Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Mol Carcinog ; 63(5): 817-833, 2024 May.
Article En | MEDLINE | ID: mdl-38299738

Chronic exposure to arsenic (As) promotes skin carcinogenesis in humans and potentially disturbs resident stem cell dynamics, particularly during maternal and early life exposure. In the present study, we demonstrate how only prenatal arsenic exposure disturbs keratinocyte stem cell (KSC) conditioning using a BALB/c mice model. Prenatal As exposure alters the normal stemness (CD34, KRT5), differentiation (Involucrin), and proliferation (PCNA) program in skin of offspring with progression of age as observed at 2, 10, and 18 weeks. Primary KSCs isolated from exposed animal at Day-2 showed increased survival (Bax:Bcl-xL, TUNEL assay), proliferation (BrdU), and differentiation (KRT5, Involucrin) potential through the activation of pro-carcinogenic IGF2R-MAPK cascade (IGF2R-G(α)q-MEK1-ERK1/2). This was associated with reduced enrichment of histone H3K27me3 and its methylase, EZH2 along with increased binding of demethylase, KDM6A at Igf2r promoter. Altered KSCs conditioning through disturbed Igf2r imprint contributed to impaired proliferation and differentiation and an aggravated tumor response in offspring.


Arsenic , Keratinocytes , Skin Neoplasms , Animals , Female , Mice , Pregnancy , Arsenic/toxicity , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , MAP Kinase Signaling System/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Receptor, IGF Type 2/drug effects , Receptor, IGF Type 2/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
2.
Biotech Histochem ; 95(3): 163-170, 2020 Apr.
Article En | MEDLINE | ID: mdl-32053010

Diabetes is a metabolic disorder that damages many organs. We investigated the effects of reperfusion using lactate Ringer's solution (LR) in a diabetic animal model. Eight-week-old rats were divided into groups: control, hemorrhagic shock induced (HS), diabetes mellitus (DM), DM plus HS (DM + HS) and DM rats that received LR after HS (DM + HS + LR). HS was induced by withdrawing blood from the femoral artery and arterial pressure was maintained at 40 mm Hg for 1 h. Animals were perfused with either withdrawn blood or LR. Rats were sacrificed and hearts were collected from all groups. Histopathological studies were performed using left ventricles and western blotting analysis was performed using protein extracted from the left ventricle. Using the TUNEL assay, we found more apoptotic cells in the DM + HS group compared to the control group, whereas in animals resuscitated with LR, the number of apoptotic cells was reduced. Western blotting showed a significant reduction in apoptotic markers, cyt c, cas 9 and cas 3, and increased survival markers, pPI3K and pAKT, in the DM + HS + LR group. Reperfusion with LR may have therapeutic effects on trauma induced HS by blocking the IGF II R facilitated apoptosis pathway in diabetic rats.


Receptor, IGF Type 2/drug effects , Reperfusion , Ringer's Lactate/pharmacology , Shock, Hemorrhagic/drug therapy , Animals , Apoptosis/drug effects , Diabetes Mellitus, Experimental , Disease Models, Animal , Rats , Shock, Hemorrhagic/metabolism
3.
Histochem Cell Biol ; 137(2): 249-59, 2012 Feb.
Article En | MEDLINE | ID: mdl-22131059

Bisphenol A (BPA), a synthetic additive used to harden polycarbonate plastics and epoxy resin, is ubiquitous in our everyday environment. Many studies have indicated detrimental effects of BPA on the mammalian reproductive abilities. This study is aimed to test the potential effects of BPA on methylation of imprinted genes during oocyte growth and meiotic maturation in CD-1 mice. Our results demonstrated that BPA exposure resulted in hypomethylation of imprinted gene Igf2r and Peg3 during oocyte growth, and enhanced estrogen receptor (ER) expression at the levels of mRNA and protein. The relationship between ER expression and imprinted gene hypomethylation was substantiated using an ER inhibitor, ICI182780. In addition, BPA promoted the primordial to primary follicle transition, thereby speeding up the depletion of the primordial follicle pool, and suppressed the meiotic maturation of oocytes because of abnormal spindle assembling in meiosis I. In conclusion, neonatal exposure to BPA inhibits methylation of imprinted genes during oogenesis via the ER signaling pathway in CD-1 mice.


DNA Methylation/drug effects , Genomic Imprinting/genetics , Kruppel-Like Transcription Factors/genetics , Oocytes/metabolism , Phenols/toxicity , Receptor, IGF Type 2/genetics , Receptors, Estrogen/metabolism , Animals , Benzhydryl Compounds , DNA Methylation/genetics , Female , Genomic Imprinting/drug effects , Kruppel-Like Transcription Factors/drug effects , Mice , Oocytes/drug effects , Oocytes/growth & development , Oogenesis/drug effects , Oogenesis/genetics , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Receptor, IGF Type 2/drug effects , Receptors, Estrogen/drug effects , Signal Transduction
4.
Diabetes ; 58(8): 1816-25, 2009 Aug.
Article En | MEDLINE | ID: mdl-19401425

