Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
3.
PLoS One ; 8(6): e67204, 2013.
Article in English | MEDLINE | ID: mdl-23826235

ABSTRACT

Traumatic brain injury (TBI) survivors often suffer from long-lasting cognitive impairment that stems from hippocampal injury. Systemic administration of insulin-like growth factor-1 (IGF-1), a polypeptide growth factor known to play vital roles in neuronal survival, has been shown to attenuate posttraumatic cognitive and motor dysfunction. However, its neuroprotective effects in TBI have not been examined. To this end, moderate or severe contusion brain injury was induced in mice with conditional (postnatal) overexpression of IGF-1 using the controlled cortical impact (CCI) injury model. CCI brain injury produces robust reactive astrocytosis in regions of neuronal damage such as the hippocampus. We exploited this regional astrocytosis by linking expression of hIGF-1 to the astrocyte-specific glial fibrillary acidic protein (GFAP) promoter, effectively targeting IGF-1 delivery to vulnerable neurons. Following brain injury, IGF-1Tg mice exhibited a progressive increase in hippocampal IGF-1 levels which was coupled with enhanced hippocampal reactive astrocytosis and significantly greater GFAP levels relative to WT mice. IGF-1 overexpression stimulated Akt phosphorylation and reduced acute (1 and 3d) hippocampal neurodegeneration, culminating in greater neuron survival at 10d after CCI injury. Hippocampal neuroprotection achieved by IGF-1 overexpression was accompanied by improved motor and cognitive function in brain-injured mice. These data provide strong support for the therapeutic efficacy of increased brain levels of IGF-1 in the setting of TBI.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/metabolism , Hippocampus/metabolism , Insulin-Like Growth Factor I/metabolism , Neuroprotection/physiology , Animals , Astrocytes/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/psychology , Cognition/physiology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Gliosis/metabolism , Gliosis/pathology , Hippocampus/pathology , Humans , Insulin-Like Growth Factor I/genetics , Memory/physiology , Mice, Transgenic , Motor Activity/physiology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
4.
J Sports Med Phys Fitness ; 52(3): 229-36, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22648460

ABSTRACT

AIM: The aim of this study, based on the interaction between two aerobic and anaerobic metabolisms with a parallel production of both aerobic and anaerobic ATP, was to develop a high intensity training programme and increase the aerobic contribution. We examined the applicability of a 16-week training programme with an ergospirometer treadmill and field tests on eight water polo players. METHODS: Tests/retests of repeated exercises to 90V (90% of maximum personal speed over 100 m freestyle) and Speed Endurance Training (SET) after eight weeks were developed. A one-way blocked ANOVA with random blocks was used and each player represented a particular block with two before-after treatments with the aim of reducing error by subtracting both the variance due to the difference between the treatments and that due to the difference between the blocks. RESULTS: A reduction (15.2%) in blood lactate was observed in response to the same absolute workload (before-after). Furthermore the anaerobic contribution to VO2max (ESCAna, Estimated Anaerobic Contribution) after eight weeks of training at 90maxV and the anaerobic contribution to VO2max (ESCAna) after speed endurance training (SET) were very significant (P<0.004) with a reduction in the anaerobic contribution of 16%. The results of the field tests show that there was a very significant reduction (P<0.001) in lactate between 90maxV and maximal aerobic power velocity (MAPv) of 24%. CONCLUSION: With 90maxV and SET, space was gained towards those velocities, which had previously required a considerable anaerobic contribution. In this way match speed was increased.


Subject(s)
Athletic Performance/physiology , Physical Education and Training/methods , Physical Endurance/physiology , Sports , Anaerobic Threshold/physiology , Analysis of Variance , Exercise Test , Humans , Male , Oxygen Consumption/physiology , Physical Fitness/physiology , Water , Young Adult
5.
Neuron ; 69(5): 893-905, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21382550

ABSTRACT

Cortical development depends on the active integration of cell-autonomous and extrinsic cues, but the coordination of these processes is poorly understood. Here, we show that the apical complex protein Pals1 and Pten have opposing roles in localizing the Igf1R to the apical, ventricular domain of cerebral cortical progenitor cells. We found that the cerebrospinal fluid (CSF), which contacts this apical domain, has an age-dependent effect on proliferation, much of which is attributable to Igf2, but that CSF contains other signaling activities as well. CSF samples from patients with glioblastoma multiforme show elevated Igf2 and stimulate stem cell proliferation in an Igf2-dependent manner. Together, our findings demonstrate that the apical complex couples intrinsic and extrinsic signaling, enabling progenitors to sense and respond appropriately to diffusible CSF-borne signals distributed widely throughout the brain. The temporal control of CSF composition may have critical relevance to normal development and neuropathological conditions.


