Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Curr Med Chem ; 18(18): 2727-42, 2011.
Article in English | MEDLINE | ID: mdl-21649580

ABSTRACT

Protein kinase B/AKT plays a central role in cancer. The serine/threonine kinase is overexpressed or constitutively active in many cancers and has been validated as a therapeutic target for cancer treatment. However, targeting the kinase activity has revealed itself to be a challenge due to non-selectivity of the compounds towards other kinases. This review summarizes other approaches scientists have developed to inhibit the activity and function of AKT. They consist in targeting the pleckstrin homology (PH) domain of AKT. Indeed, upon the generation of 3-phosphorylated phosphatidylinositol phosphates (PI3Ps) by PI3-kinase (PI3K), AKT translocates from the cytosol to the plasma membrane and binds to the PI3Ps via its PH domain. Thus, several analogs of PI3Ps (PI Analogs or PIAs), alkylphospholipids (APLs), such as edelfosine or inositol phophates (IPs) have been described that inhibit the binding of the PH domain to PI3Ps. Recent allostertic inhibitors and small molecules that do not bind the kinase domain but affect the kinase activity of AKT, presumably by interacting with the PH domain, have been also identified. Finally, several drug screening studies spawned novel chemical scaffolds that bind the PH domain of AKT. Together, these approaches have been more or less sucessfull in vitro and to some extent translated in preclinical studies. Several of these new AKT PH domain inhibitors exhibit promising anti-tumor activity in mouse models and some of them show synergy with ionizing radiation and chemotherapy. Early clinical trials have started and results will attest to the validity and efficacy of such approaches in the near future.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Proteins/antagonists & inhibitors , Phosphoproteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Blood Proteins/chemistry , Blood Proteins/metabolism , Drug Evaluation , Mice , Models, Animal , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism
2.
J Med Chem ; 44(6): 898-908, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11300871

ABSTRACT

The serine-threonine protein kinase Akt is a direct downstream target of phosphatidylinositol 3-kinase (PI3-K). The PI3-K-generated phospholipids regulate Akt activity via directly binding to the Akt PH domain. The binding of PI3-K-generated phospholipids is critical to the relocalization of Akt to the plasma membrane, which plays an important role in the process of Akt activation. Activation of the PI3-K/Akt signaling pathway promotes cell survival. To elucidate the structural basis of the interaction of PI3-K-generated phospholipids with the Akt PH domain with the objective of carrying out structure-based drug design, we modeled the three-dimensional structure of the Akt PH domain. Comparative modeling-based methods were employed, and the modeled Akt structure was used in turn to construct structural models of Akt in complex with selected PI3-K-generated phospholipids using the computational docking approach. The model of the Akt PH domain consists of seven beta-strands forming two antiparallel beta-sheets capped by a C-terminal alpha-helix. The beta1-beta2, beta3-beta4, and beta6-beta7 loops form a positively charged pocket that can accommodate the PI3-K-generated phospholipids in a complementary fashion through specific hydrogen-bonding interactions. The residues Lys14, Arg25, Tyr38, Arg48, and Arg86 form the bottom of the binding pocket and specifically interact with the 3- and 4-phophate groups of the phospholipids, while residues Thr21 and Arg23 are situated at the wall of the binding pocket and bind to the 1-phosphate group. The predicted binding mode is consistent with known site-directed mutagenesis data, which reveal that mutation of these crucial residues leads to the loss of Akt activity. Moreover, our model can be used to predict the binding affinity of PI3-K-generated phospholipids and rationalize the specificity of the Akt PH domain for PI(3,4)P2, as opposed to other phospholipids such as PI(3)P and PI(3,4,5)P3. Taken together, our modeling studies provide an improved understanding of the molecular interactions present between the Akt PH domain and the PI3-K-generated phospholipids, thereby providing a solid structural basis for the design of novel, high-affinity ligands useful in modulating the activity of Akt.


