1
|
Carver AJ, Fairbairn FM, Taylor RJ, Boggarapu S, Kamau NR, Gajmer A, Stevens HE. Placental Igf1 Overexpression Sex-Specifically Impacts Mouse Placenta Structure, Altering Offspring Striatal Development and Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.644829. [PMID: 40196637 PMCID: PMC11974794 DOI: 10.1101/2025.03.27.644829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Insulin-like growth factor 1 (IGF1) is produced primarily in the placenta in utero and is an essential hormone for neurodevelopment. Specifically, how placental IGF1 production persistently influences the brain is unclear. This study evaluated the effects of placental Igf1 overexpression on embryonic and postnatal brain development, particularly for striatum, a region highly linked to neurodevelopmental disorders. Placental Igf1 was overexpressed via placental-targeted CRISPR manipulation. This overexpression altered placenta structure and function distinctly in females and males. Early differences in placental function altered the trajectory of striatal development, as adult females showed persistent changes in striatal cell composition and striatal dependent behavior while males were less affected in brain and behavior outcomes. Overall, these results demonstrate that placental Igf1 expression alters striatal development and behavior in ways relevant to neurodevelopmental disorders. These findings expand our understanding of placental influence on neurodevelopment and will aid in identifying placental-targeted preventive interventions.
Collapse
Affiliation(s)
- Annemarie J. Carver
- Interdisciplinary Graduate Program in Genetics, University of Iowa, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Faith M. Fairbairn
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Robert J. Taylor
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Shanmukh Boggarapu
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Njenga R. Kamau
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Amrita Gajmer
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
| | - Hanna E. Stevens
- Interdisciplinary Graduate Program in Genetics, University of Iowa, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, IA, USA
- Hawk-Intellectual and Developmental Disabilities Research Center, University of Iowa, IA, USA
- Senior author
- Lead contact
| |
Collapse
|
2
|
Ye S, Wei L, Jiang Y, Yuan Y, Zeng Y, Zhu L, Xiao F. Mechanism of NO 2-induced migraine in rats: The exploration of the role of miR-653-3p/IGF1 axis. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133362. [PMID: 38157813 DOI: 10.1016/j.jhazmat.2023.133362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Migraine is a severely disabling primary neurological disorder. Although some studies have confirmed that nitrogen dioxide (NO2) pollution increases the risk of migraine, and our previous study demonstrated the role of the channel protein transient receptor potential cation channel subfamily V member 1 (TRPV1) in NO2-induced migraine, the underlying mechanisms have not been fully elucidated. This study aimed to explore the intrinsic toxicity mechanism of NO2-induced migraines using transcriptome sequencing. First, the differentially expressed genes in NO2-induced migraine, insulin-like growth factor 1 (IGF1) and miRNA miR-653-3p were identified using RNA and small RNA sequencing, and a protein interaction network was constructed using STRING to explore the possible mechanisms. Next, the targeting relationship between miR-653-3p and IGF1 was determined. NO2-induced migraine was verified by silencing miR-653-3p and IGF1, independently or in combination to regulate the protein kinase B (AKT)/TRPV1 signalling pathway through the miR-653-3p/IGF1 axis. These results indicate that the key molecular mechanism of NO2-induced migraine may be that the miR-653-3p/IGF1 axis regulates the AKT/TRPV1 signalling pathway to induce migraine. The findings of this study will further elucidate the neurotoxic mechanism of NO2-induced migraines and lay a new experimental foundation for implementing migraine-related preventive and therapeutic control measures.
