characterization and Dose-Response Relationship for Modulated Release of Nerve Growth Factor for Nerve Regeneration
- Author
-
Gregory R. D. Evans, Ralph A. Bradshaw, Darren R. Tyson, Sanjay Dhar, Juan Carlos Jimenez, Thang Nguyen, and Yousuke Hamai
- Subjects
medicine.medical_specialty ,Cellular differentiation ,Enzyme-Linked Immunosorbent Assay ,In Vitro Techniques ,Kidney ,Transfection ,Cell Line ,Internal medicine ,Nerve Growth Factor ,Neurites ,medicine ,Humans ,Dose-Response Relationship, Drug ,business.industry ,HEK 293 cells ,Kidney metabolism ,Cell Differentiation ,Embryonic stem cell ,In vitro ,Nerve Regeneration ,Cell biology ,Ecdysterone ,Endocrinology ,Nerve growth factor ,Cell culture ,Surgery ,Genetic Engineering ,business - Abstract
The development of engineered constructs to bridge nerve gaps may hold the key to improved functional outcomes in the repair of injured peripheral nerves. These constructs must be rendered bioactive by providing the growth factors required for successful peripheral nerve regeneration. Previous studies demonstrated that harvested human and rat dermal fibroblasts could be genetically engineered to release nerve growth factor (NGF) both in vitro and in vivo. The use of fibroblasts, however, has the potential to cause scarring, and the expression of NGF from those cells was transient. To overcome these potential difficulties, human embryonic kidney cells were modified for use with the ecdysone-inducible mammalian expression system. These cells (hNGF-EcR-293) have been engineered and regulated to secrete human NGF in response to the ecdysone analogue ponasterone A. HEK-293 cells were transfected with human NGF cDNA with the ecdysone-inducible mammalian expression system (Invitrogen, Carlsbad, Calif.). Stable clones were then selected. Ponasterone A, an analogue of ecdysone, was used as the inducing agent. The secretion of NGF into the medium was analyzed with two different methods. After 24 hours of exposure to the inducing agent, cell medium was transferred to PC-12 cells seeded in 12-well plates, for determination of whether the secreted NGF was bioactive. Medium from untreated or ponasterone A-treated hNGF-EcR-293 cells was deemed bioactive on the basis of its ability to induce PC-12 cell differentiation. The concentrations of secreted NGF were also quantified with an enzyme-linked immunosorbent assay, in triplicate. NGF production was measured in successive samples of the same medium during a 9-day period, with maximal release of 9.05 +/- 2.6 ng/ml at day 9. Maximal NGF production was 8.46 +/- 2.1 pg/10(3) cells at day 9. These levels were statistically significantly different from levels in noninduced samples (p
- Published
- 2004
10. The Role of Tyrosine Residues in Fibroblast Growth Factor Receptor 1 Signaling in PC12 Cells
- Author
-
Simona Raffioni, Judith Murray-Rust, Erik D. Foehr, and Ralph A. Bradshaw
- Subjects
biology ,Fibroblast growth factor receptor 1 ,Cell Biology ,Biochemistry ,Molecular biology ,Receptor tyrosine kinase ,Growth factor receptor ,biology.protein ,Phosphorylation ,Tyrosine ,Signal transduction ,Kinase activity ,Molecular Biology ,Platelet-derived growth factor receptor - Abstract
To assess the contribution of the intracellular domain tyrosine residues to the signaling capacity of fibroblast growth factor receptor 1 (FGFR1), stably transfected chimeras bearing the ectodomain of the platelet-derived growth factor receptor (PDGFR) and the endodomain of FGFR1 were systematically altered by a tyrosine to phenylalanine bloc and individual conversions. The 15 tyrosine residues of the endodomain of this construct (PFR1) were divided into four linear segments (labeled A, B, C, and D) that contained 4, 4, 2, and 5 tyrosine residues, respectively. When stimulated by platelet-derived growth factor, derivatives in which the A, B, or A + B blocs of tyrosines were mutated were about two-thirds as active as the unmodified chimera at 48 h but achieved full activity by 96 h in a neurite outgrowth assay in transfected PC12 cells. Elimination of only the two activation loop tyrosines (C bloc) also inactivated the receptor. All derivatives in which 4 (or 5) of the D bloc tyrosines were mutated were inactive in producing differentiation but showed low levels of kinase activity in in vitro assays. Derivatives in which 1, 2, or 3 tyrosines of the D bloc in different combinations were systematically changed demonstrated that 2 residues (Tyr(677) and Tyr(701), using hFGFR1 numbering) were essential for bioactivity, but the remaining 3 residues, including Tyr(766), the previously identified site for phospholipase C gamma (PLC gamma) activation, were not. Differentiation activity was paralleled by the activation (phosphorylation) of FRS2, SOS, and ERK1/2. PLC gamma activity was dependent on the presence of Tyr(766) but also required Tyr(677) and/or Tyr(701). Although fully active chimeras did not require PLC gamma, the responses of chimeras showing reduced activation of FRS2 were significantly enhanced by this activity. These results establish that PFR1 does not utilize any tyrosine residues, phosphorylated or not, to activate FRS2. However, it does require Tyr(677) and/or Tyr(701), which may function to stabilize the active conformation directly or indirectly.
- Published
- 2001
11. NF-κB Signaling Promotes Both Cell Survival and Neurite Process Formation in Nerve Growth Factor-Stimulated PC12 Cells
- Author
-
Alison O'Mahony, Warner C. Greene, Erik D. Foehr, Xin Lin, Romas Geleziunas, and Ralph A. Bradshaw
- Subjects
Programmed cell death ,Neurite ,Cell Survival ,Recombinant Fusion Proteins ,Apoptosis ,Tropomyosin receptor kinase A ,PC12 Cells ,Receptor, Nerve Growth Factor ,NF-KappaB Inhibitor alpha ,Nerve Growth Factor ,Neurites ,Animals ,Low-affinity nerve growth factor receptor ,Receptor, trkA ,ARTICLE ,Receptor ,Genes, Dominant ,Platelet-Derived Growth Factor ,biology ,Tumor Necrosis Factor-alpha ,General Neuroscience ,NF-kappa B ,Cell Differentiation ,Rats ,Cell biology ,DNA-Binding Proteins ,Nerve growth factor ,nervous system ,biology.protein ,I-kappa B Proteins ,Signal transduction ,Platelet-derived growth factor receptor ,Signal Transduction - Abstract
Nerve growth factor binds to the TrkA and p75(NTR) (p75) and generates signals leading to neuronal cell survival, differentiation, and programmed cell death. Here we describe a series of experiments involving selective activation of either TrkA or p75 in which distinct cell-signaling intermediates promote different cellular consequences. We analyzed pheochromocytoma 12 (PC12) cells stably expressing chimeras consisting of the extracellular domain of PDGF receptor (PDGFR) fused to the transmembrane and cytoplasmic segments of p75 or TrkA. Because PC12 cells lack endogenous PDGFR, addition of PDGF to these cell lines permits selective activation of the p75 or TrkA responses without stimulating endogenous receptors. Although both p75 and TrkA activated nuclear factor-kappaB (NF-kappaB), we show that distinct proximal-signaling intermediates are used by each receptor. A dominant-negative mutant of TRAF6 blocked p75- but not TrkA-mediated induction of NF-kappaB. Conversely a dominant-negative mutant of Shc inhibited TrkA but not p75 activation of NF-kappaB. Both of these distinct signaling pathways subsequently converge, leading to activation of the IkappaB kinase complex. Moreover, the activation of NF-kappaB by these distinct pathways after stimulation of either TrkA or p75 leads to different physiological consequences. Blocking p75-mediated activation of NF-kappaB by ecdysone-inducible expression of a nondegradable mutant of IkappaBalpha significantly enhanced apoptosis. In contrast, blocking NF-kappaB induction via TrkA significantly inhibited neurite process formation in PC12 cells. Together these findings indicate that, although both of these receptors lead to the activation of NF-kappaB, they proceed via distinct proximal-signaling intermediates and contribute to different cellular outcomes.
- Published
- 2000
12. Discoidin domain receptor 1 (DDR1) signaling in PC12 cells: activation of juxtamembrane domains in PDGFR/DDR/TrkA chimeric receptors
- Author
-
Simona Raffioni, Erik D. Foehr, Ralph A. Bradshaw, Anie Tatavos, Silke Goetz, Eddi Dimarco, Michele De Luca, and Eri Tanabe
- Subjects
Recombinant Fusion Proteins ,tyrosine kinase • collagen receptor • signal transduction ,PC12 Cells ,Biochemistry ,Tropomyosin receptor kinase C ,Receptor tyrosine kinase ,Substrate Specificity ,Genetics ,Animals ,Receptors, Platelet-Derived Growth Factor ,Phosphorylation ,Receptor, trkA ,Receptor ,Discoidin Domain Receptors ,Molecular Biology ,DDR1 ,biology ,Phospholipase C gamma ,Chemistry ,Cell Membrane ,Receptor Protein-Tyrosine Kinases ,Rats ,Cell biology ,Enzyme Activation ,Isoenzymes ,nervous system ,Receptors, Mitogen ,Type C Phospholipases ,ROR1 ,biology.protein ,Cancer research ,Discoidin domain-containing receptor 2 ,Platelet-derived growth factor receptor ,Discoidin domain ,Signal Transduction ,Biotechnology - Abstract
The discoidin domain receptor (DDR1) is characterized by a discoidin I motif in the extracellular domain, an unusually long cytoplasmic juxtamembrane (JM) region, and a kinase domain that is 45% identical to that of the NGF receptor, TrkA. DDR1 also has a major splice form, which has a 37 amino acid insert in the JM region with a consensus Shc PTB site that is lacking in the shorter receptor. One class of ligands for the DDR receptors has recently been identified as being derived from the collagen family, but neither native PC12 cells, which express modest amounts of DDR1, nor transfected PC12 cells, which express much larger amounts of DDR1, respond to this ligand. A chimeric receptor, containing the extracellular domain of hPDGFRbeta fused to the transmembrane and intracellular regions of DDR1, also fails to mediate neuronal-like differentiation in stably transfected PC12 cells and is only weakly autophosphorylated. However, chimeric receptors, which are composed of combinations of intracellular regions from DDR1 and TrkA (with the extracellular domain of hPDGFRbeta), in some cases provided ligand (PDGF) -inducible receptor responses. Those with the TrkA kinase domain and the DDR1 JM regions were able to produce differentiation to varying degrees, whereas the opposite combination did not. Analysis of the signaling responses of the two chimeras with DDR1 JM sequences (with and without the insert) indicated that the shorter sequence bound and activated FRS2 whereas the insert-containing form activated Shc instead. Both activated PLCgamma through the carboxyl-terminal tyrosine of the TrkA domain (Y785 in TrkA residue numbering). Mutation of this site (Y-->F) eliminated PLCgamma activation (indicating there are no other cryptic binding sites for PLCgamma in the DDR1 sequences) and markedly reduced the differentiative activity of the receptor. This is in contrast to TrkA (or PDGFRbeta/TrkA chimeras), where ablation of this pathway has no notable effect on PC12 cell morphogenic responses. Thus, the activation of FRS2 and Shc (leading to MAPK activation) is weaker in the DDR1/TrkA chimeras than in TrkA alone, and the PLCgamma contribution becomes essential for full response. Nonetheless, both DDR1 JM regions contain potentially usable signaling sites, albeit they apparently are not activated directly in DDR1 (or DDR1 chimeras) in a ligand-dependent fashion. These findings suggest that the DDR1 receptors do have signaling capacity but may require additional components or altered conditions to fully activate their kinase domains and/or sustain the activation of the JM sites.
