51
|
Sanders EJ, Harvey S. Peptide hormones as developmental growth and differentiation factors. Dev Dyn 2008; 237:1537-52. [PMID: 18498096 DOI: 10.1002/dvdy.21573] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Peptide hormones, usually considered to be endocrine factors responsible for communication between tissues remotely located from each other, are increasingly being found to be synthesized in developing tissues, where they act locally. Several hormones are now known to be produced in developing tissues that are unrelated to the endocrine gland of origin in the adult. These hormones are synthesized locally, and are active as differentiation and survival factors, before the developing adult endocrine tissue becomes functional. There is increasing evidence for paracrine and/or autocrine actions for these factors during development, thus, placing them among the conventional growth and differentiation factors. We review the evidence for the view that thyroid hormones, growth hormone, prolactin, insulin, and parathyroid hormone-related protein are developmental growth and differentiation factors.
Collapse
Affiliation(s)
- Esmond J Sanders
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.
| | | |
Collapse
|
52
|
Biochemical constitution of extracellular medium is critical for control of human breast cancer MDA-MB-231 cell motility. J Membr Biol 2008; 223:27-36. [PMID: 18575796 DOI: 10.1007/s00232-008-9110-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2007] [Accepted: 05/07/2008] [Indexed: 01/04/2023]
Abstract
Although voltage-gated sodium channel (VGSC) activity, upregulated significantly in strongly metastatic human breast cancer cells, has been found to potentiate a variety of in vitro metastatic cell behaviors, the mechanism(s) regulating channel expression/activity is not clear. As a step toward identifying possible serum factors that might be responsible for this, we tested whether medium in which fetal bovine serum (FBS) was substituted with a commercial serum replacement agent (SR-2), comprising insulin and bovine serum albumin, would influence the VGSC-dependent in vitro metastatic cell behaviors. Human breast cancer MDA-MB-231 cells were used as a model. Measurements of lateral motility, transverse migration and adhesion showed consistently that the channel's involvement in metastatic cell behaviors depended on the extracellular biochemical conditions. In normal medium (5% FBS), tetrodotoxin (TTX), a highly specific blocker of VGSCs, suppressed these cellular behaviors, as reported before. In contrast, in SR-2 medium, TTX had opposite effects. However, blocking endogenous insulin/insulin-like growth factor receptor signaling with AG1024 eliminated or reversed the anomalous effects of TTX. Insulin added to serum-free medium increased migration, and TTX increased it further. In conclusion, (1) the biochemical constitution of the extracellular medium had a significant impact upon breast cancer cells' in vitro metastatic behaviors and (2) insulin, in particular, controlled the mode of the functional association between cells' VGSC activity and metastatic machinery.
Collapse
|
53
|
Hawkes C, Amritraj A, Macdonald RG, Jhamandas JH, Kar S. Heterotrimeric G proteins and the single-transmembrane domain IGF-II/M6P receptor: functional interaction and relevance to cell signaling. Mol Neurobiol 2008; 35:329-45. [PMID: 17917122 DOI: 10.1007/s12035-007-0021-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 11/30/1999] [Accepted: 04/02/2007] [Indexed: 12/11/2022]
Abstract
The G protein-coupled receptor (GPCR) family represents the largest and most versatile group of cell surface receptors. Classical GPCR signaling constitutes ligand binding to a seven-transmembrane domain receptor, receptor interaction with a heterotrimeric G protein, and the subsequent activation or inhibition of downstream intracellular effectors to mediate a cellular response. However, recent reports on direct, receptor-independent G protein activation, G protein-independent signaling by GPCRs, and signaling of nonheptahelical receptors via trimeric G proteins have highlighted the intrinsic complexities of G protein signaling mechanisms. The insulin-like growth factor-II/mannose-6 phosphate (IGF-II/M6P) receptor is a single-transmembrane glycoprotein whose principal function is the intracellular transport of lysosomal enzymes. In addition, the receptor also mediates some biological effects in response to IGF-II binding in both neuronal and nonneuronal systems. Multidisciplinary efforts to elucidate the intracellular signaling pathways that underlie these effects have generated data to suggest that the IGF-II/M6P receptor might mediate transmembrane signaling via a G protein-coupled mechanism. The purpose of this review is to outline the characteristics of traditional and nontraditional GPCRs, to relate the IGF-II/M6P receptor's structure with its role in G protein-coupled signaling and to summarize evidence gathered over the years regarding the putative signaling of the IGF-II/M6P receptor mediated by a G protein.
Collapse
Affiliation(s)
- C Hawkes
- Department of Psychiatry, Centre for Alzheimer and Neurodegenerative Research, University of Alberta, Edmonton, AB, T6G 2B7, Canada
| | | | | | | | | |
Collapse
|
54
|
Chavarría T, Valenciano AI, Mayordomo R, Egea J, Comella JX, Hallböök F, de Pablo F, de la Rosa EJ. Differential, age-dependent MEK-ERK and PI3K-Akt activation by insulin acting as a survival factor during embryonic retinal development. Dev Neurobiol 2007; 67:1777-88. [PMID: 17659595 DOI: 10.1002/dneu.20554] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Programmed cell death is a genuine developmental process of the nervous system, affecting not only projecting neurons but also proliferative neuroepithelial cells and young neuroblasts. The embryonic chick retina has been employed to correlate in vivo and in vitro studies on cell death regulation. We characterize here the role of two major signaling pathways, PI3K-Akt and MEK-ERK, in controlled retinal organotypic cultures from embryonic day 5 (E5) and E9, when cell death preferentially affects proliferating neuroepithelial cells and ganglion cell neurons, respectively. The relative density of programmed cell death in vivo was much higher in the proliferative and early neurogenic stages of retinal development (E3-E5) than during neuronal maturation and synaptogenesis (E8-E19). In organotypic cultures from E5 and E9 retinas, insulin, as the only growth factor added, was able to completely prevent cell death induced by growth factor deprivation. Insulin activated both the PI3K-Akt and the MEK-ERK pathways. Insulin survival effect, however, was differentially blocked at the two stages. At E5, the effect was blocked by MEK inhibitors, whereas at E9 it was blocked by PI3K inhibitors. The cells which were found to be dependent on insulin activation of the MEK-ERK pathway at E5 were mostly proliferative neuroepithelial cells. These observations support a remarkable specificity in the regulation of early neural cell death.
Collapse
Affiliation(s)
- Teresa Chavarría
- Department of Cellular and Molecular Physiopathology, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
55
|
Vaught JL, Contreras PC, Glicksman MA, Neff NT. Potential utility of rhIGF-1 in neuromuscular and/or degenerative disease. CIBA FOUNDATION SYMPOSIUM 2007; 196:18-27; discussion 27-38. [PMID: 8866126 DOI: 10.1002/9780470514863.ch3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neuromuscular/neurodegenerative disorders, such as the death of spinal cord motor neurons in amyotrophic lateral sclerosis (ALS) or the degeneration of spinal cord motor neuron axons in certain peripheral neuropathies, present a unique opportunity for therapeutic intervention with neurotrophic proteins. We have found that in mixed rat embryonic spinal cord cultures or in purified motor neuron preparations, recombinant human insulin-like growth factor 1 (rhIGF-1) enhances the survival of motor neurons at EC50 concentrations of 2 nM, consistent with an interaction at the tyrosine kinase-coupled rhIGF-1 receptor. In a model of programmed cell death in ovo, administration of rhIGF-1 produces a marked survival of motor neurons. In a variety of models of predominantly motor neuron or nerve injury in rodents, administration of rhIGF-1 prevents the death of motor neurons in neonatal facial nerve lesions, attenuates the loss of cholinergic phenotype in adult hypoglossal nerve axotomy and hastens recovery from sciatic nerve crush in mice. In a genetic model of motor neuron compromise, the wobbler mouse, rhIGF-1 (1 mg/kg s.c. daily) delayed the deterioration of grip strength and provided for a more normal distribution of fibre types. In addition, rhIGF-1 (0.3-1.0 mg/kg s.c. daily) prevents the motor and/or sensory neuropathy in rodents caused by vincristine, cisplatinum or Taxol. These combined data indicate that rhIGF-1 has marked effects on the survival of compromised motor neurons and the maintenance of their axons and functional connections. They also suggest the potential utility of rhIGF-1 for the treatment of diseases such as ALS and certain neuropathies.
Collapse
Affiliation(s)
- J L Vaught
- Cephalon Inc., West Chester, PA 19380, USA
| | | | | | | |
Collapse
|
56
|
Valenciano AI, Corrochano S, de Pablo F, de la Villa P, de la Rosa EJ. Proinsulin/insulin is synthesized locally and prevents caspase- and cathepsin-mediated cell death in the embryonic mouse retina. J Neurochem 2007; 99:524-36. [PMID: 17029604 DOI: 10.1111/j.1471-4159.2006.04043.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Programmed cell death is an essential, highly regulated process in neural development. Although the role of insulin-like growth factor I in supporting the survival of neural cells has been well characterized, studies on proinsulin/insulin are scarce. Here, we characterize proinsulin/insulin effects on cell death in embryonic day 15.5 mouse retina. Both proinsulin mRNA and proinsulin/insulin immunoreactivity were found in the developing retina. Organotypic embryonic day 15.5 retinas cultured under growth factor deprivation showed an increase in cell death that was reversed by proinsulin, insulin and insulin-like growth factor I, with similar median effective concentration values via phosphatidylinositol-3-kinase activation. Although insulin and insulin-like growth factor I provoked a sustained Akt phosphorylation, proinsulin-induced phosphorylation of Akt was not found. Analysis of the growth factor deprivation-induced cell death mechanisms, using caspase and cathepsin inhibitors, demonstrated that both protease families were required for the effective execution of cell death. The insulin survival effect, which decreased the extent and distribution of cell death to levels similar to those found in vivo, was not enhanced by simultaneous treatment with caspase and cathepsin inhibitors, suggesting that insulin interferes with these protease pathways in the embryonic mouse retina. The mechanisms characterized in this study provide new details on early neural cell death and its genuine regulation by insulin/proinsulin.
