当前位置: 首页 > 期刊 > 《内分泌进展》 > 2005年第6期 > 正文
编号:11447318
The Insulin-Like Growth Factor System and Its Pleiotropic Functions in Brain
http://www.100md.com V. C. Russo, P. D. Gluckman, E. L. Feldm
    参见附件。

     Murdoch Children’s Research Institute (V.C.R., G.A.W.), Centre for Hormone Research, Royal Children’s Hospital, Parkville 3052, Victoria, Australia

    Department of Paediatrics (V.C.R., G.A.W.), University of Melbourne, Parkville 3052, Victoria, Australia

    Liggins Institute and National Research Centre for Growth and Development (P.D.G.), University of Auckland, Auckland 1020, New Zealand

    Department of Neurology (E.L.F.), University of Michigan, Ann Arbor, Michigan 48109

    Abstract

    In recent years, much interest has been devoted to defining the role of the IGF system in the nervous system. The ubiquitous IGFs, their cell membrane receptors, and their carrier binding proteins, the IGFBPs, are expressed early in the development of the nervous system and are therefore considered to play a key role in these processes. In vitro studies have demonstrated that the IGF system promotes differentiation and proliferation and sustains survival, preventing apoptosis of neuronal and brain derived cells. Furthermore, studies of transgenic mice overexpressing components of the IGF system or mice with disruptions of the same genes have clearly shown that the IGF system plays a key role in vivo.

    I. Introduction

    II. Historical Perspective

    III. Overview of the IGF System

    A. The IGF peptides

    B. The IGF receptors and their functions

    C. The IGFBPs

    IV. Expression and Pleiotropic Actions of the IGF System in the Brain

    A. IGF expression in the nervous system

    B. Type I and II IGF receptor expression in the nervous system

    C. The IGF ligands are pleiotropic factors

    D. The IGFs are neurotrophic factors

    E. IGF-I prevents neuronal apoptosis

    F. IGFBP action in the nervous system

    V. The IGF System and Neuroendocrine Cross-Talk in the Nervous System

    A. IGF-I and fibroblast growth factor (FGF)-2

    B. IGF-I and neurotrophic factors

    C. IGF-I and GH

    D. IGF-I and erythropoietin

    E. IGF-I and sex steroids

    VI. Altered Expression of the IGF System in Response to CNS Injury

    VII. Altered Expression of the IGF System in Malignancies of the Nervous System

    VIII. IGF-I Therapy in Nervous System Disease Models

    A. IGF-I therapy in acute central nervous system disease

    B. IGF-I therapy in chronic central nervous system disease

    C. IGF-I therapy in neurodegenerative disorders

    D. Neuropathy: a potential target for IGF-I therapy

    E. Targeting the IGF/IGF-IR signaling in cancer therapy

    F. The potential therapeutic use of N-terminal peptides derived from IGF-I

    IX. Perspectives for Future Directions

    I. Introduction

    THE AVAILABILITY OF animal models for brain injury and neuronal degeneration has allowed the investigation of the role of IGF-I in prevention and rescue of damaged neuronal cells. These studies have thus pointed to the potential therapeutic use of IGF-I alone or in combination with other neuroendocrine factors in the treatment of nervous system diseases.

    The main aim of this review is therefore to provide an up-to-date and comprehensive analysis of the pleiotropic functions of the IGF system in the nervous tissue. To maintain focus on the IGF system, the role and functions of the insulin system in brain and neuronal cells, also important but already amply reviewed elsewhere, will only be mentioned and discussed in brief in this review.

    II. Historical Perspective

    In 1957, Salmon and Daughaday (1) reported a serum factor (factors) that mediated the cartilage sulfation and longitudinal bone growth activity of the somatotrophic hormone (GH) (2, 3). This factor was termed "sulfation factor" and was produced by hepatic cells after exposure to GH (1). In conjunction, Dulak and Temin (4) were investigating the cell proliferative factors in serum and termed one such activity multiplication-stimulating activity. These circulating factors, which also showed insulin-like activity not suppressible by antiinsulin antibodies [nonsuppressible insulin-like activity (NSILA) I and II] (5, 6, 7, 8, 9, 10), were later found to have a similar biochemical structure to the -chain of insulin (11, 12, 13). It was apparent that these activities represented a similar group of substances with a wider biological activity than first suspected (3). NSILA I and II were two forms of an insulin-like hormone with predominant effects on cell and tissue growth (11). These small molecular mass peptides (NSILA I and II) were later renamed as somatomedins replacing the "nonsuppressible insulin-like activity" and sulfation factor terminology. Further investigation revealed that these factors mediated the actions of pituitary-derived GH, giving birth to the somatomedin hypothesis (3).

    Shortly thereafter, two mammalian somatomedins were identified by protein sequence and cDNA data (11, 14, 15, 16, 17, 18) and their structural homology with proinsulin led to their current designation of IGF-I and IGF-II.

    In the middle 1990s, Sara and co-workers (19, 20) identified a brain-specific variant of IGF-I, des(1-3) or "truncated" IGF-I, which lacks the first three amino acids and is more potent than intact IGF-I in various cell culture systems (19, 20, 21), probably due to its lower affinity for IGF binding proteins (IGFBPs) (22). This finding suggested brain synthesis of IGF-I or its truncated variant. After the isolation of IGF-I mRNA from postnatal rat brain (23), a number of in situ hybridization studies (24, 25) demonstrated that IGF-I and IGF-I receptor mRNA are synthesized in the rat brain in specific regions, namely olfactory bulb, hippocampus, and cerebellum (26). In addition IGF-carrier proteins, later named IGF binding proteins, were also found expressed in similar brain regions (27, 28, 29, 30, 31, 32, 33, 34, 35, 36). The finding of IGF-I mRNA colocalization with IGF-I receptors and the presence of IGFBPs suggested a paracrine or autocrine role for IGF-I, potentially modulated by IGFBP, in the developing brain (26, 36, 37, 38).

    The presence of IGF-I (39) and IGF-I mRNAs (40) in multiple tissues has necessitated the revision of the original somatomedin hypothesis to include both autocrine and paracrine actions of IGF in addition to its classical endocrine aspects.

    The somatomedin hypothesis has been recently challenged by several groups employing the liver-specific IGF-I deficient (LID) mice, which have reduced circulating IGF-I and elevated GH (41, 42), and the acid-labile subunit knockout ALSKO mice (43), which have reduced circulating IGF-I and IGFBP-3 but normal GH level. The LID mice, contrary to expectations (altered IGF-I-mediated GH-stimulated growth), showed normal postnatal growth and development (43, 44), whereas the ALSKO mice, despite 65% reduction in circulting IGF-I, demonstrated only 10% reduction in body weight. Interestingly, when the LID mice were crossed with the ALSKO mice (LID + ALSKO) (43), to further affect the GH/IGF-I axis, the LID-ALSKO mice (very low IGF-I and very high GH levels) showed postnatal growth retardation and osteopenia, suggesting that circulating IGF-I is important for postnatal growth and development (44). Although investigations are ongoing in this area, it is accepted at this stage that IGF-I exerts its pleiotropic functions in an endocrine, autocrine, and paracrine fashion.

    III. Overview of the IGF System

    A. The IGF peptides

    IGF-I and -II are growth-promoting peptides, members of a superfamily of related insulin-like hormones that includes insulin and relaxin in the vertebrates and bombyxin, locust insulin-related peptide, and molluscan insulin-like peptide in invertebrates (12, 13, 45, 46, 47, 48, 49). However, insulin and IGFs are the most closely related in terms of primary sequence and biological activity. The IGFs are major growth factors, wheras insulin predominantly regulates glucose uptake and cellular metabolism. They consist of A, B, C, and D domains. Large parts of the sequences within the A and B domains are homologous to the - and -chain of the human proinsulin. This sequence homology is 43% for IGF-I and 41% for IGF-II. No sequence homology exists between the C domains of IGFs and the C peptide region of human proinsulin. The C domain of the IGFs is not removed during prohormone processing; thus the mature IGF peptides are single chain polypeptides (50, 51). The gene encoding IGF-I is highly conserved (50, 52, 53, 54, 55, 56) such that 57 of 70 residues of the mature protein are identical among mammals, birds, and amphibians (50, 52, 53, 54, 55, 56). For a more extensive review of structure, functions, and regulation of the IGF-I gene, see Refs.57, 58, 59, 60, 61 .

    Expression of the Igf-1 gene is affected at many levels including gene transcription, splicing, translation, and secretion. IGF-I expression is also influenced by hormonal (GH) (1, 62, 63, 64, 65, 66), nutritional (67, 68, 69), tissue-specific and developmental factors (36, 70, 71, 72, 73).

    B. The IGF receptors and their functions

    The biological actions of the IGFs are mediated by the type I IGF receptor (IGF-IR), a glycoprotein on the cell surface that transmits IGF binding to a highly integrated intracellular signaling system (74, 75, 76, 77). Binding of the IGFs (IGF-I and II) to the IGF-IR (74, 78, 79, 80, 81, 82, 83, 84) promotes intrinsic tyrosine kinase activity that phosphorylates the insulin receptor substrates (IRS-1 to IRS-4), which then leads to the activation of two main downstream signaling cascades, the MAPK and the phosphatidylinositol 3-kinase (PI3K) cascades (85, 86, 87, 88, 89, 90). However, IGF-II binds the IGF-IR with lower affinity (74, 78, 79, 80, 81, 82, 83, 84). Expression of the IGF-IR gene (77, 91) has been detected in many tissues and is constitutively expressed in most cells (92, 93, 94); its promoter is regulated in vitro and in vivo by transcription factors such as SP1 (95, 96) and the transcription factor p53 (97, 98).

    Various IGF-I receptor subtypes that present distinct structures or binding properties have also been described. Two of these subtypes, namely hybrid and atypical IGF-I receptors, have been particularly investigated in a variety of cell types (99, 100, 101, 102). The atypical IGF receptors are characterized by their ability to bind insulin as well as IGFs with relatively high affinity (103). Hybrid insulin/IGF-I receptors have been reported in cells expressing both IGF-I and insulin receptors (104, 105, 106, 107, 108); however, the physiological significance of hybrid and atypical IGF receptors is unclear.

    The IGF-II ligand has greatest affinity for a distinct receptor, the type-II or IGF-II receptor (109, 110, 111, 112, 113, 114). This single chain polypeptide with a short cytoplasmic domain lacking tyrosine kinase activity is identical to the cation-independent mannose-6 phosphate (M6P) receptor (107, 115, 116). The IGF-II/M6P receptor binds two general classes of ligands: 1) non-M6P-containing ligands, the best characterized of which is IGF-II; and 2) M6P-containing ligands, including lysosomal enzymes. The multifunctional role of the receptor is evidenced by its function in the mediation of lysosomal enzyme trafficking, endocytosis, and lysosomal degradation of extracellular ligands, regulation of apoptotic/mitogenic effects, and possible intracellular signal transduction (115, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128).

    More recently, high-affinity binding of IGF-II to an insulin receptor isoform (IR-A) has also been reported (129, 130, 131), thus suggesting that IGF-II might also signal via the insulin receptor. However, this insulin receptor isoform (IR-A), lacking the alternative spliced exon 11, is preferentially expressed in fetal and cancer cells (130).

    C. The IGFBPs

    A family of six high-affinity IGF-binding proteins (IGFBP-1 through IGFBP-6) coordinate and regulate the biological activity of IGF in several ways: 1) transport IGF in plasma and control its diffusion and efflux from the vascular space; 2) increase the half-life and regulate clearance of the IGFs; 3) provide specific binding sites for the IGFs in the extracellular and pericellular space; and 4) modulate, inhibit, or facilitate interaction of IGFs with their receptors (111, 132, 133, 134, 135). IGFBP biological activity is regulated by posttranslational modifications such as glycosylation and phosphorylation (111) and/or differential localization of the IGFBPs in the pericellular and extracellular space (111, 134, 135, 136, 137, 138, 139). It is therefore hypothesized that IGFBPs, in addition to stabilizing and regulating levels of diffusible IGFs, might regulate IGF-I cellular responses by facilitating receptor targeting of IGF-I or modulating IGF-I bioavailability in the pericellular space (111, 135).

    The effects of the IGFBPs are further regulated by the presence of specific IGFBP proteases, which cleave the binding proteins, generating fragments with reduced or no binding affinity for the IGFs (111, 135, 136, 140). Some IGFBPs, including IGFBP-2 and -3, can induce direct cellular effects independent of the IGFs (135, 141, 142, 143, 144). IGFBP-3, similar to IGFBP-5, contains sequences with the potential for nuclear localization (145, 146) and detection of IGFBP-3 in the nuclei of dividing cells, as reported by several investigators (146, 147), strongly suggesting a role for IGFBP-3 in gene regulation. More recently, perinuclear or nuclear localization has also been reported for IGFBP-2 (148); however, the role of IGFBP-2 in this cellular compartment is yet to be determined.

    IV. Expression and Pleiotropic Actions of the IGF System in the Brain

    A. IGF expression in the nervous system

    Central nervous system (CNS) development begins in the embryo with the formation and closure of the neural tube, followed by the rapid division of pluripotential cells (stem cells), which then migrate to the periphery of the neural tube and differentiate into neural or glial cells. These cells form special structures such as nuclei, ganglia, and cerebral cortical layers, and develop a network with their cytoplasmic extensions, neurites. These complex processes are regulated by a number of growth factors including the IGFs (149, 150).

    IGF-I plays a key role in the development of the nervous system, with demonstrated effects on many stages of brain development including cell proliferation, cell differentiation, and cell survival (36, 37, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159). Although recent reports have demonstrated that postnatal circulating IGF-I might exert neurogenic/survival activity (160, 161, 162, 163, 164, 165, 166) (167, 168, 169, 170, 171), systemic IGF-I is not readily transported through the blood-brain-barrier, and therefore local production of IGF-I is considered the primary source of the ligand (autocrine and paracrine action) for brain cells.

    During embryogenesis, IGF-I mRNA expression is detectable in many brain regions (23, 25, 31, 172, 173, 174, 175, 176), with its expression being particularly high in neuronal rich regions such as the spinal cord, midbrain, cerebral cortex, hippocampus, and olfactory bulb (23, 177, 178) (Fig. 1). More precisely IGF-I is found expressed in neuronal cells with large soma and complex dendritic formations (32, 155), including sensory and projecting neurons such as the Purkinje cells (25). In most neurons, IGF-I transcription decreases significantly postnatally, a decrease that correlates with the degree of cell maturation and reaches very low levels in the adult (23, 175). However, exceptions are the mitral and tufted cells of the olfactory bulb, which undergo constant cell renewal/turnover (179); in these cells IGF-I expression persists at a high level throughout life (26, 179).

    FIG. 1. The IGF system in the rat brain olfactory bulb. IGF-I is expressed and synthesized in distinct areas of the rat brain, adjacent to regions rich in IGF-I receptors and IGFBPs (i.e., olfactory bulb is shown). This provides strong evidence for an autocrine and paracrine action of the IGFs in the nervous system. IGF action is modulated by locally expressed IGFBPs. ONL, Olfactory nerve layer; GL, glomerular cell layer; EPL, external plexiform cell layer; MI, mitral cell layer; GRL, granular cell layer. Section A-A shows a hematoxylin-eosin staining of olfactory bulb. [Modified with permission from L. W. Swanson: Brain Maps: Structures of the Rat Brain, 2nd edition, Elsevier, Amsterdam, 1998/1999 (528 ).] Area in the square is enlarged and represented in the cartoon.

    IGF-II mRNA is abundantly expressed in the embryonic rat CNS, however data are conflicting on IGF-II expression in cells of neuroepithelial origin (24, 180, 181). IGF-II is the most abundantly expressed IGF in the adult CNS, with the highest level of expression found in myelin sheaths, but also in leptomeninges, microvasculature, and the choroid plexus, all nonneuronal structures that enable diffusion of growth factors to their sites of activity (24, 25, 176, 182, 183, 184, 185).

    B. Type I and II IGF receptor expression in the nervous system

    The IGF receptors are widely expressed throughout the CNS, with high levels of expression found in specific regions and located to specific cell types (25, 26, 36, 38, 119, 153, 186, 187, 188). Given that IGF receptors are expressed from early stages of embryogenesis and throughout life, and that their ligands also show similar a "temporal-spatial" pattern of expressions, it is evident that the local brain IGF circuits are crucial modulators of the processes activated during brain development. Type I receptors are expressed throughout the rat CNS (177, 187, 189, 190, 191) with high levels of expression detected in the developing cerebellum, midbrain, olfactory bulb (Fig. 1), and in the ventral floorplate of the hindbrain (23, 24, 187). The level of IGF-IR decreases to adult levels soon after birth (192, 193) but remains relatively high in the choroid plexus, meninges, and vascular sheaths (25, 187). It is thus not surprising that knockout of the IGF-I receptor gene (194, 195, 196) produced, in addition to in utero growth retardation, a strong brain phenotype, namely a small brain.

    The type II IGF receptor, which has low affinity for IGF-I, but a high affinity for IGF-II, and also binds mannose-6-phosphate, is selectively expressed in all major brain regions (119, 183, 188, 197, 198). It is highly expressed in the pyramidal cell layers of the hippocampus, the granule layer of the dentate gyrus, olfactory bulb, the choroid plexus, and in the cerebral vasculature, ependymal cells, retina, pituitary, brainstem, and spinal cord (119, 183, 188, 197). Some of these regions undergo tissue remodelling, suggesting that the IGF-II/M6P receptor, in addition to its role in transporting lysosomal enzymes, might also participate in control of neuronal growth, differentiation, and repair, processes regulated by IGF-II/M6P receptor ligands including LIF, TGF, and retinoic acid (198).

    Early studies from our laboratory have demonstrated that some of these regions expressing both the IGF-I and IGF-II receptors are also rich in insulin receptors (199). Insulin receptor density is elevated in the choroid plexus, olfactory bulb, limbic system, and hypothalamus but is much lower in other brain regions (cortex, cerebellum, pituitary gland, thalamus), all regions concerned with olfaction, appetite, and autonomic functions (199). More recently, ablation of the insulin receptor in nestin-positive neurons (NIRKO mouse) (200) suggests a role for insulin in control of appetite suppression and reproduction (200).

    C. The IGF ligands are pleiotropic factors

    Whether the IGFs are produced locally (23, 25, 31, 172, 173, 174, 175, 176, 201) or reach brain cells systemically (160, 161, 162, 163, 164, 165, 166), these molecules exert potent neurotrophic, neurogenic, and neuroprotective/antiapoptotic activities. A large number of in vitro studies have demonstrated that IGF-I promotes mitogenesis and differentiation in glial cells (202, 203, 204, 205, 206, 207), oligodendrocytes (208, 209, 210), neuronal cells (36, 37, 211, 212), adult stem cells (213), and brain explants (21, 214, 215, 216, 217), and regulates axon myelination (218, 219, 220). IGF-I enhances growth cone motility and promotes neurite outgrowth (221, 222).

    Studies using the IGF-I –/– mice models (152, 162, 220) have clearly demonstrated that most of the IGF functions determined in vitro also apply to the in vivo situation, affecting a wide range of brain cells. The IGF-I null mice have reduced brain size and altered brain structures (151, 155) and show alteration of myelination processes (168, 223). This appears to be the result of reduced oligodendrocyte proliferation and maturation, which is also associated with reduction of axonal growth (168, 196, 223). However, some early studies from Bondy and co-workers (220) have reported instead that in IGF-I null mice, myelination is reduced but proportionate to the neuronal composition. Furthermore, ablation of the Igf-1 gene has revealed deficit in the numbers of specific neurons, oligodendrocytes in the olfactory bulb, dentate gyrus and striatum (194, 196, 220) and in cochlear ganglion neurons (169). These anatomical differences are likely to be the consequences of alteration in proliferation, survival, or differentiation caused by the absence of IGF-I during development. A study from Vicario-Abejon et al. (224) suggests that reduction of stem cell proliferation/differentiation in the IGF-I null mice might be the cause of reduced plasticity/maturation normally occuring in some brain regions (i.e., olfactory bulb) postnatally (224). In addition to gross structural brain abnormalities, the IGF null mice also show alteration of important brain metabolic functions such as reduced glucose uptake, the major source of energy of neuronal cells (225). These in vivo models thus show that the IGF system affects several steps involved in development and organization (number of cells, their connection, and the extracellular matrix) of CNS architecture.

    However, actions of IGF are not limited to fetal life (25, 149, 150, 226), but extend into postnatal (21, 217) and adult life (185, 227, 228, 229), with effects on proliferation, neuronal differentiation, and maturation maintained in some regions of the adult brain. Furthermore, alterations in levels of IGF-I have been reported in the brains of aging rats (230, 231), alterations that also correlate with deterioration of cognitive functions observed in elderly patients with low circulating IGF-I (232, 233, 234).

    D. The IGFs are neurotrophic factors

    In support of studies by the Gluckman group (235) and others on the use of IGFs as therapeutic agents (see also Section VII), many investigators have examined the cellular and molecular mechanisms of IGF action in nervous system diseases. In this regard, the IGFs have been shown to be neurotrophic factors, i.e., they promote the survival and differentiation of neuronal cells, including sensory (171, 221, 236), sympathetic (211, 237), and motor neurons (MNs) (236, 237, 238). In fact, the IGFs are the only known growth factors that support both sensory and motor nerve regeneration in adult animals (160, 238, 239, 240, 241, 242, 243). IGF-I is involved in brain plasticity processes (21, 217, 224, 244), and it specifically modulates synaptic efficacy by regulating synapse formation, neurotransmitter release, and neuronal exicitability (245, 246, 247, 248).

    IGF-I also provides constant trophic support to neuronal cells in the brain and in this way maintains appropriate neuronal function (245). Alteration of this trophic input may lead to brain disease as seen in neurodegenerative disorders such as Alzheimer’s disease (AD) (162, 249), Ataxia telangiectasia (250), Huntington’s disease (251), and Parkinson’s disease (252), all variably responsive to IGF-I treatment (see also Section VII). A recent study by Lichtenwalner et al. (253) showed that neurogenesis declines in brains of aged mice, but it is efficiently restored after IGF-I administration via intracerebroventricular infusion.

    E. IGF-I prevents neuronal apoptosis

    The ability of IGF-I (and IGF-II) to promote neuronal survival is associated with the ability of these factors to prevent apoptosis, and IGF-I appears to be a potent agent for rescuing neurons from apoptosis. For example, IGF-I prevents N-methyl-D-aspartate (NMDA)- and nitric oxide-induced apoptosis in hippocampal and cortical neurons (254, 255, 256, 257, 258).

    IGF-I has been shown to enhance the survival of rat embryo cerebellar granule cells and stimulate their terminal differentiation into cerebellar granule neurons (259, 260). Additionally, IGF-I promotes the survival of rat hypothalamic and hippocampal neurons in vitro (261). IGF-I and -II rescue chick MNs from injury-induced and developmentally regulated death (262, 263). A role for IGF-I in the regulation of Schwann cell survival has also been reported (264), as has the ability of IGF-I to prevent apoptosis in sympathetic neurons exposed to high glucose (265). Rat hippocampal neurons are also rescued by IGF-I from the induction of apoptosis by amyloid-derived peptides (162, 266, 267, 268). A similar antiapoptotic effect of IGF-I is observed in rat cerebellar neurons subjected to serum or potassium withdrawal or high KCl levels (269, 270), okadaic acid treatment (271), and in MNs during normal development or (262, 272) after axotomy or spinal transection (273, 274).

    Neuroblastoma cells are rescued from hyperosmotic shock-induced apoptosis by IGF-I (275) or metabolic stress (265, 276) including exposure to low glucose (277). IGF-I inhibits mature oligodendrocyte apoptosis during primary demyelination (278) and prevents apoptosis in neurons after nerve growth factor withdrawal (279) or serum withdrawal in brain explants (280) and neuronal cells (281).

    F. IGFBP action in the nervous system

    A growing body of evidence suggests an important role for IGFBP in the nervous system. The mRNA expression profiles and location of the most abundant IGFBP-2, -4, and -5 in the normal developing and adult CNS are well defined (31, 32, 33, 35, 282, 283, 284). IGFBP-3 (35, 284, 285) and IGFBP-6 (286) are also discretely expressed in the CNS but at lower levels, and therefore data on their mRNA expression distribution are limited. IGFBP-1 is not expressed in the CNS, but its expression is induced under certain experimental conditions (287).

    1. IGFBP-1.

    Although IGFBP-1 mRNA is normally not detectable in brain and the effects of Igfbp-1 gene deletion on CNS have not been reported, the overexpression of IGFBP-1 mRNA in brain has instead generated an interesting brain phenotype, namely small brain (288, 289, 290, 291, 292).