- Published
- 2000
13. Activation of the Stat3 Signaling Pathway Is Required for Differentiation by Interleukin-6 in PC12-E2 Cells
- Author
-
Ralph A. Bradshaw and Yvonne Y. Wu
- Subjects
STAT3 Transcription Factor ,Src Homology 2 Domain-Containing, Transforming Protein 1 ,Time Factors ,Neurite ,Cellular differentiation ,Biology ,PC12 Cells ,Biochemistry ,Stat3 Signaling Pathway ,Cell Line ,Proto-Oncogene Proteins p21(ras) ,Interferon-gamma ,Neurites ,Serine ,Animals ,Humans ,Nerve Growth Factors ,STAT1 ,Enzyme Inhibitors ,Phosphorylation ,Protein kinase A ,STAT3 ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Genes, Dominant ,Flavonoids ,Dose-Response Relationship, Drug ,Interleukin-6 ,Proteins ,JAK-STAT signaling pathway ,Cell Differentiation ,Cell Biology ,Molecular biology ,Rats ,DNA-Binding Proteins ,Adaptor Proteins, Vesicular Transport ,STAT1 Transcription Factor ,Gene Expression Regulation ,Shc Signaling Adaptor Proteins ,Mutagenesis, Site-Directed ,Trans-Activators ,biology.protein ,STAT protein ,Tyrosine ,Signal Transduction - Abstract
The role of signal transducer and activator of transcription (STAT) signaling pathways in the interleukin-6 (IL-6)-induced morphological differentiation of PC12-E2 cells was assessed using wild type and dominant negative mutants of Stat1 and Stat3, containing Tyr --Phe (YF), Ser --Ala (SA), and the double mutations (DM), respectively. FS3-YF or FS3-DM markedly inhibited the IL-6-induced response, but overexpression of FS3-SA caused only a modest inhibition. Expression of all Stat3 mutants had no effect on NGF-induced neurite outgrowth. Overexpression of wild type Stat1 protein inhibited IL-6 activated DNA binding complexes containing Stat3 homodimers, which may explain the partial negative effect of Stat1 on IL-6-induced neurite outgrowth. Specificity of these STAT constructs was confirmed using luciferase reporter gene assays, which showed that IL-6-activated transcription was blocked by expression of FS3-YF and FS3-DM and that FS1 enhanced the interferon gamma-activated transcription. Thus, in PC12-E2 cells, Stat3 homodimers are preferentially activated by IL-6, indicating a role for Stat3 in the regulation of cellular differentiation. Furthermore, IL-6 induced robust neurite outgrowth in PC12-E2 cells expressing dominant negative forms of RAS or SHC or in cells pretreated with the mitogen-activated protein kinase mitogen-activated protein kinase kinase inhibitor, PD98059. Thus, activation of the Stat3 signaling pathway, but not RAS/ERK dependent pathways, is essential for differentiation of PC12-E2 cells by IL-6.
- Published
- 2000
14. Yeast Methionine Aminopeptidase I
- Author
-
Ralph A. Bradshaw and Kenneth W. Walker
- Subjects
Alanine ,chemistry.chemical_classification ,Methionine ,biology ,Cell Biology ,Biochemistry ,chemistry.chemical_compound ,chemistry ,Methionyl Aminopeptidases ,Aspartic acid ,biology.protein ,Methionine synthase ,Asparagine ,Molecular Biology ,Amino acid synthesis ,Cysteine - Abstract
In eukaryotes, two isozymes (I and II) of methionine aminopeptidase (MetAP) catalyze the removal of the initiator methionine if the penultimate residue has a small radius of gyration (glycine, alanine, serine, threonine, proline, valine, and cysteine). Using site-directed mutagenesis, recombinant yeast MetAP I derivatives that are able to cleave N-terminal methionine from substrates that have larger penultimate residues have been expressed. A Met to Ala change at 329 (Met206 in Escherichia coli enzyme) produces an average catalytic efficiency 1.5-fold higher than the native enzyme on normal substrates and cleaves substrates containing penultimate asparagine, glutamine, isoleucine, leucine, methionine, and phenylalanine. Interestingly, the native enzyme also has significant activity with the asparagine peptide not previously identified as a substrate. Mutation of Gln356 (Gln233 inE. coli MetAP) to alanine results in a catalytic efficiency about one-third that of native with normal substrates but which can cleave methionine from substrates with penultimate histidine, asparagine, glutamine, leucine, methionine, phenylalanine, and tryptophan. Mutation of Ser195 to alanine had no effect on substrate specificity. None of the altered enzymes produced cleaved substrates with a fully charged residue (lysine, arginine, aspartic acid, or glutamic acid) or tyrosine in the penultimate position.
- Published
- 1999
15. FGF signal transduction in PC12 cells: Comparison of the responses induced by endogenous and chimeric receptors
- Author
-
Simona Raffioni, Ritsuko Fuji, Ralph A. Bradshaw, and Erik D. Foehr
- Subjects
Neurite ,Recombinant Fusion Proteins ,Blotting, Western ,Genetic Vectors ,Immunology ,PC12 Cells ,Polymerase Chain Reaction ,Receptor tyrosine kinase ,Cell Line ,Neurites ,Animals ,Humans ,Immunology and Allergy ,Receptors, Growth Factor ,Nerve Growth Factors ,Cloning, Molecular ,Insulin-like growth factor 1 receptor ,biology ,Cell Biology ,Protein-Tyrosine Kinases ,Blotting, Northern ,Receptors, Fibroblast Growth Factor ,Molecular biology ,Rats ,Cell biology ,Fibroblast growth factor receptor ,ROR1 ,biology.protein ,Electrophoresis, Polyacrylamide Gel ,GRB2 ,Signal transduction ,Platelet-derived growth factor receptor ,Signal Transduction - Abstract
Rat phaeochromocytoma (PC12) cells respond to many growth factors and produce different phenotypes, including neurite outgrowth. Receptor tyrosine kinases (RTK), which activate multiple signalling pathways in response to ligand binding, initiate many of these. One such family of receptors, the fibroblast growth factor receptor (FGFR), has four different members and expresses at least three of these in PC12 cells. A chimeric tyrosine kinase receptor, consisting of the extracellular domain of human plasma-derived growth factor receptor-beta (hPDGFR-beta) and the transmembrane and intracellular region of FGFR1 (designated PFR1), was constructed and was stably transfected into cloned PC12 cell lines. This chimera, which can be activated without stimulating endogenous RTK including other FGFR, induces neurite outgrowth in a PDGF-dependent manner. By altering the protocol for preparing the retroviral vectors, cells with a wide range of expression levels can be obtained. The amount of these chimeric receptors seems to correlate with the time and the intensity of response as observed in neurite outgrowth assays. Analysis of proteins implicated in FGFR1 signalling indicates that upon stimulation, a tyrosine phosphorylated protein designated FRS2 associates with SOS, Grb2 and the receptor. The chimeric receptor appears entirely similar to that observed for the stimulation of native PC12 cells with FGF2. These results support the view that FRS2 is the dominant FGFR1 signalling entity in PC12 cells.
- Published
- 1998
16. Differential Utilization of ShcA Tyrosine Residues and Functional Domains in the Transduction of Epidermal Growth Factor-induced Mitogen-activated Protein Kinase Activation in 293T Cells and Nerve Growth Factor-induced Neurite Outgrowth in PC12 Cells
- Author
-
Didier Thomas and Ralph A. Bradshaw
- Subjects
Phosphotyrosine binding ,Src Homology 2 Domain-Containing, Transforming Protein 1 ,Neurite ,Biology ,Kidney ,SH2 domain ,PC12 Cells ,environment and public health ,Biochemistry ,src Homology Domains ,chemistry.chemical_compound ,Epidermal growth factor ,Neurites ,Animals ,Humans ,Nerve Growth Factors ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,GRB2 Adaptor Protein ,Binding Sites ,Epidermal Growth Factor ,Membrane Proteins ,Proteins ,Tyrosine phosphorylation ,Cell Biology ,Rats ,Cell biology ,Enzyme Activation ,ErbB Receptors ,Adaptor Proteins, Vesicular Transport ,Shc Signaling Adaptor Proteins ,chemistry ,Mutagenesis ,Son of Sevenless Proteins ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Tyrosine ,GRB2 ,biological phenomena, cell phenomena, and immunity ,Phosphotyrosine-binding domain ,Protein Kinases ,Cell Division ,hormones, hormone substitutes, and hormone antagonists - Abstract
By transient expression of both truncated forms of p52(SHCA) and those with point mutations in 293T cells, it has been shown that, in addition to Tyr-317, Tyr-239/240 is a major site of phosphorylation that serves as a docking site for Grb2.Sos1 complexes. In addition, analysis of epidermal growth factor (EGF)-induced activation of mitogen-activated protein kinase in 293T cells showed that the overexpression Shc SH2 or phosphotyrosine binding (PTB) domains of ShcA alone has a more potent negative effect than the overexpression of the forms of ShcA lacking Tyr-317 or Tyr 239/240 or both. In transiently transfected PC12 cells, the ShcA PTB domain and tyrosine phosphorylation in the CH1 domain, especially on Tyr-239/240, are crucial for mediating nerve growth factor (NGF)-induced neurite outgrowth. These findings suggest that the EGF and NGF (TrkA) receptor can utilize Shc in different ways to promote their activity. For EGF-induced mitogen-activated protein kinase activation in 293T cells, both Shc PTB and SH2 domains are essential for optimal activation, indicating that a mechanism independent of Grb2 engagement with Shc may exist. For NGF-induced neurite outgrowth in PC12 cells, Shc PTB plays an essential role, and phosphorylation on Tyr-239/240, but not on Tyr-317, is required.
- Published
- 1997
17. Dissecting the roles of tyrosines 490 and 785 of TrkA protein in the induction of downstream protein phosphorylation using chimeric receptors
- Author
-
Alma L. Burlingame, Ralph A. Bradshaw, Jordane Biarc, and Robert J. Chalkley
- Subjects
Cell signaling ,Biochemistry & Molecular Biology ,animal structures ,Phosphoproteomics ,Recombinant Fusion Proteins ,1.1 Normal biological development and functioning ,Mutant Receptor ,Biochemistry ,PC12 Cells ,Medical and Health Sciences ,Receptor tyrosine kinase ,Mass Spectrometry ,Protein Phosphorylation ,Cell Signaling ,Underpinning research ,Receptors ,Low-affinity nerve growth factor receptor ,Animals ,Humans ,Receptors, Platelet-Derived Growth Factor ,Protein phosphorylation ,Receptor, trkA ,Phosphorylation ,Receptor ,Molecular Biology ,Platelet-Derived Growth Factor ,biology ,Phosphotyrosine Receptor ,Neurosciences ,Cell Biology ,Biological Sciences ,Rats ,nervous system ,Trk receptor ,trkA ,Chemical Sciences ,biology.protein ,Tyrosine ,Signal transduction ,Phosphotyrosine Signaling ,Platelet-derived growth factor receptor ,Signal Transduction - Abstract
Receptor tyrosine kinases generally act by forming phosphotyrosine-docking sites on their own endodomains that propagate signals through cascades of post-translational modifications driven by the binding of adaptor/effector proteins. The pathways that are stimulated in any given receptor tyrosine kinase are a function of the initial docking sites that are activated and the availability of downstream participants. In the case of the Trk receptors, which are activated by nerve growth factor, there are only two established phosphotyrosine-docking sites (Tyr-490 and Tyr-785 on TrkA) that are known to be directly involved in signal transduction. Taking advantage of this limited repertoire of docking sites and the availability of PC12 cell lines stably transfected with chimeric receptors composed of the extracellular domain of the PDGF receptor and the transmembrane and intracellular domains of TrkA, the downstream TrkA-induced phosphoproteome was assessed for the "native" receptor and mutants lacking Tyr-490 or both Tyr-490 and Tyr-785. Basal phosphorylation levels were compared with those formed after 20 min of stimulation with PDGF. Several thousand phosphopeptides were identified after TiO2 enrichment, and many were up- or down-regulated by receptor activation. The modified proteins in the native sample contained many of the well established participants in TrkA signaling. The results from the mutant receptors allowed grouping of these downstream targets by their dependence on the two characterized docking site(s). A clear subset that was not dependent on either Tyr-490 or Tyr-785 emerged, providing direct evidence that there are other sites on TrkA that are involved in downstream signaling.