Collapse
Affiliation(s)
- Ana I Valenciano
- Group of Growth Factors in Vertebrate Development, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | | | | | | | |
Collapse
|
57
|
Otaegi G, Yusta-Boyo MJ, Vergaño-Vera E, Méndez-Gómez HR, Carrera AC, Abad JL, González M, de la Rosa EJ, Vicario-Abejón C, de Pablo F. Modulation of the PI 3-kinase-Akt signalling pathway by IGF-I and PTEN regulates the differentiation of neural stem/precursor cells. J Cell Sci 2006; 119:2739-48. [PMID: 16787946 DOI: 10.1242/jcs.03012] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neural stem cells depend on insulin-like growth factor I (IGF-I) for differentiation. We analysed how activation and inhibition of the PI 3-kinase-Akt signalling affects the number and differentiation of mouse olfactory bulb stem cells (OBSCs). Stimulation of the pathway with insulin and/or IGF-I, led to an increase in Akt phosphorylated on residues Ser473 and Thr308 (P-Akt(Ser473) and P-Akt(Thr308), respectively) in proliferating OBSCs, and in differentiating cells. Conversely, P-Akt(Ser473) levels decreased by 50% in the OB of embryonic day 16.5-18.5 IGF-I knockout mouse embryos. Overexpression of PTEN, a negative regulator of the PI 3-kinase pathway, caused a reduction in the basal levels of P-Akt(Ser473) and P-Akt(Thr308) and a minor reduction in IGF-I-stimulated P-Akt(Ser473). Although PTEN overexpression decreased the proportion of neurons and astrocytes in the absence of insulin/IGF-I, it did not alter the proliferation or survival of OBSCs. Accordingly, overexpression of a catalytically inactive PTEN mutant promoted OBSCs differentiation. Inhibition of PI 3-kinase by LY294002 produced strong and moderate reductions in IGF-I-stimulated P-Akt(Ser473) and P-Akt(Thr308), respectively. Consequently, LY294002 reduced the proliferation of OBSCs and the number of neurons and astrocytes, and also augmented cell death. These findings indicate that OBSC differentiation is more sensitive to lower basal levels of P-Akt than proliferation or death. By regulating P-Akt levels in opposite ways, IGF-I and PTEN contribute to the fine control of neurogenesis in the olfactory bulb.
Collapse
Affiliation(s)
- Gaizka Otaegi
- Group of Growth Factors in Vertebrate Development, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, Madrid 28040, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Ashokkumar N, Pari L, Ramkumar KM. N-Benzoyl-D-phenylalanine Attenuates Brain Acetylcholinesterase in Neonatal Streptozotocin-Diabetic Rats. Basic Clin Pharmacol Toxicol 2006; 99:246-50. [PMID: 16930298 DOI: 10.1111/j.1742-7843.2006.pto_487.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The effect of hyperglycaemia due to experimental diabetes in male Wistar rats causes a decrease in the level of acetylcholinesterase (AChE) with significant increase in lipid peroxidative markers: thiobarbituric acid-reactive substances (TBARS) and hydroperoxides in brains of experimental animals. The decreased activity of both salt soluble and detergent soluble acetylcholinesterase observed in diabetes may be attributed to lack of insulin which causes specific alterations in the level of neurotransmitter, thus causing brain dysfunction. Administration of non-sulfonylurea drug N-benzoyl-D-phenylalanine (NBDP) could protect against direct action of lipid peroxidation on brain AChE and in this way it might be useful in the prevention of cholinergic neural dysfunction, which is one of the major complications in diabetes.
Collapse
Affiliation(s)
- Natarajan Ashokkumar
- Department of Biochemistry, Faculty of Science, Annamalai University, Annamalainagar-608 002, Tamil Nadu, India
| | | | | |
Collapse
|
59
|
Hernández-Sánchez C, Mansilla A, de la Rosa EJ, de Pablo F. Proinsulin in development: New roles for an ancient prohormone. Diabetologia 2006; 49:1142-50. [PMID: 16596360 DOI: 10.1007/s00125-006-0232-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2005] [Accepted: 09/27/2005] [Indexed: 10/24/2022]
Abstract
In postnatal organisms, insulin is well known as an essential anabolic hormone responsible for maintaining glucose homeostasis. Its biosynthesis by the pancreatic beta cell has been considered a model of tissue-specific gene expression. However, proinsulin mRNA and protein have been found in embryonic stages before the formation of the pancreatic primordium, and later, in extrapancreatic tissues including the nervous system. Phylogenetic studies have also confirmed that production of insulin-like peptides antecedes the morphogenesis of a pancreas, and that these peptides contribute to normal development. In recent years, other roles for insulin distinct from its metabolic function have emerged also in vertebrates. During embryonic development, insulin acts as a survival factor and is involved in early morphogenesis. These findings are consistent with the observation that, at these stages, the proinsulin gene product remains as the precursor form, proinsulin. Independent of its low metabolic activity, proinsulin stimulates proliferation in developing neuroretina, as well as cell survival and cardiogenesis in early embryos. Insulin/proinsulin levels are finely regulated during development, since an excess of the protein interferes with correct morphogenesis and is deleterious for the embryo. This fine-tuned regulation is achieved by the expression of alternative embryonic proinsulin transcripts that have diminished translational activity.
Collapse
Affiliation(s)
- C Hernández-Sánchez
- Group of Growth Factors in Vertebrate Development, Centre of Biological Investigations (CIB), Spanish Council for Research (CSIC), Ramiro de Maeztu 9, E-28040 Madrid, Spain.
| | | | | | | |
Collapse
|
60
|
Seigel GM, Lupien SB, Campbell LM, Ishii DN. Systemic IGF-I treatment inhibits cell death in diabetic rat retina. J Diabetes Complications 2006; 20:196-204. [PMID: 16632241 DOI: 10.1016/j.jdiacomp.2005.06.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2004] [Revised: 01/28/2005] [Accepted: 06/20/2005] [Indexed: 10/24/2022]
Abstract
Diabetic retinopathy can result in apoptotic cell death of retinal neurons, as well as significant visual loss. It is further known that insulin-like growth factor (IGF) levels are reduced in diabetes and that IGF-I can prevent cell death in many cell types. In this study, we tested the hypothesis that systemic treatment with IGF-I could inhibit death of neuroretinal cells in diabetic rats by examining the expression of proapoptotic markers. In diabetic rat retina, the number of TUNEL-immunoreactive cells increased approximately sixfold in the photoreceptor layer (P<.001) and eightfold in the inner nuclear layer (INL; P<.001); phospho-Akt (p-Akt; Thr 308) immunoreactivity increased eightfold in the ganglion cell layer (GCL; P<.001) and threefold in the INL (P<.01). Subcutaneous IGF-I treatment significantly reduced the number of TUNEL (P<.001) and p-Akt immunoreactive retinal cells (P<.05) in diabetic rats approximately to the level of the nondiabetic group. Qualitative results showed that caspase-3 and BAD immunoreactivities were also elevated in diabetes and reduced in IGF-I-treated animals. Elevated TUNEL and p-Akt immunoreactivities were localized to distinct cell layers in the retina of diabetic rats. Early intervention with systemic IGF-I reduced the presence of proapoptotic markers indicative of neuroretinal cell death, despite ongoing hyperglycemia and weight loss. The eye is a special sensory organ, and these data show that cell loss in the nervous system, even in uncontrolled diabetes, can be prevented by IGF-I administration.
Collapse
Affiliation(s)
- Gail M Seigel
- Department of Ophthalmology, University at Buffalo, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
61
|
Browd SR, Kenney AM, Gottfried ON, Yoon JW, Walterhouse D, Pedone CA, Fults DW. N-myc can substitute for insulin-like growth factor signaling in a mouse model of sonic hedgehog-induced medulloblastoma. Cancer Res 2006; 66:2666-72. [PMID: 16510586 DOI: 10.1158/0008-5472.can-05-2198] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Medulloblastoma is a malignant brain tumor that arises in the cerebellum in children, presumably from granule neuron precursors (GNP). Advances in patient treatment have been hindered by a paucity of animal models that accurately reflect the molecular pathogenesis of human tumors. Aberrant activation of the Sonic hedgehog (Shh) and insulin-like growth factor (IGF) pathways is associated with human medulloblastomas. Both pathways are essential regulators of GNP proliferation during cerebellar development. In cultured GNPs, IGF signaling stabilizes the oncogenic transcription factor N-myc by inhibiting glycogen synthase kinase 3beta-dependent phosphorylation and consequent degradation of N-myc. However, determinants of Shh and IGF tumorigenicity in vivo remain unknown. Here we report a high frequency of medulloblastoma formation in mice following postnatal overexpression of Shh in cooperation with N-myc. Overexpression of N-myc, alone or in combination with IGF signaling mediators or with the Shh target Gli1, did not cause tumors. Thus, Shh has transforming functions in addition to induction of N-myc and Gli1. This tumor model will be useful for testing novel medulloblastoma therapies and providing insight into mechanisms of hedgehog-mediated transformation.
Collapse
Affiliation(s)
- Samuel R Browd
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah 84132-2303, USA
| | | | | | | | | | | | | |
Collapse
|
62
|
Ster J, Colomer C, Monzo C, Duvoid-Guillou A, Moos F, Alonso G, Hussy N. Insulin-like growth factor-1 inhibits adult supraoptic neurons via complementary modulation of mechanoreceptors and glycine receptors. J Neurosci 2006; 25:2267-76. [PMID: 15745952 PMCID: PMC6726091 DOI: 10.1523/jneurosci.4053-04.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In the CNS, insulin-like growth factor-1 (IGF-1) is mainly known for its trophic effect both during development and in adulthood. Here, we show than in adult rat supraoptic nucleus (SON), IGF-1 receptor immunoreactivity is present in neurons, whereas IGF-1 immunoreactivity is found principally in astrocytes and more moderately in neurons. In vivo application of IGF-1 within the SON acutely inhibits the activity of both vasopressin and oxytocin neurons, the two populations of SON neuroendocrine cells. Recordings of acutely isolated SON neurons showed that this inhibition occurs through two rapid and reversible mechanisms, both involving the neuronal IGF-1 receptor but different intracellular messengers. IGF-1 inhibits Gd3+-sensitive and osmosensitive mechanoreceptor cation current via phosphatidylinositol-3 (PI3) kinase activation. IGF-1 also potentiates taurine-activated glycine receptor (GlyR) Cl- currents by increasing the agonist sensitivity through a extremely rapid (within a second) PI3 kinase-independent mechanism. Both mechanoreceptor channels and GlyR, which form the excitatory and inhibitory components of SON neuron osmosensitivity, are active at rest, and their respective inhibition and potentiation will both be inhibitory, leading to strong decrease in neuronal activity. It will be of interest to determine whether IGF-1 is released by neurons, thus participating in an inhibitory autocontrol, or astrocytes, then joining the growing family of glia-to-neuron transmitters that modulate neuronal and synaptic activity. Through the opposite and complementary acute regulation of mechanoreceptors and GlyR, IGF-1 appears as a new important neuromodulator in the adult CNS, participating in the complex integration of neural messages that regulates the level of neuronal excitability.