    IGFBP-1 is known to inhibit somatic linear growth, weight gain, tissue growth, and glucose metabolism (293). Increased expression of IGFBP-1 has been documented in a variety of situations, such as fetal nutritional deprivation and hypoxia, and has been considered to be a marker of metabolic disturbances that cause fetal growth retardation. Of interest, IGFBP-1 transgenic mice demonstrate both intrauterine and postnatal growth retardation phenotypes (288, 289, 290, 291, 292) including alteration of the somatotrophic axis (294).

    In a study by Doublier et al. (295), a line of transgenic mice with liver-specific expression of IGFBP-1 also showed impaired brain development and hydrocephalus.

    Furthermore, a study by Zhou and co-workers (287) described disrupted cerebellar morphogenesis and reduction in cerebellar size in mice overexpressing the hepatocyte nuclear factor-3 (HNF-3). The transgenic cerebella displayed levels of IGFBP-1 elevated to 22 times greater than those measured for wild-type cerebella, an elevation consistent with the reduction in transgenic cerebellar size.

    Together, these in vivo observations suggest that increased expression of IGFBP-1, as observed in a variety of clinical situations, may itself contribute to the growth retardation and impaired fetal brain development. However, it is suggested that the underlying mechanisms in these events involve IGFBP-1-sequestration of IGF-I.

    2. IGFBP-2.

    IGFBP-2 is expressed early in embryogenesis (296) and by embryonic d 10 is highly expressed in neuroectoderm structures including the neural tube and neuroepithelium (296). During development, the most prominent sites of IGFBP-2 expression in the CNS comprise cells with nonneuronal phenotypes including the epithelium of the choroid plexus, the floor plate, and the infundibulum (30). Later in development, IGFBP-2 mRNA is detectable throughout the brain (33, 297), particularly in brain regions undergoing continuous remodelling as is the olfactory bulb (Fig. 1), the cerebellum, and the hippocampus (33, 35, 36, 297). IGFBP-2 expression correlates and complements that of IGF-II, and both IGFBP-2 and IGF-II protein are highly abundant in the cerebrospinal fluid (CSF) and choroid plexus (36). It is also known that IGFBP-2 binds IGF-II with a moderate preferential affinity over IGF-I (135).

    IGFBP-2 associates to cell surface proteoglycans in rat brain tissue (134) and neuronal cells (136), and IGF-I/IGFBP-2/proteoglycan complexes have been identified in rat brain tissue (298), but the role of these cell membrane complexes is not completely understood. However, it has been suggested that differential localization of the IGFBPs in the pericellular and extracellular space, involving components of the extracellular matrix (111, 134, 135, 136, 137, 138, 139), might regulate levels of diffusible/free IGFs. We have recently shown that IGF-I complexes with IGFBP-2 to promote neurogenesis in adult stem cells (213) and further demonstrated that neurogenesis was inhibited by IGFBP-2 antibody blockade (213), thus suggesting a key role for IGFBP-2 in this process. Additional evidence in support of an IGF-facilitating role for IGFBP-2 in the brain comes from colocalization of injected IGF-I with IGFBP-2 (299) and subsequent neuroprotection after hypoxic-ischemic injury, an effect not seen with an IGF variant that does not bind IGFBP-2 (300). It is therefore possible that brain IGFBP-2 regulates IGF-I cellular responses by facilitating receptor targeting of IGF-I or modulating IGF-I bioavailability in the pericellular space in vivo (111, 135). Cell-associated IGFBP-2 may therefore act as a "linker" molecule allowing pericellular storage of IGF.

    This IGF-I storage might be affected by the presence of IGFBP-2 proteases (136, 281, 301, 302) that generate IGFBP-2 fragments which have markedly reduced binding affinity for the IGFs (135, 136). An IGFBP-2 fragment was identified on the cell surface of neuronal cells (136). The presence of this proteolytic fragment of IGFBP-2, capable of binding IGF-I while simultaneously being bound to the cell surface, might point to a process whereby proteolysis of membrane-bound IGFBP-2 provides a mechanism for creating peri-receptor low affinity IGF-I binding sites (136). Although we have recently demonstrated that IGFBP-2 proteolysis occurs during neuronal differentiation (281), the precise physiological significance of IGFBP-2 proteolysis in the nervous system remains to be determined.

    Thus, IGFBP-2 possesses endocrine functions elicited by modulating serum IGF half-life and transport as well as autocrine/paracrine functions that result from blocking or enhancing the availability of IGFs to bind cell surface receptors. Despite these key functions, ablation of the Igfbp-2 gene (303) generated a phenotype less dramatic than that initially predicted. Selective alterations were reported for spleen and liver size, whereas the level of other circulating IGFBPs was found to be increased in the adult animals (304). The absence of a "brain phenotype" in the Igfbp-2 –/– mouse (303) suggests functional redundancy in the IGFBP family during development of the CNS (305).

    The IGFBP-2 transgenic mouse model developed by the Hoeflich group (306) has suggested that IGFBP-2 may be a negative regulator of postnatal growth, including brain growth, in rodents, potentially by reducing the bioavailability of IGF-I. These effects were seen even when hemizygous cytomegalovirus-IGFBP-2 transgenic mice were crossed with hemizygous phosphoenolpyruvate carboxykinase-bovine GH transgenic mice (with very high GH and IGF-I serum levels) (307), thus suggesting an inhibitory role for IGFBP-2 in vivo. However, in both studies (306, 307), IGFBP-2 was constitutively overexpressed in most tissues, including the brain, resulting in very high IGFBP-2 levels, and therefore it might be possible that any specific local enhancing activities of IGFBP-2 were overridden by the high systemic level of IGFBP-2. It is therefore possible that although IGFBP-2 is abundantly expressed in the brain, expression at nonphysiological levels (up to 9-fold over the normal endogenous levels) in the transgenic mice would dramatically change local brain IGF availability leading to the reported effects on brain size.

    An example of physiological change in local brain levels of IGFBP-2 and how this might affect IGF action has been reported by Cardona-Gomez et al. (308) in a specialized group of glial cells of the third ventricle named tanycytes. These cells have the ability to accumulate IGF-I and thus regulate IGF-I availability (309, 310, 311). Cardona-Gomez’s data show that estradiol and progesterone regulate local levels of IGFBP-2, including "peri-IGF-I-receptor" IGFBP-2 (308), and that these changes affect accumulation of IGF-I in tanycytes. This accumulation process might involve interaction of IGF-I with cell surface-associated IGFBP-2 before IGF-I being intracellularly translocated (308).

    3. IGFBP-3.

    IGFBP-3 is normally expressed at a low level in the CNS, mainly in nonneuronal structures including epithelial cells (35, 37, 312, 313), and the effects of IGFBP-3 gene deletion on CNS are either unknown or have not been reported.

    Nevertheless, in a study by Ajo et al. (201), IGFBP-3 was found to be up-regulated in rat cerebral cortical cells after GH stimulation. Ajo et al. show that GH promotes proliferation of neural precursors, neurogenesis, and gliogenesis and that these responses are mediated by locally produced IGF-I and its modulator IGFBP-3.

    Conversely, a study by Rensink et al. (314), investigating the mechanisms of amyloid- (A) deposition in cerebral blood vessel walls and Abeta-induced toxicity in AD, proposed that IGFBP-3 might contribute to neuronal degeneration in AD. It is therefore possible that, as seen in other systems, IGFBP-3 might promote either enhancement or inhibition of IGF-I action in brain cells in vitro, depending on the experimental conditions.

    The function of IGFBP-3 in vivo has been in part elucidated by the IGFBP-3 transgenic mouse model. This mouse showed modest intrauterine and postnatal growth retardation despite elevated circulating IGF-I levels (315). These mice showed a significant reduction in birth weight, reduction in litter size, and postnatal reduction of both body weight and length and organ size. No change in brain growth or phenotype has been reported in IGFBP-3 transgenic mice (316).

    4. IGFBP-4.

    IGFBP-4 is normally expressed at a very low level in the CNS, where its mRNA is found in a variety of brain cell types including meningeal cells, astrocytes, and fetal neuronal cells (317). During early brain development, IGFBP-4 expression is increased, and its mRNA is easily detectable in regions such as the choroid plexus, meninges, and the basal ganglia (318). Postnatally, IGFBP-4 mRNA is found in the meningeal cell layer surrounding the developing cerebellum in the hippocampal formation and olfactory bulb (Fig. 1) (35, 317). In some of these IGFBP-4-expressing brain regions, which maintain a degree of tissue remodelling/plasticity (38), local expression for IGF-I and its receptor is also seen, thus suggesting that IGFBP-4 may play a role as a local modulator of IGF action (318).

    Overexpression of IGFBP-4 gene has only been recently investigated in smooth muscle cell-rich tissue, and therefore the effects to the nervous tissue are unknown.

    As discussed earlier, IGFBP-4 generally acts in vitro and in vivo as a potent inhibitor of IGF-I action (147, 319, 320, 321, 322), and it is therefore intriguing to find IGFBP-4 in sites where IGF-I exerts its primary mitogenic and trophic actions. A potential explanation might come from the work of Pintar et al. (305) on IGFBP-4 null mice, which indicates, contrary to expectation, that these mice have lower weight at birth. In this case, the authors argued that the absence of IGFBP-4 diminished tissue IGF storage capacity (305). This would imply that physiological levels of IGFBP-4 are indeed required for normal growth. Whether ablation of the IGFBP-4 gene also affects the CNS of these mice was not reported.

    5. IGFBP-5

    IGFBP-5 gene expression is highly abundant during brain development (32). The early expression of IGFBP-5 at embryonic d 10.5 indicates a key role of this IGFBP during embryogenesis (323). This hypothesis is further supported by a recent study from Pera et al. (150) which shows that IGFBP-5, as well as three IGFs expressed in early embryos, promoted anterior development by increasing the head region in Xenopus embryos. Thus, active IGF signals, including IGFBP-5, appear to be required for anterior neural induction in Xenopus (150). Whether IGFBP-5 has similar functions in early mammalian neural development is not known.

    In rodents, IGFBP-5 appears to be coexpressed with IGF-I in principal neurons of sensory relay systems, cerebellar cortex, hippocampal formation, and many other neuron-rich regions (32, 324), including the olfactory bulb (Fig. 1) (35). These data point to the presence of potential autocrine and paracrine interactions between IGFBP-5 and IGF-I in specific brain regions where IGFBP-5 may act as a modulator or determinant of IGF action (325).

    In addition to this spatiotemporal coexpression of IGFBP-5 and IGF-I, it is now becoming clear that IGF-I specifically regulates IGFBP-5. Using two IGF-I transgenic mice lines, Ye and D’Ercole (326) demonstrated that IGF-I up-regulates IGFBP-5 expression in vivo. This increase is specific for IGFBP-5 mRNA, because the level of expression of IGFBP-2 and IGFBP-4 mRNAs in these mice was found unchanged (326). The effects of IGFBP-5 gene ablation to the brain have not been reported.

    6. IGFBP-6.

    IGFBP-6 is poorly expressed in the nervous system, and information regarding its mRNA expression distribution, in both the developing and adult nervous system, is limited.

    IGFBP-6’s unique property of preferential binding to the IGF-II ligand (327), coupled with the fact that this ligand is the most abundantly expressed IGF in the adult CNS, suggests that the IGFBP-6/IGF-II complex has a unique role in modulating IGF-II function in the adult brain (286).

    During CNS embryogenesis, IGFBP-6 expression is tightly restricted to trigeminal ganglia and, relative to the rest of the embryo, this structure has the highest expression (286). The expression in the forebrain and cerebellum does not occur until after postnatal d 21 and then is primarily associated with GABAergic interneurons (286). The highest levels of expression in the adult animal are in the hindbrain, spinal cord, cranial ganglia, and dorsal root ganglia (286). These nuclei in the hindbrain and periphery that express IGFBP-6 are all associated with the coordination of sensorimotor function in the cerebellum, which indicates an important role for the IGFBP-6/IGF-II complex in the function and maintenance of these systems.

    As introduced earlier, IGFBP-6 preferentially binds IGF-II and is regarded as a relatively specific inhibitor of IGF-II actions (327). IGFBP-6 is often expressed in nonproliferative cells, and its expression is associated with inhibition of growth of tumor cells in vitro (281, 327, 328) and in vivo (327). These findings are also supported by overexpression of IGFBP-6 in vivo, a model recently developed by the Babajko group (329) with strong expression of IGFBP-6 in glial fibrillary acidic protein-positive cells. Preliminary analysis of the IGFBP-6 transgenic mouse shows reduced cerebellum size and weight combined with altered differentiation of astrocytes (329). Abnormalities in the hypothalamus and pituitary were also reported (329). The effects on the brain of IGFBP-6 gene ablation remain unknown.

    V. The IGF System and Neuroendocrine Cross-Talk in the Nervous System

    A. IGF-I and fibroblast growth factor (FGF)-2

    In vivo, growth factors such as IGF-I do not exist in isolation. Hence, the presence of other growth factors (Fig. 2) may further modulate the biological activity of IGFs and cellular responses. This modulation can be seen in the neurogenic activity of IGF, which in many cases requires the presence of basic FGF or FGF-2 to function (21, 330, 331).

    FIG. 2. Neuroendocrine cross-talk. In vivo, growth factors such as IGF-I do not exist in isolation. Hence, the presence of other growth factors may further modulate the biological activity and cellular responses of IGFs.

    Torres-Aleman and co-workers (332) showed that in primary cultures of hypothalamic cells, FGF-2 combined with IGF-I, significantly increased the number of neurite bearing cells, above that seen when either one of the two growth factors was added in isolation. Indeed, it appeared that the observed effect of these two growth factors was synergistic (332).

    In a study by Drago and co-workers (330), it was shown that by blocking the endogenous stimulation of IGF-I with an antibody, the ability of FGF-2 to stimulate the proliferation of precursor cells isolated from the neuroepithelium of embryonic d 10 mice was abrogated. In later work by Frodin and Gammeltoft (333), it was also found that the IGFs act synergistically with FGF-2 and nerve growth factor to promote chromaffin cell proliferation. A recent study from our laboratory (281) demonstrated, in neuroblastoma cells, that FGF-2 overrides IGF-I mitogenic and survival activity via induction of neuronal differentiation and blockade of the antiapoptotic response of IGF-I.

    Furthermore, Torres-Aleman (243) recently highlighted the importance of a number of serum growth factors (in contrast to brain-produced), which included IGF-I, FGF-2, and other blood-borne neurotrophic factors (neurotrophins), in neuroprotective surveillance. All these factors exerted a tonic trophic input on brain cells, providing defense mechanisms ranging from blockade of neuronal death to up-regulation of neurogenesis.

    B. IGF-I and neurotrophic factors

    Increased interest has recently been devoted to the interaction of IGF-I with other "classic" neurotrophic factors. A study by Corse et al. (334) has investigated the potential effects of neuroprotective factors including IGF-I, glial cell line-derived neurotrophic factor, brain-derived neurotrophic factor, neurotrophin (NT)-4 and -3 (NT-4/5, NT-3), and ciliary neurotrophic factor on mature motor neurons. Only IGF-I, glial cell line-derived neurotrophic factor, and NT-4/5 were found to be potently neuroprotective, pointing to the potential combined use of these neuroprotective factors in treatment of neurodegenerative diseases. Subsequently, the results from a number of phase III clinical trials with nerve growth factor, brain-derived neurotrophic factor, and IGF-I (335) indicate that these neurotrophic factors may find an application in degenerative disorders or injury of peripheral nerves and motor neurons (336).

    C. IGF-I and GH

    Although GH gene expression also occurs in the central and peripheral nervous system, with brain GH imunoreactivity not affected by hypophysectomy (337) (178), the potential cross-talk between the IGF system and GH in the nervous system has not been fully investigated. Several experimental models, in vivo and in vitro, support a key role for GH in brain development. The Snell dwarf mouse (Pit1) and the GHRH receptor-deficient little mouse exhibit microcephalic cerebra with hypomyelination and retarded neuronal growth with poor synaptogenesis. Postnatal GH administration normalizes neuronal growth in these mice (338, 339). Recent in vivo studies by the Williams group have demonstrated that GH is involved in neuroprotection during hypoxic/ischemic brain injury (340, 341, 342). In these studies, they also showed GH-like immunoreactivity on injured brain cells and demonstrated that GH administered intracerebroventricularly is capable of preventing brain cell loss (340, 341, 342). Whether the neurogenicprotective effects of GH involve IGF-I induction is not clear.

    However, some recent in vitro studies by Ajo et al. (337) have demonstrated that GH promotes proliferation and differentiation of fetal cerebral cortical cells in primary culture, and these effects are mediated by IGF-I. Furthermore, a study by Edmondson and co-workers (343) has demonstrated that GH and IGF-I promote growth and survival of melanocytes (neural-crest derived cells) by a process synergistic with FGF-2.

    Furthermore, there is some evidence that the GH-related hormone prolactin (PRL) also possesses neurogenic properties. In female mice during pregnancy, PRL stimulates production of neuronal progenitors in the forebrain subventricular zone (344, 345). Whether the neurogenic properties of PRL are a direct effect of its action or mediated by other factors, including IGF-I, remains to be determined.

    D. IGF-I and erythropoietin

    Erythropoietin (EPO) is traditionally known as a hematopoietic cytokine produced by the fetal liver and adult kidney in response to hypoxia (346, 347, 348, 349, 350). However, the expression of EPO and EPO-receptors in the CNS and the up-regulation of EPO by hypoxic-ischemic insult (346, 347, 348, 349, 350) suggest that this cytokine is an important mediator of the brain’s response to injury. In fact, in vivo EPO administration protects hippocampal CA1 neurons and retinal neurons from ischemic damage and prevents brain injury after a number of "insults" (346, 347, 348, 349, 350) by mechanisms involving both inhibition of apoptosis and neurotrophic actions (346, 347, 348, 349, 350, 351, 352). In the brain, EPO expression, regulated by the hypoxia-inducible factor (HIF)-1, is mainly found in astrocytes.

    Recent work by Wang et al. (353) demonstrated that IGF-I protection of primary neuronal cells exposed to low oxygen concentration correlates with activation of HIF-1 expression (351, 352). In the same studies using an in vivo model of hypoxia/ischemic brain injury, they also show that IGF-I transcriptional activation correlates with that of HIF-1, suggesting that HIF-1 might mediate some of the IGF-I responses. In another study by Chavez and LaManna (354), it was shown that IGF-I induces HIF-1 transcriptional activity in rat cerebral cortex and neuronal cells (PC12) and that this induction is abolished by a selective IGF-I receptor antagonist (JB-1) (354). Furthermore, an early study by Feldser and co-workers (355), in hepatoma cells, not only showed that insulin, IGF-I, and IGF-II induced HIF-1 expression but also demonstrated that HIF-1 is required for expression of IGF-II, IGFBP-2, and IGFBP-3 (355).

    These findings suggest the presence of a potential complex synergistic cross-talk between the IGF and the EPO system involving both activation of common intracellular signaling pathways and regulation of gene expression. The presence of synergistic cross-talk between the IGF and the EPO system in neuronal cells has recently been demonstrated by Digicaylioglu and co-workers (356), showing that EPO can exert a more immediate neuroprotective action when administered in concert with IGF-I. The neuroprotective mechanism, after coadministration of EPO and IGF-I, involved synergistic activation of the PI3K–Akt pathway (356). These findings further support the concept that the coadministration of synergistic neuroprotective agents rather than a single agent might provide improved therapeutic outcome. Thus, treatment with appropiate combinations of EPO and IGF-I could be a future therapeutic strategy for a variety of acute neurological events (356).

    E. IGF-I and sex steroids

    Among the numerous endocrine systems regulating brain physiology, sex steroids play an important role. However, it appears that part of the effects of these hormones on the brain are mediated by trophic factors including IGF-I (357, 358, 359, 360, 361). Studies from the Garcia-Segura group (357, 358, 359, 360, 361) have demonstrated that estradiol and IGF-I increase survival and differentiation of developing fetal rat hypothalamic neurons. They have demonstrated that estrogen-induced activation of the estrogen receptor requires the presence of IGF-I and that both estradiol and IGF-I use the estrogen receptor to mediate their trophic effects on hypothalamic cells. In vivo sex steroids affect IGF-I levels in the endocrine hypothalamus, arcuate nucleus, and median eminence (357, 358, 359, 360, 361).

    Furthermore, increased clinical and basic evidence suggests that gonadal steroids affect the onset and progression of several neurodegenerative diseases and schizophrenia and the recovery from traumatic neurological injury such as stroke. In the brain, similarly to the IGF system, both estrogen synthesis and estrogen receptor expression are up-regulated at sites of injury (362, 363). Once again, it is suggested that the neuroprotective effects of estrogen may be exerted independently of the classical nuclear estrogen receptors involving modulation of the IGF-I signaling (362, 363). This is supported by the fact that estrogen receptors and IGF-I receptor interact in the activation of PI3K and MAPK signaling cascades and possibly in the promotion of neuroprotection (362, 363). It is therefore possible that the decrease in estrogen and IGF-I levels with aging may thus result in an increased risk for neuronal pathological alterations. Also, in this case these findings suggest the presence of a potential complex synergistic cross-talk between the IGF and sex steroids involving activation of common intracellular signaling pathways, however the precise mechanism remains unclear.

    VI. Altered Expression of the IGF System in Response to CNS Injury

    A role for endogenous IGFs in the injured brain is suggested by a number of studies showing the induction of components of the IGF system after transient unilateral hypoxic/ischemic injury and stroke in the rat model (364, 365, 366, 367, 368, 369, 370, 371) (Fig. 3, A–C). IGF-I mRNA induction is seen within infarcted regions by 3 d after hypoxia (369) (Fig. 3A). In addition, IGF-II (365), IGFBP-2 (365, 370) (Fig. 3C), IGFBP-3 (369) (Fig. 3A), and IGFBP-5 (368) (Fig. 3B) genes are differentially induced in specific regions after hypoxic/ischemic injury in the same model, suggesting that they may modulate the actions of IGF-I in a spatiotemporal-specific manner. The induced expression of IGF-I and IGFBP-2, -3, -4, and -5 in reactive microglia, oligodendrocytes, astrocytes, and surviving neurons (313, 370) of the periinfarcted area, including areas distant from the region of cell loss, suggests a role for the local IGFBPs in transporting IGF-I from its sites of production to the sites of action (313) (Fig. 3, A–C). Furthermore, in the periinfarcted regions, IGFBP-2 was highly expressed by reactive astrocytes that were juxtaposed to surviving neurons (Fig. 3C) (313), therefore suggesting a specific role for IGFBP-2 in augmenting IGF-I action (313) (Fig. 4).

    FIG. 3. Altered expression of the IGF system in response to CNS injury. A role for endogenous IGF system in the injured brain is suggested by a number of studies showing the induction of components of the IGF system after transient unilateral hypoxic/ischemic injury and stroke in the rat model. IGF-I mRNA induction is seen within infarcted regions by 3–5 d after hypoxia/ischemic brain injury (d 5 after insult is shown). In addition, IGF-I receptor and IGFBPs genes are differentially induced in specific regions after hypoxic/ischemic injury in the same model, suggesting that they may modulate the actions of IGF-I in a spatiotemporal-specific manner (d 3 and 5 after insult are shown). In panels A–C, IGF-I, IGF-I receptor, IGFBP-3, IGFBP-5, and IGFBP-2 mRNA were detected by in situ hybridization (ISH; 5-μm paraffin sections). Immunohistochemical staining for glial fibrillary acid protein (GFAP) is shown in panels A and C, whereas staining for the 150-kDa neurofilament (150 kDa-NF) is shown in panel B. Immunoreactivity for IGFBP-2 is shown in panel C. CO, Cortex; AH, Ammon’s horn; DG, dentate gyrus; IHC, immunoistochemistry. Hematoxylin-eosin staining is shown in the left panels [obtained from L. W. Swanson: Brain Maps: Structures of the Rat Brain, 2nd edition, Elsevier, Amsterdam, 1998/1999 (528 ), with permission from Elsevier].

    FIG. 4. A model for neurotrophic and neuroprotective actions of IGF-I in brain. Secreted IGF-I exerts local autocrine (1 ) or paracrine (2 ) trophic actions. A family of IGFBPs (3 ) modulates IGF-I bioavailability. IGFBP-2, the most abundant brain IGFBP, mediates pericellular storage of IGF-I via interaction with cell surface proteoglycans (PG) (4 ) or components of the extracellular matrix. Cell surface IGF-I/IGFBP-2/PG complexes (4 ) are suggested to play a role in targeting of IGF-I to its membrane receptors. IGFBP-2 mediated IGF-I receptor targeting at the cell surface, and this event might be further potentiated by the presence of a specific IGFBP-2 protease (5 ), which generates IGFBP-2 fragments that have reduced affinity for IGF-I. In response to a number of cerebral insults (i.e., hypoxia/ischemic brain injury) (6 ), IGFBP-2 proteolysis might also affect the level of pericelluar IGF-I (7 ), therefore augmenting its neuroprotective activity (8 ). Following cerebral insult activation and recruitment of specialized brain cells (9 ) might further contribute to modulate the local IGF system (9 ).