Collapse
Affiliation(s)
- Shuzi Ye
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Lai Wei
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Yan Jiang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Yu Yuan
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Yuan Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Lemei Zhu
- School of Public Health, Changsha Medical University, Changsha 410219, Hunan, PR China
| | - Fang Xiao
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China.
| |
Collapse
|
3
|
Stoufflet J, Tielens S, Nguyen L. Shaping the cerebral cortex by cellular crosstalk. Cell 2023; 186:2733-2747. [PMID: 37352835 DOI: 10.1016/j.cell.2023.05.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/30/2023] [Accepted: 05/26/2023] [Indexed: 06/25/2023]
Abstract
The cerebral cortex is the brain's outermost layer. It is responsible for processing motor and sensory information that support high-level cognitive abilities and shape personality. Its development and functional organization strongly rely on cell communication that is established via an intricate system of diffusible signals and physical contacts during development. Interfering with this cellular crosstalk can cause neurodevelopmental disorders. Here, we review how crosstalk between migrating cells and their environment influences cerebral cortex development, ranging from neurogenesis to synaptogenesis and assembly of cortical circuits.
Collapse
Affiliation(s)
- Julie Stoufflet
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Stem Cells and GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, CHU Sart Tilman, Liège 4000, Belgium
| | - Sylvia Tielens
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Stem Cells and GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, CHU Sart Tilman, Liège 4000, Belgium
| | - Laurent Nguyen
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Stem Cells and GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, CHU Sart Tilman, Liège 4000, Belgium; Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavres, Belgium.
| |
Collapse
|
4
|
Wong-Kee-You AMB, Loveridge-Easther C, Mueller C, Simon N, Good WV. The impact of early exposure to general anesthesia on visual and neurocognitive development. Surv Ophthalmol 2022; 68:539-555. [PMID: 35970232 DOI: 10.1016/j.survophthal.2022.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/19/2022]
Abstract
Every year millions of children are exposed to general anesthesia while undergoing surgical and diagnostic procedures. In the field of ophthalmology, 44,000 children are exposed to general anesthesia annually for strabismus surgery alone. While it is clear that general anesthesia is necessary for sedation and pain minimization during surgical procedures, the possibility of neurotoxic impairments from its exposure is of concern. In animals there is strong evidence linking early anesthesia exposure to abnormal neural development. but in humans the effects of anesthesia are debated. In humans many aspects of vision develop within the first year of life, making the visual system vulnerable to early adverse experiences and potentially vulnerable to early exposure to general anesthesia. We attempt to address whether the visual system is affected by early postnatal exposure to general anesthesia. We first summarize key mechanisms that could account for the neurotoxic effects of general anesthesia on the developing brain and review existing literature on the effects of early anesthesia exposure on the visual system in both animals and humans and on neurocognitive development in humans. Finally, we conclude by proposing future directions for research that could address unanswered questions regarding the impact of general anesthesia on visual development.
Collapse
Affiliation(s)
| | - Cam Loveridge-Easther
- Smith-Kettlewell Eye Research Institute, San Francisco, CA, USA; University of Auckland, Auckland, New Zealand
| | - Claudia Mueller
- Sutter Health, San Francisco, CA, USA; Stanford Children's Health, Palo Alto, CA, USA
| | | | - William V Good
- Smith-Kettlewell Eye Research Institute, San Francisco, CA, USA.
| |
Collapse
|
5
|
IGF-1 as a Potential Therapy for Spinocerebellar Ataxia Type 3. Biomedicines 2022; 10:biomedicines10020505. [PMID: 35203722 PMCID: PMC8962315 DOI: 10.3390/biomedicines10020505] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 02/04/2023] Open
Abstract
Although the effects of growth hormone (GH) therapy on spinocerebellar ataxia type 3 (SCA3) have been examined in transgenic SCA3 mice, it still poses a nonnegligible risk of cancer when used for a long term. This study investigated the efficacy of IGF-1, a downstream mediator of GH, in vivo for SCA3 treatment. IGF-1 (50 mg/kg) or saline, once a week, was intraperitoneally injected to SCA3 84Q transgenic mice harboring a human ATXN3 gene with a pathogenic expanded 84 cytosine–adenine–guanine (CAG) repeat motif at 9 months of age. Compared with the control mice harboring a 15 CAG repeat motif, the SCA3 84Q mice treated with IGF-1 for 9 months exhibited the improvement only in locomotor function and minimized degeneration of the cerebellar cortex as indicated by the survival of more Purkinje cells with a more favorable mitochondrial function along with a decrease in oxidative stress caused by DNA damage. These findings could be attributable to the inhibition of mitochondrial fission, resulting in mitochondrial fusion, and decreased immunofluorescence staining in aggresome formation and ataxin-3 mutant protein levels, possibly through the enhancement of autophagy. The findings of this study show the therapeutic potential effect of IGF-1 injection for SCA3 to prevent the exacerbation of disease progress.