OBJECTIVE: The gluco-incretin hormones glucagon-like peptide (GLP)-1 and gastric inhibitory peptide (GIP) protect beta-cells against cytokine-induced apoptosis. Their action is initiated by binding to specific receptors that activate the cAMP signaling pathway, but the downstream events are not fully elucidated. Here we searched for mechanisms that may underlie this protective effect. RESEARCH DESIGN AND METHODS: We performed comparative transcriptomic analysis of islets from control and GipR(-/-);Glp-1-R(-/-) mice, which have increased sensitivity to cytokine-induced apoptosis. We found that IGF-1 receptor expression was markedly reduced in the mutant islets. Because the IGF-1 receptor signaling pathway is known for its antiapoptotic effect, we explored the relationship between gluco-incretin action, IGF-1 receptor expression and signaling, and apoptosis. RESULTS: We found that GLP-1 robustly stimulated IGF-1 receptor expression and Akt phosphorylation and that increased Akt phosphorylation was dependent on IGF-1 but not insulin receptor expression. We demonstrated that GLP-1-induced Akt phosphorylation required active secretion, indicating the presence of an autocrine activation mechanism; we showed that activation of IGF-1 receptor signaling was dependent on the secretion of IGF-2. We demonstrated, both in MIN6 cell line and primary beta-cells, that reducing IGF-1 receptor or IGF-2 expression or neutralizing secreted IGF-2 suppressed GLP-1-induced protection against apoptosis. CONCLUSIONS: An IGF-2/IGF-1 receptor autocrine loop operates in beta-cells. GLP-1 increases its activity by augmenting IGF-1 receptor expression and by stimulating secretion; this mechanism is required for GLP-1-induced protection against apoptosis. These findings may lead to novel ways of preventing beta-cell loss in the pathogenesis of diabetes.


Apoptosis/drug effects , Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/cytology , Receptor, IGF Type 1/genetics , Receptor, IGF Type 2/genetics , Animals , Gene Expression Regulation/drug effects , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 2/deficiency , Receptor, IGF Type 2/drug effects
5.
Brain Res ; 1131(1): 77-87, 2007 Feb 02.
Article En | MEDLINE | ID: mdl-17184742

Insulin-like growth factor-2 (IGF2) is a member of the insulin gene family with known neurotrophic properties. The actions of IGF2 are mediated via the IGF type 1 and type 2 receptors as well as through the insulin receptors, all of which are widely expressed throughout the brain. Since IGF2 is up-regulated in the brain after injury, we wanted to determine whether the absence of IGF2 can lead to any alteration on brain morphology and/or in the response of its receptor binding sites following a neurotoxic insult. No morphological differences were observed between the brains of IGF2 knockout (IGF2(-/-)) and wild-type control (IGF2(+/+)) mice. However, our in vitro receptor autoradiography results indicate that IGF2(-/-) mice had lower endogenous levels of [(125)I]IGF1 and [(125)I]insulin receptor binding sites in the hippocampus and cerebellum as compared to IGF2(+/+) mice, while endogenous [(125)I]IGF2 receptor binding showed a decrease only in the cerebellum. Seven days after kainic acid administration, the [(125)I]insulin receptor binding sites were significantly decreased in all brain regions of the IGF2(+/+) mice, while the levels of [(125)I]IGF1 and [(125)I]IGF2 binding sites were decreased only in select brain areas. The IGF2(-/-) mice, on the other hand, showed increased [(125)I]IGF1 and [(125)I]IGF2 and [(125)I]insulin receptor binding sites in selected regions such as the hippocampus and cerebellum. These results, taken together, suggest that deletion of IGF2 gene does not affect gross morphology of the brain but does selectively alter endogenous [(125)I]IGF1, [(125)I]IGF2 and [(125)I]insulin receptor binding sites and their response to neurotoxicity.


Binding, Competitive/drug effects , Brain/metabolism , Insulin-Like Growth Factor II/genetics , Neurotoxins/pharmacology , Receptor, IGF Type 2/metabolism , Age Factors , Animals , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/physiology , Brain/drug effects , Brain/embryology , Brain Damage, Chronic/genetics , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Cerebellum/drug effects , Cerebellum/embryology , Cerebellum/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Resistance/genetics , Hippocampus/drug effects , Hippocampus/embryology , Hippocampus/metabolism , Insulin-Like Growth Factor I/metabolism , Iodine Radioisotopes , Kainic Acid/pharmacology , Male , Mice , Mice, Knockout , Radioligand Assay , Receptor, IGF Type 2/drug effects , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism
6.
Neuroreport ; 17(12): 1245-9, 2006 Aug 21.
Article En | MEDLINE | ID: mdl-16951563

We have investigated the effect of scarring at a site of peripheral nerve repair by comparing regeneration of the sciatic nerve in normal mice and two transgenic strains with an increased or decreased propensity for scarring. The outcome was assessed by quantifying collagen at the repair site, recording compound action potentials and counting myelinated nerve fibres on each side of the repair. We found that higher levels of collagen scar formation were associated with smaller compound action potentials, slower conduction velocities and a reduction in fibre numbers across the repair site. We conclude that scarring impedes regeneration at sites of nerve repair and suggest that this could be amenable to therapeutic manipulation.