    A facilitative role of IGFBP-2 in IGF-I action in the nervous system is also suggested by a study from the Gluckman group (299). Using the hypoxic-ischemic brain injury rat model, we have demonstrated that centrally administrated (intracerebroventricular) 3H-IGF-I is rapidly translocated to neurons and glia in injured sites (299) and that the tritiated IGF-I signal colocalized with IGFBP-2 protein in neurons and glia from the cerebral cortex and subregions of the hippocampus. The 3H-IGF-I signal persisted for up to 6 h after administration, suggesting local storage of the IGF-I molecule (299). These data, together with their previous reports on the inefficacy of the IGF-I analog des(1-3) IGF-I to rescue hypoxic-ischemic injured neurons (300), further suggest that local IGFBPs (i.e., IGFBP-2) play a key role in IGF-I delivery to the injured site (299, 313) (Fig. 4). These findings might point to a role for cell-associated IGFBP-2 (134) to modulate local IGF-I bioavailability (Fig. 4).

    IGFBP-2 mRNA is induced throughout the injured hemisphere (313) (Fig. 3C) whereas IGFBP-3 is only moderately induced in reactive microglia and glial cells and is substantially decreased in neuronal cells of the region of injury (372) (Fig. 3A). However, in the early stages of the injury response, IGFBP-3 expression increases rapidly in vascular endothelial cells throughout the affected hemisphere with maximal expression levels at 24 h, which then become undetectable 48 h later (372). This early transient induction of IGFBP-3 in brain vascular endothelial cells is a likely mechanism by which brain vascular endothelial cells potentially regulate up-take of circulating IGFs after the hypoxic-ischemic insult (372). However, the precise mechanism involved remains to be determined. Nevertheless, these data might point to a potential role for IGFBP-3 as carrier/transporter of vascular IGF-I into the brain tissue in the early phases of the injury response, when local IGF-I availability and expression might not be sufficient to trigger the neuroprotective mechanisms (Fig. 4).

    After hypoxic-ischemic insult, IGFBP-4 mRNA is also moderately up-regulated (365, 368). This is also true for IGFBP-5 (Fig. 3B), which is up-regulated after severe hypoxic-ischemic injury in the infant rat brain (368), and its mRNA is up-regulated in cerebellar granule cells during apoptosis (325). In both cases, the increase of IGFBP-5 expression is believed to be required to maximize the availability of IGF and thus potentiate the IGF-triggered survival signaling, as has previously been described for cerebellar granule cells (325).

    After peripheral nerve injury (lumbar motoneurons), IGFBP-6 mRNA and its protein expression are strongly up-regulated in the spinal motoneurons. This increased expression of IGFBP-6 in the damaged nerve is spatiotemporally associated with a local increased expression of the IGFs (373). It is therefore likely that IGFBP-6 might play a specific role in controlling IGF availability to lesioned motoneurons and thus regulates axonal regeneration (373). These findings all suggest that the IGFs in the brain, after injury, may act as endogenous neuroprotective agents and limit the degree of neuronal and glial loss (Fig. 4).

    VII. Altered Expression of the IGF System in Malignancies of the Nervous System

    IGF-IR plays a crucial role in the induction and maintenance of the malignant phenotype. Increased expression of IGF-I, IGF-II, and IGF-IR is present in a wide range of human cancers (374), including lung, breast, thyroid, and prostate carcinomas, rhabdomyosarcomas, leukemias, and the peripheral nervous system tumor neuroblastoma (375, 376). IGF-IR overexpression leads to cellular transformation (374, 377), tumor cell proliferation (378, 379), and growth (380), whereas disruption of IGF-IR expression reverses the transformed phenotype (374, 377).

    Within the CNS, the IGF family is critical for normal development: aberrant expression and/or activation may be associated with CNS malignancy (381, 382). Components of the IGF system, in particular IGF-I, IGF-II, and the IGF-IR, are overexpressed to varying degrees in numerous CNS tumors (381, 383), including low-grade gliomas, glioblastomas, medulloblastomas, astrocytomas, ependymomas, and meningiomas.

    Expression of the IGF family in CNS malignancy has been most thoroughly investigated in glioblastoma. IGF-IR expression is higher in glioblastomas than in normal brain, and glioblastoma cell lines exhibit a dose-dependent IGF-I stimulation of both receptor autophosphorylation and thymidine incorporation, indicating a functional receptor (384). Both ligands for the IGF-IR, IGF-I and IGF-II, have increased mRNA transcript formation in glioblastoma compared with normal brain, and the major IGF-II transcript of 6.0 kb found in glioblastoma is similar to that found in fetal brain (385). The IGF ligands are also secreted into the CSF of glioblastoma patients (386). Most of the IGF binding proteins, including IGFBP-1, -2, -3, -5, and -6, are expressed in glioblastomas, with the most abundant being IGFBP-3 (387). In addition, glioblastomas express membrane-bound forms of the binding proteins, and IGFBP-3 is secreted into culture media (387). Much of the in vitro work on the role of IGF-IR in transformation has been performed in rat glioblastoma cell lines. In this work, researchers have found through mutational analyses that distinct regions of IGF-IR are critical for the biological actions of the IGF ligands, including mitogenesis, transformation, and differentiation, through the induction of specific downstream targets (388, 389, 390, 391, 392).

    IGF ligands acting through the IGF-IR are known to affect several aspects of glioblastoma tumor formation in addition to growth and transformation. IGF-I stimulates the migration (393) and invasion (394) of glioblastoma cell lines in culture, prerequisites for tumor cell metastasis. IGF-I acts as a potent survival factor in glioblastoma cell lines, even overcoming proapoptotic stimuli (394, 395). Glioblastoma cell treatment with IGF-I triple helix-forming DNA (396, 397, 398), antisense IGF-IR (399, 400, 401, 402), kinase-defective IGF-IR (403), or additional IGF-IR mutant dominant-negative constructs (404) all induce growth suppression and/or apoptosis, resulting in decreased tumor growth. These studies have led the way for future work on IGF-IR inhibition and ligand interference in glioblastoma patient therapy (382).

    As previously indicated, expression of IGF system components is also altered in other CNS tumors. In infiltrating astrocytomas, IGF-IR expression increases early in tumor development, appearing in stage II tumors and continuing into stage IV (204). IGF-IR protein levels are increased in the majority of medulloblastoma cases, with concomitant increases in phosphorylated forms of the downstream signaling molecules Erk-1, Erk-2, and Akt/PKB (405, 406). When IGF-II or activated Akt/PKB is retrovirally transferred into neural progenitors along with sonic hedgehog-patched, medulloblastomas form (407), suggesting a role for this pathway in tumor formation. In microarray analyses of ependymomas, high IGF-II expression was detected (408). High IGF-II expression occurs in meningiomas, and the IGF-II/IGFBP-2 ratio, indicating free IGF-II levels, correlates with tumor anaplastic histopathology (381). In fact, meningiomas express the highest IGF-II mRNA levels of all intracranial tumors (409). Although IGF ligand levels are increased in the majority of CNS tumors, ligand increases are not detected in the CSF, indicating that IGFs act locally in an autocrine and paracrine fashion to control tumor cell proliferation (386). This idea is supported by studies in both primary intracranial tumors and brain metastases from non-small cell lung tumors that show that interference with the IGF-I/IGF-IR autocrine signaling prevents cell growth (410, 411).

    The IGF system is also important in neuroblastomas, which typically occur peripherally but are occasionally found intracranially. IGF expression is present in all stages of primary neuroblastoma tumors (412). IGF-I or IGF-II coupled to IGF-IR promotes both autocrine and paracrine growth and survival of human neuroblastoma cell lines (413, 414, 415, 416, 417, 418). IGF-IR overexpression protects neuroblastoma from apoptosis (75, 275, 378, 419, 420, 421, 422, 423), whereas inhibition of IGF-IR using antisense strategies blocks tumor growth and induces regression of neuroblastoma tumors in mice (424). Because neuroblastoma is also thought to arise through improper differentiation, these studies coupled with the CNS tumor reports suggest a critical role for the IGF family in nervous system tumor development and growth.

    VIII. IGF-I Therapy in Nervous System Disease Models

    A. IGF-I therapy in acute central nervous system disease

    After hypoxic/ischemic brain injury in the 21-d-old rat, cell death in the pyramidal cells of the CA1/2 region, striatum, thalamus, and cortical layers 3–5 of the ligated hemisphere becomes evident within a few hours, is maximal by 24–72 h after injury, and then reduces (367). In contrast, the level of endogenous IGF-I mRNA does not rise until after 24 h from injury, becoming maximal by d 5 (235). This is compatible with earlier observations that after traumatic brain injury (425), bioassayable neurotrophic activity in cortical wounds is not induced until 6 d after injury in adult rats and 3 d after injury in infant rats. These observations of a temporal mismatch between the induction of cell death and the expression of neurotrophic activity led to the hypothesis that earlier administration of potential neurotrophins such as IGF-I might be neuroprotective.

    In adult rats with unilateral hypoperfusion injury, intracerebroventricular administration of IGF-I 2 h after injury was shown to be neuroprotective in a dose-dependent fashion (369, 426) with a marked reduction in cortical infarction, suggesting that both neurons and glia are protected (426). The window of opportunity for neuronal rescue was limited to only within 2 h of injury, but could be extended by several hours if combined with postischemic hypothermia. The latter treatment is assumed to slow the processes of delayed cell death (299, 426). The effects of IGF-I were shown to enhance functional as well as histological outcome (427). The late apoptosis that continues for many days after a stroke-like injury is also inhibited by acute postischemic IGF (427). The neuroprotective action of IGF-I is likely to be dependent on the capacity of IGFBPs to translocate IGF-I from the CSF to the site of injury. IGF-II administered in conjunction with IGF-I attenuates the neuroprotective effect of IGF-I, possibly by competing for IGFBPs (300).

    In rats, intranasal IGF-I reduced neuronal infarction after middle cerebral artery occlusion (428, 429). Other studies have confirmed the neuroprotective effect of IGF-I in spinal cord injury (430), and in the late gestation fetal sheep subject to cerebral ischemia (431). In addition, there is indirect evidence in rats of improved cognitive function after traumatic brain injury treated with IGF-I (432). Studies in the fetal sheep also suggest that IGF-I can protect oligodendrocytes and, thus, myelin production (433). This is compatible with the observation that in overexpressing IGF-I transgenic mice there is a reduced number of apoptotic oligodendrocytes (278). However, it has also been shown that IGF-I causes an increase in microglial number and increased gliosis, as indicated by the density of reactive astrocytes, and both of these cell types, in turn, express IGF-I (207, 433).

    B. IGF-I therapy in chronic central nervous system disease

    IGF-I has also been explored in chronic neurological disease. Clinical trials of IGF-I have been conducted in MN disease (434, 435, 436, 437) based on preclinical in vitro, in ovo, and in vivo observations in a variety of rodent models (438, 439, 440). The results have been conflicting. A recent systematic review concluded that although IGF-I therapy may have a modest effect, the current data are insufficient for definitive conclusions (434). It is also difficult in such studies to separate effects on the CNS from peripheral effects, for example, on muscle.

    Experimental autoimmune encephalomyelitis is used as a preclinical model for multiple sclerosis. In this model, IGF-I is reported to improve neurological and histological outcome (441, 442). More recently, it has been shown that if IGF-I is given before motor signs of encephalomyelitis are obvious, it appears to delay disease onset. Once disease takes hold, however, it shows either no beneficial effect on remyelination (443) or worsens disease outcome, particularly if the levels of bioavailable IGF-I are increased by coadministration with IGFBP-3 (444). Similarly, it has been suggested that chronic IGF-I administration may have some therapeutic role in AD (162, 445, 446). IGF-I in vitro inhibits amyloid induced neuronal death (266), induces choline acetyl-transferase (445), and affects CNS amyloid- levels (162). Studies of this kind are limited by the poor transfer of IGF-I across the intact blood-brain barrier. However, IGF-I will cross the compromised blood-brain barrier, as in multiple sclerosis or other acute syndromes (447), and new delivery approaches are in development (448).

    C. IGF-I therapy in neurodegenerative disorders

    1. MN disease.

    MN disease is a heterogeneous group of neurodegenerative disorders that selectively affect upper and/or lower MN leading to muscle atrophy and weakness. This disease of late middle age is relentlessly progressive and almost invariably lethal within 5 yr of onset of symptoms. Amytrophic lateral sclerosis (ALS) accounts for approximately 80% of all cases of MN disease and has an estimated incidence of three to five cases per 100,000 (449). Of the remaining patients, some will have a familial form of ALS, due to a mutation in cytosolic Cu/Zn superoxide dismutase (SOD1) (450). Although it is not known exactly how mutant SOD1 injures MN, mice overexpressing mutant SOD1 develop a denervating illness that resembles ALS despite normal or increased levels of wild-type SOD1 activity (449).

    Accumulating data support the therapeutic use of IGF-I in the treatment of ALS. As discussed earlier, IGF-I is essential for normal development (37, 451) and is the only known neurotrophic factor capable of supporting both sensory and motor nerve regeneration in adult animals (37, 451). The ability of IGF-I to promote neuronal survival is directly related to its potent antiapoptotic properties (376). IGF-I prevents apoptosis in MN (439, 452), glial cells (reviewed in Ref.376), and muscle cells (reviewed in Ref.453), all cell types affected by ALS (454). IGF-I serum levels are decreased in ALS patients (455) and could contribute to the development of disease (455, 456). IGF-I preserves MNs in the wobbler mouse (457), and expression of the IGF-I receptor is increased in the spinal cord of ALS patients, suggesting that there may be a compensatory increase in IGF-I receptor (458). Two placebo-controlled trials of IGF-I in ALS patients have produced mixed results. The North American ALS/IGF-I Study Group found that patients receiving IGF-I daily for 9 months had slower disease progression and reported a better quality of life than placebo-treated controls (435). However, the European ALS/IGF-I Study Group showed no benefit to IGF-I therapy in a similar paradigm (437). Thus, although the sc administration of IGF-I to ALS patients was of uncertain clinical benefit (435, 459), the data were sufficiently promising to support another multicenter clinical trial. The Great Lakes ALS Consortium, under the direction of the Mayo Clinic, is currently entering patients into a new double-blind placebo control trial of sc IGF-I in the treatment of ALS (www.alsa.org). The clinical data in man also lead investigators to employ viral gene therapy for targeted delivery of IGF-I to MN in mouse models of ALS. Retrograde delivery of IGF-I to MN occurs after im injection of IGF-I linked to adeno-associated virus and prolongs survival in a mouse model of ALS (439). The Robert Packard ALS Center at Johns Hopkins is proposing a trial of adeno-associated virus-IGF-I in ALS patients to begin within 1 yr (www.alscenter.org).

    2. AD.

    AD, the most common type of senile dementia in the elderly, is characterized neuropathologically by the presence of intracellular neurofibrillary tangles, extracellular diffuse neuritic plaques, and the loss of neurons in a defined region of the brain (460, 461, 462, 463, 464). The neuritic plaque contains amyloid filaments, dystrophic neurites, activated microglia, and astrocytes. The principal component of amyloid fibrils is the -amyloid peptide (A) (460, 462, 465). Although accumulation of amyloid plaques in the brain (463, 464) appears to be the major cause of familial AD onset, the role of this neuroanatomical alteration in sporadic forms of the disease is not fully understood. Evidence suggests that altered levels of growth factors and/or their receptors, which exert trophic and survival function in the nervous system, may underly neuronal degeneration as seen in AD brain (466, 467, 468). Metabolic and hormonal functions decline in a variety of severe age-associated pathologies, including those associated with alterations in the insulin and IGF systems (162, 164, 445, 467, 469, 470, 471, 472). AD patients show changes in circulating levels of both insulin and IGF-I, often associated with abnormal responses to insulin (473, 474, 475). Compelling evidence indicates that insulin and IGF-I have a direct effect on the metabolism and clearance of the A (162, 446, 463, 476, 477). In neuronal cells, insulin inhibits A degradation (162, 476, 477), directly increases A secretion, and decreases the intracellular level of A peptides by stimulating their intracellular trafficking (476, 477). On the other hand, serum IGF-I levels correlate with cerebral levels of A in aged rodents (162, 446). Liver-specific deletion of the igf-1 gene also has prematurely increased cerebral levels of A (162). IGF-I administration to aged rats decreases the level of A in the brain parenchyma to the levels found in young rats (162, 446), which is balanced by increased levels of A in the CSF (162, 446). IGF-I increases A clearance from the brain by enhancing transport of A-carrier proteins into the brain through the choroid plexus (162). The effects of IGF-I on A clearance are ablated by TNF, a cytokine involved in neurodegeneration-associated brain impairment (162, 446, 477).

    Decreased circulating insulin and IGF-I might influence the development of neurofibrillary tangles, another AD "marker." This process appears to be a consequence of altered tau phosphorylation, a major component of neurofibrillary tangles (477, 478, 479). Nonphosphorylated tau protein plays a key role in the assembly of tubulin monomers into microtubules to constitute the neuronal microtubule network, which is necessary to maintain neuronal cell shape and axonal transport (477, 478, 479). However, hyperphosphorylation of tau, as seen in AD, produces destabilization and dissociation of microtubules, forming abnormal filaments that aggregate in neurofibrillary tangles (477, 478, 479). Insulin and IGF-I reduce tau phosphorylation and promote tau binding to microtubules in neuronal cells (480, 481, 482, 483), whereas disruption of insulin and IGF-I signaling, as seen in the IRS-2 knockout mouse model (483), increases tau phosphorylation as seen in AD. These findings indicate a potential key role for insulin and IGF-I in regulating tau protein phosphorylation, a failure of which augments the onset of neurofibrillary-tangle AD pathology. The role of other IGF system components, such as the IGFBPs, in AD pathology remains unclear (474, 484).

    The data discussed above point to insulin and IGF-I signaling as a potential therapeutic target in AD. The effects of IGF-I on key AD markers (i.e., amyloid toxicity, tau phosphorylation) suggest the potential usefulness of this growth factor in the treatment of neurodegenerative diseases. The primary challenge in translating successful use of IGF-I in laboratory research models to growth factor therapy in the clinic is delivery of IGF-I to the brain in sufficient concentrations to influence neuronal functions (445, 448, 477). A number of recent novel approaches might offer solutions to this problem. Born et al. (485), and more recently De Rosa’s group (486), have illustrated growth factor delivery to the brain via intranasal administration. Carrascosa et al. (448) have succesfully used IGF-I microsphere therapy (sc delivery) in rodents to prevent Purkinje cell degeneration. These studies suggest that IGF-I may potentially be delivered to the brain to prevent pathophysiological changes associated with the development of AD.

    D. Neuropathy: a potential target for IGF-I therapy

    The study of the IGF system in peripheral nerve injury has focused on both crush and transection of the sciatic nerve. Sciatic nerve crush in the rat increases IGF-I mRNA distal to the crush site, whereas IGF-I expression at the crush site is not increased until 4 d after injury (487). IGF-I and IGF-IR up-regulation are observed after sciatic nerve transection (373, 488). During the first 3–7 d after transection, the expressed IGF-I is localized mainly in Schwann cells of the intact nerve and the distal stump (488, 489). IGF-I expression has also been detected in transected facial nerve 4–7 d after transection, where it is mainly localized in astrocyte processes (490).

    IGF-I supports neurite outgrowth in guinea pig myenteric plexus neurons, (491) chick sympathetic and motor neurons (272, 492, 493), and rat sensory and sympathetic neurons (265, 494). Most of the evidence for IGF-promoted neurite extension comes from studies of transected peripheral nerves. For example, rats treated with IGF-I after sciatic nerve transection exhibit increased MN survival and re-innervation of muscle, showing that IGF-I mediates functional neurite regeneration in vivo (495). Functional sciatic nerve regeneration is also promoted by IGF-I treatment in mice after sciatic nerve crush (496). The spatiotemporal regulation patterns of the IGFs and IGF-IR suggest a functional endogenous role of the IGF system in neurite regeneration after injury.

    Because of these neurite-promoting effects, IGFs may have a role in the treatment of neuropathy, particularly in the treatment of diabetic neuropathy. Decreased IGF expression may contribute to the development of diabetic neuropathy (497, 498, 499); sc IGF-II restores pain and pressure thresholds toward normal in diabetic rodents (497), whereas IGF-I and IGF-II increase sciatic nerve regeneration after injury in rodents with diabetes (500). The beneficial effects of IGF are not limited to peripheral neuropathy, with reported therapeutic effects of IGF-I in a rat model of diabetic autonomic neuropathy (501). Studies have also shown that IGFs may have a therapeutic role in chemotherapy-induced neuropathies. IGF-I can prevent vincristine-, cisplatinum-, and taxol-mediated neuropathies (502, 503).

    E. Targeting the IGF/IGF-IR signaling in cancer therapy

    Interference with IGF ligand and IGF-IR autocrine or paracrine signaling provides a novel therapeutic target for CNS malignancies (504). In fact, targeting IGF-IR expression and signaling is a new therapeutic approach to cancer treatment in numerous tumor types (380, 505, 506). A truncated IGF-IR that acts as a dominant negative receptor or antisense RNA to IGF-IR prevents metastatic breast carcinoma (399, 507). A kinase-defective IGF-IR dominant negative receptor and -IR3, the IGF-IR blocking antibody, inhibit glioblastoma tumor growth in mice, as discussed above (403). A dominant negative IGF-IR introduced using retroviral infection decreases pancreatic tumor burden, particularly when used in combination with chemotherapeutic drugs (508). Antisense IGF-IR strategies also enhance the susceptibility of Ewing’s sarcoma to doxorubicin-induced apoptosis (509), implying a use for IGF-IR disruption in combinatorial drug therapy (388). Other cancers for which IGF-IR interference is effective as a treatment strategy include melanoma, lung carcinoma, ovarian carcinoma, and rhabdomyosarcoma (reviewed in Ref.510). One problem with these treatment strategies in the CNS is delivery, although direct injection into brain tumors or implantation of antisense IGF-IR-containing cells via craniotomy (400) has proven effective. However, gene therapy approaches will likely be more advantageous and less traumatic for IGF-I inhibition treatment paradigms in CNS tumors (511). Additional human clinical trials targeting both the IGF-IR and IGF-I are ongoing (381, 382).

    F. The potential therapeutic use of N-terminal peptides derived from IGF-I

    The hypothesis that IGF-I might be degraded by removal of an N-terminal peptide to leave des-N-(1-3) IGF-I, which would have lower affinity for the IGFBPs but maintain affinity for the receptor, was first suggested by Sara and colleagues (512). However, des-IGF-I has subsequently been shown to be markedly less neuroprotective than IGF-I (300). This suggested the possibility that the N-terminal tripeptide, GPE, might itself have biological function. Sara et al. (512) previously showed that GPE stimulates dopamine and acetylcholine release, and Bourguignon and Gerard (513) have presented data suggesting that GPE might act as a NMDA receptor antagonist, followed by a report that GPE can prevent NMDA-induced neuronal toxicity in vitro (512).

    The effect of GPE in models of brain disease has since been extensively investigated. GPE given centrally or as an iv infusion has marked neuroprotective effects in rodent models of hypoxic-ischemic injury (514). The window of opportunity is considerably longer than for IGF-I and extends for greater than 11 h after injury (514). GPE protects a wide variety of neuronal phenotypes and after hypoxic-ischemic injury prevents the loss of choline acetyltransferase-expressing neurons and up-regulates neuronal nitric oxide synthase (514). It has also been demonstrated to be protective in neurotoxic models of brain disease induced by 6-hydroxydopamine (515) and by quinolinic acid (516). The actions of GPE include neuronal protection and inhibition of both caspase-3-dependent and caspase-3-independent neuronal cell death and protection of astrocytes. The tripeptide is now in phase 1 clinical studies for use as an acute neuroprotectant.

    IX. Perspectives for Future Directions

    Since the discovery of Salmon and Daughaday (1) in 1957, a considerable body of investigation has been devoted to defining the role of the IGF system in many tissues, including the brain. There is no doubt that these pleiotropic factors, in concert with their receptor and binding proteins, are involved in controlling key processes in brain development and traumatic or degenerative disorders of the nervous system.