Collapse
|
6
|
Consorti A, Di Marco I, Sansevero G. Physical Exercise Modulates Brain Physiology Through a Network of Long- and Short-Range Cellular Interactions. Front Mol Neurosci 2021; 14:710303. [PMID: 34489641 PMCID: PMC8417110 DOI: 10.3389/fnmol.2021.710303] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
In the last decades, the effects of sedentary lifestyles have emerged as a critical aspect of modern society. Interestingly, recent evidence demonstrated that physical exercise plays an important role not only in maintaining peripheral health but also in the regulation of central nervous system function. Many studies have shown that physical exercise promotes the release of molecules, involved in neuronal survival, differentiation, plasticity and neurogenesis, from several peripheral organs. Thus, aerobic exercise has emerged as an intriguing tool that, on one hand, could serve as a therapeutic protocol for diseases of the nervous system, and on the other hand, could help to unravel potential molecular targets for pharmacological approaches. In the present review, we will summarize the cellular interactions that mediate the effects of physical exercise on brain health, starting from the factors released in myocytes during muscle contraction to the cellular pathways that regulate higher cognitive functions, in both health and disease.
Collapse
Affiliation(s)
- Alan Consorti
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- NEUROFARBA, University of Florence, Florence, Italy
| | | | | |
Collapse
|
7
|
The interplay of neurovasculature and adult hippocampal neurogenesis. Neurosci Lett 2021; 760:136071. [PMID: 34147540 DOI: 10.1016/j.neulet.2021.136071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/06/2021] [Accepted: 06/15/2021] [Indexed: 01/14/2023]
Abstract
The subgranular zone of the dentate gyrus provides a local microenvironment (niche) for neural stem cells. In the adult brain, it has been established that the vascular compartment of such niches has a significant role in regulating adult hippocampal neurogenesis. More recently, evidence showed that neurovascular coupling, the relationship between blood flow and neuronal activity, also regulates hippocampal neurogenesis. Here, we review the most recent articles on addressing the intricate relationship between neurovasculature and adult hippocampal neurogenesis and a novel pathway where functional hyperemia enhances hippocampal neurogenesis. In the end, we have further reviewed recent research showing that impaired neurovascular coupling may cause declined neurogenesis and contribute to brain damage in neurodegenerative diseases.
Collapse
|
8
|
Terry TT, Cheng T, Mahjoub M, Zong H. Mosaic Analysis with Double Markers reveals IGF1R function in granule cell progenitors during cerebellar development. Dev Biol 2020; 465:130-143. [PMID: 32697974 DOI: 10.1016/j.ydbio.2020.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
During cerebellar development, granule cell progenitors (GCPs) proliferate exponentially for a fixed period, promoted by paracrine mitogenic factor Sonic Hedgehog (Shh) secreted from Purkinje cells (PCs). Dysregulation of Shh signaling leads to uncontrolled GCP proliferation and medulloblastoma. Serendipitously our previous work discovered insulin-like growth factor 1 (IGF1) as another key driver for medulloblastoma, which led to the current investigation into the role of IGF1 in GCPs during normal development. While the IGF1R conditional knockout model revealed GCP defects in anterior cerebellum, the posterior cerebellum was mostly intact, likely owing to incomplete excision of floxed alleles. To circumvent this hurdle, we enlisted a mouse genetic system called Mosaic Analysis of Double Markers (MADM), which sporadically generates homozygous null cells unequivocally labeled with GFP and their wildtype sibling cells labeled with RFP, enabling phenotypic analysis at single-cell resolution. Using MADM, we found that loss of IGF1R resulted in a 10-fold reduction of GCs in both anterior and posterior cerebellum; and that hindered S phase entry and increased cell cycle exit collectively led to this phenotype. Genetic interaction studies showed that IGF1 signaling prevents GCP cell cycle exit at least partially through suppressing the level of p27kip1, a negative regulator of cell cycle. Finally, we found that IGF1 is produced by PCs in a temporally regulated fashion: it is highly expressed early in development when GCPs proliferate exponentially, then gradually decline as GCPs commit to cell cycle exit. Taken together, our studies reveal IGF1 as a paracrine factor that positively regulates GCP cell cycle in cooperation with Shh, through dampening the level of p27 to prevent precocious cell cycle exit. Our work not only showcases the power of phenotypic analysis by the MADM system but also provides an excellent example of multi-factorial regulation of robust developmental programs.