Subject(s)
Phosphatidylinositols/chemistry , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/chemistry , Amino Acid Sequence , Hydrogen Bonding , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Protein Binding , Protein Conformation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Sequence Alignment
4.
J Invest Dermatol ; 116(1): 69-76, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11168800

ABSTRACT

Whereas previous investigations have shown that pharmacologic addition of gangliosides inhibits keratinocyte proliferation by downregulating epidermal growth factor receptor phosphorylation, the underlying biochemical basis and physiologic relevance are unknown. Using Scatchard and displacement plots, we have shown that supplemental purified gangliosides decrease the binding of (125)I-labeled epidermal growth factor to keratinocyte-derived SCC12 cells. Conversely, SCC12 cells transfected with sialidase and thus depleted of gangliosides show increased ligand binding to the epidermal growth factor receptor, which is consistent with their increased proliferation in response to epidermal growth factor and transforming growth factor-alpha, and increased phosphorylation of the epidermal growth factor receptor, and downstream signal transduction pathway components. The mechanism of the altered binding appears to involve primarily decreased numbers of available receptors within the intact membrane, but not altered receptor protein expression. These studies provide evidence that the effect of gangliosides on keratinocyte proliferation results, at least in part, from the direct binding of ganglioside to the receptor and disruption of the receptor-ligand interaction. Manipulation of membrane ganglioside content may be a powerful new means to alter epidermal growth factor receptor-dependent cell proliferation.


Subject(s)
ErbB Receptors/metabolism , DNA, Complementary/genetics , Epidermal Growth Factor/drug effects , Epidermal Growth Factor/metabolism , Gangliosides/metabolism , Gangliosides/pharmacology , Humans , Keratinocytes/chemistry , Ligands , Neuraminidase/biosynthesis , Neuraminidase/genetics , Transfection , Transforming Growth Factor alpha/drug effects , Transforming Growth Factor alpha/metabolism , Tumor Cells, Cultured
5.
J Med Chem ; 43(16): 3045-51, 2000 Aug 10.
Article in English | MEDLINE | ID: mdl-10956212

ABSTRACT

Phosphatidylinositol 3-kinase (PI3-K) phosphorylates the 3-position of phosphatidylinositol to give rise to three signaling phospholipids. Binding of the pleckstrin homology (PH) domain of Akt to membrane PI(3)P's causes the translocation of Akt to the plasma membrane bringing it into contact with membrane-bound Akt kinase (PDK1 and 2), which phosphorylates and activates Akt. Akt inhibits apoptosis by phosphorylating Bad, thus promoting its binding to and blockade of the activity of the cell survival factor Bcl-x. Herein we present the synthesis and biological activity of several novel phosphatidylinositol analogues and demonstrate the ability of the carbonate group to function as a surrogate for the phosphate moiety. Due to a combination of their PI3-K and Akt inhibitory activities, the PI analogues 2, 3, and 5 proved to be good inhibitors of the growth of various cancer cell lines with IC(50) values in the 1-10 microM range. The enhanced Akt inhibitory activity of the axial hydroxymethyl-bearing analogue 5 compared to its equatorial counterpart 6 is rationalized based upon postulated differences in the H-bonding patterns of these compounds in complex with a homology modeling generated structure of the PH domain of Akt. This work represents the first attempt to examine the effects of 3-modified PI analogues on these two crucial cell signaling proteins, PI3-K and Akt, in an effort to better understand their cell growth inhibitory properties.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Phosphatidylinositols/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Models, Molecular , Phosphatidylinositols/chemistry , Phosphatidylinositols/pharmacology , Proto-Oncogene Proteins c-akt , Structure-Activity Relationship , Tumor Cells, Cultured
6.
Exp Cell Res ; 256(1): 74-82, 2000 Apr 10.
Article in English | MEDLINE | ID: mdl-10739654