Subject(s)
Cerebral Cortex/physiology , Cerebrospinal Fluid/physiology , Neural Stem Cells/physiology , Analysis of Variance , Animals , Brain Neoplasms/cerebrospinal fluid , Cell Proliferation , Cerebral Cortex/cytology , Glioblastoma/cerebrospinal fluid , Humans , Insulin-Like Growth Factor II/metabolism , Membrane Proteins/metabolism , Mice , Neural Stem Cells/cytology , Neurons/metabolism , Nucleoside-Phosphate Kinase/metabolism , PTEN Phosphohydrolase/metabolism , Receptor, IGF Type 1/metabolism , Statistics, Nonparametric
6.
Glia ; 58(9): 1031-41, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20235220

ABSTRACT

By promoting cell proliferation, survival and maturation insulin-like growth factor (IGF)-I is essential to the normal growth and development of the central nervous system. It is clear that IGF-I actions are primarily mediated by the type I IGF receptor (IGF1R), and that phosphoinositide 3 (PI3)-Akt kinases and MAP kinases signal many of IGF-I-IGF1R actions in neural cells, including oligodendrocyte lineage cells. The precise downstream targets of these signaling pathways, however, remain to be defined. We studied oligodendroglial cells to determine whether beta-catenin, a molecule that is a downstream target of glycogen synthase kinase-3beta (GSK3beta) and plays a key role in the Wnt canonical signaling pathway, mediates IGF-I actions. We found that IGF-I increases beta-catenin protein abundance within an hour after IGF-I-induced phosphorylation of Akt and GSK3beta. Inhibiting the PI3-Akt pathway suppressed IGF-I-induced increases in beta-catenin and cyclin D1 mRNA, while suppression of GSK3beta activity simulated IGF-I actions. Knocking-down beta-catenin mRNA by RNA interference suppressed IGF-I-stimulated increases in the abundance of cyclin D1 mRNA, cell proliferation, and cell survival. Our data suggest that beta-catenin is an important downstream molecule in the PI3-Akt-GSK3beta pathway, and as such it mediates IGF-I upregulation of cyclin D1 mRNA and promotion of cell proliferation and survival in oligodendroglial cells.


Subject(s)
Cell Proliferation , Cyclin D/metabolism , Insulin-Like Growth Factor I/metabolism , Oligodendroglia/physiology , beta Catenin/metabolism , Animals , Cell Line , Cell Survival/physiology , Cells, Cultured , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction , Stem Cells/physiology , Time Factors
7.
J Neurosci Res ; 87(13): 2821-32, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19437543

ABSTRACT

Type 1 insulin-like growth factor receptor (IGF1R) signaling in neuronal development was studied in mutant mice with blunted igf1r gene expression in nestin-expressing neuronal precursors. At birth [postnatal (P) day 0] brain weights were reduced to 37% and 56% of controls in mice homozygous (nes-igf1r(-/-)) and heterozygous (nes-igf1r(-/Wt)) for the null mutation, respectively, and this brain growth retardation persisted postnatally. Stereological analysis demonstrated that the volumes of the hippocampal formation, CA fields 1-3, dentate gyrus (DG), and DG granule cell layer (GCL) were decreased by 44-54% at P0 and further by 65-69% at P90 in nes-igf1r(-/Wt) mice. In nes-igf1r(-/-) mice, volumes were 29-31% of controls at P0 and, in the two mice that survived to P90, 6-19% of controls, although the hilus could not be identified. Neuron density did not differ among the mice at any age studied; therefore, decreased volumes were due to reduced cell number. In postnatal nes-igf1r(-/Wt) mice, the percentage of apoptotic cells, as judged by activated caspase-3 immunostaining, was increased by 3.5-5.3-fold. The total number of proliferating DG progenitors (labeled by BrdU incorporation and Ki67 staining) was reduced by approximately 50%, but the percentage of these cells was similar to the percentages in littermate controls. These findings suggest that 1) the postnatal reduction in DG size is due predominantly to cell death, pointing to the importance of the IGF1R in regulating postnatal apoptosis, 2) surviving DG progenitors remain capable of proliferation despite reduced IGF1R expression, and 3) IGF1R signaling is necessary for normal embryonic brain development.