- Published
- 2013
18. A Mouse Amidase Specific for N-terminal Asparagine
- Author
-
Stuart M. Arfin, Ralph A. Bradshaw, Albert E. Stewart, Nancy A. Jenkins, Neal G. Copeland, Yong Tae Kwon, Sergei Grigoryev, and Alexander Varshavsky
- Subjects
Amidohydrolase ,Base pair ,N-end rule ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,Complementary DNA ,Asparagine ,Deamidation ,Molecular Biology ,Gene ,Peptide sequence - Abstract
The N-end rule relates the in vivo half-life of a protein to the identity of its N-terminal residue. In both fungi and mammals, the tertiary destabilizing N-terminal residues asparagine and glutamine function through their conversion, by enzymatic deamidation, into the secondary destabilizing residues aspartate and glutamate, whose destabilizing activity requires their enzymatic conjugation to arginine, one of the primary destabilizing residues. We report the isolation and analysis of a mouse cDNA and the corresponding gene (termed Ntan1) that encode a 310-residue amidohydrolase (termed NtN-amidase) specific for N-terminal asparagine. The ∼17-kilobase pair Ntan1 gene is located in the proximal region of mouse chromosome 16 and contains 10 exons ranging from 54 to 177 base pairs in length. The ∼1.4-kilobase pair Ntan1 mRNA is expressed in all of the tested mouse tissues and cell lines and is down-regulated upon the conversion of myoblasts into myotubes. The Ntan1 promoter is located ∼500 base pairs upstream of the Ntan1 start codon. The deduced amino acid sequence of mouse NtN-amidase is 88% identical to the sequence of its porcine counterpart, but bears no significant similarity to the sequence of the NTA1-encoded N-terminal amidohydrolase of the yeast Saccharomyces cerevisiae, which can deamidate either N-terminal asparagine or glutamine. The expression of mouse NtN-amidase in S. cerevisiae nta1Δ was used to verify that NtN-amidase retains its asparagine selectivity in vivo and can implement the asparagine-specific subset of the N-end rule. Further dissection of mouse Ntan1, including its null phenotype analysis, should illuminate the functions of the N-end rule, most of which are still unknown.
- Published
- 1996
19. PC12-E2 cells: A stable variant with altered responses to growth factor stimulation
- Author
-
Ralph A. Bradshaw and Yvonne Y. Wu
- Subjects
Transcription, Genetic ,Physiology ,Clinical Biochemistry ,Basic fibroblast growth factor ,Gene Expression ,Receptors, Nerve Growth Factor ,PC12 Cells ,Receptor, Nerve Growth Factor ,chemistry.chemical_compound ,Growth factor receptor ,Proto-Oncogene Proteins ,Neurites ,Animals ,Nerve Growth Factors ,RNA, Messenger ,Receptor, trkA ,Growth Substances ,Phosphotyrosine ,Mitogen-Activated Protein Kinase 1 ,Membrane Glycoproteins ,Mitogen-Activated Protein Kinase 3 ,Epidermal Growth Factor ,biology ,Fibroblast growth factor receptor 2 ,Receptor Protein-Tyrosine Kinases ,Cell Differentiation ,Tyrosine phosphorylation ,Cell Biology ,Fibroblast growth factor receptor 4 ,Fibroblast growth factor receptor 3 ,Molecular biology ,Rats ,chemistry ,Fibroblast growth factor receptor ,Calcium-Calmodulin-Dependent Protein Kinases ,Dactinomycin ,biology.protein ,Tyrosine ,Fibroblast Growth Factor 2 ,Mitogen-Activated Protein Kinases ,Platelet-derived growth factor receptor - Abstract
A variant cell line, designated E2, characterized by more rapid responses to nerve growth factor (NGF) and basic fibroblast growth factor (bFGF) and markedly more robust responses to interleukin-6 and 8-Br-cAMP, has been subcloned from the rat PC12 cell line. The enhanced responsiveness to NGF in E2 cells is not due to receptor overexpression as judged by TrkA protein levels and tyrosine kinase activity, but may be associated with the increased and prolonged tyrosine phosphorylation of ERK1 (extracellular signal regulated kinase 1) and ERK2. The rapid morphological differentiation induced by different growth factors in E2 cells is mediated in a transcription-independent manner suggesting that E2 cells may constitutively express some differentiation-associated molecules that allow direct entry into the neuronal program.
- Published
- 1995
20. Staurosporine Causes Epidermal Growth Factor to Induce Differentiation in PC12 Cells via Receptor Up-regulation
- Author
-
Simona Raffioni and Ralph A. Bradshaw
- Subjects
Biology ,Fibroblast growth factor ,PC12 Cells ,Biochemistry ,Receptor tyrosine kinase ,Mice ,Alkaloids ,Growth factor receptor ,Epidermal growth factor ,Neurites ,medicine ,Animals ,Staurosporine ,RNA, Messenger ,Phosphotyrosine ,Molecular Biology ,Protein Kinase C ,Dose-Response Relationship, Drug ,Epidermal Growth Factor ,Cell Differentiation ,Cell Biology ,Blotting, Northern ,Phosphoproteins ,Rats ,Up-Regulation ,Cell biology ,ErbB Receptors ,Kinetics ,Nerve growth factor ,Mitogen-activated protein kinase ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Tyrosine ,Signal transduction ,medicine.drug - Abstract
Although they all utilize tyrosine kinase receptors and activate signaling pathways characterized by a similar set of phosphoproteins, epidermal growth factor (EGF) promotes only cell division while fibroblast growth factor (FGF) and nerve growth factor (NGF) can induce division followed by differentiation in PC12 cells. EGF, in contrast to NGF and FGF, cannot maintain the sustained phosphorylation and activation of mitogen-activated protein (MAP) kinase kinase and MAP kinases, which may account for the difference in phenotypic response. The pretreatment of PC12 cells with staurosporine, a protein kinase inhibitor, causes a substantial increase in both receptor and MAP kinase phosphorylation that results in a differentiative response (neurite proliferation). However, neurites begin to disappear after 3 days, despite the continual presence of EGF, and are largely gone after 5 days, which is not the case with NGF and FGF. Thus, the effect of staurosporine is not permanent. Northern and Western blots indicate that the staurosporine response mainly results from a substantial up-regulation in EGF receptor synthesis, thus providing a much stronger cell surface signal and supporting the view that quantitative rather than qualitative differences distinguish the EGF versus NGF/FGF signaling pathways in these cells.
- Published
- 1995
21. Protein NH2-terminal asparagine deamidase. Isolation and characterization of a new enzyme
- Author
-
Ralph A. Bradshaw, Stuart M. Arfin, and Albert E. Stewart
- Subjects
chemistry.chemical_classification ,endocrine system ,Methionine ,Peptide ,Cell Biology ,Biochemistry ,Glutamine ,chemistry.chemical_compound ,Enzyme ,chemistry ,Amide ,Hydrolase ,Aspartic acid ,Asparagine ,Molecular Biology - Abstract
An apparently unique enzyme, designated protein NH2-terminal asparagine deamidase (PNAD), that specifically converts NH2-terminal asparagine residues of peptide and protein substrates to aspartic acid, has been isolated to homogeneity from porcine liver by an eight-step procedure. PNAD is a relatively low abundance protein, is readily solubilized, and exists as a monomeric species of approximately 33 kDa. PNAD does not act on internal asparagine residues and requires a free N alpha-amino group. It has reduced or no activity on NH2-terminal asparagine dipeptides and no activity toward free asparagine or asparagine amide. It does not act on any NH2-terminal glutamine substrates. PNAD does not show a strong pH dependence suggesting that the enzyme can act equally well on substrates with ionized or unionized alpha-amino groups. The properties and specificity of PNAD are consistent with those expected for the enzyme required for the ubiquitin-dependent turnover of intracellular proteins that initiate with Met-Asn-. Such proteins should be N alpha-acetylated on the retained initiator methionine and can subsequently be modified by the removal of N-acetyl methionine by acylaminoacid hydrolase. Conversion of the resulting NH2-terminal asparagine to aspartic acid by PNAD would render the protein susceptible to arginylation, polyubiquitinylation and degradation as specified by the N-end rule.
- Published
- 1994
22. Pro-nerve growth factor induces autocrine stimulation of breast cancer cell invasion through tropomyosin-related kinase A (TrkA) and sortilin protein
- Author
-
Yohann Demont, Robert-Alain Toillon, Geneviève Choquet-Kastylevsky, Cyril Corbet, Hubert Hondermarck, Ingrid Fliniaux, Adeline Page, Yasemin Ataman-Önal, Xuefen Le Bourhis, and Ralph A. Bradshaw
- Subjects
medicine.medical_specialty ,Biopsy ,Carbazoles ,Breast Neoplasms ,Receptors, Nerve Growth Factor ,Tropomyosin receptor kinase A ,Biology ,Biochemistry ,Indole Alkaloids ,chemistry.chemical_compound ,Internal medicine ,Cell Line, Tumor ,Nerve Growth Factor ,medicine ,Humans ,Neoplasm Invasiveness ,Enzyme Inhibitors ,Protein Precursors ,Receptor, trkA ,Autocrine signalling ,Molecular Biology ,Protein kinase B ,Molecular Bases of Disease ,Cell Biology ,Carcinoma, Ductal ,Enzyme Activation ,Adaptor Proteins, Vesicular Transport ,Autocrine Communication ,Endocrinology ,Nerve growth factor ,src-Family Kinases ,chemistry ,nervous system ,Trk receptor ,Lymphatic Metastasis ,Cancer research ,biology.protein ,Female ,K252a ,Proto-Oncogene Proteins c-akt ,Proto-oncogene tyrosine-protein kinase Src ,Neurotrophin - Abstract
The precursor of nerve growth factor (proNGF) has been described as a biologically active polypeptide able to induce apoptosis in neuronal cells, via the neurotrophin receptor p75(NTR) and the sortilin receptor. Herein, it is shown that proNGF is produced and secreted by breast cancer cells, stimulating their invasion. Using Western blotting and mass spectrometry, proNGF was detected in a panel of breast cancer cells as well as in their conditioned media. Immunohistochemical analysis indicated an overproduction of proNGF in breast tumors, when compared with benign and normal breast biopsies, and a relationship to lymph node invasion in ductal carcinomas. Interestingly, siRNA against proNGF induced a decrease of breast cancer cell invasion that was restored by the addition of non-cleavable proNGF. The activation of TrkA, Akt, and Src, but not the MAP kinases, was observed. In addition, the proNGF invasive effect was inhibited by the Trk pharmacological inhibitor K252a, a kinase-dead TrkA, and siRNA against TrkA sortilin, neurotensin, whereas siRNA against p75(NTR) and the MAP kinase inhibitor PD98059 had no impact. These data reveal the existence of an autocrine loop stimulated by proNGF and mediated by TrkA and sortilin, with the activation of Akt and Src, for the stimulation of breast cancer cell invasion.
- Published
- 2011
23. Effect of nerve growth factor and fibroblast growth factor on PC12 cells: inhibition by orthovanadate
- Author
-
Yvorme Y. Wu and Ralph A. Bradshaw
- Subjects
medicine.medical_specialty ,Neurite ,medicine.medical_treatment ,Basic fibroblast growth factor ,Gene Expression ,Protein tyrosine phosphatase ,Biology ,Medical and Health Sciences ,PC12 Cells ,Dose-Response Relationship ,Diglycerides ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Nerve Growth Factors ,Tyrosine ,Phosphorylation ,Sodium orthovanadate ,Dose-Response Relationship, Drug ,Growth factor ,Tyrosine phosphorylation ,Cell Differentiation ,Cell Biology ,Articles ,Biological Sciences ,Ornithine Decarboxylase Inhibitors ,Protein-Tyrosine Kinases ,Cell biology ,Endocrinology ,chemistry ,Acetylcholinesterase ,Fibroblast Growth Factor 2 ,Drug ,Protein Tyrosine Phosphatases ,Vanadates ,Tyrosine kinase ,Developmental Biology ,Signal Transduction - Abstract
Sodium orthovanadate, an inhibitor of protein tyrosine phosphatases, causes increased levels of tyrosine phosphorylation and blocks, at noncytotoxic concentrations, the differentiative response of rat pheochromocytoma (PC12) cells to beta-nerve growth factor (beta NGF) and basic fibroblast growth factor (bFGF) in a reversible manner. It also prevents growth factor-induced neurite proliferation in primed cells and causes the retraction of previously formed neurites, even in the presence of beta NGF or bFGF. It is equally effective in blocking neurite proliferation by 8-Br-cAMP. Zinc chloride and ammonium molybdate, two other inhibitors of tyrosine phosphatases, also cause parallel decreases in neurite proliferation. Orthovanadate generally reduces the transcription of immediate early response genes (TIS 8 and c-fos) and secondary response genes (ornithine decarboxylase (ODC), acetyl-cholinesterase (AChE) and SCG 10) induced by beta NGF, bFGF, EGF, and PMA, albeit in a variable fashion. There was no observed effect on the kinetics of expression as judged by TIS 8 induction by beta NGF and protein kinase C (PKC) downregulation did not change the levels of inhibition by orthovanadate seen in control cells. Orthovanadate does not affect the production of diacylglycerol induced by beta NGF or bFGF. These observations are consistent with the view that growth factor stimulation of differentiation in PC12 cells involves at least one other PKC independent pathway, and that cAMP and PMA (and their active analogs) activate tyrosine kinases (albeit probably secondarily), which are at least partially responsible for their actions. Although the exact site(s) of action of orthovanadate that lead to the inhibition of growth factor-induced neurite proliferation are unknown, the results presented suggest that it prolongs tyrosine phosphorylations by nonreceptor tyrosine kinases that act downstream from the receptor kinases.