ABSTRACT

Gangliosides have been described as modulators of growth factor receptors. For example, GM3 addition in cell culture medium inhibits epidermal growth factor (EGF)-stimulated receptor autophosphorylation. Furthermore, depletion of ganglioside by sialidase gene transfection appeared to increase EGF receptor (EGFR) autophosphorylation. These data suggested that changes in GM3 content may result in different responses to EGF. In this study, the ceramide analog d-threo-1-phenyl-2-decannoylamino-3-morpholino-1-propanol ([D]-PDMP), which inhibits UDP-glucose-ceramide glucosyltransferase, and addition of GM3 to the culture medium were used to study the effects of GM3 on the EGFR. Addition of 10 microM [D]-PDMP to A431 cells resulted in significant GM3 depletion. Additionally, EGFR autophosphorylation was increased after EGF stimulation. When exogenous GM3 was added in combination with [D]-PDMP, the enhanced EGFR autophosphorylation was returned to control levels. [D]-PDMP also increased EGF-induced cell proliferation, consistent with its effect on autophosphorylation. Once again, the addition of GM3 in combination with [D]-PDMP reversed these effects. These results indicate that growth factor receptor functions can be modulated by the level of ganglioside expression in cell lines. Addition of GM3 inhibits EGFR activity and decrease of GM3 levels using [D]-PDMP treatment enhances EGFR activity. Modulation of growth factor receptor function may provide an explanation for how transformation-dependent ganglioside changes contribute to the transformed phenotype.


Subject(s)
Enzyme Inhibitors/pharmacology , ErbB Receptors/physiology , G(M3) Ganglioside/physiology , Gangliosides/metabolism , Morpholines/pharmacology , Carcinoma, Squamous Cell , Cell Division/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , G(M3) Ganglioside/pharmacology , Glucosyltransferases/antagonists & inhibitors , Humans , Kinetics , Signal Transduction , Tumor Cells, Cultured
7.
Eur J Pharmacol ; 377(2-3): 241-52, 1999 Jul 21.
Article in English | MEDLINE | ID: mdl-10456437

ABSTRACT

The effects of the biguanide anti-hyperglycemic agent, metformin (N,N'-dimethyl-biguanide), on insulin signaling was studied in a human hepatoma cell line (HepG2). Cells were cultured in the absence (control cells) or in the presence of 100 microM of a cholesterol derivative, hemisuccinate of cholesterol. Cholesterol hemisuccinate-treatment alters cholesterol and lipid content of HepG2 and modulates membrane fluidity. Cholesterol hemisuccinate-treatment induces a decrease in insulin responsiveness and creates an 'insulin-resistant' state in these cells. Exposure to 100 microM of metformin resulted in a significant enhancement of insulin-stimulated lipogenesis in control and cholesterol hemisuccinate-treated cells. In control cells, metformin altered glycogenesis in a biphasic manner. In cholesterol hemisuccinate-treated cells, metformin inhibited basal glycogenesis but restored insulin-stimulated glycogenesis. Hence, to understand the mechanism of metformin action, we analyzed early steps in the insulin signaling pathway, including insulin receptor autophosphorylation, mitogen-activated-protein kinase and phosphatidylinositol 3-kinase activities, in both control and cholesterol hemisuccinate-treated cells. Overall, the results suggest that metformin may interact with the insulin receptor and/or a component involved in the early steps of insulin signal transduction.


Subject(s)
Cholesterol/pharmacology , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Receptor, Insulin/drug effects , Signal Transduction/drug effects , Cholesterol/analogs & derivatives , Drug Resistance , Humans , Insulin/pharmacology , Lipids/biosynthesis , Phosphorylation , Time Factors , Tumor Cells, Cultured
8.
Biochim Biophys Acta ; 1454(1): 38-48, 1999 May 31.
Article in English | MEDLINE | ID: mdl-10354513

ABSTRACT

The lipid content of cultured cells can be experimentally modified by supplementing the culture medium with specific lipids or by the use of phospholipases. In the case of the insulin receptor, these methods have contributed to a better understanding of lipid disorder-related diseases. Previously, our laboratory demonstrated that experimental modification of the cellular lipid composition of an insulin-sensitive rat hepatoma cell line (ZHC) resulted in an alteration in insulin receptor binding and biological action (Bruneau et al., Biochim. Biophys. Acta 928 (1987) 287-296/297-304). In this paper, we have examined the effects of lipid modification in another hepatoma cell line, HepG2. Exogenous linoleic acid (LA, n-6), eicosapentaenoic acid (EPA, n-3) or hemisuccinate of cholesterol (CHS) was added to HepG2 cells, to create a cellular model in which membrane composition was modified. In this model, we have shown that: (1) lipids were incorporated in treated HepG2 cells, but redistributed differently when compared to treated ZHC cells; (2) that insulin signaling events, such as insulin receptor autophosphorylation and the phosphorylation of the major insulin receptor substrate (IRS-1) were altered in response to the addition of membrane lipids or cholesterol derived components; and (3) different lipids affected insulin receptor signaling differently. We have also shown that the loss of insulin receptor autophosphorylation in CHS-treated cells can be correlated with a decreased sensitivity to insulin. Overall, the results suggest that the lipid environment of the insulin receptor may play an important role in insulin signal transduction.