Subject(s)
Hippocampus/growth & development , Neurogenesis/physiology , Receptor, IGF Type 1/physiology , Animals , Apoptosis , Cell Count , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Female , Gene Expression Regulation , Genes, Lethal , Genes, Reporter , Genotype , Hippocampus/embryology , Hippocampus/pathology , Hypothalamus/embryology , Hypothalamus/growth & development , Intermediate Filament Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin , Neurons/pathology , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics , Signal Transduction/physiology , Transgenes
8.
Glia ; 57(1): 1-12, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18627006

ABSTRACT

Both in vivo and in vitro studies indicate a correlation between reduced acetylation of histone core proteins and oligodendrocyte development. The nature of these histone modifications and the mechanisms mediating them remain undefined. To address these issues, we utilized OL-1 cells, a rat nontransformed oligodendrocyte cell line, and primary oligodendrocyte cultures. We found that the acetylated histone H3 at lysine 9 and lysine 14 (H3K9/K14ac) is reduced in both the myelin basic protein (MBP) and proteolipid protein (PLP) genes of maturing oligodendroglial OL-1 cells, and furthermore, this temporally correlates with increases in MBP, PLP, and histone deacetylase (HDAC) 11 expression. Disruption of developmentally-regulated histone H3 deacetylation within the MBP and PLP genes by the HDAC inhibitor trichostatin A blunts MBP and PLP expression. With its increased expression, interaction of HDAC 11 with acetylated histone H3 and recruitment of HDAC 11 to the MBP and PLP genes markedly increases in maturing OL-1 cells. Moreover, suppressing HDAC 11 expression with small interfering RNA significantly (1) increases H3K9/K14ac globally and within the MBP and PLP genes, (2) decreases MBP and PLP mRNA expression, and (3) blunts the morphological changes associated with oligodendrocyte development. Our data strongly support a specific role for HDAC 11 in histone deacetylation and in turn the regulation of oligodendrocyte-specific protein gene expression and oligodendrocyte development.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental/physiology , Histone Deacetylases/physiology , Oligodendroglia/cytology , Oligodendroglia/enzymology , Animals , Animals, Newborn , Cell Line , Cell Line, Tumor , Cells, Cultured , Humans , Rats , Stem Cells/cytology , Stem Cells/enzymology
9.
Genes Brain Behav ; 8(2): 129-42, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19016890