Collapse
Affiliation(s)
- Tiffany T Terry
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Tao Cheng
- Department of Medicine, Division of Nephrology, Washington University in St. Louis, St. Louis, MO, USA
| | - Moe Mahjoub
- Department of Medicine, Division of Nephrology, Washington University in St. Louis, St. Louis, MO, USA
| | - Hui Zong
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
9
|
Kim B, Elzinga SE, Henn RE, McGinley LM, Feldman EL. The effects of insulin and insulin-like growth factor I on amyloid precursor protein phosphorylation in in vitro and in vivo models of Alzheimer's disease. Neurobiol Dis 2019; 132:104541. [PMID: 31349033 DOI: 10.1016/j.nbd.2019.104541] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/15/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a growing problem worldwide, and there are currently no effective treatments for this devastating disease. The neurotrophic growth factors insulin and insulin-like growth factor-I (IGF-I) are currently being investigated as potential therapeutic approaches for AD in preclinical and clinical studies. However, given that the metabolic syndrome (MetS) and diabetes are risk factors for AD, it is unknown how associated insulin resistance (IR) in the brain may impact the effectiveness of these therapies for AD. In this report, we therefore investigated the mechanisms underlying the effects of insulin and IGF-I on AD-associated pathology in the context of IR, with particular emphasis on phosphorylation of amyloid precursor protein (APP), a key step in promoting amyloid plaque formation in AD. Both insulin and IGF-I decreased APP phosphorylation in cultured primary cortical neurons, supporting their therapeutic use in AD. Induction of IR blocked the beneficial effect of insulin and reduced the effect of IGF-I on APP dephosphorylation. These effects were mediated by the phosphatidylinositol 3-kinase (PI3-K)/protein kinase B (Akt) pathway, as inhibition of this pathway during IR restored the effect of IGF-I on APP dephosphorylation. Finally, we explored the translational relevance of these results in vivo by demonstrating that high fat diet fed mice, a robust model of IR and MetS, exhibited the expected increased brain APP phosphorylation. Overall, these data suggest that the beneficial therapeutic effect of insulin and IGF-I on APP phosphorylation is negatively impacted by IR, and suggest that insulin and IGF-I alone may not be appropriate therapies for AD patients with IR, MetS, or diabetes.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Sarah E Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Rosemary E Henn
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America.