- Published
- 1993
24. ChemInform Abstract: Polypeptide Growth Factors: Structure, Function, and Mechanism of Action
- Author
-
Yvonne Y. Wu, Simona Raffioni, Hubert Hondermarck, Ritsuko Fujii, Ralph A. Bradshaw, and Michael A. Yarski
- Subjects
Signal peptide ,biology ,Neurotrophic factors ,Chemistry ,Effector ,Cell surface receptor ,Extracellular ,biology.protein ,General Medicine ,Signal transduction ,Receptor ,Neurotrophin ,Cell biology - Abstract
Polypeptide growth factors are a diverse group of hormone-like agents that regulate growth and differentiation through cell surface receptors. They are generally represented by homologous families containing several members with distinct overlapping receptor interactions and hence, responsive hue specificities. Similarly, their receptors are also clustered in family groups of sequence-related proteins. The neurotrophin group, characterized by its typical prototypical member nerve growth factor (NGF), has four members which interact variably with three receptors of the family. The activation of their tyrosine bases initiates the characteristic responses. In PC12 cells, stimulation by NGF leads to activation of non-receptor tyrosine bases and several phospholipid dependent pathways. The sum of these signals induce a variety of immediate early response genes that govern the phenotypic response. However, the minimum pathway ( and its essential components) is not yet fully defined. One of the major functions of proteins is to mediate signal transduction. In higher organisms, this complex process utilizes both soluble and membrane bound ligands to initiate the responses and a wide variety of effector molecules, including cell-surface receptors, that are generally ligand specific, and the component of the intracellular machinery that translate the stimulus into the characteristic phenotypic response (ref. 1). Of the many biological responses that are controlled in this fashion, those related to growth are now amongst those that are best studied. In general, these can be subdivided into hyperplastic (increase in cell number) and hypertrophic (increase in cell size), and are, for the most part, controlled by polypeptide growth factors. These substances are basically indistinguishable in their principal functional and mechanistic properties from many of the classic hormones (ref. 2). In the main, they act through specific cell surface receptors that can initiate a variety of intracellular signals invariably involving a spectrum of protein phosphorylations (ref. 3). As with the classical hormone, the polypeptide growth factors are usually relatively small, highly soluble proteins of compact structure. Although some are glycosylated, the majority are not (ref. 4). Most also contain several disulfide bonds which lends further reinforcement to the 3-dimensional structure, and is a feature expected of proteins exported through the endoplasmic reticulum (ref. 5). There is, however, a small but significant subgroup that is devoid of signal peptides and possess only reduced half-cysteine residues, as expected for intracellular proteins. Significant quantities of these agents are often found associated with the extracellular matrix, but the manner in which they reach extracellular compartment is not presently understood. From a functional point of view, polypeptide growth factors can conveniently be divided into four categories (Table 1). The best known group are those which act on tissues and include a broad range of agents with a myriad of responsive target cells. The neurotrophic factors are a more specialized
- Published
- 2010
25. Overview of Cell–Cell and Cell–Matrix Interactions
- Author
-
E. Brad Thompson and Ralph A. Bradshaw
- Subjects
Intracrine ,Cell signaling ,Computer science ,Cell ,Local hormone ,Genomics ,Computational biology ,Biology ,Proteomics ,Juxtacrine signalling ,Cell biology ,Extracellular matrix ,Paracrine signalling ,medicine.anatomical_structure ,Pathway (interactions) ,Immunology ,medicine ,Receptor ,Autocrine signalling ,Organ system - Abstract
Publisher Summary This chapter gives an overview on cell–cell and cell–matrix interactions. Cell signaling pathways form extensive interactive networks that results in the distinctive features distinguishing many of the response properties of specific tissues and organs. An idea that chemical signals may be provided locally or regionally in tissues by a group of mechanisms that have become known as paracrine, autocrine, intracrine, and juxtacrine interactions constitutes the means for regulating tissue-specific signal responses by providing the needed signals only on a local basis. The localized signaling mechanisms can be conveniently sub-grouped into four types. Paracrine interactions induce signaling activities that occur from cell to cell within a given tissue or organ, rather than through the general circulation. This takes place as locally produced hormones or other small signaling molecules exit their cell of origin, and then, by diffusion or local circulation, act only regionally on other cells of a different type within that tissue. The local concentrations of paracrine signals can be quite high compared to the circulating levels, and thus can trigger effects by acting on low-affinity receptors or by supplying sufficient local signals to bind to high-affinity receptors even when the circulating level of a molecule that produces a similar signal is too low to do so. Two mechanisms that further extend cell signaling beyond the action of circulating messengers include juxtacrine signals, in which the signaling entity (receptor ligand) is not soluble but is membrane-bound on one cell, and is delivered by cell–cell physical approximation to the cell bearing the receptor, and intracrine signals, in which both receptor and ligand are expressed intracellularly and signals are generated without external stimuli.
- Published
- 2010
26. Isolation and characterization of the methionine aminopeptidase from porcine liver responsible for the co-translational processing of proteins
- Author
-
Ralph A. Bradshaw and R L Kendall
- Subjects
Alanine ,chemistry.chemical_classification ,Methionine ,Molecular mass ,Stereochemistry ,Methionyl aminopeptidase ,Peptide ,Cell Biology ,Biology ,Biochemistry ,chemistry.chemical_compound ,Enzyme ,chemistry ,Methionyl Aminopeptidases ,Molecular Biology ,Histidine - Abstract
A methionine aminopeptidase that specifically removes methionine residues from peptides with amino-terminal sequences of Met-Ala-, Met-Val-, Met-Ser-, Met-Gly-, and Met-Pro- but not Met-Leu- or Met-Lys- has been isolated to homogeneity from porcine liver by a procedure involving five chromatographic steps. The enzyme, whose specificity matches that predicted for the entity responsible for the co-translational amino-terminal processing of nascent polypeptide chains, has a measured molecular mass of 70,000 Da by SDS-polyacrylamide electrophoresis and 67,000 Da by gel chromatography (under nondenaturing conditions), suggesting the native molecule is a monomer. It is activated by Co2+ and inhibited by beta-mercaptoethanol and EDTA. With octapeptide substrates related to the amino-terminal portion of the beta-chain of human hemoglobin (with a histidine in position 3), the enzyme had a pH optimum of 6.0. With a synthetic peptide devoid of histidine, it showed no pH dependence from 6.0 to 8.0. This sensitivity may be due to the propensity of peptides with histidine in the third position to bind divalent cations such as Co2+. The measured Km and kappa cat values were affected by residues in the second position. The peptide corresponding to the natural sequence (Met-Val-His-) gave a kappa cat/Km value of 260 mM-1 s-1; substitution of alanine in the second position raised the kappa cat/Km to 1523 mM-1 s-1, but substitution of proline lowered the value to 130. The effects are primarily on the kappa cat. The substitution of proline (for histidine) in the third position, the mutation found in hemoglobin Long Island, prevents the removal of the methionine residue, as occurs with the mutant protein. The porcine liver enzyme is similar to methionine aminopeptidases isolated from Escherichia coli, Salmonella typhimurium, and yeast in that it also is stimulated by Co2+. However, it is much larger than these enzymes and differs somewhat in specificity, particularly with the yeast enzyme.
- Published
- 1992
27. Nerve growth factor nonresponsive pheochromocytoma cells: altered internalization results in signaling dysfunction
- Author
-
Ralph A. Bradshaw and DD Eveleth
- Subjects
medicine.medical_specialty ,media_common.quotation_subject ,Receptors, Cell Surface ,Receptors, Nerve Growth Factor ,Biology ,PC12 Cells ,Cell surface receptor ,Epidermal growth factor ,Internal medicine ,medicine ,Animals ,Nerve Growth Factors ,Receptor ,Internalization ,media_common ,Binding Sites ,Epidermal Growth Factor ,Cell Biology ,Articles ,Endocytosis ,Cell biology ,Nerve growth factor ,Endocrinology ,nervous system ,Cell culture ,Trk receptor ,Signal transduction ,Signal Transduction - Abstract
Variant rat pheochromocytoma (PC12) cells which fail to respond to nerve growth factor (NGF) (PC12nnr5) (Green, S. H., R. E. Rydel, J. L. Connoly, and L. A. Greene. 1986. J. Cell Biol. 102:830-843) bind NGF at both high and low affinity sites. Although still undefined at the molecular level, these have been referred to as type I (high) and type II (low) receptors. They are apparently composed of two membrane-bound proteins, p75 and the protooncogene trk, both of which bind NGF, and apparently contribute singularly or in concert to the two observed affinities, and to the promotion of the NGF effects. In native PC12 cells, only the high affinity receptors are apparently capable of mediating internalization and degradation. PC12nnr5 cells also display type I binding, but the subsequent internalization is not the same fashion as in the parental cell line, nor is it subjected to lysosomal degradation. Rather it is initially sequestered during the first 15 min, and is eventually released intact into the medium. In contrast, EGF is bound, internalized, and degraded by PC12nnr5 cells, albeit less efficiently than in the parent cells. These observations argue that the defect(s) preventing the PC12nnr5 variants from responding to NGF prevents competent internalization, which in the case of NGF, may be required for the full expression of activity. The absence of trk, as one alteration in PC12nnr5 cells (Loeb, D. M., J. Maragos, D. Martin-Zanca, M. V. Chao, L. F. Parada, and L. A. Greene. 1991. Cell. 66:961-966), is consistent with this conclusion.