Subject(s)
Lipids/pharmacology , Receptor, Insulin/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholesterol/analysis , Cholesterol Esters/pharmacology , Eicosapentaenoic Acid/pharmacology , Fatty Acids/analysis , Linoleic Acid/pharmacology , Lipids/isolation & purification , Membrane Fluidity/drug effects , Rats , Signal Transduction , Triglycerides/analysis , Tumor Cells, Cultured
9.
Cancer Res ; 59(1): 234-40, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9892212

ABSTRACT

Glycosphingolipids expressed in cancer cells have been implicated in the modulation of tumor cell growth through their interaction with transmembrane signaling molecules such as growth factor receptors. For glycosphingolipids to interact with growth factor receptors, the presence of sialic acid seems to be essential. Stable transfection of a gene encoding a soluble Mr 42,000 sialidase into a human epidermoid carcinoma cell line (A431) provided an approach by which the level of terminal lipid-bound sialic acid on the cell surface could be altered. In the sialidase-positive clones, the level of ganglioside GM3 was diminished, and little change was observed in protein sialylation. Sialidase-transfected cells grew faster than control cells. Sialidase expression did not modify the binding of epidermal growth factor (EGF) to its receptor but enhanced EGF receptor (EGFR) tyrosine autophosphorylation as compared to that of parental cells or cells transfected with the vector (pcDNA3) alone. Moreover, the phosphorylation of the EGFR, as well as other protein substrates, was observed at low EGF concentrations, suggesting an increase in the receptor kinase sensitivity. These data provided evidence that changes in ganglioside expression in cancer cells by appropriate gene transfection can dramatically affect EGFR kinase activity. Hence, the modulation of ganglioside expression may represent an approach to alter tumor cell growth.


Subject(s)
Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Neuraminidase/genetics , Carcinoma, Squamous Cell/genetics , ErbB Receptors/genetics , Gene Transfer Techniques , Humans , Signal Transduction/genetics , Tumor Cells, Cultured
10.
Pediatr Neurosurg ; 29(1): 1-13, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9755305

ABSTRACT

Growth factor receptors (GFRs) have been described as overexpressed in several types of brain tumors. Overexpression of these transmembrane proteins is considered to be an important part of tumorigenesis. Genetic as well as epigenetic modulation of the receptors have to be considered when trying to understand the role of GFRs in tumors or as targets for tumor therapy. GFR function can be modulated by membrane components (e.g. gangliosides) or by the change in receptor glycosylation. These types of changes and the occurrence of the expression of mutated receptor expressed in tumor cell can result in altered signaling. In this review, we have focused on GFRs, their expression and mutations in brain tumors. Recently the correlation between GFR expression and patient outcome has suggested that these tyrosine kinases and their signaling might play a decisive role in the course of patients with brain tumors. The importance of GFRs as possible targets for brain tumor therapy is also discussed.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain/metabolism , Glioma/genetics , Glioma/therapy , Receptors, Growth Factor , Animals , Brain Neoplasms/metabolism , Child , Gangliosides/metabolism , Gene Expression , Glioma/metabolism , Humans , Mutation , Rats , Receptors, Growth Factor/analysis , Receptors, Growth Factor/genetics , Treatment Outcome
11.
Ann N Y Acad Sci ; 845: 240-52, 1998 Jun 19.
Article in English | MEDLINE | ID: mdl-9668358