| |
Collapse
|
10
|
Hu T, Lu MN, Chen B, Tong J, Mao R, Li SS, Dai P, Tan YX, Xiyang YB. Electro-acupuncture-induced neuroprotection is associated with activation of the IGF-1/PI3K/Akt pathway following adjacent dorsal root ganglionectomies in rats. Int J Mol Med 2018; 43:807-820. [PMID: 30569108 PMCID: PMC6317683 DOI: 10.3892/ijmm.2018.4035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/17/2018] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to investigate the putative role and underlying mechanisms of insulin-like growth factor 1 (IGF-1) in mediating neuroplasticity in rats subjected to partial dorsal root ganglionectomies following electro-acupuncture (EA) treatment. The rats underwent bilateral removal of the L1-L4 and L6 dorsal root ganglia (DRG), sparing the L5 DRG, and were subsequently subjected to 28 days of EA treatment at two paired acupoints, zusanli (ST 36)-xuanzhong (GB 39) and futu (ST 32)-sanyinjiao (SP 6), as the EA Model group. Rats that received partial dorsal root ganglionectomies without EA treatment served as a control (Model group). Subsequently, herpes simplex virus (HSV)-IGF-1, HSV-small interfering (si) RNA-IGF-1 and the associated control vectors were injected into the L5 DRG of rats in the EA Model group. HSV-IGF-1 transfection enhanced EA-induced neuroplasticity, which manifested as partial recovery in locomotor function, remission hyperpathia, growth of DRG-derived spared fibers, increased expression of phosphorylated (p-) phosphatidylinositol 3-kinase (PI3K) and Akt, and increased pPI3K/PI3K and pAkt/Akt expression ratios. By contrast, HSV-siRNA-IGF-1 treatment attenuated these effects induced by HSV-IGF-1 transfection. The results additionally demonstrated that HSV-IGF-1 transfection augmented the outgrowth of neurites in cultured DRG neurons, and interference of the expression of IGF-1 retarded neurite outgrowth. Co-treatment with a PI3K inhibitor or Akt siRNA inhibited the aforementioned effects induced by the overexpression of IGF-1. In conclusion, the results of the present study demonstrated the crucial roles of IGF-1 in EA-induced neuroplasticity following adjacent dorsal root ganglionectomies in rats via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Tao Hu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Min-Nan Lu
- Experiment Center for Medical Science Research, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Bo Chen
- Experiment Center for Medical Science Research, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Jun Tong
- Physical Education Department, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Rui Mao
- School of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Shan-Shan Li
- Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Pin Dai
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Ya-Xin Tan
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
11
|
Attaai A, Neidert N, von Ehr A, Potru PS, Zöller T, Spittau B. Postnatal maturation of microglia is associated with alternative activation and activated TGFβ signaling. Glia 2018; 66:1695-1708. [PMID: 29575117 DOI: 10.1002/glia.23332] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022]
Abstract
Microglia are involved in a widespread set of physiological and pathological processes and further play important roles during neurodevelopmental events. Postnatal maturation of microglia has been associated with the establishment of microglia-specific gene expression patterns. The mechanisms governing microglia maturation are only partially understood but Tgfβ1 has been suggested to be one important mediator. In the present study, we demonstrate that early postnatal microglia maturation is associated with alternative microglia activation, increased engulfment of apoptotic cells as well as activated microglial Tgfβ signaling. Interestingly, microglial Tgfβ signaling preceded the induction of the microglia-specific gene expression indicating the importance of Tgfβ1 for postnatal microglia maturation. Moreover, we provide evidence that Tgfβ1 is expressed by neurons in postnatal and adult brains defining neuron-microglia communication via Tgfβ1 as an important event. Finally, we introduce the recently identified microglia marker Tmem119 as a direct Tgfβ1-Smad2 target gene. Taken together, the data presented here further increase the understanding of Tgfβ1-mediated effects in microglia and place emphasis on the importance of Tgfβ1 for microglia maturation and maintenance.