ABSTRACT

Profound impairment in social interaction is a core symptom of autism, a severe neurodevelopmental disorder. Deficits can include a lack of interest in social contact and low levels of approach and proximity to other children. In this study, a three-chambered choice task was used to evaluate sociability and social novelty preference in five lines of mice with mutations in genes implicated in autism spectrum disorders. Fmr1(tm1Cgr/Y)(Fmr1(-/y)) mice represent a model for fragile X, a mental retardation syndrome that is partially comorbid with autism. We tested Fmr1(-/y)mice on two genetic backgrounds, C57BL/6J and FVB/N-129/OlaHsd (FVB/129). Targeted disruption of Fmr1 resulted in low sociability on one measure, but only when the mutation was expressed on FVB/129. Autism has been associated with altered serotonin levels and polymorphisms in SLC6A4 (SERT), the serotonin transporter gene. Male mice with targeted disruption of Slc6a4 displayed significantly less sociability than wild-type controls. Mice with conditional overexpression of Igf-1 (insulin-like growth factor-1) offered a model for brain overgrowth associated with autism. Igf-1 transgenic mice engaged in levels of social approach similar to wild-type controls. Targeted disruption in other genes of interest, En2 (engrailed-2) and Dhcr7, was carried on genetic backgrounds that showed low levels of exploration in the choice task, precluding meaningful interpretations of social behavior scores. Overall, results show that loss of Fmr1 or Slc6a4 gene function can lead to deficits in sociability. Findings from the fragile X model suggest that the FVB/129 background confers enhanced susceptibility to consequences of Fmr1 mutation on social approach.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/psychology , Genetic Engineering , Mice, Knockout/genetics , Mice, Knockout/psychology , Social Behavior , Animals , Anxiety/psychology , Behavior, Animal/physiology , Exploratory Behavior/physiology , Female , Food Deprivation/physiology , Fragile X Mental Retardation Protein/genetics , Homeodomain Proteins/genetics , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Postural Balance/physiology , Pregnancy , Serotonin Plasma Membrane Transport Proteins/genetics , Sex Characteristics , Smell/genetics , Smell/physiology
10.
Opt Express ; 16(16): 12002-7, 2008 Aug 04.
Article in English | MEDLINE | ID: mdl-18679473

ABSTRACT

We discuss and experimentally demonstrate a scheme to achieve photorefractive solitons of arbitrary linear polarization using the quadratic electro-optic effect and describe the observation of the self-trapping of a set of linear polarized beams in different positions of a paraelectric photorefractive crystal of potassium-lithium-tantalate-niobate (KLTN) biased by the inhomogeneous field produced by two miniaturized top electrodes. The polarization of the single solitons of the set is determined by the local electrostatic configuration and the underlying tunable anisotropy, which is detected through zero-field electro-activation.


Subject(s)
Electromagnetic Phenomena/instrumentation , Electronics/instrumentation , Refractometry/instrumentation , Electromagnetic Fields , Equipment Design , Equipment Failure Analysis , Light , Linear Models , Scattering, Radiation
11.
Endocrinology ; 149(12): 5958-62, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18687773

ABSTRACT

Signaling through the type 1 IGF receptor (IGF1R) after interaction with IGF-I is crucial to the normal brain development. Manipulations of the mouse genome leading to changes in the expression of IGF-I or IGF1R significantly alters brain growth, such that IGF-I overexpression leads to brain overgrowth, whereas null mutations in either IGF-I or the IGF1R result in brain growth retardation. IGF-I signaling stimulates the proliferation, survival, and differentiation of each of the major neural lineages, neurons, oligodendrocytes, and astrocytes, as well as possibly influencing neural stem cells. During embryonic life, IGF-I stimulates neuron progenitor proliferation, whereas later it promotes neuron survival, neuritic outgrowth, and synaptogenesis. IGF-I also stimulates oligodendrocyte progenitor proliferation although inhibiting apoptosis in oligodendrocyte lineage cells and stimulating myelin production. These pleiotropic IGF-I activities indicate that other factors provide instructive signals for specific cellular events and that IGF-I acts to facilitate them. Studies of the few humans with IGF-I and/or IGF1R gene mutations indicate that IGF-I serves a similar role in man.


Subject(s)
Brain/metabolism , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Animals , Brain/cytology , Brain/growth & development , Humans , Insulin-Like Growth Factor I/physiology , Models, Biological , Myelin Sheath/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oligodendroglia/physiology , Receptor, IGF Type 1/physiology , Signal Transduction/physiology
12.
Anticancer Res ; 28(3A): 1509-16, 2008.
Article in English | MEDLINE | ID: mdl-18630505