Cicatrix/physiopathology , Nerve Regeneration/physiology , Sciatic Neuropathy/physiopathology , Wound Healing/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cicatrix/metabolism , Collagen/metabolism , Electric Stimulation/methods , Insulin-Like Growth Factor II/deficiency , Interleukin-10/deficiency , Interleukin-4/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Fibers/pathology , Nerve Fibers/physiology , Neural Conduction/genetics , Neural Conduction/radiation effects , Receptor, IGF Type 2/drug effects , Sciatic Neuropathy/genetics , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Wound Healing/genetics
7.
Org Lett ; 6(26): 4921-4, 2004 Dec 23.
Article En | MEDLINE | ID: mdl-15606100

[reaction: see text] Mannose 6-phosphate mimics locked into the alpha-configuration and bearing hydrolase-resistant phosphate surrogates were synthesized and evaluated for binding affinity to the mannose 6-phosphate/insulin-like growth factor II receptor (M6P/IGF2R). Affinity increases as the phosphate surrogate is varied in the order malonyl ether < malonate < phosphonate. An alkene cross-metathesis approach to sought-after bivalent M6P-bearing ligands is also described. These compounds were designed to map onto biantennary sectors of high-mannose-type oligosaccharides carried by glycoprotein M6P/IGF2R ligands.


Mannosephosphates/chemistry , Mannosephosphates/pharmacology , Receptor, IGF Type 2/drug effects , Carbohydrate Conformation , Carbohydrate Sequence , Glycoproteins/chemistry , Ligands , Mannosephosphates/chemical synthesis , Molecular Sequence Data , Oligosaccharides/chemistry
8.
Domest Anim Endocrinol ; 26(2): 155-75, 2004 Mar.
Article En | MEDLINE | ID: mdl-14757187

The somatotropic axis regulates growth of the gastrointestinal tract (GIT). In addition, colostrum feeding and glucocorticoids affect maturation of the GIT around birth in mammals. We have measured mRNA levels of members of the somatotropic axis to test the hypothesis that colostrum intake and dexamethasone treatment affect respective gene expression in the GIT. Calves were fed either colostrum or an isoenergetic milk-based formula, and in each feeding group, half of the calves were treated with dexamethasone (DEXA; 30 microg/kg body weight per day). Individual parameters of the somatotropic axis differed (P < 0.05) among different GIT sections and formula feeding increased (P < 0.05) mRNA levels of individual parameters at various sites of the GIT. Effects of DEXA on the somatotropic axis in the GIT partly depended on different feeding. In colostrum-fed calves, DEXA decreased (P < 0.05) mRNA levels of IGF-I (esophagus, fundus, duodenum, and ileum), IGF-II (fundus), IGFBP-2 (fundus), IGFBP-3 (fundus), IGF1R (esophagus, ileum, and colon), IGF2R (fundus), GHR (fundus), and InsR (esophagus, fundus), but in formula-fed calves DEXA increased mRNA levels of IGF-I (esophagus, rumen, jejunum, and colon). Furthermore, DEXA increased (P < 0.05) mRNA levels of IGF-II (pylorus), IGFBP-3 (duodenum), IGF2R (pylorus), and GHR (ileum), but decreased mRNA levels of IGFBP-2 (ileum), and IGF1R (fundus). Whereas formula feeding had stimulating effects, effects of DEXA treatment on the gene expression of parameters of the somatotropic axis varied among GIT sites and partly depended on feeding.


Animals, Newborn/metabolism , Cattle/metabolism , Colostrum/physiology , Dexamethasone/pharmacology , Gastrointestinal Tract/metabolism , Growth Substances/metabolism , Animals , Eating , Gastrointestinal Tract/drug effects , Gene Expression Regulation, Developmental/physiology , Glucocorticoids/pharmacology , Growth Substances/genetics , Insulin-Like Growth Factor Binding Protein 2/drug effects , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 3/drug effects , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Male , RNA, Messenger/analysis , RNA, Messenger/drug effects , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/drug effects , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , Receptor, Insulin/drug effects , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Somatotropin/drug effects , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism
9.
J Cell Physiol ; 197(3): 388-99, 2003 Dec.
Article En | MEDLINE | ID: mdl-14566968