ABSTRACT

Polypeptide growth factors and gangliosides can both be considered as trophic agents involved in almost all stages of neural cell development, differentiation, survival, and pathology. In most cases their physiological roles are still not clear due to the considerable complexity in their regulation. Several growth factors [e.g., basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF)] and one species of ganglioside (GM1) have been shown to exert interactions with each other and also to exhibit neuroprotective effects against retinal ischemia in vivo and cerebral excitotoxicity in vitro. Different experimental models are used to investigate their relevance to ischemic and excitotoxic conditions in the retina, and it is shown that (1) both bFGF and EGF show very effective neuroprotection for rat retinal neurones exposed to toxic levels of glutamate or its nonphysiological agonist kainate in vitro; (2) GM1 (10(-5M) used under the same conditions does not afford protection; (3) retinal glial cells also suffer morphological perturbations following glutamate or kainate treatment, but this effect is dependent on neuron-glial interactions, indicating the existence of intermediate neuron-derived messenger molecules; (4) these glial changes can be corrected by posttreatment with either bFGF or EGF in vitro; (5) using an in vivo animal model involving anterior chamber pressure-induced ischemia in adult rats, it is shown that either pretreatment by intraperitoneal injection of GM1, or posttreatment by intraocular injection of the same ganglioside, reduces significantly histological damage to inner nuclear regions; and (6) in cultured retinal Müller glial cells the existence of molecular and metabolic interactions between both types of trophic factors is demonstrated. Hence both these groups of trophic molecules show interesting features for retinal ischemic treatment.


Subject(s)
Gangliosides/pharmacology , Growth Substances/pharmacology , Neurons/drug effects , Neuroprotective Agents , Retina/physiology , Retinal Vessels/physiology , Animals , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , G(M1) Ganglioside/pharmacology , Ischemia/pathology , Ischemia/physiopathology , Ischemia/prevention & control , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/physiology , Neurons/cytology , Neurons/physiology , Rats , Retina/cytology , Retina/drug effects , Retinal Vessels/cytology , Retinal Vessels/drug effects
13.
J Biol Chem ; 272(14): 9275-9, 1997 Apr 04.
Article in English | MEDLINE | ID: mdl-9083062

ABSTRACT

N-linked oligosaccharides appear to be important for the function of the epidermal growth factor (EGF) receptor. In a previous study (Rebbaa, A., Yamamoto, H., Moskal, J. R., and Bremer, E. G. (1996) J. Neurochem. 67, 2265-2272), we showed that binding of the erythroagglutinating phytohemagglutin lectin from Phaseolus vulgaris to the bisecting structures on the EGF receptor from U373 MG glioma cells blocked EGF binding and receptor autophosphorylation. In this study we examined the consequences of overexpression of the bisecting structure on the EGF receptor by gene transfection of U373 MG cells with the N-acetylglucosaminyltransferase III (GnT-III). This modification leads to a significant decrease in EGF binding and EGF receptor autophosphorylation. In addition, the cellular response to EGF was found to be altered. Proliferation of U373 MG cells in serum-free medium is inhibited by EGF. In contrast, proliferation of the GnT-III-transfected cells was stimulated by EGF. These data demonstrate that changes in EGF receptor glycosylation by GnT-III transfection reduces the number of the active receptors in U373 MG cells and that this change results in change in the cellular response to EGF.


Subject(s)
Acetylglucosamine/metabolism , ErbB Receptors/metabolism , Glioma/genetics , Oligosaccharides/metabolism , Transfection , Epidermal Growth Factor/metabolism , Glioma/metabolism , Glycosylation , Humans , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation , Phytohemagglutinins/metabolism , RNA, Messenger/metabolism , Tumor Cells, Cultured
14.
Indian J Biochem Biophys ; 34(1-2): 90-6, 1997.
Article in English | MEDLINE | ID: mdl-9343935