ABSTRACT

BACKGROUND: Neuroblastoma is the third most common solid tumor in children. Treatment continues to be challenging. The pathogenesis of neuroblastoma has been related to expression of the type 1 insulin-like growth factor receptor (IGF1R) and to transcription factor MYC-N amplification. Previous studies have shown that MYC-N expression is disrupted by blockade of the IGF1R with a specific monoclonal antibody, alphaIR3. Inhibition of IGF1R signaling can be accomplished by other agents, including rapamycin or temsirolimus, which target mTOR (mammalian target of rapamycin). MATERIALS AND METHODS: BE-2(c) and IMR-32 neuroblastoma cell lines were treated with varying concentrations of alphaIR3, rapamycin and temsirolimus alone or in combination and the viable cells were counted. RESULTS: Blockade of IGF1R signaling significantly inhibited cell growth as compared to untreated controls (p < 0.05), and a combination of agents was more effective than each agent alone. CONCLUSION: The combination of rapamycin or temsirolimus with alphaIR3 blocks the IGF1R signaling pathway and has an antiproliferative effect on neuroblastoma cells warranting further investigations using inhibitors of IGF1R signaling as novel combination therapy for neuroblastoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neuroblastoma/drug therapy , Receptor, IGF Type 1/antagonists & inhibitors , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Protein Kinases/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases
13.
Growth Horm IGF Res ; 18(6): 517-25, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18550407

ABSTRACT

Our recent report on a parallel decrease in the body weights and serum IGF-I levels of weaver mice suggests that IGF-I's endocrine function may be impaired in neurodegenerative diseases. To further understand the overall effects of IGF-I deficiency on the postnatal growth, we measured bone mineral density (BMD), bone mineral content (BMC), lean body mass (LBM) and fat mass in male and female weaver mice and wild-type littermates on D21 (prepuberty), D45 (puberty), and D60 (postpuberty) using dual-energy X-ray absorptiometry (DEXA). In both male and female weaver mice, we found that the levels of circulating IGF-I paralleled those of BMD, BMC, and LBM, but not the fat mass. Male weaver mice have normal fat mass at all three ages studied, whereas female weaver mice showed a trend to increase their fat mass as they mature. To determine whether circulating IGF-I is a determinant of body composition, we crossbred IGF-I transgenic mice with homozygous weaver mice, which resulted in a significant increase in circulating IGF-I levels in both male and female weaver mice and normalization of their BMD, BMC and body weights. In summary, our results demonstrated that normal circulating IGF-I levels are important in maintaining BMD, BMC, and body composition in neurodegenerative diseases, such as hereditary cerebellar ataxia.


Subject(s)
Body Composition/physiology , Body Weight/physiology , Bone Density/physiology , Insulin-Like Growth Factor I/metabolism , Animals , Cerebellar Ataxia/genetics , Cerebellar Ataxia/physiopathology , Female , Male , Mice , Mice, Neurologic Mutants
14.
J Neurosci Res ; 86(3): 537-43, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17893925

ABSTRACT

Recent studies indicate that neural cell development in the central nervous system (CNS) correlates with a reduction in acetylation of histone core proteins. Moreover, histone hypoacetylation is thought to be important to oligodendrocyte lineage development. The mechanisms mediating the reduction in acetylation during postnatal neural development remain to be defined. To begin to understand these mechanisms, we investigated the expression of histone deacetylase 11 (HDAC11), a newly identified HDAC, in mouse brain during postnatal development. We show that HDAC11 was widely expressed in the brain and that this expression gradually increased in a region-specific pattern between birth and 4 weeks of age. At the cellular level HDAC11 protein was predominately localized in the nuclei of mature oligodendrocytes but only minimally in astrocytes. Although dentate gyrus granule neurons abundantly expressed HDAC11, granule neuron precursors in the subgranule layer exhibited little HDAC11 immunoreactivity. Double-immunostaining of the corpus callosum and dentate gyrus demonstrated that HDAC11 and Ki67, a cell-proliferating marker, are rarely colocalized in same cells. Our data show that HDAC11 was expressed in the developing brain in a temporal and spatial pattern that correlates with the maturation of neural cells, including cells of the oligodendrocyte lineage. These findings support a role for HDAC11 in CNS histone deacetylation and the development of oligodendrocytes and neurons during postnatal development.