- Published
- 1992
28. Differential induction of primary-response (TIS) genes in PC12 pheochromocytoma cells and the unresponsive variant PC12nnr5
- Author
-
David Eveleth, Joseph G. Altin, Ralph A. Bradshaw, Harvey R. Herschman, D A Kujubu, and Simona Raffioni
- Subjects
medicine.medical_specialty ,Carbachol ,Neurite ,Basic fibroblast growth factor ,Cell Biology ,Biology ,Biochemistry ,Cell biology ,chemistry.chemical_compound ,Endocrinology ,Nerve growth factor ,chemistry ,Cell culture ,Internal medicine ,medicine ,biology.protein ,Phorbol ,Signal transduction ,Molecular Biology ,medicine.drug ,Neurotrophin - Abstract
As a measure of the transmembrane signals that they transduce, two neurotrophic agents, nerve growth factor (NGF) and basic fibroblast growth factor (bFGF), and the muscarinic agonist carbachol were compared for their ability to induce TIS (tetradecanoyl phorbol acetate-inducible sequences) transcripts, representing a family of immediate early response genes, in the rat pheochromocytoma cell line PC12 and the morphologically unresponsive variant PC12nnr5. Three genes, TIS1 (also designated NGFIB), TIS8 (also designated NGFIA), and TIS21, induced in these cells by NGF (Kujubu, D.A., Lim, R.W., Varnum, B.C., and Herschman, H.R. (1987) Oncogene 1, 257-262, 1987), are also induced by bFGF and carbachol. In native PC12 cells the level of expression of TIS8 and TIS21 is similar for all three stimuli, as well as for tetradecanoyl phorbol acetate (TPA). In contrast, the induction of TIS1 by NGF and TPA is slight and is only just detectable after stimulation by bFGF, but is strong for carbachol. Thus, although all of these agents can stimulate protein kinase (PK-C), at least one TIS gene can apparently be differentially regulated by these ligands, suggesting that alternative signaling pathways must also exist. In keeping with this view, bFGF, and to a lesser degree NGF, can elicit a TIS gene response in PC12 cells in which PK-C has been down-regulated with TPA. The response to carbachol (and TPA) is effectively blocked under these conditions. Since both NGF and bFGF stimulate neurite outgrowth in such cells, PK-C is apparently not essential, i.e. does not represent the sole mechanism, for signal transduction leading to modulation of gene expression for these factors. Consistent with this model, putative protein kinase inhibitors, K252a and sphingosine, did not inhibit the TIS gene responses to bFGF. However, these agents also failed to block TIS gene responses to carbachol and TPA indicating that they were ineffective as PK-C inhibitors under these conditions. The NGF-induced response was, however, blocked by K252a indicating a unique step in the mechanism of this factor not shared by the other ligands. Sphingosine did not block TIS induction with NGF. The mutant cell line PC12 nnr5 does not respond morphologically to either NGF or bFGF. However, TIS gene responses to bFGF are unaffected, whereas those to NGF are completely abolished. The response to TPA is altered quantitatively but not qualitatively; the induction by carbachol is largely eliminated, apparently as a result of a 90% reduction in muscarinic receptors.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1991
29. Identification and initial characterization of an autocrine pheromone receptor in the protozoan ciliate Euplotes raikovi
- Author
-
Pierangelo Luporini, Ralph A. Bradshaw, Claudio Ortenzi, and Cristina Miceli
- Subjects
Electrophoresis ,Mating type ,Receptors, Peptide ,Protozoan Proteins ,Receptors, Cell Surface ,Biology ,Binding, Competitive ,Medical and Health Sciences ,Pheromones ,Cell membrane ,Competitive ,Aldesleukin ,Receptors ,medicine ,Animals ,Ciliophora ,Binding site ,Autocrine signalling ,Receptor ,Gel electrophoresis ,Polyacrylamide Gel ,Membrane Proteins ,Articles ,Cell Biology ,Binding ,Biological Sciences ,Molecular biology ,Molecular Weight ,Kinetics ,A-site ,medicine.anatomical_structure ,Biochemistry ,Receptors, Mating Factor ,Cell Surface ,Peptide ,Electrophoresis, Polyacrylamide Gel ,Peptides ,Mating Factor ,Transcription Factors ,Developmental Biology - Abstract
The polypeptide pheromone Er-1, purified from the ciliate Euplotes raikovi of mating type I and genotype mat-1/mat-1, was iodinated with 125I-Bolton-Hunter reagent to a sp act of 0.45-0.73 mu Ci/microgram of protein. This preparation of 125I-Er-1 bound specifically to high affinity binding sites on the same cells of mating type I. Binding of 125I-Er-1 occurred with an apparent Kd of 4.63 +/- 0.12 X 10(-9) M in cells in early stationary phase. It was estimated that these cells carry a total number of approximately 5 X 10(7) sites/cell, with a site density that falls in the range of 1,600-1,700/microns 2 of cell surface. Unlabeled Er-1, other homologous pheromones such as Er-2 and Er-10, antibodies specific for Er-1, and human IL-2 were shown to act as effective inhibitors of specific binding of 125I-Er-1 to mating type I cells. The "autocrine" nature of the identified specific high affinity binding sites for Er-1 was further substantiated by cross-linking experiments. These experiments revealed that mating type-I cell membranes contain one protein entity of Mr = 28,000 that is capable of reacting specifically with the homodimeric native form of Er-1.
- Published
- 1990
30. Reciprocal Modulation of Astrocyte Stellation by Thrombin and Protease Nexin-1
- Author
-
Ralph A. Bradshaw, David Gurwitz, Kathleen P. Cavanaugh, and Dennis D. Cunningham
- Subjects
Plasmin ,medicine.medical_treatment ,Hirudin ,Cell Count ,Receptors, Cell Surface ,Biology ,Ornithine Decarboxylase ,Biochemistry ,Amyloid beta-Protein Precursor ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Thrombin ,1-Methyl-3-isobutylxanthine ,medicine ,Animals ,Cells, Cultured ,Protease ,Forskolin ,Dose-Response Relationship, Drug ,Osmolar Concentration ,Cell biology ,Protease Nexins ,medicine.anatomical_structure ,Bucladesine ,chemistry ,Astrocytes ,Stellation ,Carrier Proteins ,circulatory and respiratory physiology ,medicine.drug ,Astrocyte ,Discovery and development of direct thrombin inhibitors - Abstract
When cultured astroglia are treated with agents that elevate intracellular cyclic AMP, they become process-bearing stellate cells and resemble differentiated astrocytes in vivo. Thrombin rapidly reversed the stellation induced by dibutyryl cyclic AMP, forskolin, or isoproterenol in cultured rat astrocytes; half-maximal and maximal effects occurred at 0.5 and 8 pM, respectively. The proteolytic activity of thrombin was required for stellation reversal, as thrombin derivatized at its catalytic site serine with a diisopropylphospho group was inactive. Two thrombin inhibitors, protease nexin-1 and hirudin, blocked and reversed the effect of thrombin. The stellation reversal effect of thrombin was specific, as 300–1,000-fold higher concentrations of other serine proteinases, including plasmin, urokinase, trypsin, and T cell serine proteinase-1, were ineffective. Thrombin is a mitogen for astrocytes at concentrations in excess of 30 pM. Thrombin increased both cell number and ornithine decarboxylase activity, an early marker for mitogenic stimulation, in astrocyte cultures. The lowest thrombin concentrations that completely reversed astrocyte stellation, however, did not increase ornithine decarboxylase activity. Moreover, several other mitogens for astrocytes did not reverse dibutyryl cyclic AMP-induced stellation. Thus, the stellation reversal effect of thrombin is distinct from the mitogenic response.
- Published
- 1990
31. How I became a biochemist
- Author
-
Ralph A. Bradshaw
- Subjects
0303 health sciences ,Basketball ,4. Education ,media_common.quotation_subject ,030302 biochemistry & molecular biology ,Clinical Biochemistry ,Media studies ,Pity ,Family tree ,Cell Biology ,History, 20th Century ,Biochemistry ,History, 21st Century ,United States ,Syllabus ,03 medical and health sciences ,Nothing ,Genetics ,Meaning (existential) ,Chemistry (relationship) ,Molecular Biology ,Curriculum ,030304 developmental biology ,media_common - Abstract
One of the more interesting aspects of doing genealogical research on ones family is discovering what your ancestors did for a living. Having fairly successfully retraced my family tree for quite a few generations and, in some places, back nearly 400 years, I was somewhat disappointed to learn that there was very little inclination toward science to be found in my roots. Excepting the odd pirate or cattle rustler, they were pretty much a mundane collection of farmers, merchants and laborers with a few carpenters (particularly ship builders) thrown in. Thus, I can’t cite any genetic evidence that my choice of biochemistry as a pursuit was in any way preordained or even predictable. There is, however, precedent for ‘talent’ appearing (and disappearing) from generation to generation in our family – my father was an accomplished pianist/organist (although not his main profession) and his three sons, four grandchildren, and five great grandchildren seemingly did not inherit much if any of this gift. I was born in Dorchester, Massachusetts (a part of the City of Boston) in 1941, where my father had grown up, and was educated ‘across the river’ in the Milton public school system, the town where my mother had been raised. In retrospect, my formative training was sound but uninspired and was characterized by fairly provincial instruction (my mother and her sisters had many of the same teachers that I had, with syllabi that didn’t change much). With a plethora of private and parochial schools in the area, there was also something of a ‘brain drain’ with a disproportionate number of the better students, who provide stimulation and competition in the classroom, siphoned away. Since group instruction notoriously targets the middle level of a class, the combination of these factors allowed me to pass through junior and senior high school all too facilely for my own good. There would be a price to pay for this later. My high school curriculum was the standard ‘collegebound’ course, meaning biology in the 10th grade, chemistry in the 11th and physics in the 12th with nothing that remotely smacked of biochemistry anywhere to be found. Of these courses, chemistry was by far the most interesting (and probably the best taught) although frankly I can’t recall having any revelations during this class that this was what I wanted to pursue as a career choice. And it was by no means certain that I would go to college. Neither of my parents had done so and it wasn’t until my older brother (by 3þ years) entered college did the idea begin to take hold that I might go too. I also can’t lay claim to any significant self discovery events. Indeed I was far more interested in sports (particularly baseball and basketball) for which I had some modest talents but certainly no extraordinary gifts. I may have had a chemistry set (I frankly can’t remember) but if I did, I didn’t blow up myself, my brothers (more’s the pity) or the cat, discover any new elements or otherwise perform any of the types of experiments that are the stuff of child protegees. Received 2 May 2006; accepted 8 May 2006 Address correspondence to: Ralph A. Bradshaw, Department of Physiology & Biophysics, University of California, Irvine, CA 92697, USA. E-mail: rablab@uci.edu IUBMBLife, 58(8): 495 – 498, August 2006
- Published
- 2006
32. From proteins to proteomics
- Author
-
Ralph A. Bradshaw and Alma L. Burlingame
- Subjects
Proteomics ,Spectrometry, Mass, Electrospray Ionization ,Proteome ,Clinical Biochemistry ,Protein Array Analysis ,Genomics ,Computational biology ,Biology ,Bioinformatics ,Biochemistry ,Genome ,Protein–protein interaction ,Genetics ,Animals ,Humans ,Disease markers ,Molecular Biology ,Two-dimensional gel electrophoresis ,Proteins ,Cell Biology ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Identification (biology) ,Protein Binding - Abstract
During the second half of the 20th century, biochemistry and subsequently molecular biology blossomed into the core upon which all biological and biomedical sciences now depend. A major part of these closely related disciplines has been the study of the structure and function of proteins and the diverse biological functions that they perform. Early experimentation necessarily focused on individual entities, selected mainly for their activities, but as technology improved there developed a tendency to look at proteins as larger, interactive groups or clusters. Spurred by the recent exponential production of genomic sequence data for a rapidly increasing number of species, protein chemistry has now evolved into a new discipline, proteomics. In addition to embracing the methods and approaches that have served protein scientists well in the past, it includes, and is perhaps best defined by, high-throughput analyses based in large part on 2D gel electrophoresis, MALDI and ESI mass spectrometry and combinatorial arrays. Proteomic targets include the identification of all genome products and a mapping of their interactions and expression profiles. These hold great promise for the identification of disease markers and drug targets, but are not without their challenges and pitfalls.
- Published
- 2005
33. Autocrine mitogenic activity of pheromones produced by the protozoan ciliate Euplotes raikovi
- Author
-
Ralph A. Bradshaw, Adriana Vallesi, Giovanna Giuli, and Pierangelo Luporini
- Subjects
Ciliate ,Cell type ,Multidisciplinary ,biology ,Ecology ,Molecular Sequence Data ,Protozoan Proteins ,Euplotes ,Membrane Proteins ,biology.organism_classification ,Pheromones ,Cell biology ,Paracrine signalling ,Sex pheromone ,Pheromone activity ,Animals ,Pheromone ,Amino Acid Sequence ,Mating ,Autocrine signalling ,Cell Division - Abstract
Diffusible polypeptide pheromones (formerly referred to as mating-type factors, sex factors or gamones), which distinguish otherwise morphologically identical vegetative cell (mating) types from one another, are produced by some species of ciliates. Their most striking effect can be observed by exposing cells of one type to a pheromone secreted by another co-specific cell type. In the presence of this 'non-self' signal, these cells interrupt their vegetative life to unite temporarily in mating pairs. Thus ciliate pheromones have traditionally been associated only with mating induction. However, the identification of autocrine pheromone receptors suggests a broader role, which is supported by the hypothesis that ciliates evolved their mating-type mechanism for pursuing self-recognition. We now report studies, in the cosmopolitan marine sand-dwelling protozoan ciliate Euplotes raikovi, demonstrating that these molecules promote the vegetative reproduction (mitogenic proliferation or growth) of the same cells from which they originate. As, understandably, such autocrine pheromone activity is primary to that of targeting and inducing a foreign cell to mate (paracrine functions), this finding provides an example of how the original function of a molecule can be obscured during evolution by the acquisition of a new one.