Collapse
Affiliation(s)
- Jeanne Ster
- Biologie des Neurones Endocrines, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 5101, Institut National de la Santé et de la Recherche Médicale de Pharmacologie et d'Endocrinologie, 34094 Montpellier Cedex 5, France
| | | | | | | | | | | | | |
Collapse
|
63
|
Velázquez PN, Peralta I, Bobes RJ, Romano MC. Insulin Stimulates Proliferation but Not 17β-Estradiol Production in Cultured Chick Embryo Ovarian Cells. Poult Sci 2006; 85:100-5. [PMID: 16493952 DOI: 10.1093/ps/85.1.100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The development of the chick embryo gonads is influenced by gonadotropins [follicle-stimulating hormone (FSH), luteinizing hormone, human chorionic gonadotropin (hCG)]. We have previously shown that insulin enhanced the production of androgens in the testis of the newly hatched chicken and increased the proliferation of chick embryo testis cells. In the present paper, we have studied the effect of insulin on embryonic chick embryo ovarian cells and compared them with those of human FSH and hCG. The ovaries of 18-d-old chick embryos were dissociated and cultured for different periods in Dulbecco's modified Eagle's medium in the presence and absence of insulin, human FSH, hCG, and combinations of them. 3H-thymidine incorporation was used as an indicator of cell proliferation; steroids were measured by radioimmunoassay. Results showed that insulin enhanced the proliferation of ovarian cells in a dose- and time-dependent manner. Gonadotropins did not affect significantly the ovarian cell proliferation. Insulin did not change 17beta-estradiol production. The combination of insulin and FSH or insulin and hCG decreased the stimulation of estrogen secretion caused by the addition of the gonadotropins. In some experiments, ovarian cells were cultured with or without insulin, and subpopulations were identified. The results showed that insulin but not human FSH or hCG increased the proliferation of germinal cells after 60 h in culture. Insulin and human FSH did stimulate the other 2 subpopulations. In summary, present results suggest that insulin is an important hormone in the development of the chick embryo ovary.
Collapse
Affiliation(s)
- P N Velázquez
- Dpto de Biología Celular y Tisular, Fac Medicina, UNAM, México DF
| | | | | | | |
Collapse
|
64
|
Colombo E, Giannelli SG, Galli R, Tagliafico E, Foroni C, Tenedini E, Ferrari S, Ferrari S, Corte G, Vescovi A, Cossu G, Broccoli V. Embryonic stem-derived versus somatic neural stem cells: a comparative analysis of their developmental potential and molecular phenotype. Stem Cells 2005; 24:825-34. [PMID: 16339994 DOI: 10.1634/stemcells.2005-0313] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Reliable procedures to induce neural commitment of totipotent undifferentiated embryonic stem (ES) cells have provided new tools for investigating the molecular mechanisms underlying cell fate choices. We extensively characterized the developmental potential of ES-induced neural cells obtained using an adaptation of the multistep induction protocol. We provided evidence that ES-derived neural proliferating cells are endowed with stem cell properties such as extensive self-renewal capacity and single-cell multipotency. In differentiating conditions, cells matured exclusively into neurons, astrocytes, and oligodendrocytes. All these features have been previously described in only somatic neural stem cells (NSCs). Therefore, we consider it more appropriate to rename our cells ES-derived NSCs. These similarities between the two NSC populations induced us to carefully compare their proliferation ability and differentiation potential. Although they were very similar in overall behavior, we scored specific differences. For instance, ES-derived NSCs proliferated at higher rate and consistently generated a higher number of neurons compared with somatic NSCs. To further investigate their relationships, we carried out a molecular analysis comparing their transcriptional profiles during proliferation. We observed a large fraction of shared expressed transcripts, including genes previously described to be critical in defining somatic NSC traits. Among the genes differently expressed, candidate genes possibly responsible for divergences between the two cell types were selected and further investigated. In particular, we showed that an enhanced MAPK (mitogen-activated protein kinase) signaling is acting in ES-induced NSCs, probably triggered by insulin-like growth factor-II. This may contribute to the high proliferation rate exhibited by these cells in culture.
Collapse
Affiliation(s)
- Elena Colombo
- Stem Cell Research Department, Dipartmento di Biotecnologie, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Torres Aleman I. Role of Insulin-Like Growth Factors in Neuronal Plasticity and Neuroprotection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2005; 567:243-58. [PMID: 16370142 DOI: 10.1007/0-387-26274-1_10] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
66
|
Fernandez AM, Carro EM, Lopez-Lopez C, Torres-Aleman I. Insulin-like growth factor I treatment for cerebellar ataxia: Addressing a common pathway in the pathological cascade? ACTA ACUST UNITED AC 2005; 50:134-41. [PMID: 15950289 DOI: 10.1016/j.brainresrev.2005.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2004] [Revised: 05/06/2005] [Accepted: 05/06/2005] [Indexed: 12/21/2022]
Abstract
In the present work we review evidence supporting the use of insulin-like growth factor I (IGF-I) for treatment of cerebellar ataxia, a heterogeneous group of neurodegenerative diseases of low incidence but high societal impact. Most types of ataxia display not only motor discoordination, but also additional neurological problems including peripheral nerve dysfunctions. Therefore, a feasible therapy should combine different strategies aimed to correct the various disturbances specific for each type of ataxia. For cerebellar deficits, and most probably also for other types of brain deficits, the use of a wide-spectrum neuroprotective factor such as IGF-I may prove beneficial. Intriguingly, both ataxic animals as well as human patients show altered serum IGF-I levels. While the pathogenic significance of IGF-I, if any, in this varied group of diseases is difficult to envisage, disrupted IGF-I neuroprotective signaling may constitute a common stage in the pathological cascade associated to neuronal death. Indeed, treatment with IGF-I has proven effective in animal models of ataxia. Based on this pre-clinical evidence we propose that IGF-I should be tested in clinical trials of cerebellar ataxia in those cases where either serum IGF-I deficiency (as in primary cerebellar atrophy) or loss of sensitivity to IGF-I (as in ataxia telangiectasia) has been reported. Taking advantage of the widely protective and anabolic actions of IGF-I on peripheral tissues, this neurotrophic factor may provide additional therapeutic advantages for many of the disturbances commonly associated to ataxia such as cardiopathy, muscle wasting, or immune dysfunction.
Collapse
Affiliation(s)
- A M Fernandez
- Laboratory of Neuroendocrinology, Cajal Institute, CSIC, Avda. Dr. Arce 37, 28002 Madrid, Spain
| | | | | | | |
Collapse
|
67
|
Das AV, Edakkot S, Thoreson WB, James J, Bhattacharya S, Ahmad I. Membrane properties of retinal stem cells/progenitors. Prog Retin Eye Res 2005; 24:663-81. [PMID: 15939659 DOI: 10.1016/j.preteyeres.2005.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The membrane properties of cells help integrate extrinsic information relayed through growth factors, chemokines, extracellular matrix, gap junctions and neurotransmitters towards modulating cell-intrinsic properties, which in turn determine whether cells remain quiescent, proliferate, differentiate, establish contact with other cells or remove themselves by activating programmed cell death. This review highlights some of the membrane properties of early and late retinal stem cells/progenitors, which are likely to be helpful in the identification and enrichment of these cells and in understanding mechanisms underlying their maintenance and differentiation. Understanding of membrane properties of retinal stem cells/progenitors is essential for the successful formulation of approaches to treat retinal degeneration and diseases by cell therapy.
Collapse
Affiliation(s)
- Ani V Das
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198-7691, USA
| | | | | | | | | | | |
Collapse
|
68
|
Levy YS, Gilgun-Sherki Y, Melamed E, Offen D. Therapeutic potential of neurotrophic factors in neurodegenerative diseases. BioDrugs 2005; 19:97-127. [PMID: 15807629 DOI: 10.2165/00063030-200519020-00003] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
There is a vast amount of evidence indicating that neurotrophic factors play a major role in the development, maintenance, and survival of neurons and neuron-supporting cells such as glia and oligodendrocytes. In addition, it is well known that alterations in levels of neurotrophic factors or their receptors can lead to neuronal death and contribute to the pathogenesis of neurodegenerative diseases such as Parkinson disease, Alzheimer disease, Huntington disease, amyotrophic lateral sclerosis, and also aging. Although various treatments alleviate the symptoms of neurodegenerative diseases, none of them prevent or halt the neurodegenerative process. The high potency of neurotrophic factors, as shown by many experimental studies, makes them a rational candidate co-therapeutic agent in neurodegenerative disease. However, in practice, their clinical use is limited because of difficulties in protein delivery and pharmacokinetics in the central nervous system. To overcome these disadvantages and to facilitate the development of drugs with improved pharmacotherapeutic profiles, research is underway on neurotrophic factors and their receptors, and the molecular mechanisms by which they work, together with the development of new technologies for their delivery into the brain.