Subject(s)
Aging/metabolism , Brain/enzymology , Brain/growth & development , Histone Deacetylases/metabolism , Animals , Astrocytes/enzymology , Brain/cytology , Cell Lineage , Cell Nucleus/enzymology , Cellular Senescence , Corpus Callosum/cytology , Corpus Callosum/enzymology , Corpus Callosum/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/enzymology , Dentate Gyrus/metabolism , Immunologic Techniques , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/physiology , Oligodendroglia/cytology , Oligodendroglia/enzymology , Oligodendroglia/physiology , Staining and Labeling , Stem Cells/cytology , Stem Cells/enzymology , Tissue Distribution
15.
Am J Physiol Gastrointest Liver Physiol ; 293(5): G995-1003, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17823215

ABSTRACT

Insulin-like growth factor I (IGF-I) potently stimulates intestinal growth. Insulin receptor substrate-1 (IRS-1) mediates proliferative and antiapoptotic actions of IGF-I in cell lines, but its in vivo relevance in intestine is not defined. This study tested the hypothesis that there is cell type-specific dependence on IRS-1 as a mediator of IGF-I action. Length, mass, crypt cell proliferation, and apoptosis were measured in small intestine and colon of IRS-1-null mice and wild-type (WT) littermates and in colon of IRS-1-null or WT mice expressing IGF-I transgenes. Expression of IGF-I receptor and signaling intermediates was examined in intestine of WT and IRS-1-null mice, cultured intestinal epithelial cells, and myofibroblasts. Absolute IRS-1 deficiency reduced mucosal mass in jejunum and colon, but effects were more pronounced in colon. Muscularis mass was decreased in both segments. In IGF-I transgenics, IRS-1 deficiency significantly attenuated IGF-I-stimulated growth of colonic mucosa and abolished antiapoptotic but not mitogenic effects of IGF-I transgene on crypt cells. IGF-I-induced muscularis growth was unaffected by IRS-1 deficiency. In intestinal epithelial cells, IRS-1 was expressed at higher levels than IRS-2 and was preferentially activated by IGF-I. In contrast, IGF-I activated both IRS-1 and IRS-2 in intestinal myofibroblasts and IRS-2 activation was upregulated in IRS-1-null myofibroblasts. We conclude that the intestinal epithelium but not the muscularis requires IRS-1 for normal trophic actions of IGF-I and that IRS-1 is required for antiapoptotic but not mitogenic effects of IGF-I in the intestinal crypts in vivo.


Subject(s)
Colon/cytology , Insulin-Like Growth Factor I/pharmacology , Intestinal Mucosa/cytology , Muscle, Smooth/cytology , Phosphoproteins/deficiency , Phosphoproteins/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Division/drug effects , Colon/drug effects , Colon/physiology , Insulin Receptor Substrate Proteins , Intestinal Mucosa/drug effects , Intestinal Mucosa/physiology , Mice , Mice, Knockout , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Organ Size
16.
Int J Dev Neurosci ; 25(4): 233-41, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17459648

ABSTRACT

Increased expression of insulin-like growth factor-I (IGF-I) in embryonic neural progenitors in vivo has been shown to accelerate neuron proliferation in the neocortex. In the present study, the in vivo actions of (IGF-I) on naturally occurring neuron death in the cerebral cortex were investigated during embryonic and early postnatal development in a line of transgenic (Tg) mice that overexpress IGF-I in the brain, directed by nestin genomic regulatory elements, beginning at least as early as embryonic day (E) 13. The areal density of apoptotic cells (N(A), cells/mm2) at E16 in the telencephalic wall of Tg and littermate control embryos was determined by immunostaining with an antibody specific for activated caspase-3. Stereological analyses were conducted to measure the numerical density (N(V), cells/mm3) and total number of immunoreactive apoptotic cells in the cerebral cortex of nestin/IGF-I Tg and control mice at postnatal days (P) 0 and 5. The volume of cerebral cortex and both the N(V) and total number of all cortical neurons also were determined in both cerebral hemispheres at P0, P5 and P270. Apoptotic cells were rare in the embryonic telencephalic wall at E16. However, the overall N(A) of apoptotic cells was found to be significantly less by 46% in Tg embryos. The volume of the cerebral cortex was significantly greater in Tg mice at P0 (30%), P5 (13%) and P270 (26%). The total number of cortical neurons in Tg mice was significantly increased at P0 (29%), P5 (29%) and P270 (31%), although the N(V) of cortical neurons did not differ significantly between Tg and control mice at any age. Transgenic mice at P0 and P5 exhibited significant decreases in the N(V) of apoptotic cells in the cerebral cortex (31% and 39%, respectively). The vast majority of these apoptotic cells (> 90%) were judged to be neurons by their morphological appearance. Increased expression of IGF-I inhibits naturally occurring (i.e. apoptotic) neuron death during early postnatal development of the cerebral cortex to a degree that sustains a persistent increase in total neuron number even in the adult animal.