- Published
- 1995
34. PC12 cell activation by epidermal growth factor receptor: role of autophosphorylation sites
- Author
-
Selena Larkin, Ralph A. Bradshaw, Darren R. Tyson, and Yousuke Hamai
- Subjects
Cellular differentiation ,medicine.medical_treatment ,Receptors, Prostaglandin ,Ligands ,PC12 Cells ,Developmental Neuroscience ,Epidermal growth factor ,medicine ,Neurites ,Animals ,Humans ,Epidermal growth factor receptor ,Phosphorylation ,Platelet-Derived Growth Factor ,biology ,Epidermal Growth Factor ,Chemistry ,Chimera ,Growth factor ,Autophosphorylation ,Phosphotransferases ,Cell Differentiation ,Cell biology ,Rats ,Enzyme Activation ,ErbB Receptors ,biology.protein ,GRB2 ,Signal transduction ,Platelet-derived growth factor receptor ,Developmental Biology - Abstract
PC12 cells have been used as a model system for neuronal differentiation due to their ability to alter their phenotype to a sympathetic neuron-like cell in response to nerve growth factor or fibroblast growth factor. Under some conditions, epidermal growth factor (EGF) can also induce PC12 cells to differentiate. To study signaling from the EGF receptor without the confounding effects of endogenous EGF receptors we generated a chimeric receptor comprised of the ectodomain of platelet-derived growth factor (PDGF) receptor in-frame with the transmembrane and cytoplasmic domains of EGF receptor, termed PER. Expression of PER in PC12 cells confers the ability of PDGF to induce differentiation whereas PDGF has no effect on untransfected PC12 cells. This response is kinase activity-dependent since a kinase-deficient mutant (K721M) fails to induce differentiation in response to PDGF. Mutation of five tyrosine residues that are autophosphorylated in response to EGF either individually or in combination had minimal effects on the ability of these receptors to induce morphological PC12 cell differentiation. The PER mutant with all five autophosphorylation sites mutated to phenylalanine (5YF) was equivalently capable of interacting with several important signaling molecules, including Shc, Grb2, Gab1, phospholipase Cgamma, and Cbl. Furthermore, both the phosphatidylinositol 3-kinase (PI3K)/Akt and Ras/Erk pathways were activated in a sustained manner when PER or 5YF-expressing cells were stimulated with PDGF. Our results show that the five autophosphorylation sites in the extra-kinase C-terminal domain of EGFR are not required for the ability of EGFR to induce morphological differentiation of PC12 cells.
- Published
- 2003
35. Transmembrane Receptor Oligomerization
- Author
-
Darren R. Tyson and Ralph A. Bradshaw
- Subjects
Janus kinase 1 ,Chemistry ,Class C GPCR ,Immune receptor ,Biology ,Glycoprotein 130 ,Ligand (biochemistry) ,Rhodopsin-like receptors ,Receptor tyrosine kinase ,Transmembrane protein ,Cell biology ,Chemokine receptor ,Biochemistry ,biology.protein ,Biophysics ,Receptor ,Flux (metabolism) ,Function (biology) ,G protein-coupled receptor - Abstract
Publisher Summary This chapter provides a brief description on how oligomerization is utilized by the major cell transmembrane receptor classes. The cell-surface receptors possessing intrinsic tyrosine kinase activity (RTK) minimally require a dimeric state for their full activity. RTK dimerization occurs as a result of ligand binding, and the ligand-bound monomer then recruits another monomer into the complex, allowing for the interaction of the kinase domains and their subsequent transphosphorylation. The initial phosphorylation events usually occur on the activation loop and help stabilize the open conformation, thereby allowing access of adenosine triphosphate (ATP) and substrate to their respective binding sites within the kinase domain. The cytokine receptors are composed of numerous kinds of transmembrane receptors that are characterized by conserved patterns of amino acid residues in their ectodomains and the lack of enzymatic activity within their endodomains. They associate with non-receptor tyrosine kinases such as the Janus kinases (JAK) or Src family kinases. The class I cytokine receptors are the growth hormone receptor (GHR) and erythropoietin receptor (EPOR) that function as homodimers. EPOR appears to exist at the cell surface as an inactive dimer and this state is mediated by the transmembrane domain. Type II cytokine receptors consist primarily of receptors for interferons and interleukin-10 (IL-10). The receptors for interferon-γ (IFNγR) and IL-10 (IL-10R) have a similar structure in that they are each composed of two type 1 receptors and two type 2 receptors forming a heterotetramer.
- Published
- 2003
36. The protease inhibitory properties of the Alzheimer's beta-amyloid precursor protein
- Author
-
Melih Arici, R W Blacher, H F Dovey, Michael Blaber, William C. Mobley, Ralph A. Bradshaw, Pamela J. Ward, Peter Seubert, Ivan Lieberburg, and Sukanto Sinha
- Subjects
Cathepsin ,Protease ,biology ,Chemistry ,medicine.medical_treatment ,Cell Biology ,Trypsin ,Biochemistry ,Fusion protein ,Thrombin ,Protein A/G ,medicine ,biology.protein ,Amyloid precursor protein ,Kunitz domain ,Molecular Biology ,medicine.drug - Abstract
We have expressed the 57-amino acid Kunitz domain of the Alzheimer's beta-amyloid precursor protein (APP751) as a bacterial fusion protein. The protease inhibitory properties of the purified fusion protein, BX9, were virtually identical in all respects tested to those of purified secreted APP751. Both proteins strongly inhibited pancreatic trypsin (Kis = 0.2 and 0.3 nM) and less well epidermal growth factor-binding protein (Kis = 1 and 3.5 nM), alpha-chymotrypsin (Kis = 3 and 6 nM), and the gamma-subunit of nerve growth factor (Kis = 8 and 9 M). Neither protein appreciably inhibited plasma and pancreatic kallikreins, thrombin, lung tryptase, neutrophil elastase, or cathepsin G. The remarkable similarity of the protease inhibitory profile of BX9 to that of secreted APP751 suggests that proper intramolecular disulfide bond formation has occurred in the bacterial fusion protein and leads to the conclusion that the amyloid precursor protein Kunitz domain is a relatively specific inhibitor of only a few trypsin-like arginine esterases.
- Published
- 1990
37. Chimeras of the native form or achondroplasia mutant (G375C) of human fibroblast growth factor receptor 3 induce ligand-dependent differentiation of PC12 cells
- Author
-
Simona Raffioni, Ralph A. Bradshaw, John J. Wasmuth, and Leslie M. Thompson
- Subjects
medicine.medical_treatment ,Recombinant Fusion Proteins ,Biology ,PC12 Cells ,Achondroplasia ,Structure-Activity Relationship ,Growth factor receptor ,medicine ,Neurites ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 3 ,Growth factor receptor inhibitor ,Receptors, Platelet-Derived Growth Factor ,Phosphorylation ,Phosphotyrosine ,Molecular Biology ,Insulin-like growth factor 1 receptor ,Growth factor ,Cell Differentiation ,Cell Biology ,Fibroblast growth factor receptor 4 ,Fibroblast growth factor receptor 3 ,Protein-Tyrosine Kinases ,Molecular biology ,Receptors, Fibroblast Growth Factor ,Rats ,Cartilage ,Fibroblast growth factor receptor ,biology.protein ,Platelet-derived growth factor receptor ,Research Article - Abstract
Mutations in the gene for human fibroblast growth factor receptor 3 (hFGFR3) cause a variety of skeletal dysplasias, including the most common genetic form of dwarfism, achondroplasia (ACH). Evidence indicates that these phenotypes are not due to simple haploinsufficiency of FGFR3 but are more likely related to a role in negatively regulating skeletal growth. The effects of one of these mutations on FGFR3 signaling were examined by constructing chimeric receptors composed of the extracellular domain of human platelet-derived growth factor receptor beta (hPDGFR beta) and the transmembrane and intracellular domains of hFGFR3 or of an ACH (G375C) mutant. Following stable transfection in PC12 cells, which lack platelet-derived growth factor (PDGF) receptors, all clonal cell lines, with either type of chimera, showed strong neurite outgrowth in the presence of PDGF but not in its absence. Antiphosphotyrosine immunoblots showed ligand-dependent autophosphorylation, and both receptor types stimulated strong phosphorylation of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase, an event associated with the differentiative response of these cells. In addition, ligand-dependent phosphorylation of phospholipase Cgamma and Shc was also observed. All of these responses were comparable to those observed from ligand activation, such as by nerve growth factor, of the native PC12 cells used to prepare the stable transfectants. The cells with the chimera bearing the ACH mutation were more rapidly responsive to ligand with less sustained MAPK activation, indicative of a preactivated or primed condition and consistent with the view that these mutations weaken ligand control of FGFR3 function. However, the full effect of the mutation likely depends in part on structural features of the extracellular domain. Although FGFR3 has been suggested to act as a negative regulator of long-bone growth in chrondrocytes, it produces differentiative signals similar to those of FGFR1, to which only positive effects have been ascribed, in PC12 cells. Therefore, its regulatory effects on bone growth likely result from cellular contexts and not the induction of a unique FGFR3 signaling pathway.
- Published
- 1997
38. Nerve growth factor receptors
- Author
-
Ralph A. Bradshaw and Hubert Hondermarck
- Subjects
MAPK/ERK pathway ,Nerve growth factor ,nervous system ,STAT protein ,Low-affinity nerve growth factor receptor ,Biology ,Tropomyosin receptor kinase A ,Signal transduction ,Receptor ,Transcription factor ,Cell biology - Abstract
Publisher Summary This chapter discusses the nerve growth factor (NGF) receptors. NGF recognition by specific cell surface receptors is provided by two independent entities. These differ in both structure and function. TrkA clearly induces tyrosine phosphorylations that are important in many responses, including differentiation in PC12 cells, and is the principal effector in neuronally responsive cells. TrkE may serve a similar function in non-neuronal cells, although its rigorous identity as a NGF-specific receptor remains to be satisfied. p75, although more abundant and widely distributed, is less understood in physiological terms. Evidence supports the view that it can induce some intracellular signals and it plays a role in modulating TrkA activity. However, such effects seem more related to overall cellular responses than they are to changes in molecular events—that is, modification of signal transduction cascades. Non-signaling role, have also been suggested. In the sense that the same signaling pathway may be utilized in different cells to different ends, it is also likely that other pathway, not directly induced by NGF via TrkA, may produce NGF-like responses. A PC12 cell variant (PC12-E2), responds to IL-6 in a manner quite similar to NGF, but it does not activate the Ras- mitogen-activated protein kinase (MAPK) pathway. It is likely that a pathway involving signal transducer and activator of transcription (STAT) proteins, transcription factors that are activated in the cytoplasm by receptor mediated phosphorylations and translocated to the nucleus, are involved. This emphasizes the multiplicity of mechanisms available to accomplish similar biological phenomena.