Collapse
Affiliation(s)
- Yossef S Levy
- Laboratory of Neuroscineces, Felsenstein Medical Research Center, Israel
| | | | | | | |
Collapse
|
69
|
Carro E, Trejo JL, Spuch C, Bohl D, Heard JM, Torres-Aleman I. Blockade of the insulin-like growth factor I receptor in the choroid plexus originates Alzheimer's-like neuropathology in rodents: new cues into the human disease? Neurobiol Aging 2005; 27:1618-31. [PMID: 16274856 DOI: 10.1016/j.neurobiolaging.2005.09.039] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 09/02/2005] [Accepted: 09/27/2005] [Indexed: 11/29/2022]
Abstract
The possibility that perturbed insulin/insulin-like growth factor I (IGF-I) signalling is involved in development of late-onset forms of Alzheimer's disease (AD) is gaining increasing attention. We recently reported that circulating IGF-I participates in brain amyloid beta (Abeta) clearance by modulating choroid plexus function. We now present evidence that blockade of the IGF-I receptor in the choroid plexus originates changes in brain that are reminiscent of those found in AD. In rodents, IGF-I receptor impairment led to brain amyloidosis, cognitive disturbance, and hyperphosphorylated tau deposits together with other changes found in Alzheimer's disease such as gliosis and synaptic protein loss. While these disturbances were mostly corrected by restoring receptor function, blockade of the IGF-I receptor exacerbated AD-like pathology in old mutant mice already affected of brain amyloidosis and cognitive derangement. These findings may provide new cues into the causes of late-onset Alzheimer's disease in humans giving credence to the notion that an abnormal age-associated decline in IGF-I input to the choroid plexus may contribute to development of AD in genetically prone subjects.
Collapse
Affiliation(s)
- Eva Carro
- Laboratory of Neuroendocrinology, Cajal Institute, CSIC, Avda. Dr. Arce 37, 28002 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
70
|
Harris PW, Brimble MA, Muir VJ, Lai MY, Trotter NS, Callis DJ. Synthesis of proline-modified analogues of the neuroprotective agent glycyl-l-prolyl-glutamic acid (GPE). Tetrahedron 2005. [DOI: 10.1016/j.tet.2005.08.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
71
|
Hernández-Sánchez C, Bártulos O, de Pablo F. Proinsulin: much more than a hormone precursor in development. Rev Endocr Metab Disord 2005; 6:211-6. [PMID: 16151625 DOI: 10.1007/s11154-005-3052-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Catalina Hernández-Sánchez
- Group of Growth Factors in Vertebrate Development, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, E-28040, Madrid, Spain.
| | | | | |
Collapse
|
72
|
Nagaraja TN, Patel P, Gorski M, Gorevic PD, Patlak CS, Fenstermacher JD. In normal rat, intraventricularly administered insulin-like growth factor-1 is rapidly cleared from CSF with limited distribution into brain. Cerebrospinal Fluid Res 2005; 2:5. [PMID: 16045806 PMCID: PMC1190198 DOI: 10.1186/1743-8454-2-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Accepted: 07/26/2005] [Indexed: 01/23/2023] Open
Abstract
Background Putatively active drugs are often intraventricularly administered to gain direct access to brain and circumvent the blood-brain barrier. A few studies on the normal central nervous system (CNS) have shown, however, that the distribution of materials after intraventricular injections is much more limited than presumed and their exit from cerebrospinal fluid (CSF) is more rapid than generally believed. In this study, we report the intracranial distribution and the clearance from CSF and adjacent CNS tissue of radiolabeled insulin-like growth factor-1 after injection into one lateral ventricle of the normal rat brain. Methods Under barbiturate anesthesia, 125I-labeled insulin-like growth factor-1 (IGF-1) was injected into one lateral ventricle of normal Sprague-Dawley rats. The subsequent distribution of IGF-1 through the cerebrospinal fluid (CSF) system and into brain, cerebral blood vessels, and systemic blood was measured over time by gamma counting and quantitative autoradiography (QAR). Results Within 5 min of infusion, IGF-1 had spread from the infused lateral ventricle into and through the third and fourth ventricles. At this time, 25% of the infused IGF-1 had disappeared from the CSF-brain-meningeal system; the half time of this loss was 12 min. The plasma concentration of cleared IGF-1 was, however, very low from 2 to 9 min and only began to rise markedly after 20 min. This delay between loss and gain plus the lack of radiotracer in the cortical subarachnoid space suggested that much of the IGF-1 was cleared into blood via the cranial and/or spinal nerve roots and their associated lymphatic systems rather than periventricular tissue and arachnoid villi. Less than 10% of the injected radioactivity remained in the CSF-brain system after 180 min. The CSF and arteries and arterioles within the subarachnoid cisterns were labeled with IGF-1 within 10 min. Between 60 and 180 min, most of the radioactivity within the cranium was retained within and around these blood vessels and by periaqueductal gray matter. Tissue profiles at two sites next to ventricular CSF showed that IGF-1 penetrated less than 1.25 mm into brain tissue and appreciable 125I-activity remained at the tissue-ventricular CSF interface after 180 min. Conclusion Our findings suggest that entry of IGF-1 into normal brain parenchyma after lateral ventricle administration is limited by rapid clearance from CSF and brain and slow movement, apparently by diffusion, into the periventricular tissue. Various growth factors and other neuroactive agents have been reported to be neuroprotective within the injured brain after intraventricular administration. It is postulated that the delivery of such factors to neurons and glia in the injured brain may be facilitated by abnormal CSF flow. These several observations suggest that the flow of CSF and entrained solutes may differ considerably between normal and abnormal brain and even among various neuropathologies.
Collapse
Affiliation(s)
| | - Padma Patel
- Department of Anesthesiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Martin Gorski
- Department of Anesthesiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Peter D Gorevic
- Department of Medicine, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Clifford S Patlak
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
73
|
Duenker N, Valenciano AI, Franke A, Hernández-Sánchez C, Dressel R, Behrendt M, De Pablo F, Krieglstein K, de la Rosa EJ. Balance of pro-apoptotic transforming growth factor-β and anti-apoptotic insulin effects in the control of cell death in the postnatal mouse retina. Eur J Neurosci 2005; 22:28-38. [PMID: 16029193 DOI: 10.1111/j.1460-9568.2005.04183.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Transforming growth factor (TGF)-beta and insulin display opposite effects in regulating programmed cell death during vertebrate retina development; the former induces apoptosis while the latter prevents it. In the present study we investigated coordinated actions of TGF-beta and insulin in an organotypic culture system of early postnatal mouse retina. Addition of exogenous TGF-beta resulted in a significant increase in cell death whereas exogenous insulin attenuated apoptosis and was capable of blocking TGF-beta-induced apoptosis. This effect appeared to be modulated via insulin-induced transcriptional down-regulation of TGF-beta receptor II levels. The analysis of downstream signalling molecules also revealed opposite effects of both factors; insulin provided survival signalling by increasing the level of anti-apoptotic Bcl-2 protein expression and phosphorylation and down-regulating caspase 3 activity whereas pro-apoptotic TGF-beta signalling reduced Bcl-2 mRNA levels and Bcl-2 phosphorylation and induced the expression of TGF-induced immediate-early gene (TIEG), a Krüppel-like zinc-finger transcription factor, mimicking TGF-beta activity.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Apoptosis/physiology
- Caspase 3
- Caspases/genetics
- Caspases/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cells, Cultured
- DNA-Binding Proteins/drug effects
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Down-Regulation/drug effects
- Down-Regulation/physiology
- Drug Interactions/physiology
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Insulin/metabolism
- Insulin/pharmacology
- Mice
- Mice, Inbred C57BL
- Neurons/drug effects
- Neurons/metabolism
- Organ Culture Techniques
- Organogenesis/drug effects
- Organogenesis/physiology
- Phosphorylation/drug effects
- Protein Serine-Threonine Kinases
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/drug effects
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Retina/drug effects
- Retina/growth & development
- Retina/metabolism
- Smad Proteins
- Trans-Activators/drug effects
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcriptional Activation/drug effects
- Transcriptional Activation/genetics
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- N Duenker
- Centre of Anatomy, Department of Neuroanatomy, University of Göttingen, Göttingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Hartmann W, Koch A, Brune H, Waha A, Schüller U, Dani I, Denkhaus D, Langmann W, Bode U, Wiestler OD, Schilling K, Pietsch T. Insulin-like growth factor II is involved in the proliferation control of medulloblastoma and its cerebellar precursor cells. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1153-62. [PMID: 15793295 PMCID: PMC1602379 DOI: 10.1016/s0002-9440(10)62335-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Medulloblastomas (MBs), the most frequent malignant brain tumors of childhood, presumably originate from cerebellar neural precursor cells. An essential fetal mitogen involved in the pathogenesis of different embryonal tumors is insulin-like growth factor II (IGF-II). We screened human MB biopsies of the classic (CMB) and desmoplastic (DMB) variants for IGF2 transcripts of the four IGF2 promoters. We found IGF2 transcription from the imprinted promoter P3 to be significantly increased in the desmoplastic variant compared to the classic subgroup. This was not a result of loss of imprinting of IGF2 in desmoplastic tumors. We next examined the interaction of IGF-II and Sonic hedgehog (Shh), which serves as a critical mitogen for cerebellar granule cell precursors (GCPs) in the external granule cell layer from which DMBs are believed to originate. Mutations of genes encoding components of the Shh-Patched signaling pathway occur in approximately 50% of DMBs. To analyze the effects of IGF-II on Hedgehog signaling, we cultured murine GCP and human MB cells in the presence of Shh and Igf-II. In GCPs, a synergistic effect of Shh and Igf-II on proliferation and gli1 and cyclin D1 mRNA expression was found. Igf-II, but not Shh, induced phosphorylation of Akt and its downstream target Gsk-3beta. In six of nine human MB cell lines IGF-II displayed a growth-promoting effect that was mediated mainly through the IGF-I receptor. Together, our data point to an important role of IGF-II for the proliferation control of both cerebellar neural precursors and MB cells.