Collapse
Affiliation(s)
- Abdelraheim Attaai
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University Freiburg, Freiburg, Germany.,Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Nicolas Neidert
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander von Ehr
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Phani Sankar Potru
- Institute of Anatomy, University of Rostock, Germany.,Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tanja Zöller
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Björn Spittau
- Institute of Anatomy, University of Rostock, Germany.,Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Pfisterer U, Khodosevich K. Neuronal survival in the brain: neuron type-specific mechanisms. Cell Death Dis 2017; 8:e2643. [PMID: 28252642 PMCID: PMC5386560 DOI: 10.1038/cddis.2017.64] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/24/2017] [Accepted: 01/31/2017] [Indexed: 12/19/2022]
Abstract
Neurogenic regions of mammalian brain produce many more neurons that will eventually survive and reach a mature stage. Developmental cell death affects both embryonically produced immature neurons and those immature neurons that are generated in regions of adult neurogenesis. Removal of substantial numbers of neurons that are not yet completely integrated into the local circuits helps to ensure that maturation and homeostatic function of neuronal networks in the brain proceed correctly. External signals from brain microenvironment together with intrinsic signaling pathways determine whether a particular neuron will die. To accommodate this signaling, immature neurons in the brain express a number of transmembrane factors as well as intracellular signaling molecules that will regulate the cell survival/death decision, and many of these factors cease being expressed upon neuronal maturation. Furthermore, pro-survival factors and intracellular responses depend on the type of neuron and region of the brain. Thus, in addition to some common neuronal pro-survival signaling, different types of neurons possess a variety of 'neuron type-specific' pro-survival constituents that might help them to adapt for survival in a certain brain region. This review focuses on how immature neurons survive during normal and impaired brain development, both in the embryonic/neonatal brain and in brain regions associated with adult neurogenesis, and emphasizes neuron type-specific mechanisms that help to survive for various types of immature neurons. Importantly, we mainly focus on in vivo data to describe neuronal survival specifically in the brain, without extrapolating data obtained in the PNS or spinal cord, and thus emphasize the influence of the complex brain environment on neuronal survival during development.
Collapse
Affiliation(s)
- Ulrich Pfisterer
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Wrigley S, Arafa D, Tropea D. Insulin-Like Growth Factor 1: At the Crossroads of Brain Development and Aging. Front Cell Neurosci 2017; 11:14. [PMID: 28203146 PMCID: PMC5285390 DOI: 10.3389/fncel.2017.00014] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/16/2017] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor 1 (IGF1) is a polypeptide hormone structurally similar to insulin. It is central to the somatotropic axis, acting downstream of growth hormone (GH). It activates both the mitogen-activated protein (MAP) kinase and PI3K signaling pathways, acting in almost every tissue in the body to promote tissue growth and maturation through upregulation of anabolic processes. Overall GH and IGF1 signaling falls with age, suggesting that it is this reduced IGF1 activity that leads to age-related changes in organisms. However, mutations that reduce IGF1-signaling activity can dramatically extend the lifespan of organisms. Therefore, the role of IGF1 in the overall aging process is unclear. This review article will focus on the role of IGF1 in brain development and aging. The evidence points towards a role for IGF1 in neurodevelopment both prenatally and in the early post-natal period, and in plasticity and remodeling throughout life. This review article will then discuss the hallmarks of aging and cognitive decline associated with falls in IGF1 levels towards the end of life. Finally, the role of IGF1 will be discussed within the context of both neuropsychiatric disorders caused by impaired development of the nervous system, and neurodegenerative disorders associated with aging. IGF1 and its derivatives are shown to improve the symptoms of certain neuropsychiatric disorders caused by deranged neurodevelopment and these effects have been correlated with changes in the underlying biology in both in vitro and in vivo studies. On the other hand, studies looking at IGF1 in neurodegenerative diseases have been conflicting, supporting both a role for increased and decreased IGF1 signaling in the underlying pathogenesis of these diseases.