    Although the development of in vivo models such as the transgenic and knockout mice or disease/injury models has contributed to providing unique information about the growth promoting, neurogenic and neuroprotective actions of the IGF system, much research remains. An area of study that is currently receiving more interest from a large number of investigators is that related to stem cell research in the developing and adult brain. It is well known that progenitor cells in the dentate gyrus of hippocampus and the subventricular zone of lateral ventricles generate new neurons throughout the life of mammals, however very little is known about how stem and progenitor cells "decide" whether to proliferate or exit the cell cycle. Some of these events are directly or indirectly controlled by the IGFs, and therefore detailed studies directed to understand the mechanisms by which the IGFs control neuronal proliferation and differentiation and the complex intracellular signaling cross-talk activated by these ligands are required. Knowledge in this area could be of potential relevance to neurodegenerative disorders such as AD.

    Intriguingly, and in contrast to the mentioned role of IGF-I in normal physiology, recent data on the role of IGF and insulin signaling pathways on longevity in Caenorhabditis elegans, Drosophila, and rodents suggest that partial blockade of these signaling systems dramatically extends life span (517, 518, 519, 520, 521, 522, 523, 524). In C. elegans, restoration of IGF signaling in neuronal cells, but not other cell systems, reverses the extended longevity, suggesting that IGF signaling in the nervous system is critical in this process (517, 518, 525). Reduced nutrition, which lowers circulating and brain IGF content is similarly effective in enhancing longevity (518), whereas in C. elegans selective ablation of nutritionally responsive olfactory and gustatory neurons (IGF receptor-rich regions in mammals) similarly extends longevity (526, 527).

    The mechanisms by which IGF and nutritional responsive regions of the brain regulate longevity remain unclear, but paradoxically too much IGF in the brain may enhance neuronal survival but may reduce the life span of the organism.

    Although there is an increasing body of evidence for the potent antiapoptotic and neuroprotective effects of IGF-I, we still lack a clear understanding of the in vivo cellular and molecular mechanisms involved. This is a critical goal to be achieved for the development of IGF-I based preventive protective and regenerative therapies. However, it should be remembered that in vivo, IGF-I does not act in isolation and other growth factors might potentially modulate its biological action (Fig. 2). It is critical therefore to develop improved in vitro and in vivo models for the investigation of these potential interactions and the mechanisms involved.

    Acknowledgments

    We thank Dr. Leon Bach (University of Melbourne, Melbourne, Australia) and Dr. Chris Williams (University of Auckland, Auckland, New Zealand) for their helpful comments and suggestions. We also thank Ms. Suzana Metaxas and Miss Elena Andaloro from the Centre for Hormone Research for their assistance with the preparation of this manuscript.

    Footnotes

    This work was supported by Grants 209067 and 149219 from the National Health and Medical Research Council of Australia (to V.C.R. and G.A.W.), Juvenile Diabetes Research Foundation International Grant 1-2004-501, Diabetes Australia, and Murdoch Children’s Research Institute. We also acknowledge the support from the National Institutes of Health (Grant NS38849), the Juvenile Diabetes Research Foundation Center for the Study of Complications in Diabetes, and the Program for Understanding Neurological Diseases.

    First Published Online August 30, 2005

    Abbreviations: A, -Amyloid peptide; AD, Alzheimer’s disease; ALS, amytrophic lateral sclerosis; CNS, central nervous system; CSF, cerebrospinal fluid; EPO, erythropoietin; FGF, fibroblast growth factor; GPE, N-terminal tripeptide (glycine-proline-glutamate); HIF, hypoxia-inducible factor; IGF-IR, type I IGF receptor; IRS, insulin receptor substrate(s); LID, liver-specific IGF-I deficient; MN, motor neuron; M6P, mannose-6 phosphate; NMDA, N-methyl-D-aspartate; NSILA, nonsuppressible insulin-like activity; NT, neurotrophin; PI3K, phosphatidylinositol 3-kinase; PRL, prolactin; SOD1, Cu/Zn superoxide dismutase.

    References

    Salmon WD, Daughaday W 1957 A hormonally controlled serum factor which stimulates sulfate incorporation by cartilage in vitro. J Lab Clin Med 49:825–836

    Rinderknecht E, Humbel RE 1976 Polypeptides with nonsuppressible insulin-like and cell-growth promoting activities in human serum: isolation, chemical characterization, and some biological properties of forms I and II. Proc Natl Acad Sci USA 73:2365–2369

    Daughaday WH, Hall K, Raben MS, Salmon Jr WD, van den Brande JL, van Wyk JJ 1972 Somatomedin: proposed designation for sulphation factor. Nature 235:107

    Dulak NC, Temin HM 1973 A partially purified polypeptide fraction from rat liver cell conditioned medium with multiplication-stimulating activity for embryo fibroblasts. J Cell Physiol 81:153–160

    Burgi H, Muller WA, Humbel RE, Labhart A, Froesch ER 1966 Non-suppressible insulin-like activity of human serum. I. Physicochemical properties, extraction and partial purification. Biochim Biophys Acta 121:349–359

    Jakob A, Burgi H, Froesch ER, Labhart A 1967 Nonsuppressible insulin-like activity of human serum. Ger Med Mon 12:286–292

    Zingg AE, Froesch ER 1973 Effects of partially purified preparations with nonsuppressible insulin-like activity (NSILA-S) on sulfate incorporation into rat and chicken cartilage. Diabetologia 9:472–476

    Megyesi K, Kahn CR, Roth J, Froesch ER, Humbel RE, Zapf J, Neville Jr DM 1974 Insulin and non-suppressible insulin-like activity (NSILA-s): evidence for separate plasma membrane receptor sites. Biochem Biophys Res Commun 57:307–315

    Froesch ER, Jakob A, Zahnd GR, Simon E 1966 Immunoreactive insulin, suppressible, and nonsuppressible insulin-like activities in rat after administration of glucose and induction of hyperglycaemia by mannoheptulose. Diabetologia 2:265–268

    Froesch ER, Zapf J, Meuli C, Mader M, Waldvogel M, Kaufmann U, Morell B 1975 Biological properties of NSILA-S. Adv Metab Disord 8:211–235

    Rinderknecht E, Humbel RE 1976 Amino-terminal sequences of two polypeptides from human serum with nonsuppressible insulin-like and cell-growth-promoting activities: evidence for structural homology with insulin B chain. Proc Natl Acad Sci USA 73:4379–4381

    Rinderknecht E, Humbel RE 1978 The amino acid sequence of human insulin-like growth factor I and its structural homology with proinsulin. J Biol Chem 253:2769–2776

    Rinderknecht E, Humbel RE 1978 Primary structure of human insulin-like growth factor II. FEBS Lett 89:283–286

    Humbel RE 1990 Insulin-like growth factors I and II. Eur J Biochem 190:445–462

    Hintz RL, Rinderknecht E 1980 Somatomedin-C shares the carboxy-terminal antigenic determinants with insulin-like growth factor-I. J Clin Endocrinol Metab 51:672–673

    Jansen M, van Schaik FM, van Tol H, Van den Brande JL, Sussenbach JS 1985 Nucleotide sequences of cDNAs encoding precursors of human insulin-like growth factor II (IGF-II) and an IGF-II variant. FEBS Lett 179:243–246

    Jansen M, van Schaik FM, Ricker AT, Bullock B, Woods DE, Gabbay KH, Nussbaum AL, Sussenbach JS, Van den Brande JL1983 Sequence of cDNA encoding human insulin-like growth factor I precursor. Nature 306:609–611

    de Pagter-Holthuizen P, Jansen M, Bovenberg W, Van den Brande JL, Sussenbach JS 1989 The human somatomedin/insulin-like growth factor genes: organization and development-specific expression. J Intern Med 225:37–42

    Giacobini MM, Olson L, Hoffer BJ, Sara VR 1990 Truncated IGF-1 exerts trophic effects on fetal brain tissue grafts. Exp Neurol 108:33–37

    Sara VR, Carlsson-Skwirut C, Drakenberg K, Giacobini MB, Hakansson L, Mirmiran M, Nordberg A, Olson L, Reinecke M, Stahlbom PA, Sandberg-Nordquist AC 1993 The biological role of truncated insulin-like growth factor-1 and the tripeptide GPE in the central nervous system. Ann NY Acad Sci 692:183–191

    Russo VC, Werther GA 1994 Des(1-3) IGF-I potently enhances differentiated cell growth in olfactory bulb organ culture. Growth Factors 11:301–311

    Oh Y, Müller HL, Lee DY, Fielder PJ, Rosenfeld RG 1993 Characterization of the affinities of insulin-like growth factor (IGF)-binding proteins 1–4 for IGF-I, IGF-II, IGF-I/insulin hybrid, and IGF-I analogs. Endocrinology 132:1337–1344

    Rotwein P, Burgess SK, Milbrandt JD, Krause JE 1988 Differential expression of insulin-like growth factor genes in rat central nervous system. Proc Natl Acad Sci USA 85:265–269

    Bondy CA, Werner H, Roberts Jr CT, LeRoith D 1990 Cellular pattern of insulin-like growth factor-I (IGF-I) and type I IGF receptor gene expression in early organogenesis: comparison with IGF-II gene expression. Mol Endocrinol 4:1386–1398

    Bondy C, Werner H, Roberts Jr CT, LeRoith D 1992 Cellular pattern of type-I insulin-like growth factor receptor gene expression during maturation of the rat brain: comparison with insulin-like growth factors I and II. Neuroscience 46:909–923

    Werther GA, Abate M, Hogg A, Cheesman H, Oldfield B, Hards D, Hudson P, Power B, Freed K, Herington AC 1990 Localization of insulin-like growth factor-I mRNA in rat brain by in situ hybridization—relationship to IGF-I receptors. Mol Endocrinol 4:773–778

    Binoux M, Hossenlopp P, Lassarre C, Hardouin N 1981 Production of insulin-like growth factors and their carrier by rat pituitary gland and brain explants in culture. FEBS Lett 124:178–184

    Smith EP, Sadler TW, D’Ercole AJ 1987 Somatomedins/insulin-like growth factors, their receptors and binding proteins are present during mouse embryogenesis. Development 101:73–82

    Lamson G, Pham H, Oh Y, Ocrant I, Schwander J, Rosenfeld RG 1989 Expression of the BRL-3A insulin-like growth factor binding protein (rBP-30) in the rat central nervous system. Endocrinology 125:1100–1102

    Wood TL, Brown AL, Rechler MM, Pintar JE 1990 The expression pattern of an insulin-like growth factor (IGF)-binding protein gene is distinct from IGF-II in the midgestational rat embryo. Mol Endocrinol 4:1257–1263

    Lee WH, Javedan S, Bondy CA 1992 Coordinate expression of insulin-like growth factor system components by neurons and neuroglia during retinal and cerebellar development. J Neurosci 12:4737–4744

    Bondy C, Lee WH 1993 Correlation between insulin-like growth factor (IGF)-binding protein 5 and IGF-I gene expression during brain development. J Neurosci 13:5092–5104

    Lee WH, Michels KM, Bondy CA 1993 Localization of insulin-like growth factor binding protein-2 messenger RNA during postnatal brain development: correlation with insulin-like growth factors I and II. Neuroscience 53:251–265

    Pons S, Rejas MT, Torres-Aleman I 1991 Ontogeny of insulin-like growth factor I, its receptor, and its binding proteins in the rat hypothalamus. Brain Res Dev Brain Res 62:169–175

    Russo VC, Edmondson SR, Mercuri FA, Buchanan CR, Werther GA 1994 Identification, localization, and regulation of insulin-like growth factor binding proteins and their messenger ribonucleic acids in the newborn rat olfactory bulb. Endocrinology 135:1437–1446

    D’Ercole AJ, Ye P, Calikoglu AS, Gutierrez-Ospina G 1996 The role of the insulin-like growth factors in the central nervous system. Mol Neurobiol 13:227–255

    Leventhal PS, Russel JW, Feldman EL 1999 IGF and the nervous system. In: Rosenfeld RG, Roberts CTJ, eds. The IGF system. Totowa, NJ: Humana Press; 425–455

    Werther GA, Russo V, Baker N, Butler G 1998 The role of the insulin-like growth factor system in the developing brain. Horm Res 49(Suppl 1):37–40

    D’Ercole AJ, Stiles AD, Underwood LE 1984 Tissue concentrations of somatomedin C: further evidence for multiple sites of synthesis and paracrine or autocrine mechanisms of action. Proc Natl Acad Sci USA 81:935–939

    Lund PK, Moats-Staats BM, Hynes MA, Simmons JG, Jansen M, D’Ercole AJ, Van Wyk JJ 1986 Somatomedin-C/insulin-like growth factor-I and insulin-like growth factor-II mRNAs in rat fetal and adult tissues. J Biol Chem 261:14539–14544

    Yakar S, Liu JL, Stannard B, Butler A, Accili D, Sauer B, LeRoith D 1999 Normal growth and development in the absence of hepatic insulin-like growth factor I. Proc Natl Acad Sci USA 96:7324–7329

    Sjogren K, Liu JL, Blad K, Skrtic S, Vidal O, Wallenius V, LeRoith D, Tornell J, Isaksson OG, Jansson JO, Ohlsson C 1999 Liver-derived insulin-like growth factor I (IGF-I) is the principal source of IGF-I in blood but is not required for postnatal body growth in mice. Proc Natl Acad Sci USA 96:7088–7092

    Ueki I, Ooi GT, Tremblay ML, Hurst KR, Bach LA, Boisclair YR 2000 Inactivation of the acid labile subunit gene in mice results in mild retardation of postnatal growth despite profound disruptions in the circulating insulin-like growth factor system. Proc Natl Acad Sci USA 97:6868–6873

    Haluzik M, Yakar S, Gavrilova O, Setser J, Boisclair Y, LeRoith D 2003 Insulin resistance in the liver-specific IGF-1 gene-deleted mouse is abrogated by deletion of the acid-labile subunit of the IGF-binding protein-3 complex: relative roles of growth hormone and IGF-1 in insulin resistance. Diabetes 52:2483–2489

    Isaacs N, James R, Niall H, Bryant-Greenwood G, Dodson G, Evans A, North AC 1978 Relaxin and its structural relationship to insulin. Nature 271:278–281

    Kawakami A, Kataoka H, Oka T, Mizoguchi A, Kimura-Kawakami M, Adachi T, Iwami M, Nagasawa H, Suzuki A, Ishizaki H 1990 Molecular cloning of the Bombyx mori prothoracicotropic hormone. Science 247:1333–1335

    Kawakami A, Iwami M, Nagasawa H, Suzuki A, Ishizaki H 1989 Structure and organization of four clustered genes that encode bombyxin, an insulin-related brain secretory peptide of the silkmoth Bombyx mori. Proc Natl Acad Sci USA 86:6843–6847

    Smit AB, Vreugdenhil E, Ebberink RH, Geraerts WP, Klootwijk J, Joosse J 1988 Growth-controlling molluscan neurons produce the precursor of an insulin-related peptide. Nature 331:535–538

    Lagueux M, Lwoff L, Meister M, Goltzene F, Hoffmann JA 1990 cDNAs from neurosecretory cells of brains of Locusta migratoria (Insecta, Orthoptera) encoding a novel member of the superfamily of insulins. Eur J Biochem 187:249–254

    Zapf J, Froesch ER 1986 Insulin-like growth factors/somatomedins: structure, secretion, biological actions and physiological role. Horm Res 24:121–130

    Daughaday WH, Rotwein P 1989 Insulin-like growth factors I and II. Peptide messenger ribonucleic acid and gene structures, serum, and tissue concentrations. Endocr Rev 10:68–91

    Shimatsu A, Rotwein P 1987 Mosaic evolution of the insulin-like growth factors. Organization, sequence, and expression of the rat insulin-like growth factor I gene. J Biol Chem 262:7894–7900

    Kajimoto Y, Rotwein P 1991 Structure of the chicken insulin-like growth factor I gene reveals conserved promoter elements. J Biol Chem 266:9724–9731

    Perfetti R, Scott LA, Shuldiner AR 1994 The two nonallelic insulin-like growth factor-I genes in Xenopus laevis are differentially regulated during development. Endocrinology 135:2037–2044

    Koval A, Kulik V, Duguay S, Plisetskaya E, Adamo ML, Roberts Jr CT, Leroith D, Kavsan V 1994 Characterization of a salmon insulin-like growth factor I promoter. DNA Cell Biol 13:1057–1062

    Chan SJ, Cao QP, Steiner DF 1990 Evolution of the insulin superfamily: cloning of a hybrid insulin/insulin-like growth factor cDNA from amphioxus. Proc Natl Acad Sci USA 87:9319–9323

    Foyt HL, LeRoith D, Roberts Jr CT 1991 Differential association of insulin-like growth factor I mRNA variants with polysomes in vivo. J Biol Chem 266:7300–7305

    Rotwein P 1991 Structure, evolution, expression and regulation of insulin-like growth factors I and II. Growth Factors 5:3–18

    Jansen E, Steenbergh PH, van Schaik FM, Sussenbach JS 1992 The human IGF-I gene contains two cell type-specifically regulated promoters. Biochem Biophys Res Commun 187:1219–1226

    Rotwein P, Bichell DP, Kikuchi K 1993 Multifactorial regulation of IGF-I gene expression. Mol Reprod Dev 35:358–363

    LeRoith D, Roberts Jr CT 1993 Insulin-like growth factors. Ann NY Acad Sci 692:1–9

    Bichell DP, Kikuchi K, Rotwein P 1992 Growth hormone rapidly activates insulin-like growth factor I gene transcription in vivo. Mol Endocrinol 6:1899–1908

    Gronowski AM, Rotwein P 1995 Rapid changes in gene expression after in vivo growth hormone treatment. Endocrinology 136:4741–4748

    Butler AA, Ambler GR, Breier BH, LeRoith D, Roberts Jr CT, Gluckman PD 1994 Growth hormone (GH) and insulin-like growth factor-I (IGF-I) treatment of the GH-deficient dwarf rat: differential effects on IGF-I transcription start site expression in hepatic and extrahepatic tissues and lack of effect on type I IGF receptor mRNA expression. Mol Cell Endocrinol 101:321–330

    Benbassat C, Shoba LN, Newman M, Adamo ML, Frank SJ, Lowe Jr WL 1999 Growth hormone-mediated regulation of insulin-like growth factor I promoter activity in C6 glioma cells. Endocrinology 140:3073–3081

    Meton I, Boot EP, Sussenbach JS, Steenbergh PH 1999 Growth hormone induces insulin-like growth factor-I gene transcription by a synergistic action of STAT5 and HNF-1. FEBS Lett 444:155–159

    Thissen JP, Ketelslegers JM, Underwood LE 1994 Nutritional regulation of the insulin-like growth factors. Endocr Rev 15:80–101

    Rabkin R 1997 Nutrient regulation of insulin-like growth factor-I. Miner Electrolyte Metab 23:157–160

    Muaku SM, Beauloye V, Thissen JP, Underwood LE, Fossion C, Gerard G, Ketelslegers JM, Maiter D 1996 Long-term effects of gestational protein malnutrition on postnatal growth, insulin-like growth factor (IGF)-I, and IGF-binding proteins in rat progeny. Pediatr Res 39:649–655

    D’Ercole AJ 1987 Somatomedins/insulin-like growth factors and fetal growth. J Dev Physiol 9:481–495

    Sara VR, Carlsson-Skwirut C 1986 The biosynthesis of somatomedins and their role in the fetus. Acta Endocrinol Suppl (Copenh) 279:82–85

    Milner RD, Hill DJ 1989 Fetal growth signals. Arch Dis Child 64(1 Spec No):53–57

    Rappolee DA, Werb Z 1991 Endogenous IGF-II mediates growth in preimplantation mouse embryos. In: Spencer EM, ed. Modern concepts of insulin-like growth factors. New York: Elsevier; 3–8

    Sepp-Lorenzino L 1998 Structure and function of the insulin-like growth factor I receptor. Breast Cancer Res Treat 47:235–253

    Kim B, van Golen CM, Feldman EL 2004 Insulin-like growth factor-I signaling in human neuroblastoma cells. Oncogene 23:130–141

    Van Obberghen E, Ksauga M, Le Cam A, Hedo JA, Itin A, Harrison LC 1981 Biosynthetic labeling of insulin receptor: studies of subunits in cultured human IM-9 lymphocytes. Proc Natl Acad Sci USA 78:1052–1056

    Ullrich A, Gray A, Tam AW, Yang-Feng T, Tsubokawa M, Collins C, Henzel W, Le Bon T, Kathuria S, Chen E 1986 Insulin-like growth factor I receptor primary structure: comparison with insulin receptor suggests structural determinants that define functional specificity. EMBO J 5:2503–2512

    Gual P, Baron V, Alengrin F, Van Obberghen E 1995 A conformational change in the -subunit of the insulin-like growth factor I receptor identified by antipeptide antibodies. Endocrinology 136:5298–5304

    Sun XJ, Rothenberg P, Kahn CR, Backer JM, Araki E, Wilden PA, Cahill DA, Goldstein BJ, White MF 1991 Structure of the insulin receptor substrate IRS-1 defines a unique signal transduction protein. Nature 352:73–77

    Araki E, Lipes MA, Patti ME, Bruning JC, Haag 3rd B, Johnson RS, Kahn CR 1994 Alternative pathway of insulin signalling in mice with targeted disruption of the IRS-1 gene. Nature 372:186–190

    Sun XJ, Wang LM, Zhang Y, Yenush L, Myers Jr MG, Glasheen E, Lane WS, Pierce JH, White MF 1995 Role of IRS-2 in insulin and cytokine signalling. Nature 377:173–177

    Rubin R, Baserga R 1995 Insulin-like growth factor-I receptor. Its role in cell proliferation, apoptosis, and tumorigenicity. Lab Invest 73:311–331

    White MF, Kahn CR 1994 The insulin signaling system. J Biol Chem 269:1–4

    LeRoith D 2000 Insulin-like growth factor I receptor signaling—overlapping or redundant pathways Endocrinology 141:1287–1288

    van der Geer P, Hunter T, Lindberg RA 1994 Receptor protein-tyrosine kinases and their signal transduction pathways. Ann Rev Cell Biol 10:251–337

    Guan KL 1994 The mitogen activated protein kinase signal transduction pathway: from the cell surface to the nucleus. Cell Signal 6:581–589

    Chakravarthy MV, Abraha TW, Schwartz RJ, Fiorotto ML, Booth FW 2000 Insulin-like growth factor-I extends in vitro replicative life span of skeletal muscle satellite cells by enhancing G1/S cell cycle progression via the activation of phosphatidylinositol 3'-kinase/Akt signaling pathway. J Biol Chem 275:35942–35952

    Prisco M, Romano G, Peruzzi F, Valentinis B, Baserga R 1999 Insulin and IGF-I receptors signaling in protection from apoptosis. Horm Metab Res 31:80–89

    Baserga R 1999 The IGF-I receptor in cancer research. Exp Cell Res 253:1–6

    Melmed S, Yamashita S, Yamasaki H, Fagin J, Namba H, Yamamoto H, Weber M, Morita S, Webster J, Prager D 1996 IGF-I receptor signalling: lessons from the somatotroph. Recent Prog Horm Res 51:189–215

    Sundaresan S, Francke U 1989 Insulin-like growth factor I receptor gene is concordant with c-Fes protooncogene and mouse chromosome 7 in somatic cell hybrids. Somat Cell Mol Genet 15:373–376

    Werner H, Woloschak M, Adamo M, Shen-Orr Z, Roberts Jr CT, LeRoith D 1989 Developmental regulation of the rat insulin-like growth factor I receptor gene. Proc Natl Acad Sci USA 86:7451–7455

    Caro JF, Poulos J, Ittoop O, Pories WJ, Flickinger EG, Sinha MK 1988 Insulin-like growth factor I binding in hepatocytes from human liver, human hepatoma, and normal, regenerating, and fetal rat liver. J Clin Invest 81:976–981

    Santos A, Yusta B, Fernandez-Moreno MD, Blazquez E 1994 Expression of insulin-like growth factor-I (IGF-I) receptor gene in rat brain and liver during development and in regenerating adult rat liver. Mol Cell Endocrinol 101:85–93

    Werner H, Bach MA, Stannard B, Roberts Jr CT, LeRoith D 1992 Structural and functional analysis of the insulin-like growth factor I receptor gene promoter. Mol Endocrinol 6:1545–1558

    Beitner-Johnson D, Werner H, Roberts Jr CT, LeRoith D 1995 Regulation of insulin-like growth factor I receptor gene expression by Sp1: physical and functional interactions of Sp1 at GC boxes and at a CT element. Mol Endocrinol 9:1147–1156