- Published
- 1997
39. Induction of neurite outgrowth by interleukin-6 is accompanied by activation of Stat3 signaling pathway in a variant PC12 cell (E2) line
- Author
-
Yvonne Y. Wu and Ralph A. Bradshaw
- Subjects
STAT3 Transcription Factor ,Neurite ,Basic fibroblast growth factor ,Molecular Sequence Data ,Biology ,Fibroblast growth factor ,Biochemistry ,PC12 Cells ,Stat3 Signaling Pathway ,chemistry.chemical_compound ,Neurites ,Animals ,Humans ,Nerve Growth Factors ,Phosphorylation ,STAT3 ,Molecular Biology ,Cell Nucleus ,Base Sequence ,Interleukin-6 ,Tyrosine phosphorylation ,Cell Biology ,DNA ,Molecular biology ,Cell biology ,Rats ,DNA-Binding Proteins ,Fibroblast Growth Factors ,Kinetics ,Nerve growth factor ,STAT1 Transcription Factor ,nervous system ,chemistry ,biology.protein ,Trans-Activators ,Tyrosine ,Leukemia inhibitory factor ,Protein Kinases ,Acute-Phase Proteins ,Protein Binding ,Signal Transduction - Abstract
PC12-E2 cells, a stable variant subcloned from native cell populations, produce neurites in a rapid, transcription-independent manner upon exposure to nerve growth factor (NGF) or basic fibroblast growth factor (bFGF). They also give a similar morphological response to interleukin-6 (IL-6), which is, however, transcription-dependent and with a slower onset, a phenomenon basically not observed in native PC12 cells. The response profile of PC12-E2 cells to NGF and bFGF is similar to that observed for native PC12 cells pre-exposed (primed) to NGF, and such cells also respond to IL-6 in a fashion indistinguishable from PC12-E2 cells. Mechanistically, NGF and bFGF induce a sustained phosphorylation and activation of ERK1 and ERK2 in both cells, while IL-6 produces only a transient and weak tyrosine phosphorylation. However, it does stimulate a prolonged and biphasic tyrosine phosphorylation and nuclear translocation of Stat3 (signal transducers and activators of transcription 3; at least 24 h) and, to a lesser extent, Stat1. Gel shift and supershift analyses confirm that IL-6 predominantly activates Stat3 (and some Stat1) and stimulates sis-inducible element binding activity. Other members of the same cytokine subfamily, including ciliary neurotrophic factor and leukemia inhibitory factor, also cause a transient initial phase of tyrosine phosphorylation and activation of Stat1 and Stat3 (up to 1 h) but fail to stimulate a second phase of response and do not produce significant neurites. These results suggest that sustained signaling of either STAT or ERK pathways in PC12-E2 cells leads to induction of neuronal differentiation. However, only the latter is effective in native PC12 cells as the activation of Stat3 and Stat1 in native PC12 cells by IL-6 fails to induce neuronal differentiation. Thus, the response of PC12-E2 cells to IL-6 suggests the constitutive expression of a required factor(s) for differentiation, that is induced in native PC12 cells by NGF or bFGF (possibly by ERK activation), but not by IL-6 via Janus kinase/STAT activation. This factor(s), which has a sufficient half-life to allow primed cells to remain responsive to IL-6 for several days, is necessary but not sufficient for differentiation (as measured by neurite proliferation) to occur.
- Published
- 1996
40. Synergistic induction of neurite outgrowth by nerve growth factor or epidermal growth factor and interleukin-6 in PC12 cells
- Author
-
Yvonne Y. Wu and Ralph A. Bradshaw
- Subjects
MAPK/ERK pathway ,Neurite ,Nerve Tissue Proteins ,Biochemistry ,PC12 Cells ,chemistry.chemical_compound ,GAP-43 Protein ,Epidermal growth factor ,Neurites ,Animals ,Nerve Growth Factors ,RNA, Messenger ,Phosphorylation ,STAT3 ,Molecular Biology ,Membrane Glycoproteins ,biology ,Epidermal Growth Factor ,Kinase ,Interleukin-6 ,Membrane Proteins ,Tyrosine phosphorylation ,Drug Synergism ,Cell Biology ,Molecular biology ,Rats ,Nerve growth factor ,nervous system ,chemistry ,biology.protein ,Microtubule Proteins ,Tyrosine ,Carrier Proteins ,Tyrosine kinase ,Protein Binding - Abstract
Native PC12 cells respond differentially to nerve growth factor (NGF) but not interleukin-6 (IL-6); PC12-E2 cells, a stable variant, respond to both stimuli (and more rapidly to NGF). Neither responds to epidermal growth factor (EGF). NGF primarily induces the RAS/extracellular signal-regulated kinase (ERK) pathway and IL-6 activates a JAK (Janus tyrosine kinase)/STAT (signal transducers and activators of transcription) response. EGF also stimulates RAS/ERK but in a transient manner. When either cell type is treated with combinations of NGF, EGF, and IL-6, at concentrations that produce modest or no response, a substantial augmentation of neurite outgrowth is observed. With PC12-E2 cells, a subthreshold concentration of IL-6 increases NGF response by approximately 2-3-fold after 1-2 days; the increase with EGF is more pronounced. Native PC12 cells show even greater synergistic effects with NGF and IL-6. The most dramatic effect was observed with low levels of EGF, where IL-6 increased the percentage of responsive cells from zero to approximately 60% after 3 days. In addition, two neural-specific transcripts, GAP-43 and SCG-10, are synergistically increased by the combinations of growth factors. Importantly, IL-6 does not enhance ERK phosphorylation in the presence of either NGF or EGF. In contrast, NGF and EGF, in the presence or absence of IL-6, cause mobility shifts of Stat3 that are consistent with serine phosphorylations. Although these modifications do not lead to activation and translocation by themselves, in the presence of the tyrosine phosphorylation induced by IL-6, they may play a role in the synergistic responses. These observations suggest a differentially regulated two-stage mechanism for the differentiative response of PC12 cells to NGF.
- Published
- 1996
41. Src homologous and collagen (Shc) protein binds to F-actin and translocates to the cytoskeleton upon nerve growth factor stimulation in PC12 cells
- Author
-
Scott D. Patterson, Ralph A. Bradshaw, and Didier Thomas
- Subjects
Src Homology 2 Domain-Containing, Transforming Protein 1 ,Membrane ruffling ,Molecular Sequence Data ,Arp2/3 complex ,macromolecular substances ,Biology ,SH2 domain ,environment and public health ,Biochemistry ,PC12 Cells ,Oncogene Protein pp60(v-src) ,chemistry.chemical_compound ,Animals ,Amino Acid Sequence ,Nerve Growth Factors ,Cytoskeleton ,Molecular Biology ,Actin ,Cytochalasin D ,Adaptor Proteins, Signal Transducing ,Proteins ,Biological Transport ,Cell Biology ,Precipitin Tests ,Actins ,Cell biology ,Rats ,Adaptor Proteins, Vesicular Transport ,chemistry ,Shc Signaling Adaptor Proteins ,biology.protein ,Collagen ,biological phenomena, cell phenomena, and immunity ,hormones, hormone substitutes, and hormone antagonists ,Proto-oncogene tyrosine-protein kinase Src ,Protein Binding - Abstract
Immunoprecipitates of metabolically labeled PC12 cells consistently contained a 43-kDa protein that was associated with Shc, a signal-transducing protein with a single SH2 domain. Following affinity chromatography with immobilized recombinant glutathione S-transferase (GST)-Shc fusion protein, the 43-kDa protein was identified as actin by mass spectrometry and immunoblotting. Cosedimentation experiments using purified actin and GST-Shc showed that Shc binds directly to F-actin, confirming Shc-actin interaction in vivo. Various GST-truncated Shc fusion proteins were prepared and used in actin cosedimentation assays. Constructs containing the SH2 and collagen homology domains were not precipitated, and those containing the amino-terminal domain were. Thus, Shc-actin interactions do not occur in the region of tyrosine phosphorylation and leave the SH2 domain free to bind to other tyrosine-phosphorylated molecules. Although the major pool of Shc in unstimulated PC12 cells is soluble, two other pools are associated with the cytoskeleton and the submembranous cytoskeleton. Upon nerve growth factor stimulation, approximately 50% of the soluble Shc translocates to both cytoskeleton environments within 2 min, decreasing thereafter. When cells were pretreated with cytochalasin D, a drug that disrupts actin filaments, Shc translocation to the cytoskeleton was abolished. However, in the submembranous fraction, the Shc level was elevated in resting cells following cytochalasin D treatment. The kinetics of translocation, compared to mitogen-activated protein kinase activation, and the nature of the Shc-actin interaction suggest that the cytoskeletal association of Shc, induced by growth factors, may be related to membrane ruffling and actin fiber reorganization.
- Published
- 1995
42. Neuronal differentiation signals are controlled by nerve growth factor receptor/Trk binding sites for SHC and PLC gamma
- Author
-
Axel Choidas, Klaus Seedorf, Axel Ullrich, A Obermeier, Ralph A. Bradshaw, and Joseph Schlessinger
- Subjects
Molecular Sequence Data ,Receptors, Nerve Growth Factor ,Biology ,Transfection ,PC12 Cells ,General Biochemistry, Genetics and Molecular Biology ,Receptor tyrosine kinase ,Phosphatidylinositol 3-Kinases ,Cell surface receptor ,Animals ,Nerve Growth Factors ,Molecular Biology ,Neurons ,Platelet-Derived Growth Factor ,General Immunology and Microbiology ,Phospholipase C ,Base Sequence ,General Neuroscience ,Cell Differentiation ,Protein-Tyrosine Kinases ,Recombinant Proteins ,Cell biology ,Phosphotransferases (Alcohol Group Acceptor) ,Nerve growth factor ,Biochemistry ,nervous system ,Trk receptor ,Type C Phospholipases ,biology.protein ,Signal transduction ,Neurotrophin ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction ,Research Article - Abstract
Differentiation and survival of neuronal cell types requires the action of neurotrophic polypeptides such as nerve growth factor (NGF). In the central and peripheral nervous system and the phaeochromocytoma cell model PC12, NGF exerts its effects through the activation of the signalling capacity of Trk, a receptor tyrosine kinase (RTK) which upon interaction with NGF becomes phosphorylated on tyrosines and thereby acquires the potential to interact with signal-transducing proteins such as phospholipase C-gamma (PLC gamma), phosphatidylinositol-3'-kinase (PI3'-K) and SHC. Mutagenesis of the specific binding sites for these src homology 2 (SH2) domain-containing substrates within the Trk cytoplasmic domain suggests a non-essential function of PI3'-K and reveals a major role for the signal controlled by the SHC binding site at tyrosine 490 and a co-operative function of the PLC gamma-mediated pathway for neuronal differentiation of PC12 cells.
- Published
- 1994
43. Co-translational Modification, Stability and Turnover of Eukaryotic Proteins
- Author
-
Jose Sy, Hubert Hondermarck, Ralph A. Bradshaw, Richard L. Kendall, Stuart M. Arfin, and Albert E. Stewart
- Subjects
Folding (chemistry) ,Cellular activity ,Chemistry ,Porcine liver ,Biological activity ,Eukaryotic cell ,Function (biology) ,Cell biology - Abstract
Eukaryotic function is tightly controlled through the complex mechanisms that regulate transcriptional and translational events. These processes are variously augmented by co- and post-translational modifications that affect function, location and ultimately turnover for each protein. Among the least well understood aspects are stability, including the process of folding, and degradation of both normal and damaged proteins. Since proteolytic destruction of proteins is as important as synthesis in determining the level of a biological activity, it represents a major cellular activity whose dissection is essential for the full appreciation of the regulation of eukaryotic cell function.
- Published
- 1994
44. Nerve growth factor revisited
- Author
-
Judith Murray-Rust, Ralph A. Bradshaw, R. Lapatto, Tom L. Blundell, and Neil Q. McDonald
- Subjects
medicine.medical_specialty ,Protein Conformation ,Molecular Sequence Data ,Receptors, Nerve Growth Factor ,Biology ,Biochemistry ,Proto-Oncogene Mas ,Structure-Activity Relationship ,Protein structure ,Neurotrophic factors ,Internal medicine ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Nerve Growth Factors ,Receptor ,Molecular Biology ,Peptide sequence ,Cell biology ,Endocrinology ,Nerve growth factor ,Trk receptor ,biology.protein ,Tyrosine kinase ,Neurotrophin - Abstract
Recent studies on nerve growth factor have revealed important new insights into the structure, function and evolution of this prototypical neurotrophic factor. Some of its features are (1) it has a unique three-dimensional fold that has since been found in two other growth factors, (2) it uses the trk proto-oncogene product, which has a tyrosine kinase, as a receptor and (3) it shares homology with at least three other factors, now collectively called neurotrophins, which have a spectrum of target cells.