Zinc (Zn(2+)) is a multifunctional micronutrient. The list of functions for this micronutrient expanded with the recent discovery that Zn(2+) retains insulin-like growth factors binding proteins (IGFBPs) on the surface of cultured cells, lowers the affinity of cell-associated IGFBPs, and increases the affinity of the cell surface insulin-like growth factor (IGF)-type 1 receptor (IGF-1R). However, currently there is no information concerning the effect of Zn(2+) on soluble IGFBPs. In the current study, the soluble IGFBP-5 secreted by BC(3)H-1 cells is shown to bind approximately 50% more [(125)I]-IGF-II than [(125)I]-IGF-I at pH 7.4. Zn(2+) is shown to depress the binding of both IGF-I and IGF-II to soluble secreted IGFBP-5; [(125)I]-IGF-I binding is affected more so than [(125)I]-IGF-II binding. Zn(2+) acts by lowering the affinity (K(a)) of IGFBP-5 for the IGFs. Scatchard plots are non-linear indicating the presence of high and low affinity binding sites; Zn(2+) affects only binding to the high affinity site. In contrast, Zn(2+) increases the affinity by which either [(125)I]-IGF-I or [(125)I]-R(3)-IGF-I binds to the IGF-1R, but depresses [(125)I]-IGF-II binding to the IGF-type 2 receptor (IGF-2R) on BC(3)H-1 cells. By depressing the association of the IGFs with soluble IGFBPs, Zn(2+) is shown to repartition either [(125)I]-IGF-I or [(125)I]-IGF-II from soluble IGFBP-5 onto cell surface IGF receptors. Zn(2+) was active at physiological doses depressing IGF binding to IGFBP-5 and the IGF-2R at 15-20 microM. Hence, a novel mechanism is further characterized by which the trace micronutrient Zn(2+) could regulate IGF activity.


Insulin-Like Growth Factor Binding Protein 5/metabolism , Receptors, Cell Surface/metabolism , Somatomedins/metabolism , Zinc/metabolism , Animals , Binding Sites/drug effects , Binding Sites/physiology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Insulin-Like Growth Factor Binding Protein 5/drug effects , Macromolecular Substances , Mice , Models, Molecular , Muscle Cells/drug effects , Muscle Cells/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/drug effects , Receptor, IGF Type 2/metabolism , Receptors, Cell Surface/drug effects , Somatomedins/drug effects , Zinc/pharmacology
10.
Exp Dermatol ; 12(4): 426-34, 2003 Aug.
Article En | MEDLINE | ID: mdl-12930299

In this study, we have examined the effects of retinoic acid (RA) on the human immortalized keratinocyte cell line (HaCaT). A significant twofold (P < 0.01) increase in apoptotic cell death compared with the control was found within 24 h of treatment with 10-5 M of RA. Apoptosis was confirmed by flow cytometry. Cycloheximide did not inhibit this acute RA-induced apoptosis. Interestingly, insulin-like growth factor-II (IGF-II, 50 ng/ml) was able to significantly (67.3%; P < 0.05) reduce RA effects, whereas IGF-I (50 ng/ml) and insulin (75 ng/ml) were without effect. Furthermore, analogues of IGF-II [leu27 IGF-II and Des(1-6) IGF-II], with altered affinities for the IGF-I receptor and IGF-binding proteins (IGFBPs), but retained affinities for the IGF-II receptor, also completely inhibited (100%; P < 0.01) RA-induced apoptosis, while an IGF-I receptor antagonist did not reduce the survival effects of IGF-II. Insulin pretreatment negates the survival effect of IGF-II. In contrast, mannose 6 phosphate (M6P) did not alter RA or IGF-II actions. These results indicate that rapid induction of cell death by RA is independent of production or secretion of new proteins. The inhibition of RA action by IGF-II was independent of its ability to signal through the IGF-I receptor or to interact with IGFBPs.


Apoptosis/drug effects , Insulin-Like Growth Factor II/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Receptor, IGF Type 2/metabolism , Tretinoin/pharmacology , Cell Division/drug effects , Cell Line , Cycloheximide/pharmacology , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Keratinocytes/cytology , Mannosephosphates/pharmacology , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/drug effects , Recombinant Proteins/pharmacology
11.
J Cell Biol ; 160(2): 223-33, 2003 Jan 20.
Article En | MEDLINE | ID: mdl-12538642

The 280-kD cation-independent mannose-6-phosphate receptor (MPR) has been shown to play a role in endocytic uptake of granzyme B, since target cells overexpressing MPR have an increased sensitivity to granzyme B-mediated apoptosis. On this basis, it has been proposed that cells lacking MPR are poor targets for cytotoxic lymphocytes that mediate allograft rejection or tumor immune surveillance. In the present study, we report that the uptake of granzyme B into target cells is independent of MPR. We used HeLa cells overexpressing a dominant-negative mutated (K44A) form of dynamin and mouse fibroblasts overexpressing or lacking MPR to show that the MPR/clathrin/dynamin pathway is not required for granzyme B uptake. Consistent with this observation, cells lacking the MPR/clathrin pathway remained sensitive to granzyme B. Exposure of K44A-dynamin-overexpressing and wild-type HeLa cells to granzyme B with sublytic perforin resulted in similar apoptosis in the two cell populations, both in short and long term assays. Granzyme B uptake into MPR-overexpressing L cells was more rapid than into MPR-null L cells, but the receptor-deficient cells took up granzyme B through fluid phase micropinocytosis and remained sensitive to it. Contrary to previous findings, we also demonstrated that mouse tumor allografts that lack MPR expression were rejected as rapidly as tumors that overexpress MPR. Entry of granzyme B into target cells and its intracellular trafficking to induce target cell death in the presence of perforin are therefore not critically dependent on MPR or clathrin/dynamin-dependent endocytosis.