    Werner H, Karnieli E, Rauscher FJ, LeRoith D 1996 Wild-type and mutant p53 differentially regulate transcription of the insulin-like growth factor I receptor gene. Proc Natl Acad Sci USA 93:8318–8323

    Ohlsson C, Kley N, Werner H, LeRoith D 1998 p53 regulates insulin-like growth factor-I (IGF-I) receptor expression and IGF-I-induced tyrosine phosphorylation in an osteosarcoma cell line. Endocrinology 139:1101–1107

    Burant CF, Treutelaar MK, Allen KD, Sens DA, Buse MG 1987 Comparison of insulin and insulin-like growth factor I receptors from rat skeletal muscle and L-6 myocytes. Biochem Biophys Res Commun 147:100–107

    Milazzo G, Yip CC, Maddux BA, Vigneri R, Goldfine ID 1992 High-affinity insulin binding to an atypical insulin-like growth factor-I receptor in human breast cancer cells. J Clin Inv 89:899–908

    Jonas HA, Cox AJ, Harrison LC 1989 Delineation of atypical insulin receptors from classical insulin and type I insulin-like growth factor receptors in human placenta. Biochem J 257:101–107

    Jonas HA, Cox AJ 1990 Insulin-like growth factor binding to the atypical insulin receptors of a human lymphoid-derived cell line (IM-9). Biochem J 66:737–742

    Siddle K, Soos MA, Field CE, Nave BT 1994 Hybrid and atypical insulin/insulin-like growth factor I receptors. Horm Res 41(Suppl 2):56–64

    Soos MA, Siddle K 1989 Immunological relationships between receptors for insulin and insulin-like growth factor I. Evidence for structural heterogeneity of insulin-like growth factor I receptors involving hybrids with insulin receptors. Biochem J 263:553–563

    Soos MA, Whittaker J, Lammers R, Ullrich A, Siddle K 1990 Receptors for insulin and insulin-like growth factor-I can form hybrid dimers. Characterisation of hybrid receptors in transfected cells. Biochem J 270:383–390

    Moxham CP, Duronio V, Jacobs S 1989 Insulin-like growth factor I receptor -subunit heterogeneity. Evidence for hybrid tetramers composed of insulin- like growth factor I and insulin receptor heterodimers. J Biol Chem 264:13238–13244

    LeRoith D, Werner H, Faria TN, Kato H, Adamo M, Roberts Jr CT 1993 Insulin-like growth factor receptors. Implications for nervous system function. Ann NY Acad Sci 692:22–32

    Pandini G, Frasca F, Mineo R, Sciacca L, Vigneri R, Belfiore A 2002 Insulin/insulin-like growth factor I hybrid receptors have different biological characteristics depending on the insulin receptor isoform involved. J Biol Chem 277:39684–39695

    Sakano K, Enjoh T, Numata F, Fujiwara H, Marumoto Y, Higashihashi N, Sato Y, Perdue JF, Fujita-Yamaguchi Y 1991 The design, expression, and characterization of human insulin-like growth factor II (IGF-II) mutants specific for either the IGF-II/cation-independent mannose 6-phosphate receptor or IGF-I receptor. J Biol Chem 266:20626–20635

    Oh Y, Beukers MW, Pham HM, Smanik PA, Smith MC, Rosenfeld RG 1991 Altered affinity of insulin-like growth factor II (IGF-II) for receptors and IGF-binding proteins, resulting from limited modifications of the IGF-II molecule. Biochem J 278:249–254

    Jones JI, Clemmons DR 1995 Insulin-like growth factors and their binding proteins: biological actions. Endocr Rev 16:3–34

    Dore S, Kar S, Quirion R 1997 Presence and differential internalization of two distinct insulin-like growth factor receptors in rat hippocampal neurons. Neuroscience 78:373–383

    Braulke T 1999 Type-2 IGF receptor: a multi-ligand binding protein. Horm Metab Res 31:242–246

    Kiess W, Yang Y, Kessler U, Hoeflich A 1994 Insulin-like growth factor II (IGF-II) and the IGF-II/mannose-6-phosphate receptor: the myth continues. Horm Res 41(Suppl 2):66–73

    Morgan DO, Edman JC, Standring DN, Fried VA, Smith MC, Roth RA, Rutter WJ 1987 Insulin-like growth factor II receptor as a multifunctional binding protein. Nature 329:301–307

    Blanchard F, Duplomb L, Raher S, Vusio P, Hoflack B, Jacques Y, Godard A 1999 Mannose 6-phosphate/insulin-like growth factor II receptor mediates internalization and degradation of leukemia inhibitory factor but not signal transduction. J Biol Chem 274:24685–24693

    von Figura K, Hasilik A 1986 Lysosomal enzymes and their receptors. Ann Rev Biochem 55:167–193

    Ghahary A, Tredget EE, Shen Q, Kilani RT, Scott PG, Houle Y 2000 Mannose-6-phosphate/IGF-II receptors mediate the effects of IGF- 1-induced latent transforming growth factor 1 on expression of type I collagen and collagenase in dermal fibroblasts. Growth Factors 17:167–176

    Hawkes C, Kar S 2003 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 458:113–127

    Ludwig T, Eggenschwiler J, Fisher P, D’Ercole AJ, Davenport ML, Efstratiadis A 1996 Mouse mutants lacking the type 2 IGF receptor (IGF2R) are rescued from perinatal lethality in Igf2 and Igf1r null backgrounds. Dev Biol 177:517–535

    Ghahary A, Tredget EE, Mi L, Yang L 1999 Cellular response to latent TGF-1 is facilitated by insulin-like growth factor-II/mannose-6-phosphate receptors on MS-9 cells. Exp Cell Res 251:111–120

    Hoeflich A, Yang Y, Rascher W, Blum WF, Huber S, Koepf G, Kolb HJ, Kiess W 1996 Coordinate expression of insulin-like growth factor II (IGF-II) and IGF-II/mannose-6-phosphate receptor mRNA and stable expression of IGF-I receptor mRNA during differentiation of human colon carcinoma cells (Caco-2). Eur J Endocrinol 135:49–59

    Mottola C, Czech MP 1984 The type II insulin-like growth factor receptor does not mediate increased DNA synthesis in H-35 hepatoma cells. J Biol Chem 259:12705–12713

    Hartmann H, Meyer-Alber A, Braulke T 1992 Metabolic actions of insulin-like growth factor II in cultured adult rat hepatocytes are not mediated through the insulin-like growth factor II receptor. Diabetologia 35:216–223

    Kiess W, Haskell JF, Lee L, Greenstein LA, Miller BE, Aarons AL, Rechler MM, Nissley SP 1987 An antibody that blocks insulin-like growth factor (IGF) binding to the type II IGF receptor is neither an agonist nor an inhibitor of IGF-stimulated biologic responses in L6 myoblasts. J Biol Chem 262:12745–12751

    Adashi EY, Resnick CE, Rosenfeld RG 1990 Insulin-like growth factor-I (IGF-I) and IGF-II hormonal action in cultured rat granulosa cells: mediation via type I but not type II IGF receptors. Endocrinology 126:216–222

    Stracke ML, Engel JD, Wilson LW, Rechler MM, Liotta LA, Schiffmann E 1989 The type I insulin-like growth factor receptor is a motility receptor in human melanoma cells. J Biol Chem 264:21544–21549

    Hirai K, Miyamasu M, Yamaguchi M, Nakajima K, Ohtoshi T, Koshino T, Takaishi T, Morita Y, Ito K 1993 Modulation of human basophil histamine release by insulin-like growth factors. J Immunol 150:1503–1508

    Denley A, Bonython ER, Booker GW, Cosgrove LJ, Forbes BE, Ward CW, Wallace JC 2004 Structural determinants for high-affinity binding of insulin-like growth factor II to insulin receptor (IR)-A, the exon 11 minus isoform of the IR. Mol Endocrinol 18:2502–2512

    Frasca F, Pandini G, Scalia P, Sciacca L, Mineo R, Costantino A, Goldfine ID, Belfiore A, Vigneri R 1999 Insulin receptor isoform A, a newly recognized, high-affinity insulin-like growth factor II receptor in fetal and cancer cells. Mol Cell Biol 19:3278–3288

    Seino S, Bell GI 1989 Alternative splicing of human insulin receptor messenger RNA. Biochem Biophys Res Commun 159:312–316

    Rajaram S, Baylink DJ, Mohan S 1997 Insulin-like growth factor-binding proteins in serum and other biological fluids: regulation and functions. Endocr Rev 18:801–831

    Bach LA, Rechler MM 1995 Insulin like growth factor binding proteins. Diabetes Rev 3:38–61

    Russo VC, Bach LA, Fosang AJ, Baker NL, Werther GA 1997 Insulin-like growth factor binding protein-2 binds to cell surface proteoglycans in the rat brain olfactory bulb. Endocrinology 138:4858–4867

    Firth SM, Baxter RC 2002 Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev 23:824–854

    Russo VC, Rekaris G, Baker NL, Bach LA, Werther GA 1999 Basic fibroblast growth factor induces proteolysis of secreted and cell membrane-associated insulin-like growth factor binding protein-2 in human neuroblastoma cells. Endocrinology 140:3082–3090

    Parker A, Clarke JB, Busby Jr WH, Clemmons DR 1996 Identification of the extracellular matrix binding sites for insulin-like growth factor-binding protein 5. J Biol Chem 271:13523–13529

    Parker A, Rees C, Clarke J, Busby Jr WH, Clemmons DR 1998 Binding of insulin-like growth factor (IGF)-binding protein-5 to smooth-muscle cell extracellular matrix is a major determinant of the cellular response to IGF-I. Mol Biol Cell 9:2383–2392

    Rees C, Clemmons DR 1998 Inhibition of IGFBP-5 binding to extracellular matrix and IGF-I-stimulated DNA synthesis by a peptide fragment of IGFBP-5. J Cell Biochem 71:375–381

    Binoux M, Lalou C, Mohseni-Zadeh S 1999 Biological action of proteolytic fragments of IGF binding proteins. In: Rosenfeld RG, Roberts CTJ, eds. Contemporary endocrinology: the IGF system. Totowa, NJ: Humana Press; 281–313

    Oh Y, Muller HL, Lamson G, Rosenfeld RG 1993 Insulin-like growth factor (IGF)-independent action of IGF- binding protein-3 in Hs578T human breast cancer cells. Cell surface binding and growth inhibition. J Biol Chem 268:14964–14971

    Oh Y, Muller HL, Pham H, Rosenfeld RG 1993 Demonstration of receptors for insulin-like growth factor binding protein-3 on Hs578T human breast cancer cells. J Biol Chem 268:26045–26048

    Yamanaka Y, Fowlkes JL, Wilson EM, Rosenfeld RG, Oh Y 1999 Characterization of insulin-like growth factor binding protein-3 (IGFBP-3) binding to human breast cancer cells: kinetics of IGFBP- 3 binding and identification of receptor binding domain on the IGFBP-3 molecule. Endocrinology 140:1319–1328

    Schutt BS, Langkamp M, Rauschnabel U, Ranke MB, Elmlinger MW 2004 Integrin-mediated action of insulin-like growth factor binding protein-2 in tumor cells. J Mol Endocrinol 32:859–868

    Radulescu RT 1994 Nuclear localization signal in insulin-like growth factor-binding protein type 3. Trends Biochem Sci 19:278

    Schedlich LJ, Young TF, Firth SM, Baxter RC 1998 Insulin-like growth factor-binding protein (IGFBP)-3 and IGFBP-5 share a common nuclear transport pathway in T47D human breast carcinoma cells. J Biol Chem 273:18347–18352

    Wraight CJ, Liepe IJ, White PJ, Hibbs AR, Werther GA 1998 Intranuclear localization of insulin-like growth factor binding protein-3 (IGFBP-3) during cell division in human keratinocytes. J Invest Dermatol 111:239–242

    Hoeflich A, Reisinger R, Schuett BS, Elmlinger MW, Russo VC, Vargas GA, Jehle PM, Lahm H, Renner-Muller I, Wolf E 2004 Peri/nuclear localization of intact insulin-like growth factor binding protein-2 and a distinct carboxyl-terminal IGFBP-2 fragment in vivo. Biochem Biophys Res Commun 324:705–710

    Richard-Parpaillon L, Heligon C, Chesnel F, Boujard D, Philpott A 2002 The IGF pathway regulates head formation by inhibiting Wnt signaling in Xenopus. Dev Biol 244:407–417

    Pera EM, Wessely O, Li SY, De Robertis EM 2001 Neural and head induction by insulin-like growth factor signals. Dev Cell 1:655–665

    Cheng CM, Cohen M, Tseng V, Bondy CA 2001 Endogenous IGF1 enhances cell survival in the postnatal dentate gyrus. J Neurosci Res 64:341–347

    Bondy CA, Cheng CM 2002 Insulin-like growth factor-1 promotes neuronal glucose utilization during brain development and repair processes. Int Rev Neurobiol 51:189–217

    Bondy CA, Cheng CM 2004 Signaling by insulin-like growth factor 1 in brain. Eur J Pharmacol 490:25–31

    Smith A, Wang J, Cheng CM, Zhou J, Weickert CS, Bondy CA 2004 High-level expression of Dok-1 in neurons of the primate prefrontal cortex and hippocampus. J Neurosci Res 75:218–224

    Cheng CM, Mervis RF, Niu SL, Salem Jr N, Witters LA, Tseng V, Reinhardt R, Bondy CA 2003 Insulin-like growth factor 1 is essential for normal dendritic growth. J Neurosci Res 73:1–9

    Varela-Nieto I, Morales-Garcia JA, Vigil P, Diaz-Casares A, Gorospe I, Sanchez-Galiano S, Canon S, Camarero G, Contreras J, Cediel R, Leon Y 2004 Trophic effects of insulin-like growth factor-I (IGF-I) in the inner ear. Hear Res 196:19–25

    Aguado-Llera D, Arilla-Ferreiro E, Campos-Barros A, Puebla-Jimenez L, Barrios V 2005 Protective effects of insulin-like growth factor-I on the somatostatinergic system in the temporal cortex of -amyloid-treated rats. J Neurochem 92:607–615

    Sun LY, Evans MS, Hsieh J, Panici J, Bartke A 2005 Increased neurogenesis in dentate gyrus of long-lived Ames dwarf mice. Endocrinology 146:1138–1144

    Guan J, Thomas GB, Lin H, Mathai S, Bachelor DC, George S, Gluckman PD 2004 Neuroprotective effects of the N-terminal tripeptide of insulin-like growth factor-1, glycine-proline-glutamate (GPE) following intravenous infusion in hypoxic-ischemic adult rats. Neuropharmacology 47:892–903

    Trejo JL, Carro E, Torres-Aleman I 2001 Circulating insulin-like growth factor I mediates exercise-induced increases in the number of new neurons in the adult hippocampus. J Neurosci 21:1628–1634

    Trejo JL, Carro E, Nunez A, Torres-Aleman I 2002 Sedentary life impairs self-reparative processes in the brain: the role of serum insulin-like growth factor-I. Rev Neurosci 13:365–374

    Carro E, Trejo JL, Gomez-Isla T, LeRoith D, Torres-Aleman I 2002 Serum insulin-like growth factor I regulates brain amyloid- levels. Nat Med 8:1390–1397

    Carro E, Trejo JL, Busiguina S, Torres-Aleman I 2001 Circulating insulin-like growth factor I mediates the protective effects of physical exercise against brain insults of different etiology and anatomy. J Neurosci 21:5678–5684

    Busiguina S, Fernandez AM, Barrios V, Clark R, Tolbert DL, Berciano J, Torres-Aleman I 2000 Neurodegeneration is associated to changes in serum insulin-like growth factors. Neurobiol Dis 7:657–665

    Popken GJ, Hodge RD, Ye P, Zhang J, Ng W, O’Kusky JR, D’Ercole AJ 2004 In vivo effects of insulin-like growth factor-I (IGF-I) on prenatal and early postnatal development of the central nervous system. Eur J Neurosci 19:2056–2068

    Pulford BE, Ishii DN 2001 Uptake of circulating insulin-like growth factors (IGFs) into cerebrospinal fluid appears to be independent of the IGF receptors as well as IGF-binding proteins. Endocrinology 142:213–220

    Torres-Aleman I, Pons S, Arevalo MA 1994 The insulin-like growth factor I system in the rat cerebellum: developmental regulation and role in neuronal survival and differentiation. J Neurosci Res 39:117–126

    Ye P, Li L, Richards RG, DiAugustine RP, D’Ercole AJ 2002 Myelination is altered in insulin-like growth factor-I null mutant mice. J Neurosci 22:6041–6051

    Camarero G, Avendano C, Fernandez-Moreno C, Villar A, Contreras J, de Pablo F, Pichel JG, Varela-Nieto I 2001 Delayed inner ear maturation and neuronal loss in postnatal Igf-1-deficient mice. J Neurosci 21:7630–7641

    Bunn RC, King WD, Winkler MK, Fowlkes JL 2005 Early developmental changes in IGF-I, IGF-II, IGF binding protein-1, and IGF binding protein-3 concentration in the cerebrospinal fluid of children. Pediatr Res 58:89–93

    Carro E, Trejo JL, Nunez A, Torres-Aleman I 2003 Brain repair and neuroprotection by serum insulin-like growth factor I. Mol Neurobiol 27:153–162

    Han VK, Lund PK, Lee DC, D’Ercole AJ 1988 Expression of somatomedin/insulin-like growth factor messenger ribonucleic acids in the human fetus: identification, characterization, and tissue distribution. J Clin Endocrinol Metab 66:422–429

    Han VK, Hill DJ, Strain AJ, Towle AC, Lauder JM, Underwood LE, D’Ercole AJ 1987 Identification of somatomedin/insulin-like growth factor immunoreactive cells in the human fetus. Pediatr Res 22:245–249

    Han VK, D’Ercole AJ, Lund PK 1987 Cellular localization of somatomedin (insulin-like growth factor) messenger RNA in the human fetus. Science 236:193–197

    Andersson IK, Edwall D, Norstedt G, Rozell B, Skottner A, Hansson HA 1988 Differing expression of insulin-like growth factor I in the developing and in the adult rat cerebellum. Acta Physiol Scand 132:167–173

    Ayer-le Lievre C, Stahlbom PA, Sara VR 1991 Expression of IGF-I and -II mRNA in the brain and craniofacial region of the rat fetus. Development 111:105–115

    Garcia-Segura LM, Perez J, Pons S, Rejas MT, Torres-Aleman I 1991 Localization of insulin-like growth factor I (IGF-I)-like immunoreactivity in the developing and adult rat brain. Brain Res 560:167–174

    Edmondson SR, Werther GA, Russell A, Le Roith D, Roberts Jr CT, Beck F 1995 Localization of growth hormone receptor/binding protein messenger ribonucleic acid (mRNA) during rat fetal development: relationship to insulin-like growth factor-I mRNA. Endocrinology 136:4602–4609

    Graziadei PPC, Monti-Graziadei GA 1978 Continuous nerve cell renewal in the olfactory system. In: Jacobson M, ed. Handbook of sensory physiology. Berlin: Springer; 55–83

    Hirvonen H, Sandberg M, Kalimo H, Hukkanen V, Vuorio E, Salmi TT, Alitalo K 1989 The N-myc proto-oncogene and IGF-II growth factor mRNAs are expressed by distinct cells in human fetal kidney and brain. J Cell Biol 108:1093–1104

    Caelers A, Schmid AC, Hrusovsky A, Reinecke M 2003 Insulin-like growth factor II mRNA is expressed in neurones of the brain of the bony fish Oreochromis mossambicus, the tilapia. Eur J Neurosci 18:355–363

    Logan A, Gonzalez AM, Hill DJ, Berry M, Gregson NA, Baird A 1994 Coordinated pattern of expression and localization of insulin-like growth factor-II (IGF-II) and IGF-binding protein-2 in the adult rat brain. Endocrinology 135:2255–2264

    Couce ME, Weatherington AJ, McGinty JF 1992 Expression of insulin-like growth factor-II (IGF-II) and IGF-II/mannose-6-phosphate receptor in the rat hippocampus: an in situ hybridization and immunocytochemical study. Endocrinology 131:1636–1642

    Stylianopoulou F, Herbert J, Soares MB, Efstratiadis A 1988 Expression of the insulin-like growth factor II gene in the choroid plexus and the leptomeninges of the adult rat central nervous system. Proc Natl Acad Sci USA 85:141–145

    Aberg MAI, Aberg ND, Palmer TD, Alborn AM, Carlsson-Skwirut C, Bang P, Rosengren LE, Olsson T, Gage FH, Eriksson PS 2003 IGF-I has a direct proliferative effect in adult hippocampal progenitor cells. Mol Cell Neurosci 24:23–40

    Werther GA, Russo V, Rekaris G, Baker N, Fosang AJ, Bach LA 1998 Insulin-like growth factors: implications for the CNS. J Endocrinol Invest 21:116–121

    Werther GA, Hogg A, McKinley M, Oldfield B, Figdor R, Mendelsohn FAO 1989 Localization and characterization of insulin-like growth factor-I receptors in rat brain and pituitary gland, using in vitro autoradiography and computerised densitometry, a distinct distribution from insulin receptors. J Neuroendocrinol 1:369–377

    Wilczak N, De Bleser P, Luiten P, Geerts A, Teelken A, De Keyser J 2000 Insulin-like growth factor II receptors in human brain and their absence in astrogliotic plaques in multiple sclerosis. Brain Res 863:282–288

    Gammeltoft S, Haselbacher GK, Humbel RE, Fehlmann M, Van Obberghen E 1985 Two types of receptors for insulin-like growth factors in mammalian brain. EMBO J 4:3407–3412

    Gammeltoft S, Ballotti R, Nielsen FC, Kowalski A, Van Obberghen E 1988 Receptors for insulin-like growth factors in the central nervous system: structure and function. Horm Metab Res 20:436–442

    Bondy CA, Lee WH 1993 Patterns of insulin-like growth factor and IGF receptor gene expression in the brain. Functional implications. Ann NY Acad Sci 692:33–43

    Baron-Van Evercooren A, Olichon-Berthe C, Kowalski A, Visciano G, Van Obberghen E 1991 Expression of IGF-I and insulin receptor genes in the rat central nervous system: a developmental, regional, and cellular analysis. J Neurosci Res 28:244–253

    Kar S, Chabot JG, Quirion R 1993 Quantitative autoradiographic localization of [125I]insulin-like growth factor I, [125I]insulin-like growth factor II, and [125I]insulin receptor binding sites in developing and adult rat brain. J Comp Neurol 333:375–397

    Liu JP, Baker J, Perkins AS, Robertson EJ, Efstratiadis A 1993 Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF receptor (Igf1r). Cell 75:59–72

    Baker J, Liu JP, Robertson EJ, Efstratiadis A 1993 Role of insulin-like growth factors in embryonic and postnatal growth. Cell 75:73–82

    Beck KD, Powell-Braxton L, Widmer HR, Valverde J, Hefti F 1995 Igf1 gene disruption results in reduced brain size, CNS hypomyelination, and loss of hippocampal granule and striatal parvalbumin-containing neurons. Neuron 14:717–730

    Valentino KL, Ocrant I, Rosenfeld RG 1990 Developmental expression of insulin-like growth factor-II receptor immunoreactivity in the rat central nervous system. Endocrinology 126:914–920

    Hawkes C, Kar S 2004 The insulin-like growth factor-II/mannose-6-phosphate receptor: structure, distribution and function in the central nervous system. Brain Res Brain Res Rev 44:117–140

    Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen AM, Mendelsohn FA 1987 Localization and characterization of insulin receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry. Endocrinology 121:1562–1570

    Bruning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban PC, Klein R, Krone W, Muller-Wieland D, Kahn CR 2000 Role of brain insulin receptor in control of body weight and reproduction. Science 289:2122–2125

    Ajo R, Cacicedo L, Navarro C, Sanchez-Franco F 2003 Growth hormone action on proliferation and differentiation of cerebral cortical cells from fetal rat. Endocrinology 144:1086–1097

    Han VK, Lauder JM, D’Ercole AJ 1987 Characterization of somatomedin/insulin-like growth factor receptors and correlation with biologic action in cultured neonatal rat astroglial cells. J Neurosci 7:501–511

    Tranque PA, Calle R, Naftolin F, Robbins R 1992 Involvement of protein kinase-C in the mitogenic effect of insulin-like growth factor-I on rat astrocytes. Endocrinology 131:1948–1954

    Hirano H, Lopes MB, Laws Jr ER, Asakura T, Goto M, Carpenter JE, Karns LR, VandenBerg SR 1999 Insulin-like growth factor-1 content and pattern of expression correlates with histopathologic grade in diffusely infiltrating astrocytomas. Neuro-oncol 1:109–119