Subject(s)
Apoptosis/physiology , Cerebral Cortex , Gene Expression Regulation, Developmental/physiology , Insulin-Like Growth Factor Binding Protein 1/physiology , Neural Inhibition/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/genetics , Caspase 3/metabolism , Cell Count , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Humans , Insulin-Like Growth Factor Binding Protein 1/genetics , Intermediate Filament Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin
17.
J Neurosci Res ; 85(8): 1618-27, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17455296

ABSTRACT

Insulin-like growth factor-1 (IGF-1) is essential to hippocampal neurogenesis and the neuronal response to hypoxia/ischemia injury. IGF (IGF-1 and -2) signaling is mediated primarily by the type 1 IGF receptor (IGF-1R) and modulated by six high-affinity binding proteins (IGFBP) and the type 2 IGF receptor (IGF-2R), collectively termed IGF system proteins. Defining the precise cells that express each is essential to understanding their roles. With the exception of IGFBP-1, we found that mouse hippocampus expresses mRNA for each of these proteins during the first 2 weeks of postnatal life. Compared to postnatal day 14 (P14), mRNA abundance at P5 was higher for IGF-1, IGFBP-2, -3, and -5 (by 71%, 108%, 100%, and 98%, respectively), lower for IGF-2, IGF-2R, and IGFBP-6 (by 65%, 78%, and 44%, respectively), and unchanged for IGF-1R and IGFBP-4. Using laser capture microdissection (LCM), we found that granule neurons and pyramidal neurons exhibited identical patterns of expression of IGF-1, IGF-1R, IGF-2R, IGFBP-2, and -4, but did not express other IGF system genes. We then compared IGF system expression in mature granule neurons and their progenitors. Progenitors exhibited higher mRNA levels of IGF-1 and IGF-1R (by 130% and 86%, respectively), lower levels of IGF-2R (by 72%), and similar levels of IGFBP-4. Our data support a role for IGF in hippocampal neurogenesis and provide evidence that IGF actions are regulated within a defined in vivo milieu.


Subject(s)
Hippocampus/metabolism , Insulin-Like Growth Factor Binding Proteins/biosynthesis , Neurons/metabolism , Receptor, IGF Type 1/biosynthesis , Animals , Animals, Newborn , Gene Expression Regulation, Developmental , Hippocampus/growth & development , Insulin-Like Growth Factor Binding Proteins/genetics , Lasers , Mice , Mice, Inbred C57BL , Microdissection , Pyramidal Cells/metabolism , RNA, Messenger/biosynthesis , Receptor, IGF Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
19.
Int J Dev Neurosci ; 25(2): 95-105, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17306496