Apoptosis/immunology , Cell Membrane/immunology , Endocytosis/immunology , Killer Cells, Natural/enzymology , Receptor, IGF Type 2/deficiency , Serine Endopeptidases/immunology , T-Lymphocytes, Cytotoxic/enzymology , Animals , Apoptosis/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Clathrin/drug effects , Clathrin/genetics , Clathrin/metabolism , Dynamins/drug effects , Dynamins/genetics , Dynamins/metabolism , Endocytosis/drug effects , Female , Graft Rejection/genetics , Graft Rejection/immunology , Granzymes , HeLa Cells , Humans , Killer Cells, Natural/immunology , Male , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neoplasms/immunology , Neoplasms/metabolism , Perforin , Pore Forming Cytotoxic Proteins , Receptor, IGF Type 2/drug effects , Receptor, IGF Type 2/genetics , Serine Endopeptidases/deficiency , Serine Endopeptidases/pharmacology , T-Lymphocytes, Cytotoxic/immunology
12.
J Clin Endocrinol Metab ; 86(8): 3665-74, 2001 Aug.
Article En | MEDLINE | ID: mdl-11502794

We have earlier shown that migration and invasiveness of first trimester human extravillous trophoblast cells are stimulated by IGF-II, independently of IGF type 1 receptor and that migration stimulation is the primary reason for increased extravillous trophoblast cell invasiveness induced by IGF-II. In the present study we examined the functional role of IGF type II receptor in IGF-II stimulation of extravillous trophoblast cell migration and the underlying signal transduction pathways including the participation of inhibitory G protein(s) and MAPK. The migratory ability of a well characterized in vitro propagated human first trimester extravillous trophoblast cell line expressing the phenotype of extravillous trophoblast cells in situ was quantitated with a Transwell migration assay under different experimental conditions. We found that the extravillous trophoblast cells expressed an abundance of IGF type 2 receptor as detected by immunostaining and Western blots, and recombinant human IGF-II promoted their migration in a dose- and time-dependent manner. Both polyclonal and monoclonal IGF type 2 receptor-blocking antibodies blocked migration-stimulating effects of IGF-II. Two synthetic IGF-II analogs ([Leu27]IGF-II, which can bind to IGF type 2 receptor and IGF-binding proteins, but not IGF type 1 receptor, and [QAYL-Leu27]IGF-II, which can bind to IGFR-II, but neither IGFR-I nor IGF-binding proteins) both stimulated extravillous trophoblast cell migration to levels higher than those induced by wild-type IGF-II. These results reveal that IGF-II action was mediated by IGF type 2 receptor, independently of IGF type 1 receptor and IGF-binding proteins. Treatment of extravillous trophoblast cell membrane preparations with IGF-II decreased adenylyl cyclase activity in a concentration-dependant manner, indicating the participation of inhibitory G proteins in IGF-II action. This was substantiated further with the findings that increasing intracellular cAMP using forskolin or (Bu)2cAMP inhibited basal extravillous trophoblast cell migration and blocked IGF-II stimulation of migration. IGF-II treatment rapidly stimulated phosphorylation of MAPK (ERK-1 and -2), which was blocked by pretreatment of extravillous trophoblast cells with the MAPK kinase (MEK) inhibitor PD98059. Treatment with this inhibitor also blocked extravillous trophoblast cell migration in the presence or absence of IGF-II. These results, taken together, reveal that IGF-II stimulates extravillous trophoblast cell migration by signaling through IGF type 2 receptor, involving inhibitory G proteins and activating the MAPK pathway.


GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Insulin-Like Growth Factor II/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Receptor, IGF Type 2/physiology , Trophoblasts/physiology , Adenylyl Cyclases/metabolism , Antibodies/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Line , Cell Movement/drug effects , Cell Movement/physiology , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Humans , Insulin-Like Growth Factor II/analogs & derivatives , Kinetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Pregnancy , Receptor, IGF Type 2/drug effects , Receptor, IGF Type 2/immunology , Trophoblasts/cytology , Trophoblasts/drug effects
13.
Thromb Haemost ; 85(5): 903-7, 2001 May.
Article En | MEDLINE | ID: mdl-11372686

Antiphospholipid antibodies (APLA) are associated with thrombophilia and recurrent pregnancy loss. Different mechanisms have been proposed to explain their pathogenic effects and among them, we have previously shown that APLA accumulate in late endosomes of human umbilical vein endothelial cells (HUVEC) leading to a redistribution of the cation-independent mannose-6-phosphate receptor (CI-M6PR). Because many APLA are directed towards beta2-glycoprotein 1 (beta2GP1)phospholipid complexes, we investigated the localisation of beta2GP1 in HUVEC. By immunofluorescence analysis, using monoclonal and polyclonal anti-beta2GP1 antibodies, we detected beta2GP1 at the cell surface and in late endosomes. Incubation of HUVEC with anti-beta2GP1 antibodies resulted in antibody accumulation at the cell surface and within late endosomes and in a redistribution of the CI-M6PR from the Golgi apparatus to late endosomes. The anti-beta2GP1 antibodies remained detectable in late endosomes even after several days of incubation in antibody-free medium. The accumulation of anti-beta2GP1 antibodies in late endosomes of endothelial cells and the resulting modification of intracellular protein trafficking may contribute to the pathogenic effects of these antibodies.