    O’Donnell SL, Frederick TJ, Krady JK, Vannucci SJ, Wood TL 2002 IGF-I and microglia/macrophage proliferation in the ischemic mouse brain. Glia 39:85–97

    Aberg ND, Blomstrand F, Aberg MA, Bjorklund U, Carlsson B, Carlsson-Skwirut C, Bang P, Ronnback L, Eriksson PS 2003 Insulin-like growth factor-I increases astrocyte intercellular gap junctional communication and connexin43 expression in vitro. J Neurosci Res 74:12–22

    Cao Y, Gunn AJ, Bennet L, Wu D, George S, Gluckman PD, Shao XM, Guan J 2003 Insulin-like growth factor (IGF)-1 suppresses oligodendrocyte caspase-3 activation and increases glial proliferation after ischemia in near-term fetal sheep. J Cereb Blood Flow Metab 23:739–747

    Carson MJ, Behringer RR, Brinster RL, McMorris FA 1993 Insulin-like growth factor I increases brain growth and central nervous system myelination in transgenic mice. Neuron 10:729–740

    McMorris FA, Mozell RL, Carson MJ, Shinar Y, Meyer RD, Marchetti N 1993 Regulation of oligodendrocyte development and central nervous system myelination by insulin-like growth factors. Ann NY Acad Sci 692:321–334

    McMorris FA, McKinnon RD 1996 Regulation of oligodendrocyte development and CNS myelination by growth factors: prospects for therapy of demyelinating disease. Brain Pathol 6:313–329

    Recio-Pinto E, Rechler MM, Ishii DN 1986 Effects of insulin, insulin-like growth factor-II, and nerve growth factor on neurite formation and survival in cultured sympathetic and sensory neurons. J Neurosci 6:1211–1219

    Morrione A, Romano G, Navarro M, Reiss K, Valentinis B, Dews M, Eves E, Rosner MR, Baserga R 2000 Insulin-like growth factor I receptor signaling in differentiation of neuronal H19–7 cells. Cancer Res 60:2263–2272

    Brooker GJ, Kalloniatis M, Russo VC, Murphy M, Werther GA, Bartlett PF 2000 Endogenous IGF-1 regulates the neuronal differentiation of adult stem cells. J Neurosci Res 59:332–341

    Giacobini MM, Zetterstrom RH, Young D, Hoffer B, Sara V, Olson L 1994 IGF-1 influences olfactory bulb maturation. Evidence from anti-IGF-1 antibody treatment of developing grafts in oculo. Brain Res Dev Brain Res 84:67–76

    Giacobini MM, Zetterstrom R, Sara V, Olson L 1993 Differential effects of insulin-like growth factor-1 in the developing rat olfactory bulb and cerebral cortex. Evidence from intraocular grafting. Ann NY Acad Sci 692:294–296

    Werther GA, Cheesman H, Russo V 1993 Olfactory bulb organ culture is supported by combined insulin-like growth factor-I and basic fibroblast growth factor. Brain Res 617:339–342

    Russo VC, Cheesman H, Werther GA 1994 Organ culture: an in vitro system for the sustained growth of neonatal olfactory bulb. Neurosci Protocols 3:1–11

    Ye P, Carson J, D’Ercole AJ 1995 In vivo actions of insulin-like growth factor-I (IGF-I) on brain myelination: studies of IGF-I and IGF binding protein-1 (IGFBP-1) transgenic mice. J Neurosci 15:7344–7356

    Cheng HL, Russell JW, Feldman EL 1999 IGF-I promotes peripheral nervous system myelination. Ann NY Acad Sci 883:124–130

    Cheng CM, Joncas G, Reinhardt RR, Farrer R, Quarles R, Janssen J, McDonald MP, Crawley JN, Powell-Braxton L, Bondy CA 1998 Biochemical and morphometric analyses show that myelination in the insulin-like growth factor 1 null brain is proportionate to its neuronal composition. J Neurosci 18:5673–5681

    Feldman EL, Sullivan KA, Kim B, Russell JW 1997 Insulin-like growth factors regulate neuronal differentiation and survival. Neurobiol Dis 4:201–214

    Puglianiello A, Germani D, Rossi P, Cianfarani S 2000 IGF-I stimulates chemotaxis of human neuroblasts. Involvement of type 1 IGF receptor, IGF binding proteins, phosphatidylinositol-3 kinase pathway and plasmin system. J Endocrinol 165:123–131

    Ye P, Li L, Lund PK, D’Ercole AJ 2002 Deficient expression of insulin receptor substrate-1 (IRS-1) fails to block insulin-like growth factor-I (IGF-I) stimulation of brain growth and myelination. Brain Res Dev Brain Res 136:111–121

    Vicario-Abejon C, Yusta-Boyo MJ, Fernandez-Moreno C, de Pablo F 2003 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 23:895–906

    Cheng CM, Reinhardt RR, Lee WH, Joncas G, Patel SC, Bondy CA 2000 Insulin-like growth factor 1 regulates developing brain glucose metabolism. Proc Natl Acad Sci USA 97:10236–10241

    Bondy C, Chin E 1991 IGF-I mRNA localization in trigeminal and sympathetic nerve target zones during rat embryonic development. Adv Exp Med Biol 293:431–437

    Lopez-Lopez C, LeRoith D, Torres-Aleman I 2004 Insulin-like growth factor I is required for vessel remodeling in the adult brain. Proc Natl Acad Sci USA 101:9833–9838

    Trejo JL, Carro E, Lopez-Lopez C, Torres-Aleman I 2004 Role of serum insulin-like growth factor I in mammalian brain aging. Growth Horm IGF Res 14(Suppl A):S39–S43

    Daftary SS, Gore AC 2004 The hypothalamic insulin-like growth factor-1 receptor and its relationship to gonadotropin-releasing hormones neurones during postnatal development. J Neuroendocrinol 16:160–169

    Sonntag WE, Lynch CD, Bennett SA, Khan AS, Thornton PL, Cooney PT, Ingram RL, McShane T, Brunso-Bechtold JK 1999 Alterations in insulin-like growth factor-1 gene and protein expression and type 1 insulin-like growth factor receptors in the brains of ageing rats. Neuroscience 88:269–279

    Poe BH, Linville C, Riddle DR, Sonntag WE, Brunso-Bechtold JK 2001 Effects of age and insulin-like growth factor-1 on neuron and synapse numbers in area CA3 of hippocampus. Neuroscience 107:231–238

    Rollero A, Murialdo G, Fonzi S, Garrone S, Gianelli MV, Gazzerro E, Barreca A, Polleri A 1998 Relationship between cognitive function, growth hormone and insulin-like growth factor I plasma levels in aged subjects. Neuropsychobiology 38:73–79

    Kalmijn S, Janssen JAMJL, Pols HAP, Lamberts SWJ, Breteler MMB 2000 A prospective study on circulating insulin-like growth factor I (IGF-I), IGF-binding proteins, and cognitive function in the elderly. J Clin Endocrinol Metab 85:4551–4555

    Sytze van Dam P, Aleman A 2004 Insulin-like growth factor-I, cognition and brain aging. Eur J Pharmacol 490:87–95

    Guan J, Bennet L, Gluckman PD, Gunn AJ 2003 Insulin-like growth factor-1 and post-ischemic brain injury. Prog Neurobiol 70:443–462

    Oorschot DE, McLennan IS 1998 The trophic requirements of mature motoneurons. Brain Res 789:315–321

    Sendtner M 1995 Molecular biology of neurotrophic factors. Baillieres Clin Neurol 4:575–591

    Rabinovsky ED, Gelir E, Gelir S, Lui H, Kattash M, DeMayo FJ, Shenaq SM, Schwartz RJ 2003 Targeted expression of IGF-1 transgene to skeletal muscle accelerates muscle and motor neuron regeneration. FASEB J 17:53–55

    Thanos PK, Okajima S, Tiangco DA, Terzis JK 1999 Insulin-like growth factor-I promotes nerve regeneration through a nerve graft in an experimental model of facial paralysis. Restor Neurol Neurosci 15:57–71

    Kerns JM, Shott S, Brubaker L, Sakamoto K, Benson JT, Fleischer AE, Coleman ME 2003 Effects of IGF-I gene therapy on the injured rat pudendal nerve. Int Urogynecol J Pelvic Floor Dysfunct 14:2–7; discussion 8

    Tiangco DA, Papakonstantinou KC, Mullinax KA, Terzis JK 2001 IGF-I and end-to-side nerve repair: a dose-response study. J Reconstr Microsurg 17:247–256

    Di Giulio AM, Germani E, Lesma E, Muller E, Gorio A 2000 Glycosaminoglycans co-administration enhance insulin-like growth factor-I neuroprotective and neuroregenerative activity in traumatic and genetic models of motor neuron disease: a review. Int J Dev Neurosci 18:339–346

    Torres-Aleman I 2000 Serum growth factors and neuroprotective surveillance: focus on IGF-1. Mol Neurobiol 21:153–160

    Arsenijevic Y, Weiss S, Schneider B, Aebischer P 2001 Insulin-like growth factor-I is necessary for neural stem cell proliferation and demonstrates distinct actions of epidermal growth factor and fibroblast growth factor-2. J Neurosci 21:7194–7202

    Torres-Aleman I 1999 Insulin-like growth factors as mediators of functional plasticity in the adult brain. Horm Metab Res 31:114–119

    Anderson MF, Aberg MA, Nilsson M, Eriksson PS 2002 Insulin-like growth factor-I and neurogenesis in the adult mammalian brain. Brain Res Dev Brain Res 134:115–122

    O’Kusky JR, Ye P, D’Ercole AJ 2000 Insulin-like growth factor-I promotes neurogenesis and synaptogenesis in the hippocampal dentate gyrus during postnatal development. J Neurosci 20:8435–8442

    Saura J, Curatolo L, Williams CE, Gatti S, Benatti L, Peeters C, Guan J, Dragunow M, Post C, Faull RL, Gluckman PD, Skinner SJ 1999 Neuroprotective effects of Gly-Pro-Glu, the N-terminal tripeptide of IGF-1, in the hippocampus in vitro. Neuroreport 10:161–164

    Dore S, Kar S, Quirion R 1997 Rediscovering an old friend, IGF-I: potential use in the treatment of neurodegenerative diseases. Trends Neurosci 20:326–331

    Peretz S, Jensen R, Baserga R, Glazer PM 2001 ATM-dependent expression of the insulin-like growth factor-I receptor in a pathway regulating radiation response. Proc Natl Acad Sci USA 98:1676–1681

    Humbert S, Bryson EA, Cordelieres FP, Connors NC, Datta SR, Finkbeiner S, Greenberg ME, Saudou F 2002 The IGF-1/Akt pathway is neuroprotective in Huntington’s disease and involves Huntingtin phosphorylation by Akt. Dev Cell 2:831–837

    Offen D, Shtaif B, Hadad D, Weizman A, Melamed E, Gil-Ad I 2001 Protective effect of insulin-like-growth-factor-1 against dopamine-induced neurotoxicity in human and rodent neuronal cultures: possible implications for Parkinson’s disease. Neurosci Lett 316:129–132

    Lichtenwalner RJ, Forbes ME, Bennett SA, Lynch CD, Sonntag WE, Riddle DR 2001 Intracerebroventricular infusion of insulin-like growth factor-I ameliorates the age-related decline in hippocampal neurogenesis. Neuroscience 107:603–613

    Tagami M, Yamagata K, Nara Y, Fujino H, Kubota A, Numano F, Yamori Y 1997 Insulin-like growth factors prevent apoptosis in cortical neurons isolated from stroke-prone spontaneously hypertensive rats. Lab Invest 76:603–612

    Heck S, Lezoualc’h F, Engert S, Behl C 1999 Insulin-like growth factor-1-mediated neuroprotection against oxidative stress is associated with activation of nuclear factor B. J Biol Chem 274:9828–9835

    Olson BR, Scott DC, Wetsel WC, Elliot SJ, Tomic M, Stojilkovic S, Nieman LK, Wray S 1995 Effects of insulin-like growth factors I and II and insulin on the immortalized hypothalamic GTI-7 cell line. Neuroendocrinology 62:155–165

    Sortino MA, Canonico PL 1996 Neuroprotective effect of insulin-like growth factor I in immortalized hypothalamic cells. Endocrinology 137:1418–1422

    Zheng WH, Kar S, Quirion R 2002 Insulin-like growth factor-1-induced phosphorylation of transcription factor FKHRL1 is mediated by phosphatidylinositol 3-kinase/Akt kinase and role of this pathway in insulin-like growth factor-1-induced survival of cultured hippocampal neurons. Mol Pharmacol 62:225–233

    Linseman DA, Phelps RA, Bouchard RJ, Le SS, Laessig TA, McClure ML, Heidenreich KA 2002 Insulin-like growth factor-I blocks Bcl-2 interacting mediator of cell death (Bim) induction and intrinsic death signaling in cerebellar granule neurons. J Neurosci 22:9287–9297

    Gleichmann M, Weller M, Schulz JB 2000 Insulin-like growth factor-1-mediated protection from neuronal apoptosis is linked to phosphorylation of the pro-apoptotic protein BAD but not to inhibition of cytochrome c translocation in rat cerebellar neurons. Neurosci Lett 282:69–72

    Zhang J, Popken GJ, Ye P, D’Ercole AJ 2003 Down-regulation of 14–3-3 eta gene expression by IGF-I in mouse cerebellum during postnatal development. Brain Res Dev Brain Res 143:199–206

    Neff NT, Prevette D, Houenou LJ, Lewis ME, Glicksman MA, Yin QW, Oppenheim RW 1993 Insulin-like growth factors: putative muscle-derived trophic agents that promote motoneuron survival. J Neurobiol 24:1578–1588

    Yin QW, Johnson J, Prevette D, Oppenheim RW 1994 Cell death of spinal motoneurons in the chick embryo following deafferentation: rescue effects of tissue extracts, soluble proteins, and neurotrophic agents. J Neurosci 14:7629–7640

    Syroid DE, Zorick TS, Arbet-Engels C, Kilpatrick TJ, Eckhart W, Lemke G 1999 A role for insulin-like growth factor-I in the regulation of Schwann cell survival. J Neurosci 19:2059–2068

    Russell JW, Feldman EL 1999 Insulin-like growth factor-I prevents apoptosis in sympathetic neurons exposed to high glucose. Horm Metab Res 31:90–96

    Dore S, Kar S, Quirion R 1997 Insulin-like growth factor I protects and rescues hippocampal neurons against -amyloid- and human amylin-induced toxicity. Proc Natl Acad Sci USA 94:4772–4777

    Niikura T, Hashimoto Y, Okamoto T, Abe Y, Yasukawa T, Kawasumi M, Hiraki T, Kita Y, Terashita K, Kouyama K, Nishimoto I 2001 Insulin-like growth factor I (IGF-I) protects cells from apoptosis by Alzheimer’s V642I mutant amyloid precursor protein through IGF-I receptor in an IGF-binding protein-sensitive manner. J Neurosci 21:1902–1910

    Dore S, Bastianetto S, Kar S, Quirion R 1999 Protective and rescuing abilities of IGF-I and some putative free radical scavengers against -amyloid-inducing toxicity in neurons. Ann NY Acad Sci 890:356–364

    D’Mello SR, Borodezt K, Soltoff SP 1997 Insulin-like growth factor and potassium depolarization maintain neuronal survival by distinct pathways: possible involvement of PI 3-kinase in IGF-1 signaling. J Neurosci 17:1548–1560

    Galli C, Meucci O, Scorziello A, Werge TM, Calissano P, Schettini G 1995 Apoptosis in cerebellar granule cells is blocked by high KCl, forskolin, and IGF-1 through distinct mechanisms of action: the involvement of intracellular calcium and RNA synthesis. J Neurosci 15:1172–1179

    Fernandez-Sanchez MT, Garcia-Rodriguez A, Diaz-Trelles R, Novelli A 1996 Inhibition of protein phosphatases induces IGF-1-blocked neurotrophin-insensitive neuronal apoptosis. FEBS Lett 398:106–112

    D’Costa AP, Prevette DM, Houenou LJ, Wang S, Zackenfels K, Rohrer H, Zapf J, Caroni P, Oppenheim RW 1998 Mechanisms of insulin-like growth factor regulation of programmed cell death of developing avian motoneurons. J Neurobiol 36:379–394

    Kermer P, Klocker N, Labes M, Bahr M 2000 Insulin-like growth factor-I protects axotomized rat retinal ganglion cells from secondary death via PI3-K-dependent Akt phosphorylation and inhibition of caspase-3 in vivo. J Neurosci 20:2–8

    Pu SF, Zhuang HX, Marsh DJ, Ishii DN 1999 Insulin-like growth factor-II increases and IGF is required for postnatal rat spinal motoneuron survival following sciatic nerve axotomy. J Neurosci Res 55:9–16

    Van Golen CM, Feldman EL 2000 Insulin-like growth factor I is the key growth factor in serum that protects neuroblastoma cells from hyperosmotic-induced apoptosis. J Cell Physiol 182:24–32

    Delaney CL, Russell JW, Cheng HL, Feldman EL 2001 Insulin-like growth factor-I and over-expression of Bcl-xL prevent glucose-mediated apoptosis in Schwann cells. J Neuropathol Exp Neurol 60:147–160

    Russo VC, Kobayashi K, Najdovska S, Baker NL, Werther GA 2004 Neuronal protection from glucose deprivation via modulation of glucose transport and inhibition of apoptosis: a role for the insulin-like growth factor system. Brain Res 1009:40–53

    Mason JL, Ye P, Suzuki K, D’Ercole AJ, Matsushima GK 2000 Insulin-like growth factor-1 inhibits mature oligodendrocyte apoptosis during primary demyelination. J Neurosci 20:5703–5708

    Russell JW, Windebank AJ, Schenone A, Feldman EL 1998 Insulin-like growth factor-I prevents apoptosis in neurons after nerve growth factor withdrawal. J Neurobiol 36:455–467

    Ring BD, Scully S, Davis CR, Baker MB, Cullen MJ, Pelleymounter MA, Danilenko DM 2000 Systemically and topically administered leptin both accelerate wound healing in diabetic ob/ob mice. Endocrinology 141:446–449

    Russo VC, Andaloro E, Fornaro SA, Najdovska S, Newgreen DF, Bach LA, Werther GA 2004 Fibroblast growth factor-2 over-rides insulin-like growth factor-I induced proliferation and cell survival in human neuroblastoma cells. J Cell Physiol 199:371–380

    Ocrant I 1991 Insulin-like growth factor binding proteins in the nervous system. Adv Exp Med Biol 293:471–482

    Ocrant I 1993 Insulin-like growth factor binding proteins in nervous tissue-derived cells. Ann NY Acad Sci 692:44–50

    Han VK, Matsell DG, Delhanty PJ, Hill DJ, Shimasaki S, Nygard K 1996 IGF-binding protein mRNAs in the human fetus: tissue and cellular distribution of developmental expression. Horm Res 45:160–166

    Ocrant I, Fay CT, Parmelee JT 1990 Characterization of insulin-like growth factor binding proteins produced in the rat central nervous system. Endocrinology 127:1260–1267

    Naeve GS, Vana AM, Eggold JR, Verge G, Ling N, Foster AC 2000 Expression of rat insulin-like growth factor binding protein-6 in the brain, spinal cord, and sensory ganglia. Brain Res Mol Brain Res 75:185–197

    Zhou H, Hughes DE, Major ML, Yoo K, Pesold C, Costa RH 2001 Atypical mouse cerebellar development is caused by ectopic expression of the forkhead box transcription factor HNF-3. Gene Expr 9:217–236

    D’Ercole AJ, Dai Z, Xing Y, Boney C, Wilkie MB, Lauder JM, Han VK, Clemmons DR 1994 Brain growth retardation due to the expression of human insulin like growth factor binding protein-1 in transgenic mice: an in vivo model for the analysis of igf function in the brain. Brain Res Dev Brain Res 82:213–222

    Murphy LJ, Rajkumar K, Molnar P 1995 Phenotypic manifestations of insulin-like growth factor binding protein-1 (IGFBP-1) and IGFBP-3 overexpression in transgenic mice. Prog Growth Factor Res 6:425–432

    Ni W, Rajkumar K, Nagy JI, Murphy LJ 1997 Impaired brain development and reduced astrocyte response to injury in transgenic mice expressing IGF binding protein-1. Brain Res 769:97–107

    Lee KH, Calikoglu AS, Ye P, D’Ercole AJ 1999 Insulin-like growth factor-I (IGF-I) ameliorates and IGF binding protein-1 (IGFBP-1) exacerbates the effects of undernutrition on brain growth during early postnatal life: studies in IGF-I and IGFBP-1 transgenic mice. Pediatr Res 45:331–336

    Murphy LJ 2000 Overexpression of insulin-like growth factor binding protein-1 in transgenic mice. Pediatr Nephrol 14:567–571

    Lee PD, Giudice LC, Conover CA, Powell DR 1997 Insulin-like growth factor binding protein-1: recent findings and new directions. Proc Soc Exp Biol Med 216:319–357

    Seurin D, Froment P, Bluet-Pajot MT, Epelbaum J, Monget P, Binoux M 2002 Functional alteration of the somatotrophic axis in transgenic mice with liver-specific expression of human insulin-like growth factor binding protein-1. Pediatr Res 52:168–174

    Doublier S, Duyckaerts C, Seurin D, Binoux M 2000 Impaired brain development and hydrocephalus in a line of transgenic mice with liver-specific expression of human insulin-like growth factor binding protein-1. Growth Horm IGF Res 10:267–274

    Wood TL, Streck RD, Pintar JE 1992 Expression of the IGFBP-2 gene in post-implantation rat embryos. Development 114:59–66

    Liu X, Yao DL, Bondy CA, Brenner M, Hudson LD, Zhou J, Webster HD 1994 Astrocytes express insulin-like growth factor-I (IGF-I) and its binding protein, IGFBP-2, during demyelination induced by experimental autoimmune encephalomyelitis. Mol Cell Neurosci 5:418–430

    Russo VC, Schutt BS, Andaloro E, Ymer S, Hoeflich A, Fosang AJ, Ranke M, Bach LA, Werther GA, IGFBP-2/IGF-I/proteoglycan complexes in the rat brain olfactory bulb. Proc 12th International Congress of Endocrinology, Lisbon, Portugal, 2004. Medimont International Proceedings ISBN 88-7587-071-3:817–820

    Guan J, Beilharz EJ, Skinner SJ, Williams CE, Gluckman PD 2000 Intracerebral transportation and cellular localisation of insulin-like growth factor-1 following central administration to rats with hypoxic-ischemic brain injury. Brain Res 853:163–173

    Guan J, Williams CE, Skinner SJ, Mallard EC, Gluckman PD 1996 The effects of insulin-like growth factor (IGF)-1, IGF-2, and des-IGF-1 on neuronal loss after hypoxic-ischemic brain injury in adult rats: evidence for a role for IGF binding proteins. Endocrinology 137:893–898

    Ishikawa K, Ohe Y, Tatemoto K 1995 Synthesis and secretion of insulin-like growth factor (IGF)-II and IGF binding protein-2 by cultivated brain meningeal cells. Brain Res 697:122–129

    Menouny M, Binoux M, Babajko S 1997 Role of insulin-like growth factor binding protein-2 and its limited proteolysis in neuroblastoma cell proliferation: modulation by transforming growth factor- and retinoic acid. Endocrinology 138:683–690

    Wood TL, Rogler L, Streck RD, Cerro J, Green B, Grewal A, Pintar JE 1993 Targeted disruption of IGFBP-2 gene. Growth Regul 3:5–8

    Wood TL, Rogler LE, Czick ME, Schuller AG, Pintar JE 2000 Selective alterations in organ sizes in mice with a targeted disruption of the insulin-like growth factor binding protein-2 gene. Mol Endocrinol 14:1472–1482

    Pintar JE, Schuller A, Cerro JA, Czick M, Grewal A, Green B 1995 Genetic ablation of IGFBP-2 suggests functional redundancy in the IGFBP family. Prog Growth Factor Res 6:437–445

    Hoeflich A, Wu M, Mohan S, Foll J, Wanke R, Froehlich T, Arnold GJ, Lahm H, Kolb HJ, Wolf E 1999 Overexpression of insulin-like growth factor-binding protein-2 in transgenic mice reduces postnatal body weight gain. Endocrinology 140:5488–5496

    Hoeflich A, Nedbal S, Blum WF, Erhard M, Lahm H, Brem G, Kolb HJ, Wanke R, Wolf E 2001 Growth inhibition in giant growth hormone transgenic mice by overexpression of insulin-like growth factor-binding protein-2. Endocrinology 142:1889–1898