- Published
- 1993
45. Dipeptide inhibitors of ubiquitin-mediated protein turnover prevent growth factor-induced neurite outgrowth in rat pheochromocytoma PC12 cells
- Author
-
Ralph A. Bradshaw, Stuart M. Arfin, Jose Sy, and Hubert Hondermarck
- Subjects
Reticulocytes ,Neurite ,Cellular differentiation ,Proteolysis ,medicine.medical_treatment ,Biophysics ,Biology ,Biochemistry ,PC12 Cells ,Structure-Activity Relationship ,Reticulocyte ,Protein biosynthesis ,medicine ,Neurites ,Animals ,Nerve Growth Factors ,Molecular Biology ,Ubiquitins ,medicine.diagnostic_test ,Growth factor ,Protein turnover ,Proteins ,Cell Biology ,Dipeptides ,Recombinant Proteins ,Cell biology ,Rats ,Kinetics ,medicine.anatomical_structure ,nervous system ,Cell culture ,Fibroblast Growth Factor 2 - Abstract
Dipeptide inhibitors of the ubiquitin-dependent proteolysis pathway governed by N-terminal recognition (N-end rule) in reticulocyte lysates significantly suppress NGF- and bFGF-induced neurite outgrowth in rat pheochromocytoma PC12 cells, but do not cause retraction of already formed neurites. Peptides which do not inhibit proteolysis are also without effect on PC12 cell differentiation. Suppression of neurite outgrowth is readily reversible upon removal of the inhibitors. These data demonstrate a requirement for specific protein turnover in the process of neuron-like differentiation in PC12 cells and provide the first demonstration of a physiological role for the N-end rule.
- Published
- 1992
46. Activation of phosphatidylinositol 3-kinase by epidermal growth factor, basic fibroblast growth factor, and nerve growth factor in PC12 pheochromocytoma cells
- Author
-
Ralph A. Bradshaw and Simona Raffioni
- Subjects
medicine.medical_treatment ,Basic fibroblast growth factor ,Biology ,PC12 Cells ,chemistry.chemical_compound ,Phosphatidylinositol 3-Kinases ,Epidermal growth factor ,Cell surface receptor ,medicine ,Animals ,Growth factor receptor inhibitor ,Phosphatidylinositol ,Nerve Growth Factors ,Phosphotyrosine ,Multidisciplinary ,Epidermal Growth Factor ,Growth factor ,Phosphotransferases ,Tyrosine phosphorylation ,Cell biology ,Rats ,Enzyme Activation ,Kinetics ,chemistry ,Liver ,biology.protein ,Tyrosine ,Fibroblast Growth Factor 2 ,Mitogens ,Platelet-derived growth factor receptor ,Cell Division ,Research Article ,Thymidine - Abstract
Epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), and nerve growth factor (NGF), which stimulate the phosphorylation of proteins on tyrosine in PC12 cells, initiate these modifications through ligand-specific cell surface receptors that contain the causative tyrosine kinases. One apparent substrate for these enzymes is phosphatidylinositol 3-kinase (PI 3-kinase), an enzyme that phosphorylates the D-3 position of the inositol ring and associates with several protein tyrosine kinases, as indicated by the fact that it is immunoprecipitated from EGF-, bFGF-, and NGF-stimulated PC12 cells by an anti-phosphotyrosine antibody. All three growth factors increase immunoprecipitable PI 3-kinase activity after 2 min of addition at concentrations able to stimulate either mitogenic or neurotrophic responses in PC12 cells. The level of stimulation of PI 3-kinase activity by EGF, bFGF, and NGF is 15- to 20-fold, 2- to 3-fold, and 8- to 10-fold, respectively. Moreover, tyrosine phosphorylation of PI 3-kinase was detected in EGF-, bFGF-, and NGF-stimulated PC12 cells, and the amount of the phosphorylation correlated with the level of stimulation of enzyme activity. In contrast, phosphatidylinositol 4-kinase, which produces the inositol phospholipids cleaved by phospholipase C-gamma to yield diacylglycerol and inositol-1,4,5-trisphosphate, is not affected by these growth factors. The pattern of stimulation of PI 3-kinase does not correlate with the induction of neurite outgrowth but rather with the mitotic responses, suggesting that PI 3-kinase and its products may be more important for signaling in cell division than in trophic processes. However, the levels of phosphatidylinositol 3-phosphate do not coincide with the stimulation of [3H]thymidine incorporation by these growth factors, rendering its role in mitotic functions, at least in PC12 cells, also uncertain.
- Published
- 1992
47. Testing the in vivo role of protein kinase C and c-fos in neurite outgrowth by microinjection of antibodies into PC12 cells
- Author
-
Richard Wetts, Joseph G. Altin, Ralph A. Bradshaw, and K T Riabowol
- Subjects
Neurite ,Microinjections ,Basic fibroblast growth factor ,Biology ,Fibroblast growth factor ,c-Fos ,PC12 Cells ,Antibodies ,chemistry.chemical_compound ,Neurites ,Animals ,Nerve Growth Factors ,Molecular Biology ,Microinjection ,Protein kinase C ,Protein Kinase C ,Antibodies, Monoclonal ,Cell Biology ,Molecular biology ,Cell biology ,Rats ,Fibroblast Growth Factors ,Nerve growth factor ,chemistry ,biology.protein ,Signal transduction ,Proto-Oncogene Proteins c-fos ,Cell Division ,Research Article - Abstract
To define the molecular bases of growth factor-induced signal transduction pathways, antibodies known to block the activity of either protein kinase C (PKC) or the fos protein were introduced into PC12 cells by microinjection. The antibody against PKC significantly inhibited neurite outgrowth when scored 24 h after microinjection and exposure to nerve growth factor (NGF). Microinjection of antibodies to fos significantly increased the percentage of neurite-bearing cells after exposure to either NGF or basic fibroblast growth factor (bFGF) but inhibited the stimulation of DNA synthesis by serum, suggesting that in PC12 cells, fos is involved in cellular proliferation. Thus, activation of PKC is involved in the induction of neurite outgrowth by NGF, but expression of the fos protein, which is induced by both NGF and bFGF, is not necessary and inhibits neurite outgrowth.
- Published
- 1992
48. Localization of acidic fibroblast growth factor within the mouse brain using biochemical and immunocytochemical techniques
- Author
-
Jerry Di Salvo, Guillermo Giménez-Gallego, Kenneth A. Thomas, Richard S. Morrison, Ralph A. Bradshaw, Kim B. Seroogy, Sandra E. Loughlin, Philippe Cioff, and James H. Fallon
- Subjects
Male ,medicine.medical_specialty ,Cerebellum ,Clinical Biochemistry ,Immunocytochemistry ,Blotting, Western ,Molecular Sequence Data ,Hypothalamus ,Enzyme-Linked Immunosorbent Assay ,Biology ,Cross Reactions ,Reticular formation ,Basal Ganglia ,Mice ,Endocrinology ,Thalamus ,Antibody Specificity ,Internal medicine ,Cortex (anatomy) ,Pons ,medicine ,Animals ,Amino Acid Sequence ,Medulla ,Brain Chemistry ,Cerebral Cortex ,Medulla Oblongata ,Immune Sera ,Cell Biology ,Olfactory Bulb ,Cell biology ,medicine.anatomical_structure ,nervous system ,Spinal Cord ,Fibroblast Growth Factor 1 ,Brainstem ,Peptides - Abstract
The localization of acidic fibroblast growth factor (aFGF) in the male mouse brain was studied with biochemical and immunocytochemical techniques. Using two peptide-based aFGF antisera directed against independent epitopes, Western gel analysis of dissected brain demonstrated significant levels of aFGF immunoreactivity in the pons-medulla, hypothalamus and cerebellum. The cortex contained much less immunoreactivity. Consistent with the biochemical data, immunocytochemical analysis with the same two antisera demonstrated that aFGF immunoreactivity is localized in neuronal cell bodies in these regions. Numerous immunoreactive neurons were observed in the reticular formation of the pons and medulla, as well as in several other brainstem nuclei and areas. Immunoreactive neurons were also present in the lateral and medial hypothalamus, and some thalamic, subthalamic and epithalamic nuclei. In the basal ganglia, immunoreactive neurons were present in the amygdala and septum. Few intensely stained immunoreactive neurons were observed in the striatum, pallidum and neocortex. Limbic cortices contained more numerous immunoreactive neurons than neocortex. These results support the concept that aFGF is present in the brain, where it is heterogeneously distributed in neuronal cell bodies in regions involved in sensory, extrapyramidal motor, limbic and autonomic functions. The results are consistent with various neurotrophic, mitogenic, and neuromodulatory functions associated with aFGF in the mammalian central nervous system.
- Published
- 1992
49. Acidic fibroblast growth factor is present in regenerating limb blastemas of axolotls and binds specifically to blastema tissues
- Author
-
Didier Thomas, Susan V. Bryant, Bénoni Boilly, Kathleen P. Cavanaugh, Ralph A. Bradshaw, and Hubert Hondermarck
- Subjects
DNA Replication ,Neurite ,Mesenchyme ,medicine.medical_treatment ,Blotting, Western ,Enzyme-Linked Immunosorbent Assay ,Biology ,Sodium Chloride ,Fibroblast growth factor ,Ambystoma ,Binding, Competitive ,Culture Techniques ,medicine ,Animals ,Regeneration ,Binding site ,Molecular Biology ,Polysaccharide-Lyases ,integumentary system ,Chondroitin Lyases ,Growth factor ,Regeneration (biology) ,Cell Differentiation ,Extremities ,Cell Biology ,Hydrogen-Ion Concentration ,Heparin lyase ,Axons ,Cell biology ,Fibroblast Growth Factors ,medicine.anatomical_structure ,Heparin Lyase ,Immunology ,Blastema ,Cell Division ,Developmental Biology - Abstract
The growth of regenerating limbs of amphibians depends upon proliferation of the blastema cells that accumulate beneath the epidermal cap. The epidermal cap is known to be mitogenic for the blastema cells. We have extracted a mitogenic activity from both the mesenchymal and epidermal (epidermal cap) components of cone stage blastemas which is retained on heparin-Sepharose and elutes with 1.15 M NaCl. This fraction stimulates neurite outgrowth of PC12 cells and [3H]thymidine incorporation into CCL 39 cells and is potentiated by heparin. The 2 M fraction was inactive. The heparin-Sepharose-purified growth factor cross-reacts with bovine acidic FGF polyclonal antibodies and shows a Mr of 16,000 on Western blots. Blastema membranes contain specific high affinity binding sites (Kd = 25 pM; capacity = 30 fmole/mg protein) and low affinity binding sites (Kd = 18 nM; capacity = 30 pmole/mg protein) for aFGF as revealed by Scatchard analysis. 125I-aFGF which is bound specifically by both the epidermal cap and mesenchyme of blastema frozen sections is displaced by an excess of unlabeled factor and inhibited by heparin. Heparinase treatment and 2 M NaCl washing which decreased the binding was fourfold more efficient for epidermal cap than for mesenchyme suggesting the presence of high affinity receptors in the latter tissue. The presence of aFGF (or a closely related molecule) in blastemas is consistent with our earlier results that showed stimulation of proliferation of cultured blastema cells by acidic or basic FGF or heparin alone. These results suggest the possibility that aFGF is stored in the epidermal cap during limb regeneration and that it stimulates the proliferation of the underlaying mesenchyme. © 1991.
- Published
- 1991
50. Isolation and Characterization of Growth Factors
- Author
-
Ralph A. Bradshaw and K. P. Cavanaugh
- Subjects
Nerve growth factor ,Cell division ,Cell surface receptor ,Growth factor ,medicine.medical_treatment ,Gene expression ,medicine ,Biology ,Gene ,Transmembrane protein ,Cell biology ,Hormone - Abstract
Polypeptide growth factors represent a diverse group of hormone-like agents that affect a variety of cellular processes including metabolic regulation, cell division, extension of processes (and other morphological changes), and the maintenance of viability (James and Bradshaw 1984). They differ from their more classical counterparts in that they are often synthesized in a variety of cells and usually enjoy a spectrum of target tissues. They also often use non-systemic transport mechanisms. However, they are highly similar to (even indistinguishable from) other classic polypeptide hormones in the manner in which they interact with target tissues. These similarities include the obligatory requirement for cell surface receptors, the nature of the transmembrane signals generated and their effects on gene expression including the genes affected. Therefore, while it is convenient to consider this group of substances in a separate category (hence the retention of the term “polypeptide growth factor”), it is clear that they are an important part of the endocrine system.
- Published
- 1991
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.