ABSTRACT

Ganglioside (GG) and neurotrophic growth factor (GF) interactions in retinal neuronal and glial cells have been very little studied. Rat retinas were mechanically separated into outer (photoreceptor or PR) and inner (other neurons, IR) halves by planar vibratome sectioning and retinal Müller glial (RMG) cells were isolated and cultured according to previously published methods. The distribution on a percent molar basis of individual GG was different between the two halves: PR were dominated by GD3 (48% total GG) and contained only trace amounts (< 4%) of complex species (GT1b, GQ); IR was more typical of mature brain tissue, exhibiting substantial amounts (approximately 25%) of more complex GG. The GG profile of RMG cells was also simple, dominated by GM3 (60%) and GD1a (20%). A single addition to the medium of 500 pM bFGF or EGF for 48 hr to cultured RMG cells led to significant increases in total GG levels of 30-40%. Such treatments by both growth factors induced increases in GM3, whereas longer exposure (96 hr) of confluent RMG to these factors additionally stimulated synthesis of more complex GG. Incubations of RMG with [3H]-glucosamine showed that GG synthesis was 2-fold stimulated by growth factors. We also tested the effect of GM3 on one of the bFGF receptor transduction pathways, namely PI-3 kinase activation. To our knowledge these data constitute the first demonstration of neurotrophic factor stimulation of GG levels in cells of CNS in vitro. Such complex interactions may have particularly important consequences for neural physiopathology.


Subject(s)
Gangliosides/metabolism , Nerve Growth Factors/pharmacology , Retina/drug effects , Animals , Cells, Cultured , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Lipid Metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Rats , Retina/cytology , Retina/metabolism
15.
J Lipid Mediat Cell Signal ; 14(1-3): 277-88, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8906573

ABSTRACT

Gangliosides have long been implicated in cell growth regulation and play an important role as modulators in protein phosphorylation. In order to better understand how glycosphingolipids and growth factors interact, we examined the modulation of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) effects on retinal Müller glial cells (RMG), following modification of their GG composition. Treatment of MG cells with GG (GM1, GT1b) and asialoGM1 resulted in modifications of several aspects of cellular responses to EGF- and FGF-receptor (R) activation: mitogenesis, cell migration, tyrosine phosphorylation of the EGF-R and FGF-R and even their cellular substrates were particularly influenced by GG. Indeed GG caused modifications of EGF-R and FGF-R autophosphorylation kinetics. GG long term effects (mitogenesis and migration) correlate with short term effects (tyrosine phosphorylation) and differences in receptor tyrosine kinase signalling could explain the specificity in growth factor responses.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Fibroblast Growth Factor 2/metabolism , Gangliosides/pharmacology , Receptors, Fibroblast Growth Factor/metabolism , Retinal Ganglion Cells/metabolism , Signal Transduction/drug effects , Animals , Cells, Cultured , Rats
16.
Glia ; 17(3): 206-16, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8840162

ABSTRACT

Polypeptide growth factors and membrane-bound gangliosides are involved in cell signaling, including that observed in cells of neural origin. To analyze possible interactions between these two systems, we investigated the modulation of short- and long-term responses to basic fibroblast and epidermal growth factor (bFGF and EGF, respectively) in cultured retinal Müller glial cells following experimental modification of their ganglioside composition. These glial cells readily incorporated exogenously administered GM3 ganglioside, which was not substantially metabolized within 24 h. Such treatments significantly inhibited bFGF-induced DNA replication and cell migration, while having much less effect on analogous EGF-mediated behaviors. To explore GM3/growth factor interactions further, different aspects of glial metabolism in response to bFGF or EGF stimulation were examined: membrane fluidity, growth factor binding, global and individual changes in growth factor-induced phosphotyrosine levels, and growth factor-induced activation of mitogen-activated protein kinase. GM3 reduced the intensity of immunocytochemical labeling of phosphotyrosine-containing proteins within bFGF-stimulated cells and down-regulated FGF receptor activation and tyrosine phosphorylation of its cellular substrates, whereas similar parameters in EGF-stimulated cells were much less affected. Hence the data reveal a complex relationship in normal neural cells between polypeptide growth factors and membrane-bound gangliosides, which may participate in retinal cellular physiology in vivo.


Subject(s)
Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gangliosides/pharmacology , Retinal Ganglion Cells/drug effects , Animals , Binding, Competitive , Cells, Cultured , Dose-Response Relationship, Drug , Immunohistochemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...