    Cardona-Gomez GP, Chowen JA, Garcia-Segura LM 2000 Estradiol and progesterone regulate the expression of insulin-like growth factor-I receptor and insulin-like growth factor binding protein-2 in the hypothalamus of adult female rats. J Neurobiol 43:269–281

    Fernandez-Galaz MC, Torres-Aleman I, Garcia-Segura LM 1996 Endocrine-dependent accumulation of IGF-I by hypothalamic glia. Neuroreport 8:373–377

    Garcia-Segura LM, Chowen JA, Parducz A, Naftolin F 1994 Gonadal hormones as promoters of structural synaptic plasticity: cellular mechanisms. Prog Neurobiol 44:279–307

    Duenas M, Luquin S, Chowen JA, Torres-Aleman I, Naftolin F, Garcia-Segura LM 1994 Gonadal hormone regulation of insulin-like growth factor-I-like immunoreactivity in hypothalamic astroglia of developing and adult rats. Neuroendocrinology 59:528–538

    Lee WH, Bondy C 1993 Insulin-like growth factors and cerebral ischemia. Ann NY Acad Sci 679:418–422

    Beilharz EJ, Russo VC, Butler G, Baker NL, Connor B, Sirimanne ES, Dragunow M, Werther GA, Gluckman PD, Williams CE, Scheepens A 1998 Co-ordinated and cellular specific induction of the components of the IGF/IGFBP axis in the rat brain following hypoxic-ischemic injury. Brain Res Mol Brain Res 59:119–134

    Rensink AA, Gellekink H, Otte-Holler I, ten Donkelaar HJ, de Waal RM, Verbeek MM, Kremer B 2002 Expression of the cytokine leukemia inhibitory factor and pro-apoptotic insulin-like growth factor binding protein-3 in Alzheimer’s disease. Acta Neuropathol (Berl) 104:525–533

    Modric T, Silha JV, Shi Z, Gui Y, Suwanichkul A, Durham SK, Powell DR, Murphy LJ 2001 Phenotypic manifestations of insulin-like growth factor-binding protein-3 overexpression in transgenic mice. Endocrinology 142:1958–1967

    Murphy LJ, Molnar P, Lu X, Huang H 1995 Expression of human insulin-like growth factor-binding protein-3 in transgenic mice. J Mol Endocrinol 15:293–303

    Chernausek SD, Murray MA, Cheung PT 1993 Expression of insulin-like growth factor binding protein-4 (IGFBP-4) by rat neural cells–comparison to other IGFBPs. Regul Pept 48:123–132

    Brar AK, Chernausek SD 1993 Localization of insulin-like growth factor binding protein-4 expression in the developing and adult rat brain: analysis by in situ hybridization. J Neurosci Res 35:103–114

    LaTour D, Mohan S, Linkhart TA, Baylink DJ, Strong DD 1990 Inhibitory insulin-like growth factor-binding protein: cloning, complete sequence, and physiological regulation. Mol Endocrinol 4:1806–1814

    Kassem M, Okazaki R, De Leon D, Harris SA, Robinson JA, Spelsberg TC, Conover CA, Riggs BL 1996 Potential mechanism of estrogen-mediated decrease in bone formation: estrogen increases production of inhibitory insulin-like growth factor-binding protein-4. Proc Assoc Am Physicians 108:155–164

    Conover CA 1996 Regulation and physiological role of insulin-like growth factor binding proteins. Endocr J 43(Suppl):S43—S48

    Zhou R, Flaswinkel H, Schneider MR, Lahm H, Hoeflich A, Wanke R, Wolf E 2004 Insulin-like growth factor-binding protein-4 inhibits growth of the thymus in transgenic mice. J Mol Endocrinol 32:349–364

    Green BN, Jones SB, Streck RD, Wood TL, Rotwein P, Pintar JE 1994 Distinct expression patterns of insulin-like growth factor binding proteins 2 and 5 during fetal and postnatal development. Endocrinology 134:954–962

    Cheng HL, Sullivan KA, Feldman EL 1996 Immunohistochemical localization of insulin-like growth factor binding protein-5 in the developing rat nervous system. Brain Res Dev Brain Res 92:211–218

    Roschier M, Kuusisto E, Suuronen T, Korhonen P, Kyrylenko S, Salminen A 2001 Insulin-like growth factor binding protein 5 and type-1 insulin-like growth factor receptor are differentially regulated during apoptosis in cerebellar granule cells. J Neurochem 76:11–20

    Ye P, D’Ercole J 1998 Insulin-like growth factor I (IGF-I) regulates IGF binding protein-5 gene expression in the brain. Endocrinology 139:65–71

    Bach LA 1999 Insulin-like growth factor binding protein-6: the "forgotten" binding protein Horm Metab Res 31:226–234

    Babajko S, Leneuve P, Loret C, Binoux M 1997 IGF-binding protein-6 is involved in growth inhibition in SH- SY5Y human neuroblastoma cells: its production is both IGF- and cell density-dependent. J Endocrinol 152:221–227

    Bienvenu G, Seurin D, Grellier P, Froment P, Baudrimont M, Monget P, Le Bouc Y, Babajko S 2004 Insulin-like growth factor binding protein-6 transgenic mice: postnatal growth, brain development, and reproduction abnormalities. Endocrinology 145:2412–2420

    Drago J, Murphy M, Carroll SM, Harvey RP, Bartlett PF 1991 Fibroblast growth factor-mediated proliferation of central nervous system precursors depends on endogenous production of insulin-like growth factor I. Proc Natl Acad Sci USA 88:2199–2203

    Lavenius E, Parrow V, Nanberg E, Pahlman S 1994 Basic FGF and IGF-I promote differentiation of human SH-SY5Y neuroblastoma cells in culture. Growth Factors 10:29–39

    Torres-Aleman I, Naftolin F, Robbins RJ 1990 Trophic effects of basic fibroblast growth factor on fetal rat hypothalamic cells: interactions with insulin-like growth factor I. Brain Res Dev Brain Res 52:253–257

    Frodin M, Gammeltoft S 1994 Insulin-like growth factors act synergistically with basic fibroblast growth factor and nerve growth factor to promote chromaffin cell proliferation. Proc Natl Acad Sci USA 91:1771–1775

    Corse AM, Bilak MM, Bilak SR, Lehar M, Rothstein JD, Kuncl RW 1999 Preclinical testing of neuroprotective neurotrophic factors in a model of chronic motor neuron degeneration. Neurobiol Dis 6:335–346

    Yuen EC 2001 The role of neurotrophic factors in disorders of peripheral nerves and motor neurons. Phys Med Rehabil Clin N Am 12:293–306, viii

    Tuszynski MH, Blesch A 2004 Nerve growth factor: from animal models of cholinergic neuronal degeneration to gene therapy in Alzheimer’s disease. Prog Brain Res 146:441–449

    Ajo R, Cacicedo L, Navarro C, Sanchez-Franco F 2003 Growth hormone action on proliferation and differentiation of cerebral cortical cells from fetal rat. Endocrinology 144:1086–1097

    Sugisaki T, Noguchi T, Tsukada Y 1985 Cerebral myelinogenesis in the Snell dwarf mouse: stimulatory effects of GH and T4 restricted to the first 20 days of postnatal life. Neurochem Res 10:767–778

    Noguchi T, Sugisaki T, Tsukada Y 1984 Stimulation of Snell dwarf mouse neuronal growth by GH and T4. Neurochem Pathol 2:123–138

    Scheepens A, Sirimanne E, Beilharz E, Breier BH, Waters MJ, Gluckman PD, Williams CE 1999 Alterations in the neural growth hormone axis following hypoxic-ischemic brain injury. Brain Res Mol Brain Res 68:88–100

    Scheepens A, Sirimanne ES, Breier BH, Clark RG, Gluckman PD, Williams CE 2001 Growth hormone as a neuronal rescue factor during recovery from CNS injury. Neuroscience 104:677–687

    Scheepens A, Williams CE, Breier BH, Guan J, Gluckman PD 2000 A role for the somatotropic axis in neural development, injury and disease. J Pediatr Endocrinol Metab 13(Suppl 6):1483–1491

    Edmondson SR, Russo VC, McFarlane AC, Wraight CJ, Werther GA 1999 Interactions between growth hormone, insulin-like growth factor I, and basic fibroblast growth factor in melanocyte growth. J Clin Endocrinol Metab 84:1638–1644

    Lennington JB, Yang Z, Conover JC 2003 Neural stem cells and the regulation of adult neurogenesis. Reprod Biol Endocrinol 1:99

    Bridges RS, Grattan DR 2003 Prolactin-induced neurogenesis in the maternal brain. Trends Endocrinol Metab 14:199–201

    Ghezzi P, Brines M 2004 Erythropoietin as an antiapoptotic, tissue-protective cytokine. Cell Death Differ 11(Suppl 1):S37–S44

    Strunk T, Hartel C, Schultz C 2004 Does erythropoietin protect the preterm brain Arch Dis Child Fetal Neonatal Ed 89:F364—F366

    Fischer S, Wiesnet M, Marti HH, Renz D, Schaper W 2004 Simultaneous activation of several second messengers in hypoxia-induced hyperpermeability of brain derived endothelial cells. J Cell Physiol 198:359–369

    Grasso G, Sfacteria A, Cerami A, Brines M 2004 Erythropoietin as a tissue-protective cytokine in brain injury: what do we know and where do we go Neuroscientist 10:93–98

    Buemi M, Caccamo C, Nostro L, Cavallaro E, Floccari F, Grasso G 2004 Brain and cancer: the protective role of erythropoietin. Med Res Rev 25:245–259

    Sakanaka M, Wen TC, Matsuda S, Masuda S, Morishita E, Nagao M, Sasaki R 1998 In vivo evidence that erythropoietin protects neurons from ischemic damage. Proc Natl Acad Sci USA 95:4635–4640

    Wen TC, Sadamoto Y, Tanaka J, Zhu PX, Nakata K, Ma YJ, Hata R, Sakanaka M 2002 Erythropoietin protects neurons against chemical hypoxia and cerebral ischemic injury by up-regulating Bcl-xL expression. J Neurosci Res 67:795–803

    Wang X, Deng J, Boyle DW, Zhong J, Lee WH 2004 Potential role of IGF-I in hypoxia tolerance using a rat hypoxic-ischemic model: activation of hypoxia-inducible factor 1. Pediatr Res 55:385–394

    Chavez JC, LaManna JC 2002 Activation of hypoxia-inducible factor-1 in the rat cerebral cortex after transient global ischemia: potential role of insulin-like growth factor-1. J Neurosci 22:8922–8931

    Feldser D, Agani F, Iyer NV, Pak B, Ferreira G, Semenza GL 1999 Reciprocal positive regulation of hypoxia-inducible factor 1 and insulin-like growth factor 2. Cancer Res 59:3915–3918

    Digicaylioglu M, Garden G, Timberlake S, Fletcher L, Lipton SA 2004 Acute neuroprotective synergy of erythropoietin and insulin-like growth factor I. Proc Natl Acad Sci USA 101:9855–9860

    Garcia-Segura LM, Duenas M, Fernandez-Galaz MC, Chowen JA, Argente J, Naftolin F, Torres-Aleman I 1996 Interaction of the signalling pathways of insulin-like growth factor-I and sex steroids in the neuroendocrine hypothalamus. Horm Res 46:160–164

    Azcoitia I, Sierra A, Garcia-Segura LM 1999 Neuroprotective effects of estradiol in the adult rat hippocampus: interaction with insulin-like growth factor-I signalling. J Neurosci Res 58:815–822

    Cardona-Gomez GP, Mendez P, DonCarlos LL, Azcoitia I, Garcia-Segura LM 2001 Interactions of estrogens and insulin-like growth factor-I in the brain: implications for neuroprotection. Brain Res Brain Res Rev 37:320–334

    Shingo AS, Kito S 2003 Estrogen induces insulin-like growth factor-1 mRNA expression in the immortalized hippocampal cell: determination by quantitative real-time polymerase chain reaction. Neurochem Res 28:1379–1383

    Quesada A, Micevych PE 2004 Estrogen interacts with the IGF-1 system to protect nigrostriatal dopamine and maintain motoric behavior after 6-hydroxdopamine lesions. J Neurosci Res 75:107–116

    Azcoitia I, Doncarlos LL, Garcia-Segura LM 2002 Estrogen and brain vulnerability. Neurotox Res 4:235–245

    Mendez P, Azcoitia I, Garcia-Segura LM 2003 Estrogen receptor forms estrogen-dependent multimolecular complexes with insulin-like growth factor receptor and phosphatidylinositol 3-kinase in the adult rat brain. Brain Res Mol Brain Res 112:170–176

    Breese CR, D’Costa A, Rollins YD, Adams C, Booze RM, Sonntag WE, Leonard S 1996 Expression of insulin-like growth factor-1 (IGF-1) and IGF-binding protein 2 (IGF-BP2) in the hippocampus following cytotoxic lesion of the dentate gyrus. J Comp Neurol 369:388–404

    Sandberg Nordqvist AC, von Holst H, Holmin S, Sara VR, Bellander BM, Schalling M 1996 Increase of insulin-like growth factor (IGF)-1, IGF binding protein-2 and -4 mRNAs following cerebral contusion. Brain Res Mol Brain Res 38:285–293

    Walter HJ, Berry M, Hill DJ, Logan A 1997 Spatial and temporal changes in the insulin-like growth factor (IGF) axis indicate autocrine/paracrine actions of IGF-I within wounds of the rat brain. Endocrinology 138:3024–3034

    Beilharz EJ, Bassett NS, Sirimanne ES, Williams CE, Gluckman PD 1995 Insulin-like growth factor II is induced during wound repair following hypoxic-ischemic injury in the developing rat brain. Brain Res 29:81–91

    Beilharz EJ, Klempt ND, Klempt M, Sirimanne E, Dragunow M, Gluckman PD 1993 Differential expression of insulin-like growth factor binding proteins (IGFBP) 4 and 5 mRNA in the rat brain after transient hypoxic-ischemic injury. Brain Res 18:209–215

    Gluckman PD, Klempt ND, Guan J, Mallard C, Sirimanne E, Dragunow M, Klempt M, Singh K, Williams C, Nikolics K 1992 A role for IGF-I in the rescue of CNS neurons following hypoxic-ischemic injury. Biochem Biophys Res Commun 182:593–599

    Klempt M, Klempt ND, Gluckman PD 1994 Hypoxia and hypoxia/ischemia affect the expression of insulin-like growth factor binding protein 2 in the developing rat brain. Brain Res Mol Brain Res 17:347–350

    Yao DL, Liu X, Hudson LD, Webster HD 1995 Insulin-like growth factor I treatment reduces demyelination and up-regulates gene expression of myelin-related proteins in experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA 92:6190–6194

    Lee WH, Wang GM, Yang XL, Seaman LB, Vannucci SI 1999 Perinatal hypoxia-ischemia decreased neuronal but increased cerebral vascular endothelial IGFBP3 expression. Endocrine 11:181–188

    Hammarberg H, Risling M, Hokfelt T, Cullheim S, Piehl F 1998 Expression of insulin-like growth factors and corresponding binding proteins (IGFBP 1–6) in rat spinal cord and peripheral nerve after axonal injuries. J Comp Neurol 400:57–72

    Adams TE, Epa VC, Garrett TP, Ward CW 2000 Structure and function of the type 1 insulin-like growth factor receptor. Cell Mol Life Sci 57:1050–1093

    Nechushtan A, Smith CL, Hsu YT, Youle RJ 1999 Conformation of the Bax C-terminus regulates subcellular location and cell death. EMBO J 18:2330–2341

    Leinninger GM, Meyer GE, Feldman EL 2002 IGFs and the nervous system. In: LeRoith D, Zumkeller W, Baxter R, eds. Insulin-like growth factors. Eurekah.com/Landes Bioscience and Kluwer Academic/Plenum Publishers; 158–187

    Baserga R 2000 The contradictions of the insulin-like growth factor 1 receptor. Oncogene 19:5574–5581

    Singleton JR, Randolph AE, Feldman EL 1996 Insulin-like growth factor I receptor prevents apoptosis and enhances neuroblastoma tumorigenesis. Cancer Res 56:4522–4529

    Surmacz E, Guvakova MA, Nolan MK, Nicosia RF, Sciacca L 1998 Type I insulin-like growth factor receptor function in breast cancer. Breast Cancer Res Treat 47:255–267

    Butler AA, Blakesley VA, Poulaki V, Tsokos M, Wood TL, LeRoith D 1998 Stimulation of tumor growth by recombinant human insulin-like growth factor-I (IGF-I) is dependent on the dose and the level of IGF-I receptor expression. Cancer Res [Erratum (1998) 58:5630] 58:3021–3027

    Zumkeller W, Westphal M 2001 The IGF/IGFBP system in CNS malignancy. Mol Pathol 54:227–229

    Zumkeller W 2002 IGFs and IGF-binding proteins as diagnostic markers and biological modulators in brain tumors. Expert Rev Mol Diagn 2:473–477

    Glick RP, Lichtor T, Unterman TG 1997 Insulin-like growth factors in central nervous system tumors. J Neurooncol 35:315–325

    Merrill MJ, Edwards NA 1990 Insulin-like growth factor-I receptors in human glial tumors. J Clin Endocrinol Metab 71:199–209

    Sandberg AC, Engberg C, Lake M, von Holst H, Sara VR 1988 The expression of insulin-like growth factor I and insulin-like growth factor II genes in the human fetal and adult brain and in glioma. Neurosci Lett 93:114–119

    Glick RP, Unterman TG 1995 Radioimmunoassay of insulin-like growth factors I and II in the cerebrospinal fluid of patients with pituitary and other central nervous system tumors. Neurosurgery 36:556–563; discussion, 563–564

    Zumkeller W, Muller D, Muller S, Gunther C, Westphal M 1998 Expression and synthesis of insulin-like growth factor-binding proteins in human glioma cell lines. Int J Oncol 12:129–135

    Li S, Ferber A, Miura M, Baserga R 1994 Mitogenicity and transforming activity of the insulin-like growth factor-I receptor with mutations in the tyrosine kinase domain. J Biol Chem 269:32558–32564

    Hongo A, D’Ambrosio C, Miura M, Morrione A, Baserga R 1996 Mutational analysis of the mitogenic and transforming activities of the insulin-like growth factor I receptor. Oncogene 12:1231–1238

    Surmacz E, Sell C, Swantek J, Kato H, Roberts CT Jr, LeRoith D, Baserga R 1995 Dissociation of mitogenesis and transforming activity by C-terminal truncation of the insulin-like growth factor-I receptor. Exp Cell Res 218:370–380

    Li S, Resnicoff M, Baserga R 1996 Effect of mutations at serines 1280–1283 on the mitogenic and transforming activities of the insulin-like growth factor I receptor. J Biol Chem 271:12254–12260

    Miura M, Surmacz E, Burgaud JL, Baserga R 1995 Different effects on mitogenesis and transformation of a mutation at tyrosine 1251 of the insulin-like growth factor I receptor. J Biol Chem 270:22639–22644

    Brockmann MA, Ulbricht U, Gruner K, Fillbrandt R, Westphal M, Lamszus K 2003 Glioblastoma and cerebral microvascular endothelial cell migration in response to tumor-associated growth factors. Neurosurgery 52:1391–1399

    Chakravarti A, Loeffler JS, Dyson NJ 2002 Insulin-like growth factor receptor I mediates resistance to anti-epidermal growth factor receptor therapy in primary human glioblastoma cells through continued activation of phosphoinositide 3-kinase signaling. Cancer Res 62:200–207

    Naidu KA, Tang JL, Prockop LD, Nicosia SV, Coppola D 2001 Antiproliferative and apoptotic effect of ascorbyl stearate in human glioblastoma multiforme cells: modulation of insulin-like growth factor-I receptor (IGF-IR) expression. J Neurooncol 54:15–22

    Ly A, Duc HT, Kalamarides M, Trojan LA, Pan Y, Shevelev A, Francois JC, Noel T, Kane A, Henin D, Anthony DD, Trojan J 2001 Human glioma cells transformed by IGF-I triple helix technology show immune and apoptotic characteristics determining cell selection for gene therapy of glioblastoma. Mol Pathol 54:230–239

    Trojan LA, Kopinski P, Wei MX, Ly A, Glogowska A, Czarny J, Shevelev A, Przewlocki R, Henin D, Trojan J 2002 IGF-I: from diagnostic to triple-helix gene therapy of solid tumors. Acta Biochim Pol 49:979–990

    Trojan LA, Kopinski P, Mazurek A, Chyczewski L, Ly A, Jarocki P, Niklinski J, Shevelev A, Trzos R, Pan Y, Gitis DJ, Bierwagen M, Czapiewska JL, Wei MX, Michalkiewicz J, Henin D, Popiela T, Evrard F, Kasprzak H, Anthony D, Trojan J 2003 IGF-I triple helix gene therapy of rat and human gliomas. Rocz Akad Med Bialymst 48:18–27

    Chernicky CL, Yi L, Tan H, Gan SU, Ilan J 2000 Treatment of human breast cancer cells with antisense RNA to the type I insulin-like growth factor receptor inhibits cell growth, suppresses tumorigenesis, alters the metastatic potential, and prolongs survival in vivo. Cancer Gene Ther 7:384–395

    Andrews DW, Resnicoff M, Flanders AE, Kenyon L, Curtis M, Merli G, Baserga R, Iliakis G, Aiken RD 2001 Results of a pilot study involving the use of an antisense oligodeoxynucleotide directed against the insulin-like growth factor type I receptor in malignant astrocytomas. J Clin Oncol 19:2189–2200

    Resnicoff M 1998 Antitumor effects elicited by antisense-mediated downregulation of the insulin-like growth factor I receptor (review). Int J Mol Med 1:883–888

    Resnicoff M, Li W, Basak S, Herlyn D, Baserga R, Rubin R 1996 Inhibition of rat C6 glioblastoma tumor growth by expression of insulin-like growth factor I receptor antisense mRNA. Cancer Immunol Immunother 42:64–68

    Seely BL, Samimi G, Webster NJ 2002 Retroviral expression of a kinase-defective IGF-I receptor suppresses growth and causes apoptosis of CHO and U87 cells in vivo. BMC Cancer 2:15

    Burgaud JL, Resnicoff M, Baserga R 1995 Mutant IGF-I receptors as dominant negatives for growth and transformation. Biochem Biophys Res Commun 214:475–481

    Del Valle L, Wang JY, Lassak A, Peruzzi F, Croul S, Khalili K, Reiss K 2002 Insulin-like growth factor I receptor signaling system in JC virus T antigen-induced primitive neuroectodermal tumors–medulloblastomas. J Neurovirol 8(Suppl 2):138–147

    Wang JY, Del Valle L, Gordon J, Rubini M, Romano G, Croul S, Peruzzi F, Khalili K, Reiss K 2001 Activation of the IGF-IR system contributes to malignant growth of human and mouse medulloblastomas. Oncogene 20:3857–3868

    Rao G, Pedone CA, Valle LD, Reiss K, Holland EC, Fults DW 2004 Sonic hedgehog and insulin-like growth factor signaling synergize to induce medulloblastoma formation from nestin-expressing neural progenitors in mice. Oncogene 23:6156–6162

    Korshunov A, Neben K, Wrobel G, Tews B, Benner A, Hahn M, Golanov A, Lichter P 2003 Gene expression patterns in ependymomas correlate with tumor location, grade, and patient age. Am J Pathol 163:1721–1727

    Hultberg BM, Haselbacher G, Nielsen FC, Wulff BS, Gammeltoft S 1993 Gene expression of insulin-like growth factor II in human intracranial meningioma. Cancer 72:3282–3286

    Patti R, Reddy CD, Geoerger B, Grotzer MA, Raghunath M, Sutton LN, Phillips PC 2000 Autocrine secreted insulin-like growth factor-I stimulates MAP kinase-dependent mitogenic effects in human primitive neuroectodermal tumor/medulloblastoma. Int J Oncol 16:577–584

    Hwang CC, Fang K, Li L, Shih SH 1995 Insulin-like growth factor-I is an autocrine regulator for the brain metastatic variant of a human non-small cell lung cell line. Cancer Lett 94:157–163

    Sullivan KA, Castle VP, Hanash SM, Feldman EL 1995 Insulin-like growth factor II in the pathogenesis of human neuroblastoma. Am J Pathol 147:1790–1798

    El-Badry OM, Helman LJ, Chatten J, Steinberg SM, Evans AE, Israel MA 1991 Insulin-like growth factor II-mediated proliferation of human neuroblastoma. J Clin Invest 87:648–657