Collapse
Affiliation(s)
- Wolfgang Hartmann
- Department of Neuropathology, University of Bonn Medical Center, Sigmund-Freud-St. 25, D-53105 Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Smith A, Chan SJ, Gutiérrez J. Autoradiographic and immunohistochemical localization of insulin-like growth factor-I receptor binding sites in brain of the brown trout, Salmo trutta. Gen Comp Endocrinol 2005; 141:203-13. [PMID: 15804507 DOI: 10.1016/j.ygcen.2004.12.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2004] [Revised: 11/26/2004] [Accepted: 12/16/2004] [Indexed: 11/22/2022]
Abstract
Insulin-like growth factor-I (IGF-I), a peptide closely related to insulin, is known to play crucial roles in brain development. While the central sites of action of IGF-I in higher vertebrates are now well established, surprisingly little is known in the teleost model where the brain undergoes continual, indeterminate, growth. In this study, we have mapped the distribution of putative IGF-I receptor (IGF-IR) binding sites in the brain of the brown trout using both ligand binding in vitro autoradiography and immunohistochemistry. The presence of IGF binding proteins (IGFBPs) was further studied by competitive inhibition using unlabelled IGF-I and des-(1-3)-IGF-I. In both juvenile and adult trout brain, [125I]IGF-I binding was highest in cerebellum and optic tectum, both regions of the teleost brain known to grow the most actively throughout life. At the cellular level, IGF-IR immunoreactivity was confirmed on cell bodies and dendrites, particularly of larger presumptive neurons including purkinje cells and dendritic fibres throughout the molecular layer of the cerebellum. Abundant IGF-IR expression in hypothalamic regions may further be related to neuron growth while a possible hypophysiotropic role will require further investigation. Competitive inhibition studies employing des-(1-3)-IGF-I also suggest IGFBPs are present in all regions exhibiting high [125I]IGF-I ligand binding and confirms the presence of this important regulatory component of the IGF-I system in the teleost brain. The importance of the IGF-I system in brain development, particularly in regions such as the cerebellum, together with the continual lifetime growth of the fish central nervous system, suggest the teleost brain is an extremely useful site for studying the actions of IGF-I in relation to neuron proliferation, growth, and survival in an adult brain.
Collapse
Affiliation(s)
- Alastair Smith
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Private Bag 11222, Palmerston North, New Zealand
| | | | | |
Collapse
|
76
|
Diaz-Casares A, Leon Y, de la Rosa EJ, Varela-Nieto I. Regulation of Vertebrate Sensory Organ Development: A Scenario for Growth Hormone and Insulin-Like Growth Factors Action. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2005; 567:221-42. [PMID: 16370141 DOI: 10.1007/0-387-26274-1_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Amelia Diaz-Casares
- Instituto de Investigaciones Biomedicas Alberto Sols, Consejo Superior de Investigaciones Cientificas-Universidad Autonoma de Madrid, Spain
| | | | | | | |
Collapse
|
77
|
Lai MYH, Brimble MA, Callis DJ, Harris PWR, Levi MS, Sieg F. Synthesis and pharmacological evaluation of glycine-modified analogues of the neuroprotective agent glycyl-l-prolyl-l-glutamic acid (GPE). Bioorg Med Chem 2005; 13:533-48. [PMID: 15598574 DOI: 10.1016/j.bmc.2004.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Revised: 10/01/2004] [Accepted: 10/04/2004] [Indexed: 11/18/2022]
Abstract
The synthesis of 10 G*PE analogues, wherein the glycine residue has been modified, is described by coupling readily accessible dibenzyl-L-prolyl-L-glutamate 2 with various analogues of glycine. Pharmacological evaluation of the novel compounds was undertaken to further understand the role of the glycine residue on the observed neuroprotective properties of the endogenous tripeptide GPE.
Collapse
Affiliation(s)
- Michelle Y H Lai
- Neuren Pharmaceuticals Medicinal Chemistry Group, Department of Chemistry, University of Auckland, 23 Symonds Street, Auckland 1000, New Zealand
| | | | | | | | | | | |
Collapse
|
78
|
Trotter NS, Brimble MA, Harris PWR, Callis DJ, Sieg F. Synthesis and neuroprotective activity of analogues of glycyl-l-prolyl-l-glutamic acid (GPE) modified at the α-carboxylic acid. Bioorg Med Chem 2005; 13:501-17. [PMID: 15598572 DOI: 10.1016/j.bmc.2004.10.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Revised: 10/01/2004] [Accepted: 10/04/2004] [Indexed: 11/16/2022]
Abstract
The synthesis of nine GPE* analogues, wherein the alpha-carboxylic acid group of glutamic acid has been modified, is described by coupling readily accessible N-benzyloxycarbonyl-glycyl-L-proline 2 with various analogues of glutamic acid. Pharmacological evaluation of the novel compounds was undertaken to further understand the role of the glutamate residue on the observed neuroprotective properties of the endogenous tripeptide GPE.
Collapse
Affiliation(s)
- Nicholas S Trotter
- Neuren Pharmaceuticals Medicinal Chemistry Group, Department of Chemistry, University of Auckland, 23 Symonds Street, Auckland 1000, New Zealand
| | | | | | | | | |
Collapse
|
79
|
Brimble MA, Trotter NS, Harris PWR, Sieg F. Synthesis and pharmacological evaluation of side chain modified glutamic acid analogues of the neuroprotective agent glycyl-l-prolyl-l-glutamic acid (GPE). Bioorg Med Chem 2005; 13:519-32. [PMID: 15598573 DOI: 10.1016/j.bmc.2004.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Revised: 10/01/2004] [Accepted: 10/04/2004] [Indexed: 11/16/2022]
Abstract
The synthesis of eight GPE* analogues, wherein the gamma-carboxylic moiety of the glutamic residue has been modified, is described by coupling readily accessible N-benzyloxycarbonyl-glycyl-L-proline with various analogues of glutamic acid. Pharmacological evaluation of the novel compounds was undertaken to further understand the role of the glutamate residue on the observed neuroprotective properties of the endogenous tripeptide GPE.
Collapse
Affiliation(s)
- Margaret A Brimble
- Neuren Pharmaceuticals, Medicinal Chemistry Group, Department of Chemistry, University of Auckland, 23 Symonds Street, Auckland 1000, New Zealand.
| | | | | | | |
Collapse
|
80
|
Milosevic A, Goldman JE. Potential of progenitors from postnatal cerebellar neuroepithelium and white matter: lineage specified vs. multipotent fate. Mol Cell Neurosci 2004; 26:342-53. [PMID: 15207858 DOI: 10.1016/j.mcn.2004.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2003] [Revised: 02/20/2004] [Accepted: 02/20/2004] [Indexed: 10/26/2022] Open
Abstract
Progenitors that migrate through the white matter of the postnatal cerebellum give rise to interneurons, astrocytes, and oligodendrocytes. To investigate the lineage potential of progenitors from the neuroepithelium and the white matter, we performed an in vitro clonal analysis in the presence or absence of various growth factors. Clonal progeny of cells labeled with a green fluorescent protein (GFP)-expressing retrovirus was characterized using morphological features and lineage markers. The large majority of clones were homogeneous, containing astrocytes, oligodendrocytes, neurons, or hybrid progenitors-cells labeled with markers for astrocytes and oligodendrocytes. Heterogeneous clones consisted of astrocytes and oligodendrocytes, with only a few mixed glial-neuronal clones. The neuroepithelium contains a higher number of multipotent progenitors than the white matter, pointing to a lineage specification of most of the cerebellar progenitors before their migration to the white matter.
Collapse
Affiliation(s)
- Ana Milosevic
- Division of Neuropathology, Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | | |
Collapse
|
81
|
Broccolini A, Ricci E, Pescatori M, Papacci M, Gliubizzi C, D'Amico A, Servidei S, Tonali P, Mirabella M. Insulin-like growth factor I in inclusion-body myositis and human muscle cultures. J Neuropathol Exp Neurol 2004; 63:650-9. [PMID: 15217093 DOI: 10.1093/jnen/63.6.650] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Possible pathogenic mechanisms of sporadic inclusion-body myositis (sIBM) include abnormal production and accumulation of amyloid beta (A beta), muscle aging, and increased oxidative stress. Insulin-like growth factor I (IGF-I), an endocrine and autocrine/paracrine trophic factor, provides resistance against A beta toxicity and oxidative stress in vitro and promotes cell survival. In this study we analyzed the IGF-I signaling pathway in sIBM muscle and found that 16.2% +/- 2.5% of nonregenerating fibers showed increased expression of IGF-I, phosphatidylinositide 3'OH-kinase, and Akt. In the majority of sIBM abnormal muscle fibers, increased IGF-I mRNA and protein correlated with the presence of A beta cytoplasmic inclusions. To investigate a possible relationship between A beta toxicity and IGF-I upregulation, normal primary muscle cultures were stimulated for 24 hours with the A beta(25-35) peptide corresponding to the biologically active domain of A beta. This induced an increase of IGF-I mRNA and protein in myotubes at 6 hours, followed by a gradual reduction thereafter. The level of phosphorylated Akt showed similar changes. We suggest that in sIBM. IGF-I overexpression represents a reactive response to A beta toxicity, possibly providing trophic support to vulnerable fibers. Understanding the signaling pathways activated by IGF-I in sIBM may lead to novel therapeutic strategies for the disease.
Collapse
|
82
|
Hawkes C, Kar S. The insulin-like growth factor-II/mannose-6-phosphate receptor: structure, distribution and function in the central nervous system. ACTA ACUST UNITED AC 2004; 44:117-40. [PMID: 15003389 DOI: 10.1016/j.brainresrev.2003.11.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2003] [Indexed: 01/25/2023]
Abstract
The insulin-like growth factor-II/mannose-6-phosphate (IGF-II/M6P) receptor is a multifunctional single transmembrane glycoprotein which, along with the cation-dependent M6P (CD-M6P) receptor, mediates the trafficking of M6P-containing lysosomal enzymes from the trans-Golgi network (TGN) to lysosomes. Cell surface IGF-II/M6P receptors also function in the degradation of the non-glycosylated IGF-II polypeptide hormone, as well as in the capture and activation/degradation of extracellular M6P-bearing ligands. In recent years, the multifaceted role of the receptor has become apparent, as several lines of evidence have indicated that in addition to its role in lysosomal enzyme trafficking, clearance and/or activation of a variety of growth factors and endocytosis-mediated degradation of IGF-II, the IGF-II/M6P receptor may also mediate transmembrane signal transduction in response to IGF-II binding under certain conditions. However, very little is known about the physiological significance of the receptor in the function of the central nervous system (CNS). This review aims to delineate what is currently known about IGF-II/M6P receptor structure, its ligand binding properties and role in lysosomal enzyme transport. It also summarizes the recent data regarding the role of the receptor in the CNS, including its distribution, possible importance for normal and activity-dependent functioning as well as its implications in neurodegenerative disorders such as Alzheimer's disease (AD).