Collapse
Affiliation(s)
- Sarah Wrigley
- School of Medicine, Trinity College Dublin Dublin, Ireland
| | - Donia Arafa
- School of Medicine, Trinity College Dublin Dublin, Ireland
| | - Daniela Tropea
- Neuropsychiatric Genetics, Trinity Translational Medicine Institute St. James HospitalDublin, Ireland; Institute of Neuroscience, Trinity College DublinDublin, Ireland
| |
Collapse
|
14
|
Rauskolb S, Dombert B, Sendtner M. Insulin-like growth factor 1 in diabetic neuropathy and amyotrophic lateral sclerosis. Neurobiol Dis 2017; 97:103-113. [DOI: 10.1016/j.nbd.2016.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/29/2016] [Accepted: 04/29/2016] [Indexed: 12/12/2022] Open
|
15
|
Kobayashi K, Sano H, Kato S, Kuroda K, Nakamuta S, Isa T, Nambu A, Kaibuchi K, Kobayashi K. Survival of corticostriatal neurons by Rho/Rho-kinase signaling pathway. Neurosci Lett 2016; 630:45-52. [PMID: 27424794 DOI: 10.1016/j.neulet.2016.07.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/07/2016] [Accepted: 07/13/2016] [Indexed: 01/10/2023]
Abstract
Developing cortical neurons undergo a number of sequential developmental events including neuronal survival/apoptosis, and the molecular mechanism underlying each characteristic process has been studied in detail. However, the survival pathway of cortical neurons at mature stages remains largely uninvestigated. We herein focused on mature corticostriatal neurons because of their important roles in various higher brain functions and the spectrum of neurological and neuropsychiatric disorders. The small GTPase Rho is known to control diverse and essential cellular functions through some effector molecules, including Rho-kinase, during neural development. In the present study, we investigated the role of Rho signaling through Rho-kinase in the survival of corticostriatal neurons. We performed the conditional expression of Clostridium botulinum C3 ADP-ribosyltransferase (C3 transferase) or dominant-negative form for Rho-kinase (Rho-K DN), a well-known inhibitor of Rho or Rho-kinase, respectively, in corticostriatal neurons using a dual viral vector approach combining a neuron-specific retrograde gene transfer lentiviral vector and an adeno-associated virus vector. C3 transferase markedly decreased the number of corticostriatal neurons, which was attributed to caspase-3-dependent enhanced apoptosis. In addition, Rho-K DN produced phenotypic defects similar to those caused by C3 transferase. These results indicate that the Rho/Rho-kinase signaling pathway plays a crucial role in the survival of corticostriatal neurons.
Collapse
Affiliation(s)
- Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan.
| | - Hiromi Sano
- SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan; Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Keisuke Kuroda
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tadashi Isa
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan; Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Atsushi Nambu
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan; Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| |
Collapse
|
16
|
Dyer AH, Vahdatpour C, Sanfeliu A, Tropea D. The role of Insulin-Like Growth Factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience 2016; 325:89-99. [DOI: 10.1016/j.neuroscience.2016.03.056] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/29/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
|
17
|
Nieto-Estévez V, Defterali Ç, Vicario-Abejón C. IGF-I: A Key Growth Factor that Regulates Neurogenesis and Synaptogenesis from Embryonic to Adult Stages of the Brain. Front Neurosci 2016; 10:52. [PMID: 26941597 PMCID: PMC4763060 DOI: 10.3389/fnins.2016.00052] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/05/2016] [Indexed: 12/28/2022] Open
Abstract
The generation of neurons in the adult mammalian brain requires the activation of quiescent neural stem cells (NSCs). This activation and the sequential steps of neuron formation from NSCs are regulated by a number of stimuli, which include growth factors. Insulin-like growth factor-I (IGF-I) exert pleiotropic effects, regulating multiple cellular processes depending on their concentration, cell type, and the developmental stage of the animal. Although IGF-I expression is relatively high in the embryonic brain its levels drop sharply in the adult brain except in neurogenic regions, i.e., the hippocampus (HP) and the subventricular zone-olfactory bulb (SVZ-OB). By contrast, the expression of IGF-IR remains relatively high in the brain irrespective of the age of the animal. Evidence indicates that IGF-I influences NSC proliferation and differentiation into neurons and glia as well as neuronal maturation including synapse formation. Furthermore, recent studies have shown that IGF-I not only promote adult neurogenesis by regulating NSC number and differentiation but also by influencing neuronal positioning and migration as described during SVZ-OB neurogenesis. In this article we will revise and discuss the actions reported for IGF-I signaling in a variety of in vitro and in vivo models, focusing on the maintenance and proliferation of NSCs/progenitors, neurogenesis, and neuron integration in synaptic circuits.