    Martin DM, Feldman EL 1993 Regulation of insulin-like growth factor-II expression and its role in autocrine growth of human neuroblastoma cells. J Cell Physiol 155:290–300

    Meghani MA, Martin DM, Singleton JR, Feldman EL 1993 Effects of serum and insulin-like growth factors on human neuroblastoma cell growth. Regul Pept 48:217–224

    Martin DM, Singleton JR, Meghani MA, Feldman EL 1993 IGF receptor function and regulation in autocrine human neuroblastoma cell growth. Regul Pept 48:225–232

    Leventhal PS, Randolph AE, Vesbit TE, Schenone A, Windebank A, Feldman EL 1995 Insulin-like growth factor-II as a paracrine growth factor in human neuroblastoma cells. Exp Cell Res 221:179–186

    Kiess W, Koepf G, Christiansen H, Blum WF 1997 Human neuroblastoma cells use either insulin-like growth factor-I or insulin-like growth factor-II in an autocrine pathway via the IGF-I receptor: variability of IGF, IGF binding protein (IGFBP) and IGF receptor gene expression and IGF and IGFBP secretion in human neuroblastoma cells in relation to cellular proliferation. Regul Pept 72:19–29

    Singleton JR, Dixit VM, Feldman EL 1996 Type I insulin-like growth factor receptor activation regulates apoptotic proteins. J Biol Chem 271:31791–31794

    Matthews CC, Odeh HM, Feldman EL 1997 Insulin-like growth factor-I is an osmoprotectant in human neuroblastoma cells. Neuroscience 79:525–534

    van Golen CM, Castle VP, Feldman EL 2000 IGF-I receptor activation and BCL-2 overexpression prevent early apoptotic events in human neuroblastoma. Cell Death Differ 7:654–665

    van Golen CM, Schwab TS, Ignatoski KM, Ethier SP, Feldman EL 2001 PTEN/MMAC1 overexpression decreases insulin-like growth factor-I-mediated protection from apoptosis in neuroblastoma cells. Cell Growth Differ 12:371–378

    Leinninger GM, Russell JW, van Golen CM, Berent A, Feldman EL 2004 Insulin-like growth factor-I regulates glucose-induced mitochondrial depolarization and apoptosis in human neuroblastoma. Cell Death Differ 11:885–896

    Liu X, Turbyville T, Fritz A, Whitesell L 1998 Inhibition of insulin-like growth factor I receptor expression in neuroblastoma cells induces the regression of established tumors in mice. Cancer Res 58:5432–5438

    Nieto-Sampedro M, Lewis ER, Cotman CW, Manthorpe M, Skaper SD, Barbin G, Longo FM, Varon S 1982 Brain injury causes a time-dependent increase in neuronotrophic activity at the lesion site. Science 217:860–861

    Guan J, Williams C, Gunning M, Mallard C, Gluckman P 1993 The effects of IGF-1 treatment after hypoxic-ischemic brain injury in adult rats. J Cereb Blood Flow Metab 13:609–616

    Guan J, Miller OT, Waugh KM, McCarthy DC, Gluckman PD 2001 Insulin-like growth factor-1 improves somatosensory function and reduces the extent of cortical infarction and ongoing neuronal loss after hypoxia-ischemia in rats. Neuroscience 105:299–306

    Liu XF, Fawcett JR, Thorne RG, Frey 2nd WH 2001 Non-invasive intranasal insulin-like growth factor-I reduces infarct volume and improves neurologic function in rats following middle cerebral artery occlusion. Neurosci Lett 308:91–94

    Liu XF, Fawcett JR, Thorne RG, DeFor TA, Frey 2nd WH 2001 Intranasal administration of insulin-like growth factor-I bypasses the blood-brain barrier and protects against focal cerebral ischemic damage. J Neurol Sci 187:91–97

    Sharma HS, Nyberg F, Westman J, Alm P, Gordh T, Lindholm D 1998 Brain derived neurotrophic factor and insulin like growth factor-1 attenuate upregulation of nitric oxide synthase and cell injury following trauma to the spinal cord. An immunohistochemical study in the rat. Amino Acids 14:121–129

    Johnston BM, Mallard EC, Williams CE, Gluckman PD 1996 Insulin-like growth factor-1 is a potent neuronal rescue agent after hypoxic-ischemic injury in fetal lambs. J Clin Invest 97:300–308

    Saatman KE, Contreras PC, Smith DH, Raghupathi R, McDermott KL, Fernandez SC, Sanderson KL, Voddi M, McIntosh TK 1997 Insulin-like growth factor-1 (IGF-1) improves both neurological motor and cognitive outcome following experimental brain injury. Exp Neurol 147:418–427

    Guan J, Bennet L, George S, Wu D, Waldvogel HJ, Gluckman PD, Faull RL, Crosier PS, Gunn AJ 2001 Insulin-like growth factor-1 reduces postischemic white matter injury in fetal sheep. J Cereb Blood Flow Metab 21:493–502

    Mitchell JD, Wokke JH, Borasio GD 2002 Recombinant human insulin-like growth factor I (rhIGF-I) for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev:CD002064

    Lai EC, Felice KJ, Festoff BW, Gawel MJ, Gelinas DF, Kratz R, Murphy MF, Natter HM, Norris FH, Rudnicki SA 1997 Effect of recombinant human insulin-like growth factor-I on progression of ALS. A placebo-controlled study. The North America ALS/IGF-I Study Group. Neurology 49:1621–1630

    Lange DJ, Felice KJ, Festoff BW, Gawel MJ, Gelinas DF, Kratz R, Lai EC, Murphy MF, Natter HM, Norris FH, Rudnicki S 1996 Recombinant human insulin-like growth factor-I in ALS: description of a double-blind, placebo-controlled study. North American ALS/IGF-I Study Group. Neurology 47(4 Suppl 2):S93—S94; discussion, S94—S95

    Borasio GD, Robberecht W, Leigh PN, Emile J, Guiloff RJ, Jerusalem F, Silani V, Vos PE, Wokke JH, Dobbins T 1998 A placebo-controlled trial of insulin-like growth factor-I in amyotrophic lateral sclerosis. European ALS/IGF-I Study Group. Neurology 51:583–586

    Lewis ME, Neff NT, Contreras PC, Stong DB, Oppenheim RW, Grebow PE, Vaught JL 1993 Insulin-like growth factor-I: potential for treatment of motor neuronal disorders. Exp Neurol 124:73–88

    Kaspar BK, Llado J, Sherkat N, Rothstein JD, Gage FH 2003 Retrograde viral delivery of IGF-1 prolongs survival in a mouse ALS model. Science 301:839–842

    Vergani L, Losa M, Lesma E, Di Giulio AM, Torsello A, Muller EE, Gorio A 1999 Glycosaminoglycans boost insulin-like growth factor-I-promoted neuroprotection: blockade of motor neuron death in the wobbler mouse. Neuroscience 93:565–572

    Liu X, Linnington C, Webster HD, Lassmann S, Yao DL, Hudson LD, Wekerle H, Kreutzberg GW 1997 Insulin-like growth factor-I treatment reduces immune cell responses in acute non-demyelinative experimental autoimmune encephalomyelitis. J Neurosci Res 47:531–538

    Yao DL, Liu X, Hudson LD, Webster HD 1996 Insulin-like growth factor-I given subcutaneously reduces clinical deficits, decreases lesion severity and upregulates synthesis of myelin proteins in experimental autoimmune encephalomyelitis. Life Sci 58:1301–1306

    Cannella B, Pitt D, Capello E, Raine CS 2000 Insulin-like growth factor-1 fails to enhance central nervous system myelin repair during autoimmune demyelination. Am J Pathol 157:933–943

    Lovett-Racke AE, Bittner P, Cross AH, Carlino JA, Racke MK 1998 Regulation of experimental autoimmune encephalomyelitis with insulin-like growth factor (IGF-1) and IGF-1/IGF-binding protein-3 complex (IGF-1/IGFBP3). J Clin Invest 101:1797–1804

    Dore S, Kar S, Zheng WH, Quirion R 2000 Rediscovering good old friend IGF-I in the new millenium: possible usefulness in Alzheimer’s disease and stroke. Pharm Acta Helv 74:273–280

    Carro E, Torres-Aleman I 2004 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 490:127–133

    Partridge L, Gems D 2002 The evolution of longevity. Curr Biol 12:R544–R546

    Carrascosa C, Torres-Aleman I, Lopez-Lopez C, Carro E, Espejo L, Torrado S, Torrado JJ 2004 Microspheres containing insulin-like growth factor I for treatment of chronic neurodegeneration. Biomaterials 25:707–714

    Feldman EL 2004 Amyotrophic lateral sclerosis and other motor neuron diseases. In: Goldman L, Ausiello D, eds. Cecil textbook of medicine. Philadelphia: W.B. Saunders; 2316–2319

    Price DL, Borchelt DR, Wong PC, Pardo CA, Thinakaran G, Lee MK, Cleveland DW, Sisodia SS 1996 Neurodegenerative diseases and model systems. Cold Spring Harb Symp Quant Biol 61:725–738

    Anlar B, Sullivan KA, Feldman EL 1999 Insulin-like growth factor-I and central nervous system development. Horm Metab Res 31:120–125

    Vincent AM, Mobley BC, Hiller A, Feldman EL 2004 IGF-I prevents glutamate-induced motor neuron programmed cell death. Neurobiol Dis 16:407–416

    Singleton JR, Feldman EL 2001 Insulin-like growth factor-I in muscle metabolism and myotherapies. Neurobiol Dis 8:541–554

    Cleveland DW, Rothstein JD 2001 From Charcot to Lou Gehrig: deciphering selective motor neuron death in ALS. Nat Rev Neurosci 2:806–819

    Torres-Aleman I, Barrios V, Berciano J 1998 The peripheral insulin-like growth factor system in amyotrophic lateral sclerosis and in multiple sclerosis. Neurology 50:772–776

    Wilczak N, de Vos RA, De Keyser J 2003 Free insulin-like growth factor (IGF)-I and IGF binding proteins 2, 5, and 6 in spinal motor neurons in amyotrophic lateral sclerosis. Lancet 361:1007–1011

    Vaught JL, Contreras PC, Glicksman MA, Neff NT 1996 Potential utility of rhIGF-1 in neuromuscular and/or degenerative disease. Ciba Found Symp 196:18–27; discussion, 27–38

    Adem A, Ekblom J, Gillberg PG, Jossan SS, Hoog A, Winblad B, Aquilonius SM, Wang LH, Sara V 1994 Insulin-like growth factor-1 receptors in human spinal cord: changes in amyotrophic lateral sclerosis. J Neural Transm Gen Sect 97:73–84

    Ackerman SJ, Sullivan EM, Beusterien KM, Natter HM, Gelinas DF, Patrick DL 1999 Cost effectiveness of recombinant human insulin-like growth factor I therapy in patients with ALS. Pharmacoeconomics 15:179–195

    Hardy J 1997 Amyloid, the presenilins and Alzheimer’s disease. Trends Neurosci 20:154–159

    Wisniewski T, Ghiso J, Frangione B 1997 Biology of A amyloid in Alzheimer’s disease. Neurobiol Dis 4:313–328

    Selkoe DJ 1999 Translating cell biology into therapeutic advances in Alzheimer’s disease. Nature 399:A23–A31

    Selkoe DJ 2001 Clearing the brain’s amyloid cobwebs. Neuron 32:177–180

    Clark CM, Karlawish JH 2003 Alzheimer disease: current concepts and emerging diagnostic and therapeutic strategies. Ann Intern Med 138:400–410

    Cotman CW, Su JH 1996 Mechanisms of neuronal death in Alzheimer’s disease. Brain Pathol 6:493–506

    Lindsay RM, Wiegand SJ, Altar CA, DiStefano PS 1994 Neurotrophic factors: from molecule to man. Trends Neurosci 17:182–190

    Connor B, Dragunow M 1998 The role of neuronal growth factors in neurodegenerative disorders of the human brain. Brain Res Brain Res Rev 27:1–39

    Connor B, Beilharz EJ, Williams C, Synek B, Gluckman PD, Faull RL, Dragunow M 1997 Insulin-like growth factor-I (IGF-I) immunoreactivity in the Alzheimer’s disease temporal cortex and hippocampus. Brain Res Mol Brain Res 49:283–290

    Amaducci L, Tesco G 1994 Aging as a major risk for degenerative diseases of the central nervous system. Curr Opin Neurol 7:283–286

    Arvat E, Broglio F, Ghigo E 2000 Insulin-like growth factor I: implications in aging. Drugs Aging 16:29–40

    Ghigo E, Arvat E, Gianotti L, Lanfranco F, Broglio F, Aimaretti G, Maccario M, Camanni F 2000 Hypothalamic growth hormone-insulin-like growth factor-I axis across the human life span. J Pediatr Endocrinol Metab 13(Suppl 6):1493–1502

    Frolich L, Blum-Degen D, Bernstein HG, Engelsberger S, Humrich J, Laufer S, Muschner D, Thalheimer A, Turk A, Hoyer S, Zochling R, Boissl KW, Jellinger K, Riederer P 1998 Brain insulin and insulin receptors in aging and sporadic Alzheimer’s disease. J Neural Transm 105:423–438

    Mustafa A, Lannfelt L, Lilius L, Islam A, Winblad B, Adem A 1999 Decreased plasma insulin-like growth factor-I level in familial Alzheimer’s disease patients carrying the Swedish APP 670/671 mutation. Dement Geriatr Cogn Disord 10:446–451

    Tham A, Nordberg A, Grissom FE, Carlsson-Skwirut C, Viitanen M, Sara VR 1993 Insulin-like growth factors and insulin-like growth factor binding proteins in cerebrospinal fluid and serum of patients with dementia of the Alzheimer type. J Neural Transm Park Dis Dement Sect 5:165–176

    Xie L, Helmerhorst E, Taddei K, Plewright B, Van Bronswijk W, Martins R 2002 Alzheimer’s -amyloid peptides compete for insulin binding to the insulin receptor. J Neurosci 22:RC221

    Gasparini L, Gouras GK, Wang R, Gross RS, Beal MF, Greengard P, Xu H 2001 Stimulation of -amyloid precursor protein trafficking by insulin reduces intraneuronal -amyloid and requires mitogen-activated protein kinase signaling. J Neurosci 21:2561–2570

    Gasparini L, Xu H 2003 Potential roles of insulin and IGF-1 in Alzheimer’s disease. Trends Neurosci 26:404–406

    Stoothoff WH, Johnson GV 2005 Tau phosphorylation: physiological and pathological consequences. Biochim Biophys Acta 1739:280–297

    Buee L, Bussiere T, Buee-Scherrer V, Delacourte A, Hof PR 2000 Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res Brain Res Rev 33:95–130

    Hong M, Lee VM 1997 Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurons. J Biol Chem 272:19547–19553

    Lesort M, Jope RS, Johnson GV 1999 Insulin transiently increases tau phosphorylation: involvement of glycogen synthase kinase-3 and Fyn tyrosine kinase. J Neurochem 72:576–584

    Lesort M, Johnson GV 2000 Insulin-like growth factor-1 and insulin mediate transient site-selective increases in tau phosphorylation in primary cortical neurons. Neuroscience 99:305–316

    Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL, Farhang-Fallah J, Dikkes P, Warot XM, Rio C, Corfas G, White MF 2003 Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J Neurosci 23:7084–7092

    Ikonen M, Liu B, Hashimoto Y, Ma L, Lee KW, Niikura T, Nishimoto I, Cohen P 2003 Interaction between the Alzheimer’s survival peptide humanin and insulin-like growth factor-binding protein 3 regulates cell survival and apoptosis. Proc Natl Acad Sci USA 100:13042–13047

    Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL 2002 Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci 5:514–516

    De Rosa R, Garcia AA, Braschi C, Capsoni S, Maffei L, Berardi N, Cattaneo A 2005 Intranasal administration of nerve growth factor (NGF) rescues recognition memory deficits in AD11 anti-NGF transgenic mice. Proc Natl Acad Sci USA 102:3811–3816

    Pu SF, Zhuang HX, Ishii DN 1995 Differential spatio-temporal expression of the insulin-like growth factor genes in regenerating sciatic nerve. Brain Res Mol Brain Res 34:18–28

    Cheng HL, Randolph A, Yee D, Delafontaine P, Tennekoon G, Feldman EL 1996 Characterization of insulin-like growth factor-I and its receptor and binding proteins in transected nerves and cultured Schwann cells. J Neurochem 66:525–536

    Zochodne DW, Cheng C 2000 Neurotrophins and other growth factors in the regenerative milieu of proximal nerve stump tips. J Anat 196:279–283

    Gehrmann J, Yao DL, Bonetti B, Bondy CA, Brenner M, Zhou J, Kreutzberg GW, Webster HD 1994 Expression of insulin-like growth factor-I and related peptides during motoneuron regeneration. Exp Neurol 128:202–210

    Mulholland MW, Romanchuk G, Simeone DM, Flowe K 1992 Stimulation of myenteric plexus neurite outgrowth by insulin and insulin-like growth factors I and II. Life Sci 51:1789–1796

    Zackenfels K, Oppenheim RW, Rohrer H 1995 Evidence for an important role of IGF-I and IGF-II for the early development of chick sympathetic neurons. Neuron 14:731–741

    Caroni P, Grandes P 1990 Nerve sprouting in innervated adult skeletal muscle induced by exposure to elevated levels of insulin-like growth factors. J Cell Biol 110:1307–1317

    Prager D, Melmed S 1993 Insulin and insulin-like growth factor I receptors: are there functional distinctions Endocrinology 132:1419–1420

    Vergani L, Di Giulio AM, Losa M, Rossoni G, Muller EE, Gorio A 1998 Systemic administration of insulin-like growth factor decreases motor neuron cell death and promotes muscle reinnervation. J Neurosci Res 54:840–847

    Contreras PC, Steffler C, Yu E, Callison K, Stong D, Vaught JL 1995 Systemic administration of rhIGF-I enhanced regeneration after sciatic nerve crush in mice. J Pharmacol Exp Ther 274:1443–1449

    Zhuang HX, Wuarin L, Fei ZJ, Ishii DN 1997 Insulin-like growth factor (IGF) gene expression is reduced in neural tissues and liver from rats with non-insulin-dependent diabetes mellitus, and IGF treatment ameliorates diabetic neuropathy. J Pharmacol Exp Ther 283:366–374

    Wuarin L, Guertin DM, Ishii DN 1994 Early reduction in insulin-like growth factor gene expression in diabetic nerve. Exp Neurol 130:106–114

    Migdalis IN, Kalogeropoulou K, Kalantzis L, Nounopoulos C, Bouloukos A, Samartzis M 1995 Insulin-like growth factor-I and IGF-I receptors in diabetic patients with neuropathy. Diabet Med 12:823–827

    Ishii DN, Lupien SB 1995 Insulin-like growth factors protect against diabetic neuropathy: effects on sensory nerve regeneration in rats. J Neurosci Res 40:138–144

    Schmidt RE, Dorsey DA, Beaudet LN, Plurad SB, Parvin CA, Miller MS 1999 Insulin-like growth factor I reverses experimental diabetic autonomic neuropathy. Am J Pathol 155:1651–1660

    Contreras PC, Vaught JL, Gruner JA, Brosnan C, Steffler C, Arezzo JC, Lewis ME, Kessler JA, Apfel SC 1997 Insulin-like growth factor-I prevents development of a vincristine neuropathy in mice. Brain Res 774:20–26

    Songyang Z, Baltimore D, Cantley LC, Kaplan DR, Franke TF 1997 Interleukin 3-dependent survival by the Akt protein kinase. Proc Natl Acad Sci USA 94:11345–11350

    Reiss K 2002 Insulin-like growth factor-I receptor—a potential therapeutic target in medulloblastomas. Expert Opin Ther Targets 6:539–544

    Blakesley VA, Stannard BS, Kalebic T, Helman LJ, LeRoith D 1997 Role of the IGF-I receptor in mutagenesis and tumor promotion. J Endocrinol 152:339–344

    Werner H 1998 Dysregulation of the type 1 IGF receptor as a paradigm in tumor progression. Mol Cell Endocrinol 141:1–5

    Dunn SE, Ehrlich M, Sharp NJ, Reiss K, Solomon G, Hawkins R, Baserga R, Barrett JC 1998 A dominant negative mutant of the insulin-like growth factor-I receptor inhibits the adhesion, invasion, and metastasis of breast cancer. Cancer Res 58:3353–3361

    Min Y, Adachi Y, Yamamoto H, Ito H, Itoh F, Lee CT, Nadaf S, Carbone DP, Imai K 2003 Genetic blockade of the insulin-like growth factor-I receptor: a promising strategy for human pancreatic cancer. Cancer Res 63:6432–6441

    Scotlandi K, Maini C, Manara MC, Benini S, Serra M, Cerisano V, Strammiello R, Baldini N, Lollini PL, Nanni P, Nicoletti G, Picci P 2002 Effectiveness of insulin-like growth factor I receptor antisense strategy against Ewing’s sarcoma cells. Cancer Gene Ther 9:296–307

    Baserga R 1996 Controlling IGF-receptor function: a possible strategy for tumor therapy. Trends Biotechnol 14:150–152

    Culver KW 1996 Gene therapy for malignant neoplasms of the CNS. Bone Marrow Transplant 18(Suppl 3):S6–S9

    Sara VR, Carlsson-Skwirut C, Bergman T, Jornvall H, Roberts PJ, Crawford M, Hakansson LN, Civalero I, Nordberg A 1989 Identification of Gly-Pro-Glu (GPE), the aminoterminal tripeptide of insulin-like growth factor 1 which is truncated in brain, as a novel neuroactive peptide. Biochem Biophys Res Commun 165:766–771

    Bourguignon J, Gerard A 1999 Role of insulin-like growth factor binding proteins in limitation of IGF-I degradation into the N-methyl-D-aspartate receptor antagonist GPE: evidence from gonadotrophin-releasing hormone secretion in vitro at two developmental stages. Brain Res 847:247–252

    Guan J, Waldvogel HJ, Faull RL, Gluckman PD, Williams CE 1999 The effects of the N-terminal tripeptide of insulin-like growth factor-1, glycine-proline-glutamate in different regions following hypoxic-ischemic brain injury in adult rats. Neuroscience 89:649–659

    Krishnamurthi R, Stott S, Maingay M, Faull RL, McCarthy D, Gluckman P, Guan J 2004 N-terminal tripeptide of IGF-1 improves functional deficits after 6-OHDA lesion in rats. Neuroreport 15:1601–1604

    Alexi T, Hughes PE, van Roon-Mom WM, Faull RL, Williams CE, Clark RG, Gluckman PD 1999 The IGF-I amino-terminal tripeptide glycine-proline-glutamate (GPE) is neuroprotective to striatum in the quinolinic acid lesion animal model of Huntington’s disease. Exp Neurol 159:84–97

    Barbieri M, Bonafe M, Franceschi C, Paolisso G 2003 Insulin/IGF-I-signaling pathway: an evolutionarily conserved mechanism of longevity from yeast to humans. Am J Physiol Endocrinol Metab 285:E1064–E1071

    Richardson A, Liu F, Adamo ML, Remmen HV, Nelson JF 2004 The role of insulin and insulin-like growth factor-I in mammalian ageing. Best Pract Res Clin Endocrinol Metab 18:393–406

    Rea S, Johnson TE 2003 A metabolic model for life span determination in Caenorhabditis elegans. Dev Cell 5:197–203

    Tatar M, Bartke A, Antebi A 2003 The endocrine regulation of aging by insulin-like signals. Science 299:1346–1351

    Antebi A 2004 Inside insulin signaling, communication is key to long life. Sci Aging Knowledge Environ 2004:pe25

    Bluher M, Kahn BB, Kahn CR 2003 Extended longevity in mice lacking the insulin receptor in adipose tissue. Science 299:572–574

    Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC, Cervera P, Le Bouc Y 2003 IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421:182–187

    Hwangbo DS, Gershman B, Tu MP, Palmer M, Tatar M 2004 Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body. Nature 429:562–566

    Wolkow CA, Kimura KD, Lee MS, Ruvkun G 2000 Regulation of C. elegans life-span by insulinlike signaling in the nervous system. Science 290:147–150

    Apfeld J, Kenyon C 1999 Regulation of lifespan by sensory perception in Caenorhabditis elegans. Nature 402:804–809

    Alcedo J, Kenyon C 2004 Regulation of C. elegans longevity by specific gustatory and olfactory neurons. Neuron 41:45–55

    Swanson LW 1998/1999 Brain maps: structures of the rat brain. 2nd ed. Amsterdam: Elsevier

您现在查看是摘要介绍页,详见ORG附件