Collapse
Affiliation(s)
- C Hawkes
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H4H 1R3
| | | |
Collapse
|
83
|
Carro E, Torres-Aleman I. The role of insulin and insulin-like growth factor I in the molecular and cellular mechanisms underlying the pathology of Alzheimer's disease. Eur J Pharmacol 2004; 490:127-33. [PMID: 15094079 DOI: 10.1016/j.ejphar.2004.02.050] [Citation(s) in RCA: 201] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2004] [Indexed: 11/26/2022]
Abstract
Cellular and molecular processes leading to abnormal accumulation of beta amyloid in the brain are slowly being uncovered. A potential involvement of insulin and insulin-like growth factor I (IGF-I) in this plausible pathogenic process in Alzheimer's disease has recently been proposed. Evidence favoring this idea stems from the ability of both hormones to stimulate beta amyloid release from neurons as well as by the stimulatory effect that IGF-I exerts on brain amyloid clearance. In addition, insulin and IGF-I levels are altered in Alzheimer's patients and, probably in close association to these changes, cell sensitivity towards insulin--and possibly also IGF-I--is decreased in these patients. We now review evidence that disturbed insulin/IGF-I signaling to brain cells, initiated at the level of the blood-brain barriers is probably instrumental in development of brain amyloidosis. Furthermore, insulin and IGF-I are potent neuroprotective factors and can regulate levels of phosphorylated tau, a major component of neurofibrillary tangles found in Alzheimer's brains. Therefore, a decrease in trophic support to neurons together with increased tau phosphorylation will follow loss of sensitivity towards insulin and IGF-I. Altogether, this supports the notion that a single pathogenic event, i.e., brain resistance to insulin/IGF-I, accounts for neuronal atrophy/death, tangle formation and brain amyloidosis typical of Alzheimer's pathology.
Collapse
Affiliation(s)
- Eva Carro
- Laboratory of Neuroendocrinology, Cajal Institute, CSIC, Avenida Doctor Arce 37, Madrid ES 28002, Spain
| | | |
Collapse
|
84
|
Abstract
Following unilateral transection (pedunculotomy) of the neonatal rat olivocerebellar pathway, the remaining inferior olive reinnervates the denervated hemicerebellum with correct topography. The critical period for this transcommissural reinnervation closes between postnatal days 7 and 10 but can be extended by injection of growth factors. Whether growth factor treatment can extend developmental plasticity into a mature, myelinated milieu remains unknown. Rats aged 11-30 days, underwent unilateral pedunculotomy followed 24 h later by injection of insulin-like growth factor 1 (IGF-1) into the denervated cerebellum. In all animals, IGF-1 induced transcommissural olivocerebellar reinnervation, which displayed organisation consistent with normal olivocerebellar topography even following pedunculotomy up to day 20. Thus IGF-1 can reproduce developmental neuroplasticity to promote appropriate target reinnervation in a mature myelinated environment.
Collapse
Affiliation(s)
- Rachel M Sherrard
- Developmental Neuroplasticity Laboratory, Department of Anatomy, School of Biomedical Science, James Cook University, Queensland 4811, Australia.
| | | |
Collapse
|
85
|
Hernández-Sánchez C, Mansilla A, de la Rosa EJ, Pollerberg GE, Martínez-Salas E, de Pablo F. Upstream AUGs in embryonic proinsulin mRNA control its low translation level. EMBO J 2004; 22:5582-92. [PMID: 14532130 PMCID: PMC213770 DOI: 10.1093/emboj/cdg515] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Proinsulin is expressed prior to development of the pancreas and promotes cell survival. Here we study the mechanism affecting the translation efficiency of a specific embryonic proinsulin mRNA. This transcript shares the coding region with the pancreatic form, but presents a 32 nt extended leader region. Translation of proinsulin is markedly reduced by the presence of two upstream AUGs within the 5' extension of the embryonic mRNA. This attenuation is lost when the two upstream AUGs are mutated to AAG, leading to translational efficiency similar to that of the pancreatic mRNA. The upstream AUGs are recognized as initiator codons, because expression of upstream ORF is detectable from the embryonic transcript, but not from the mutated or the pancreatic mRNAs. Strict regulation of proinsulin biosynthesis appears to be necessary, since exogenous proinsulin added to embryos in ovo decreased apoptosis and generated abnormal developmental traits. A novel mechanism for low level proinsulin expression thus relies on upstream AUGs within a specific form of embryonic proinsulin mRNA, emphasizing its importance as a tightly regulated developmental signal.
Collapse
Affiliation(s)
- Catalina Hernández-Sánchez
- Group of Growth Factors in Vertebrate Development, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, E-28040 Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
86
|
Varela-Nieto I, de la Rosa EJ, Valenciano AI, León Y. Cell death in the nervous system: lessons from insulin and insulin-like growth factors. Mol Neurobiol 2003; 28:23-50. [PMID: 14514984 DOI: 10.1385/mn:28:1:23] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2002] [Accepted: 02/28/2003] [Indexed: 12/11/2022]
Abstract
Programmed cell death is an essential process for proper neural development. Cell death, with its similar regulatory and executory mechanisms, also contributes to the origin or progression of many or even all neurodegenerative diseases. An understanding of the mechanisms that regulate cell death during neural development may provide new targets and tools to prevent neurodegeneration. Many studies that have focused mainly on insulin-like growth factor-I (IGF-I), have shown that insulin-related growth factors are widely expressed in the developing and adult nervous system, and positively modulate a number of processes during neural development, as well as in adult neuronal and glial physiology. These factors also show neuroprotective effects following neural damage. Although some specific actions have been demonstrated to be anti-apoptotic, we propose that a broad neuroprotective role is the foundation for many of the observed functions of the insulin-related growth factors, whose therapeutical potential for nervous system disorders may be greater than currently accepted.
Collapse
Affiliation(s)
- Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Arturo Duperier 4, E-28029 Madrid, Spain.
| | | | | | | |
Collapse
|
87
|
Kenney AM, Widlund HR, Rowitch DH. Hedgehog and PI-3 kinase signaling converge on Nmyc1 to promote cell cycle progression in cerebellar neuronal precursors. Development 2003; 131:217-28. [PMID: 14660435 DOI: 10.1242/dev.00891] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neuronal precursor cells in the developing cerebellum require activity of the sonic hedgehog (Shh) and phosphoinositide-3-kinase (PI3K) pathways for growth and survival. Synergy between the Shh and PI3K signaling pathways are implicated in the cerebellar tumor medulloblastoma. Here, we describe a mechanism through which these disparate signaling pathways cooperate to promote proliferation of cerebellar granule neuron precursors. Shh signaling drives expression of mRNA encoding the Nmyc1 oncoprotein (previously N-myc), which is essential for expansion of cerebellar granule neuron precursors. The PI3K pathway stabilizes Nmyc1 protein via inhibition of GSK3-dependent Nmyc1 phosphorylation and degradation. The effects of PI3K activity on Nmyc1 stabilization are mimicked by insulin-like growth factor, a PI3K agonist with roles in central nervous system precursor growth and tumorigenesis. These findings indicate that Shh and PI3K signaling pathways converge on N-Myc to regulate neuronal precursor cell cycle progression. Furthermore, they provide a rationale for therapeutic targeting of PI3K signaling in medulloblastoma.
Collapse
Affiliation(s)
- Anna Marie Kenney
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
88
|
Hawkes C, Kar S. Insulin-like growth factor-II/mannose-6-phosphate receptor: widespread distribution in neurons of the central nervous system including those expressing cholinergic phenotype. J Comp Neurol 2003; 458:113-27. [PMID: 12596253 DOI: 10.1002/cne.10578] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The insulin-like growth factor-II/mannose-6-phosphate (IGF-II/M6P) receptor is single transmembrane glycoprotein that plays a critical role in the trafficking of lysosomal enzymes and the internalization of circulating IGF-II. At present, there is little information regarding the cellular distribution of the IGF-II/M6P receptor within the adult rat brain. With the use of immunoblotting and immunocytochemical methods, we found that the IGF-II/M6P receptor is widely but selectively expressed in all major brain areas, including the olfactory bulb, striatum, cortex, hippocampus, thalamus, hypothalamus, cerebellum, brainstem, and spinal cord. Intense IGF-II/M6P receptor immunoreactivity was apparent on neuronal cell bodies within the striatum, deeper layers (layers IV and V) of the cortex, pyramidal and granule cell layers of the hippocampal formation, selected thalamic nuclei, Purkinje cells of the cerebellum, pontine nucleus and motoneurons of the brainstem as well as in the spinal cord. Moderate neuronal labeling was evident in the olfactory bulb, basal forebrain areas, hypothalamus, superior colliculus, midbrain areas, granule cells of the cerebellum and in the intermediate regions of the spinal gray matter. We also observed dense neuropil labeling in many regions, suggesting that this receptor is localized in dendrites and/or axon terminals. Double-labeling studies further indicated that a subset of IGF-II/M6P receptor colocalizes with cholinergic cell bodies and fibers in the septum, striatum, diagonal band complex, nucleus basalis, cortex, hippocampus, and motoneurons of the brainstem and spinal cord. The observed widespread distribution and colocalization of IGF-II/M6P receptor in the adult rat brain provide an anatomic basis to suggest a multifunctional role for the receptor in a wide-spectrum of central nervous system neurons, including those expressing a cholinergic phenotype.
Collapse
Affiliation(s)
- Cheryl Hawkes
- Douglas Hospital Research Center, Department of Neurology and Neurosurgery, Verdun, Quebec H4H 1R3, Canada
| | | |
Collapse
|
89
|
Trejo JL, Carro E, Nuñez A, Torres-Aleman I. Sedentary life impairs self-reparative processes in the brain: the role of serum insulin-like growth factor-I. Rev Neurosci 2003; 13:365-74. [PMID: 12542262 DOI: 10.1515/revneuro.2002.13.4.365] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Regular exercise has long being recognized as an important contributor to appropriate health status and is currently recommended to reduce the incidence of many diseases. More recent is the notion that sedentary life may also be a risk factor for neurodegenerative diseases even though for the last decade the beneficial effects of exercise on brain function have been widely documented. In the brain, exercise exerts both acute and long-term changes that can be interpreted as beneficial, such as increased levels of various neurotrophic factors or enhanced cognition. However, the signals involved in exercise-induced changes in the brain are not yet well known. It is generally thought that they arise from the periphery as a direct consequence of increased metabolic activity and aim to elicit adaptive changes in brain function. However, body-to-brain signaling induced by exercise also underlies a different aspect. Exercise induces changes in the brain that are essential for proper brain function. In this view, sedentarism, a relatively new cultural trait, negates the beneficial effects of exercise and paves the way to pathological derangement. A critical step in this process is exercise-induced uptake by the brain of insulin-like growth factor-I (IGF-I), a circulating hormone with potent neurotrophic activity. We summarize the evidence supporting the hypothesis that serum IGF-I is a neuroprotective hormone within a neuroprotective network modulated by physical activity.