Endosomes/chemistry , Endothelium, Vascular/cytology , Glycoproteins/metabolism , Antibodies/metabolism , Antibodies/pharmacology , Antibodies, Antiphospholipid , Anticoagulants/immunology , Anticoagulants/metabolism , Antiphospholipid Syndrome/etiology , Endothelium, Vascular/ultrastructure , Glycoproteins/immunology , Humans , Microscopy, Fluorescence , Receptor, IGF Type 2/drug effects , Receptor, IGF Type 2/metabolism , Umbilical Veins/cytology , beta 2-Glycoprotein I
14.
Am J Physiol Regul Integr Comp Physiol ; 280(4): R1230-9, 2001 Apr.
Article En | MEDLINE | ID: mdl-11247849

Insulin-like growth factor (IGF)-I and -II have been cloned from a number of teleost species, but their cellular actions in fish are poorly defined. In this study, we show that both IGF-I and -II stimulated zebrafish embryonic cell proliferation and DNA synthesis in a concentration-dependent manner, whereas insulin had little mitogenic activity. Affinity cross-linking and immunoblotting studies revealed the presence of IGF receptors with the characteristics of the mammalian type I IGF receptor. Competitive binding assay results indicated that the binding affinities of the zebrafish IGF-I receptors to IGF-I, IGF-II, and insulin are 1.9, 2.6, and >190 nM, indicating that IGF-I and -II bind to the IGF-I receptor(s) with approximately equal high affinity. To further investigate the cellular mechanism of IGF actions, we have studied the effects of IGFs on two major signal transduction pathways: mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3 kinase). IGFs activated MAPK in zebrafish embryonic cells in a dose-dependent manner. This activation occurred within 5 min of IGF-I stimulation and disappeared after 1 h. IGF-I also caused a concentration-dependent activation of protein kinase B, a downstream target of PI3 kinase, this activation being sustained for several hours. Inhibition of MAPK activation by the MAPK kinase inhibitor PD-98059 inhibited the IGF-I-stimulated DNA synthesis. Similarly, use of the PI3 kinase inhibitor LY-294002 also inhibited IGF-I-stimulated DNA synthesis. When both the MAPK and PI3 kinase pathways were inhibited using a combination of these compounds, the IGF-I-stimulated DNA synthesis was completely negated. These results indicate that both IGF-I and -II are potent mitogens for zebrafish embryonic cells and that activation of both the MAPK and PI3 kinase-signaling pathways is required for the mitogenic action of IGFs in zebrafish embryonic cells.


Insulin-Like Growth Factor II/pharmacology , Insulin-Like Growth Factor I/pharmacology , MAP Kinase Signaling System/physiology , Phosphatidylinositol 3-Kinases/metabolism , Receptor, IGF Type 1/physiology , Receptor, IGF Type 2/physiology , Signal Transduction/physiology , Animals , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Chromones/pharmacology , Embryo, Nonmammalian , Flavonoids/pharmacology , Humans , Insulin/pharmacology , Kinetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 2/drug effects , Recombinant Proteins/pharmacology , Salmon , Signal Transduction/drug effects , Zebrafish
15.
Int J Mol Med ; 5(1): 33-41, 2000 Jan.
Article En | MEDLINE | ID: mdl-10601571

The Hep3B cell line analyzed in the present study is a widely used in vitro model in studies characterizing pathogenetic, functional, and therapeutic aspects of human hepatocellular carcinoma (HCC). Here we have determined the chromosomal composition using a combination of cytogenetic techniques. In agreement with the original description for this cell line, Hep3B was found to have a hypotriploid chromosome content carrying 59-63 chromosomes and no cytogenetic differences were demonstrated between early and late passages suggesting that this cell line has remained stable after repeated subculturing. Mutations and alterations of the IGF-axis as well as of chromosome 1p34, where the genes for histone deacetylase 1 (HDAC1) and transforming growth factor beta receptor interacting protein-1 (TRIP-1) map, are frequent events in hepatocarcinogenesis. This study characterizes the Hep3B cell line in detail at the karyotypic level, using comparative genomic hybridization (CGH), spectral karyotyping (SKY), G-banding and FISH techniques. We have also examined the effects of the histone deacetylase inhibitor trichostatin A (TSA) on members of the IGF-axis, and analysed them with regard to the karyotype. The results show that expression of one member of the IGF-axis, IGFBP-3, is greatly upregulated by treatment of Hep3B cells with TSA. As IGFBP-3 has been shown to induce apoptosis, these results suggest a possible use for histone deacetylase inhibitors and/or IGFBP-3 in the treatment of HCC.