ABSTRACT

The process by which oligodendrocyte progenitors differentiate into mature oligodendrocytes is complex and incompletely understood in part because of the paucity of oligodendrocyte precursors cell lines that can be studied in culture. We have developed a non-immortalized rat oligodendrocyte precursor line, called OL-1, which behaves in a fashion consistent with developing oligodendrocytes in vivo. This OL-1 line provides a model for the study of oligodendrocyte development and offers an alternative to the CG-4 cell line. When OL-1 cells are propagated in conditioned growth media, they have morphology consistent with immature oligodendrocytes and exhibit A2B5 antigen positive and myelin basic protein-negative immunoreactivity. Withdrawal of conditioned growth media and culture in serum-free medium results in OL-1 cell maturation, manifested by a shift to myelin basic protein-positive immunoreactivity, A2B5 antigen-negative immunoreactivity, decreased NG2 mRNA expression, increased expression of proteolipid protein mRNA, and increased expression of CNP protein. In addition, the expression of proteolipid protein and its splicing variant DM-20 exhibit a pattern that is similar to brain proteolipid protein expression during development. When OL-1 cells are exposed to Insulin-like growth factor-I, there are significant increases in proteolipid protein mRNA expression (p<0.05), the number of cell processes (p<0.05), and cell number (p<0.05). Treatment with the caspase inhibitors Z-DEVD-FMK and Z-VAD-FMK (inhibitors of caspases 3, 6, 7, 8, 10 and 1, 3, 4, respectively), Insulin-like growth factor-I, or both, results in a similar increase in cell number. Because Insulin-like growth factor-I does not substantially increase the BrdU labeling of OL-1 cells, these data collectively indicate that Insulin-like growth factor-I increases OL-1 cell number predominately by promoting survival, rather than stimulating proliferation. This non-immortalized oligodendrocyte precursor cell line, therefore, exhibits behavior consistent with the in vivo development of oligodendrocytes and provides an excellent model for the study of developing oligodendrocytes.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/embryology , Central Nervous System/growth & development , Insulin-Like Growth Factor I/metabolism , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Antigens/genetics , Antigens, Surface/immunology , Caspase Inhibitors , Caspases/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Line , Cell Shape/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Central Nervous System/cytology , Culture Media, Conditioned/pharmacology , Enzyme Inhibitors/pharmacology , Insulin-Like Growth Factor I/pharmacology , Myelin Basic Protein/immunology , Myelin Proteolipid Protein/genetics , Oligodendroglia/cytology , Oligodendroglia/drug effects , Proteoglycans/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Receptors, Atrial Natriuretic Factor/immunology , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/drug effects
20.
J Neurosci Res ; 85(4): 712-22, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17279553

ABSTRACT

Our groups have reported that tumor necrosis factor-alpha (TNF-alpha) causes myelin damage and apoptosis of oligodendrocytes and their precursors in vitro and in vivo. We also have reported that insulin-like growth factor-I (IGF-I) can protect cultured oligodendrocytes and their precursors from TNF-alpha-induced damage. In this study, we investigated whether IGF-I can protect oligodendrocytes and myelination from TNF-alpha-induced damage in vivo by cross-breeding TNF-alpha transgenic (Tg) mice with IGF-I Tg mice that overexpress IGF-I exclusively in brain. At 8 weeks of age, compared with those of wild-type (WT) mice, the brain weights of TNF-alpha Tg mice were decreased by approximately 20%, and those of IGF-I Tg mice were increased by approximately 20%. The brain weights of mice that carry both TNF-alpha and IGF-I transgenes (TNF-alpha/IGF-I Tg mice) did not differ from those of WT mice. As judged by histochemical staining and immunostaining, myelin content in the cerebellum of TNF-alpha/IGF-I Tg mice was similar to that in WT mice and much more than that in TNF-alpha Tg mice. Consistently, Western immunoblot analysis showed that myelin basic protein (MBP) abundance in the cerebellum of TNF-alpha/IGF-I Tg mice was double that in TNF-alpha Tg mice. In comparison with WT mice, the number of oligodendrocytes was decreased by approximately 36% in TNF-alpha Tg mice, whereas it was increased in IGF-I Tg mice by approximately 40%. Oligodendrocyte number in TNF-alpha/IGF-I Tg mice was almost twice that in TNF-alpha Tg mice. Furthermore, IGF-I overexpression significantly reduced TNF-alpha-induced increases in apoptotic cell number, active caspase-3 abundance, and degradaion of MBP. Our results indicate that IGF-I is capable of protecting myelin and oligodendrocytes from TNF-alpha-induced damage in vivo.


Subject(s)
Demyelinating Diseases/prevention & control , Insulin-Like Growth Factor I/physiology , Tumor Necrosis Factor-alpha , Animals , Antigens/metabolism , Caspase 3/metabolism , Cell Count/methods , Cerebellum/metabolism , Cerebellum/pathology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Female , Gene Expression Regulation/physiology , Glutathione S-Transferase pi/metabolism , Immunohistochemistry , In Situ Nick-End Labeling/methods , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Transgenic , Myelin Basic Protein/metabolism , Organ Size/genetics , Proteoglycans/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...