Collapse
Affiliation(s)
- Jose Luis Trejo
- Laboratory of Neuroendocrinology, Instituto Cajal, CSIC, Madrid, Spain
| | | | | | | |
Collapse
|
90
|
Yokoo H, Saitoh T, Shiraishi S, Yanagita T, Sugano T, Minami SI, Kobayashi H, Wada A. Distinct effects of ketone bodies on down-regulation of cell surface insulin receptor and insulin receptor substrate-1 phosphorylation in adrenal chromaffin cells. J Pharmacol Exp Ther 2003; 304:994-1002. [PMID: 12604674 DOI: 10.1124/jpet.102.044115] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Treatment (>/=24 h) of cultured bovine adrenal chromaffin cells with ketoacidosis-related concentrations (>/=3 mM) of acetoacetate (but not beta-hydroxybutyrate, acetone, and acidic medium) caused a time- and concentration-dependent reduction of cell surface (125)I-insulin binding by ~38%, with no change in the K(d) value. The reduction of (125)I-insulin binding returned to control nontreated level at 24 h after the washout of acetoacetate-treated cells. Acetoacetate did not increase the internalization rate of cell surface insulin receptor (IR), as measured in the presence of brefeldin A, an inhibitor of cell surface vesicular exit from the trans-Golgi network. Acetoacetate (10 mM for 24 h) lowered cellular levels of the immunoreactive IR precursor molecule (approximately 190 kDa) and IR by 22 and 28%, respectively. Acetoacetate decreased IR mRNA levels by approximately 23% as early as 6 h, producing their maximum plateau reduction at 12 and 24 h. The half-life of IR mRNA was shortened by acetoacetate from 13.6 to 9.5 h. Immunoprecipitation followed by immunoblot analysis revealed that insulin-induced (100 nM for 10 min) tyrosine-phosphorylation of insulin receptor substrate-1 (IRS-1) was attenuated by 56% in acetoacetate-treated cells, with no change in IRS-1 level. These results suggest that chronic treatment with acetoacetate selectively down-regulated the density of cell surface functional IR via lowering IR mRNA levels and IR synthesis, thereby retarding insulin-induced activation of IRS-1.
Collapse
Affiliation(s)
- Hiroki Yokoo
- Department of Pharmacology, Miyazaki Medical College, Miyazaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Locally born olfactory bulb stem cells proliferate in response to insulin-related factors and require endogenous insulin-like growth factor-I for differentiation into neurons and glia. J Neurosci 2003. [PMID: 12574418 DOI: 10.1523/jneurosci.23-03-00895.2003] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
After late embryogenesis, new neurons are continuously added to the olfactory bulb (OB) from stem cells located in the forebrain subventricular zone. Nonetheless, stem cells have not been described within the embryonic olfactory bulb. Here we report the isolation of local olfactory bulb stem cells from the embryonic day 12.5-14.5 mouse embryo. These cells were 99.2% nestin positive and proliferated extensively in culture to at least 150 cell doublings. Clonal analysis demonstrated that neurons (TuJ1(+)), astrocytes (GFAP(+)), and oligodendrocytes (O4(+)) could be generated from single-plated cells, indicating that they are multipotent. At least 90% of proliferating cells expressed insulin-like growth factor-I (IGF-I), (pro)insulin, and their cognate receptors; these growth factors collaborated with fibroblast growth factor-2 plus epidermal growth factor (EGF) to promote stem cell proliferation and sphere formation. Cells from Igf-I(-)/- mice, however, proliferated as extensively as did Igf-I(+/+) cells. Differentiation and survival of stem cell-generated neurons and glia showed strong dependence on exogenous IGF-I, but oligodendrocyte differentiation also required insulin at low concentration. Furthermore, the percentages of stem cell-generated neurons, astrocytes, and oligodendrocytes were markedly lower in the cultures prepared from the Igf-I(-)/- mice compared with those of Igf-I(+/+). Concordantly, lack of IGF-I resulted in abnormal formation of the olfactory bulb mitral cell layer and altered radial glia morphology. These results support the presence within the embryonic mouse olfactory bulb of stem cells with specific requirements for insulin-related growth factors for proliferation or differentiation. They demonstrate that IGF-I is an endogenous factor regulating the differentiation of stem and other precursor cells within the olfactory bulb.
Collapse
|
92
|
Gago N, Avellana-Adalid V, Baron-Van Evercooren A, Schumacher M. Control of cell survival and proliferation of postnatal PSA-NCAM(+) progenitors. Mol Cell Neurosci 2003; 22:162-78. [PMID: 12676527 DOI: 10.1016/s1044-7431(02)00030-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In the present work, we studied the effects of several growth factors on survival and proliferation of freshly isolated neural progenitors expressing the polysialylated form of neural cell adhesion molecule (PSA-NCAM). Cells were obtained from postnatal day 2 rat forebrain, using isolation method. We found that (1) insulin-like growth factor 1 (IGF-1) exerts a powerful survival effect by inhibiting apoptotic cell death, (2) epidermal growth factor (EGF) strongly increases cell proliferation, (3) the combination of IGF-1 plus EGF promotes cellular expansion, (4) basic fibroblast growth factor displays only a weak mitogenic effect, and (5) platelet-derived growth factor-AA (PDGF-AA) has no effect on cell survival and proliferation. These results suggest that the postnatal PSA-NCAM(+) progenitors characterized in the present work may represent a transitional stage, between the embryonic EGF-responsive neural progenitors and the postnatal PSA-NCAM(+) progenitors already described that are PDGF-responsive. For these "early PSA-NCAM(+) progenitors," insulin-like growth factor 1 and EGF seem to play a pivotal role in the control of cell death and cell proliferation.
Collapse
Affiliation(s)
- Nathalie Gago
- INSERM U 488, 80, rue du Général Leclerc, 94276, Bicêtre, France.
| | | | | | | |
Collapse
|
93
|
Seto D, Zheng WH, McNicoll A, Collier B, Quirion R, Kar S. Insulin-like growth factor-I inhibits endogenous acetylcholine release from the rat hippocampal formation: possible involvement of GABA in mediating the effects. Neuroscience 2003; 115:603-12. [PMID: 12421625 DOI: 10.1016/s0306-4522(02)00450-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Evidence suggests that insulin-like growth factor-I (IGF-I) plays an important role during brain development and in the maintenance of normal as well as activity-dependent functioning of the adult brain. Apart from its trophic effects, IGF-I has also been implicated in the regulation of brain neurotransmitter release thus indicating a neuromodulatory role for this growth factor in the central nervous system. Using in vitro slice preparations, we have earlier reported that IGF-I potently inhibits K(+)-evoked endogenous acetylcholine (ACh) release from the adult rat hippocampus and cortex but not from the striatum. The effects of IGF-I on hippocampal ACh release was sensitive to the Na(+) channel blocker tetrodotoxin, suggesting that IGF-I might act indirectly via the release of other transmitters/modulators. In the present study, we have characterized the possible involvement of GABA in IGF-I-mediated inhibition of ACh release and measured the effects of this growth factor on choline acetyltransferase (ChAT) activity and high-affinity choline uptake in the hippocampus of the adult rat brain. Prototypical agonists of GABA(A) and GABA(B) receptors (i.e. 10 microM muscimol and 10 microM baclofen) inhibited, whereas the antagonists of the respective receptors (i.e. 10 microM bicuculline and 10 microM phaclofen) potentiated K(+)-evoked ACh release from rat hippocampal slices. IGF-I (10 nM) inhibited K(+)- as well as veratridine-evoked ACh release from rat hippocampal slices and the effect is possibly mediated via the activation of a typical IGF-I receptor and the subsequent phosphorylation of the insulin receptor substrate-1 (IRS-1). The inhibitory effects of IGF-I on hippocampal ACh release were not additive to those of either muscimol or baclofen, but were attenuated by GABA antagonists, bicuculline and phaclofen. Additionally, in contrast to ACh release, IGF-I did not alter either the activity of the enzyme ChAT or the uptake of choline in the hippocampus. These results, taken together, indicate that IGF-I, under acute conditions, can decrease hippocampal ACh release by acting on the typical IGF-I/IRS receptor complex while having no direct effect on ChAT activity or the uptake of choline. Furthermore, the evidence that effects of IGF-I could be modulated, at least in part, by GABA antagonists suggest that the release of GABA and the activation of its receptors may possibly be involved in mediating the inhibitory effects of IGF-I on hippocampal ACh release.
Collapse
Affiliation(s)
- D Seto
- Douglas Hospital Research Center, Department of Psychiatry, 6875 La Salle Boulevard, Verdun, QC, Canada H4H 1R3
| | | | | | | | | | | |
Collapse
|
94
|
Pérez-Martín M, Cifuentes M, Grondona JM, Bermúdez-Silva FJ, Arrabal PM, Pérez-Fígares JM, Jiménez AJ, García-Segura LM, Férnandez-Llebrez P. Neurogenesis in explants from the walls of the lateral ventricle of adult bovine brain: role of endogenous IGF-1 as a survival factor. Eur J Neurosci 2003; 17:205-11. [PMID: 12542656 DOI: 10.1046/j.1460-9568.2003.02432.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Previous studies have shown the existence of proliferating cells in explants from bovine (Bos Taurus) lateral ventricle walls that were maintained for several days in vitro in the absence of serum and growth factors. In this study we have characterized the nature of new cells and have assessed whether the insulin-like growth factor-1 (IGF-1) receptor regulates their survival and/or proliferation. The explants were composed of the ependymal layer and attached subependymal cells. Ependymal cells in culture were labelled with glial markers (S-100, vimentin, GFAP, BLBP, 3A7 and 3CB2) and did not incorporate bromodeoxiuridine when this molecule was added to the culture media. Most subependymal cells were immunoreactive for beta III-tubulin, a neuronal marker, and did incorporate bromodeoxiuridine. Subependymal neurons displayed immunoreactivity for IGF-1 and its receptor and expressed IGF-1 mRNA, indicating that IGF-1 is produced in the explants and may act on new neurons. Addition to the culture media of an IGF-1 receptor antagonist, the peptide JB1, did not affect the incorporation of bromodeoxiuridine to proliferating subependymal cells. However, JB1 significantly increased the number of TUNEL positive cells in the subependymal zone, suggesting that IGF-1 receptor is involved in the survival of subependymal neurons. In conclusion, these findings indicate that neurogenesis is maintained in explants from the lateral cerebral ventricle of adult bovine brains and that IGF-1 is locally produced in the explants and may regulate the survival of the proliferating neurons.