Carcinoma, Hepatocellular/genetics , Hydroxamic Acids/pharmacology , Insulin-Like Growth Factor Binding Protein 3/genetics , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/therapy , Chromosome Banding , Chromosome Mapping , Eukaryotic Initiation Factor-3 , Humans , In Situ Hybridization, Fluorescence , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor II/pharmacology , Karyotyping , Liver Neoplasms/therapy , Ploidies , Proteins/genetics , Receptor, IGF Type 2/drug effects , Ribonucleases/metabolism , Tumor Cells, Cultured
16.
J Nutr ; 128(2): 158-65, 1998 Feb.
Article En | MEDLINE | ID: mdl-9446837

Epidemiologic data and animal models have demonstrated a correlation between dietary fat composition and colon cancer risk. We have previously found that dietary fat alters cell proliferation in rat colon, which may influence the risk of colon cancer. Growth factors, including insulin-like growth factor (IGF) I and II, regulate the cell cycle in most mammalian tissues. Hence, we measured IGF-I and IGF-II receptor expression in colonocytes from Sprague-Dawley rats fed diets containing either beef tallow (BT) or corn oil (CO) at 12, 30 or 37% of energy for 4 wk. Quantitative reverse transcriptase polymerase chain reaction (RT-PCR) using an internal standard was used to examine the relative expression of both IGF-I and II receptor mRNA in three sections of the colon. The IGF-I receptor protein was also measured by Western immunoblot. In the distal colon, IGF-I receptor gene expression and protein increased significantly as the percentage of CO increased. In both proximal and middle colon, an increased percentage of BT resulted in significantly increased IGF-II receptor expression. In the proximal colon, IGF-II receptor expression decreased with increasing CO concentration, whereas in the middle colon, rats fed 37% CO had significantly higher IGF-II receptor expression than rats fed 12 or 30% CO. IGF-II receptor gene expression in proximal colon decreased with increased fat quantity, independently of fat source, whereas in the middle colon, increased fat quantity resulted in increased IGF-II receptor expression. Thus IGF-I and IGF-II receptor mRNA and IGF-I receptor protein level in colon mucosa were significantly altered by dietary fat source and quantity, thereby suggesting a potential influence of dietary fat on the endocrine regulation of colon cell mitogenesis.


Colon/drug effects , Dietary Fats/pharmacology , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 2/drug effects , Animals , Cattle , Colon/metabolism , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Corn Oil/administration & dosage , Corn Oil/pharmacology , Dietary Fats/administration & dosage , Dose-Response Relationship, Drug , Gene Expression , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism
17.
Proc Natl Acad Sci U S A ; 94(12): 6232-7, 1997 Jun 10.
Article En | MEDLINE | ID: mdl-9177200

The insulin-like growth factor II (IGF-II)/mannose-6-phosphate (M-6-P) receptor is known to participate in endocytosis as well as sorting of lysosomal enzymes and is involved in membrane trafficking through rapid cycling between cytosolic membrane compartments and the plasma membrane. Here we demonstrate that IGF-II, acting through the IGF-II/M-6-P receptor, promotes exocytosis of insulin in the pancreatic beta cell. The effect of IGF-II was evoked at nonstimulatory concentrations of glucose, was mediated by a pertussis toxin sensitive GTP-binding protein, was dependent on protein kinase C-induced phosphorylation, and was independent of changes in cytoplasmic free Ca2+ concentration. Since the applied concentration of IGF-II is within the range normally found free in circulation in humans, this novel signaling pathway for the IGF-II/M-6-P receptor is likely to be involved in modulation of insulin exocytosis under physiological conditions.


Exocytosis/drug effects , Insulin-Like Growth Factor II/pharmacology , Insulin/metabolism , Islets of Langerhans/physiology , Receptor, IGF Type 2/physiology , Signal Transduction , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Glucose/pharmacology , Insulin Secretion , Insulin-Like Growth Factor II/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Mice , Mice, Obese , Protein Kinase C/metabolism , Receptor, IGF Type 2/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Tolbutamide/pharmacology
18.
Mol Biol Cell ; 8(4): 577-82, 1997 Apr.
Article En | MEDLINE | ID: mdl-9247639

Mannose 6-phosphate receptors carry newly synthesized lysosomal hydrolases from the trans-Golgi network to endosomes, then return to the trans-Golgi network for another round of enzyme delivery. Wortmannin, an inhibitor of phosphatidylinositol 3-kinase, interferes with the delivery of newly synthesized lysosomal enzymes to lysosomes. We used two independent assays of mannose 6-phosphate receptor trafficking to determine the precise step that is blocked by wortmannin. Using an assay that monitors resialylation of desialylated cell surface 300-kDa mannose 6-phosphate receptors, we found that receptor endocytosis and transport to the trans-Golgi network were not inhibited by 2 microM wortmannin. In addition, this concentration of drug had no effect on the transport of the mannose 6-phosphate receptor from late endosomes to the trans-Golgi network using a system that reconstitutes this transport process in cell extracts. Under the same conditions, wortmannin significantly inhibited the generation of mature cathepsin D. In addition, the structurally unrelated phosphatidylinositol 3-kinase inhibitor, LY294002, was also without effect when added to in vitro endosome-trans-Golgi network transport reactions. These experiments demonstrate that the interruption in lysosomal enzyme targeting is most likely due to a wortmannin-sensitive process required for the export of these receptors from the trans-Golgi network, consistent with the established role of phosphatidylinositol 3-kinase in the equivalent transport process in Saccharomyces cerevisiae.


Endosomes/metabolism , Golgi Apparatus/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptor, IGF Type 2/metabolism , Androstadienes/pharmacology , Animals , CHO Cells/drug effects , CHO Cells/metabolism , Cathepsin D/drug effects , Cathepsin D/metabolism , Chromones/pharmacology , Cricetinae , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Receptor, IGF Type 2/drug effects , Wortmannin
19.
Arch Toxicol ; 71(7): 409-15, 1997.
Article En | MEDLINE | ID: mdl-9209685

The effects of phenobarbitone and methylclofenapate were studied on the expression of growth factor and growth factor receptors in livers of male Wistar rats. The major findings were: (1) a significant reduction in epidermal growth factor receptor (EGFR) protein observed with both treatments, and (2) levels of EGFR transcripts were only slightly decreased with both compounds. The reduction in the receptor level therefore does not occur via regulation of transcription. Mannose-6-phosphate receptors (M6PR, also called insulin-like growth factor II receptor) and M6PR transcripts remained unchanged in both experimental groups. Hepatocyte growth factor receptor (HGFR) transcripts were also unchanged in both experimental groups. Transcript levels of transforming growth factor-beta 1 (TGF-beta 1) were lower in both treatment groups compared with the control; the reduction was significant in the methylclofenapate group. This may have relevance to the finding by others that nafenopin, another peroxisome proliferator, suppresses rat hepatocyte apoptosis. Another finding of general interest was that the three "housekeeping genes", namely albumin, actin and glyceraldehyde-3-phosphate dehydrogenase, were influenced by both treatments thus limiting their use as controls for gel loading. The adaptation of a growth regulatory mechanism via EGFR and its ligands may provide conditions such that cells with aberrant growth control have a selective growth advantage over normal cells thus promoting tumorigenesis.


Clofenapate/administration & dosage , Growth Substances/genetics , Liver/drug effects , Liver/metabolism , Phenobarbital/administration & dosage , Proteins/genetics , RNA, Messenger/metabolism , Receptors, Growth Factor/drug effects , Receptors, Growth Factor/genetics , Animals , Body Weight/drug effects , ErbB Receptors/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Growth Substances/metabolism , Male , Organ Size/drug effects , Proteins/drug effects , RNA, Messenger/drug effects , Rats , Rats, Wistar , Receptor, IGF Type 2/drug effects , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , Receptors, Growth Factor/metabolism , Transcription, Genetic/drug effects
20.
Proc Natl Acad Sci U S A ; 94(25): 13671-6, 1997 Dec 09.
Article En | MEDLINE | ID: mdl-9391084

Retinoic acid (RA) exerts diverse biological effects in the control of cell growth in embryogenesis and oncogenesis. These effects of RA are thought to be mediated by the nuclear retinoid receptors. Mannose-6-phosphate (M6P)/insulin-like growth factor-II (IGF-II) receptor is a multifunctional membrane glycoprotein that is known to bind both M6P and IGF-II and function primarily in the binding and trafficking of lysosomal enzymes, the activation of transforming growth factor-beta, and the degradation of IGF-II. M6P/IGF-II receptor has recently been implicated in fetal development and carcinogenesis. Despite the functional similarities between RA and the M6P/IGF-II receptor, no direct biochemical link has been established. Here, we show that the M6P/IGF-II receptor also binds RA with high affinity at a site that is distinct from those for M6P and IGF-II, as identified by a photoaffinity labeling technique. We also show that the binding of RA to the M6P/IGF-II receptor enhances the primary functions of this receptor. The biological consequence of the interaction appears to be the suppression of cell proliferation and/or induction of apoptosis. These findings suggest that the M6P/IGF-II receptor mediates a RA response pathway that is important in cell growth regulation. This discovery of the interaction of RA with the M6P/IGF-II receptor may have important implications for our understanding of the roles of RA and the M6P/IGF-II receptor in development, carcinogenesis, and lysosomal enzyme-related diseases.


Receptor, IGF Type 2/metabolism , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Animals , Binding Sites , Blood Proteins/metabolism , Cells, Cultured , Insulin-Like Growth Factor II/metabolism , Kinetics , Mannosephosphates/metabolism , Membrane Proteins/metabolism , Myocardium/metabolism , Photoaffinity Labels , Rats , Receptor, IGF Type 2/drug effects , Receptors, Retinoic Acid/drug effects , Tretinoin/pharmacology
...