Collapse
Affiliation(s)
- M Pérez-Martín
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29071 Malaga, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Cardona-Gomez GP, Mendez P, Garcia-Segura LM. Synergistic interaction of estradiol and insulin-like growth factor-I in the activation of PI3K/Akt signaling in the adult rat hypothalamus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 107:80-8. [PMID: 12414126 DOI: 10.1016/s0169-328x(02)00449-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Estradiol and insulin-like growth factor-I (IGF-I) interact in the hypothalamus to regulate neuronal function, synaptic plasticity and neuroendocrine events. However, the molecular mechanisms involved in these interactions are still unknown. In the present study, the effect of estradiol on the signaling pathways of IGF-I receptor has been assessed in the hypothalamus of young adult ovariectomized rats, using specific antibodies for the phosphorylated forms of extracellular-signal regulated kinase (ERK) 1 and ERK2 and Akt/protein kinase B (Akt/PKB). Estradiol treatment resulted, between 6 and 24 h after systemic administration, in dose-dependent effects on the phosphorylation of ERK and Akt/PKB. Estradiol did not modify the level of ERK phosphorylation induced by intracerebroventricular administration of IGF-I. However, both hormones had a synergistic effect on the phosphorylation of Akt/PKB. These findings suggest that estrogen effects in the hypothalamus may be mediated in part by the activation of the signaling pathways of the IGF-I receptor.
Collapse
|
96
|
Camarero G, Villar MA, Contreras J, Fernández-Moreno C, Pichel JG, Avendaño C, Varela-Nieto I. Cochlear abnormalities in insulin-like growth factor-1 mouse mutants. Hear Res 2002; 170:2-11. [PMID: 12208536 DOI: 10.1016/s0378-5955(02)00447-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Insulin-like growth factor 1 (IGF-1) modulates inner ear cell proliferation, differentiation and survival in culture. Its function in human hearing was first evidenced by a report of a boy with a homozygous deletion of the Igf-1 gene, who showed severe sensorineural deafness [Woods et al., New Engl. J. Med. 335 (1996) 1363-1367]. To better understand the in vivo role of IGF-1 during inner ear differentiation and maturation, we studied the cochleae of Igf-1 gene knockout mice by performing morphometric stereological analyses, immunohistochemistry and electron microscopy on postnatal days 5 (P5), P8 and P20. At P20, but not at P5, the volumes of the cochlea and cochlear ganglion were significantly reduced in mutant mice, although the reduction was less severe than whole body dwarfism. A significant decrease in the number and average size of auditory neurons was also evident at P20. IGF-1-deficient cochlear neurons showed increased apoptosis, along with altered expression of neurofilament 200 kDa and vimentin. The eighth nerve, the cochlear ganglion and the fibers innervating the sensory cells of the organ of Corti of the P20 mouse mutants presented increased expression of vimentin, whereas the expression of neurofilament was decreased. In addition, the myelin sheath was severely affected in ganglion neurons. In conclusion, IGF-1 deficit in mice severely affects postnatal survival, differentiation and maturation of the cochlear ganglion cells.
Collapse
Affiliation(s)
- Guadalupe Camarero
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier 4, 28029, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
97
|
Gutiérrez-Ospina G, Gutiérrez de la Barrera A, Larriva J, Giordano M. Insulin-like growth factor I partly prevents axon elimination in the neonate rat optic nerve. Neurosci Lett 2002; 325:207-10. [PMID: 12044657 DOI: 10.1016/s0304-3940(02)00293-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Developmental neuronal death ensues after access of innervating neurons to target-derived neurotrophic factors is restricted. Recent evidence suggests, however, that growth factors such as those of the insulin family modulate neuronal death through autocrine/paracrine mechanisms. In rats, retinal ganglion neurons (RGNs) undergo massive death during early postnatal life. During this same period, the expression of various members of the insulin-like growth factor I (IGF-I) protein family is down regulated. To evaluate whether ocular IGF-I might modulate RGN death, we administered IGF-I in the posterior chamber of the eye of newborn rats. Optic nerve fiber number was estimated in control and IGF-I treated animals at postnatal day 5 when RGN death peaks. Intraocular IGF-I treatment at birth partly prevented optic nerve fiber elimination. Because the axon number in the optic nerve correlates to some extent with the RGN number, these results suggest that IGF-I may modulate RGN death in vivo through local interactions.
Collapse
Affiliation(s)
- Gabriel Gutiérrez-Ospina
- Department of Cell Biology and Physiology, Biomedical Research Institute, National University of México, Mexico D.F., 04510, Mexico.
| | | | | | | |
Collapse
|
98
|
Abstract
Studies in the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans have revealed that components of the insulin signaling pathway have been highly conserved during evolution. Genetic analysis in Drosophila suggests that structural conservation also extends to the functional level. Flies carrying mutations that reduce insulin signaling have a growth deficiency phenotype similar to that seen in mice with disruptions of genes encoding insulin-like growth factors (IGFs) or the IGF-I receptor. Recent studies in flies have demonstrated a role for the insulin signaling pathway in the regulation of metabolism, reproduction and lifespan via modulation of central neuroendocrine pathways. Similarly, mice with loss of brain insulin receptors or insulin receptor substrate 2 deficiency exhibit neuroendocrine defects and female infertility. These parallels suggest that the insulin system has multiple conserved roles, acting directly to modulate growth and indirectly, via the neuroendocrine system, to modulate peripheral physiology in response to changes in nutrient availability.
Collapse
Affiliation(s)
- Robert S Garofalo
- Dept Cardiovascular and Metabolic Diseases, Pfizer Global Research and Development, MS-3220-3082, Groton, CT 06340, USA.
| |
Collapse
|
99
|
Chowen JA, Goya L, Ramos S, Busiguina S, García-Segura LM, Argente J, Pascual-Leone AM. Effects of early undernutrition on the brain insulin-like growth factor-I system. J Neuroendocrinol 2002; 14:163-9. [PMID: 11849376 DOI: 10.1046/j.0007-1331.2001.00758.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Undernutrition reduces circulating concentrations of insulin-like growth factor (IGF)-I, but how it affects the brain IGF system, especially during development, is largely unknown. We have studied IGF-I, IGF-II, IGF receptor and IGF binding protein (BP)-2 mRNA expression in the hypothalamus, cerebellum and cerebral cortex of neonatal rats that were food restricted beginning on gestational day 16. One group was refed starting on postnatal day 14. Rats were killed on postnatal day 8 or 22. Undernutrition did not produce an overall reduction in brain weight at either age but, at 22 days, both the cerebellum and hypothalamus weighed significantly less. At 8 days, no change was detected in the central IGF axis in response to undernutrition. However, in 22-day-old undernourished rats, IGF-I and IGF receptor mRNA expression were increased in both the hypothalamus and cerebellum, while IGFBP-2 was decreased, but only in the hypothalamus. Refeeding had no effect on any of these parameters. These results suggest that the hypothalamus and cerebellum respond to malnutrition and the decrease in circulating IGF-I, a peptide fundamental for growth and development, by increasing the local production of both the growth factor and its receptor in attempt to maintain normal development.
Collapse
Affiliation(s)
- J A Chowen
- Department of Endocrinology, Laboratory of Investigation, Autonoma University, University Children's Hospital Niño Jesús, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
100
|
Sizonenko SV, Sirimanne ES, Williams CE, Gluckman PD. Neuroprotective effects of the N-terminal tripeptide of IGF-1, glycine-proline-glutamate, in the immature rat brain after hypoxic-ischemic injury. Brain Res 2001; 922:42-50. [PMID: 11730700 DOI: 10.1016/s0006-8993(01)03148-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Insulin growth factor 1 (IGF-1) has an important role in brain development and is strongly expressed during recovery after a hypoxic-ischemic injury. Some of its central actions could be mediated through the N-terminal tripeptide fragment of IGF-1: Gly-Pro-Glu (GPE). The neuroprotective properties of GPE given after a moderate injury in the developing rat brain were evaluated and the binding sites of [(3)H]GPE characterised by autoradiography. After right unilateral injury, GPE or vehicle (V) was injected in the right lateral ventricle (i.c.v.) or in the peritoneal cavity (i.p.) of 21-day-old rats. The percentage of surviving neurons in CA1-2 of the hippocampus was higher in the animals treated with 30 microg of GPE i.c.v. (V: 7.7+/-4.9%, GPE: 26.4+/-7.5%, P=0.02) and 300 microg i.p. (V: 30.2+/-9.1%, GPE: 68.8+/-10.6%, P=0.02) than in animals receiving vehicle. I.p. injection of 300 microg of GPE (V: 78.4+/-7.5%, GPE: 88.4+/-3.2%, P=0.04) was also neuroprotective in the lateral cortex. I.c.v. injection of [(3)H]GPE suggested binding to glial cells in the white matter tracts, the cortex and striatum as opposed to neurons. Although the precise mode of action of GPE is unknown, this study suggests that local administration of GPE is neuroprotective after brain HI injury via glial cells. In addition, systemic administration of GPE showed a more widespread neuroprotective effect. GPE may represent a complementary pathway for central and systemic IGF-1's antiapoptotic effects.
Collapse
Affiliation(s)
- S V Sizonenko
- Liggins Institute, Faculty of Medicine and Health Science, The University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | | | | | | |
Collapse
|