Collapse
Affiliation(s)
- Vanesa Nieto-Estévez
- Consejo Superior de Investigaciones Científicas (CSIC), Instituto CajalMadrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)Madrid, Spain
| | - Çağla Defterali
- Consejo Superior de Investigaciones Científicas (CSIC), Instituto CajalMadrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)Madrid, Spain
| | - Carlos Vicario-Abejón
- Consejo Superior de Investigaciones Científicas (CSIC), Instituto CajalMadrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)Madrid, Spain
| |
Collapse
|
18
|
McGinley LM, Sims E, Lunn JS, Kashlan ON, Chen KS, Bruno ES, Pacut CM, Hazel T, Johe K, Sakowski SA, Feldman EL. Human Cortical Neural Stem Cells Expressing Insulin-Like Growth Factor-I: A Novel Cellular Therapy for Alzheimer's Disease. Stem Cells Transl Med 2016; 5:379-91. [PMID: 26744412 PMCID: PMC4807660 DOI: 10.5966/sctm.2015-0103] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/19/2015] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent age-related neurodegenerative disorder and a leading cause of dementia. Current treatment fails to modify underlying disease pathologies and very little progress has been made to develop effective drug treatments. Cellular therapies impact disease by multiple mechanisms, providing increased efficacy compared with traditional single-target approaches. In amyotrophic lateral sclerosis, we have shown that transplanted spinal neural stem cells (NSCs) integrate into the spinal cord, form synapses with the host, improve inflammation, and reduce disease-associated pathologies. Our current goal is to develop a similar "best in class" cellular therapy for AD. Here, we characterize a novel human cortex-derived NSC line modified to express insulin-like growth factor-I (IGF-I), HK532-IGF-I. Because IGF-I promotes neurogenesis and synaptogenesis in vivo, this enhanced NSC line offers additional environmental enrichment, enhanced neuroprotection, and a multifaceted approach to treating complex AD pathologies. We show that autocrine IGF-I production does not impact the cell secretome or normal cellular functions, including proliferation, migration, or maintenance of progenitor status. However, HK532-IGF-I cells preferentially differentiate into gamma-aminobutyric acid-ergic neurons, a subtype dysregulated in AD; produce increased vascular endothelial growth factor levels; and display an increased neuroprotective capacity in vitro. We also demonstrate that HK532-IGF-I cells survive peri-hippocampal transplantation in a murine AD model and exhibit long-term persistence in targeted brain areas. In conclusion, we believe that harnessing the benefits of cellular and IGF-I therapies together will provide the optimal therapeutic benefit to patients, and our findings support further preclinical development of HK532-IGF-I cells into a disease-modifying intervention for AD.
Collapse
Affiliation(s)
- Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Erika Sims
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - J Simon Lunn
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Osama N Kashlan
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin S Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Elizabeth S Bruno
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Crystal M Pacut
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Tom Hazel
- Neuralstem, Inc., Germantown, Maryland, USA
| | - Karl Johe
- Neuralstem, Inc., Germantown, Maryland, USA
| | - Stacey A Sakowski
- A. Alfred Taubman Medical Research Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA A. Alfred Taubman Medical Research Institute, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Insulin-Like Growth Factor-1 Receptor Is Differentially Distributed in Developing Cerebellar Cortex of Rats Born to Diabetic Mothers. J Mol Neurosci 2015; 58:221-32. [DOI: 10.1007/s12031-015-0661-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 09/30/2015] [Indexed: 01/04/2023]
|
20
|
Lu-Nguyen NB, Broadstock M, Yáñez-Muñoz RJ. Efficient Expression of Igf-1 from Lentiviral Vectors Protects In Vitro but Does Not Mediate Behavioral Recovery of a Parkinsonian Lesion in Rats. Hum Gene Ther 2015. [PMID: 26222254 DOI: 10.1089/hum.2015.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gene therapy approaches delivering neurotrophic factors have offered promising results in both preclinical and clinical trials of Parkinson's disease (PD). However, failure of glial cell line-derived neurotrophic factor in phase 2 clinical trials has sparked a search for other trophic factors that may retain efficacy in the clinic. Direct protein injections of one such factor, insulin-like growth factor (IGF)-1, in a rodent model of PD has demonstrated impressive protection of dopaminergic neurons against 6-hydroxydopamine (6-OHDA) toxicity. However, protein infusion is associated with surgical risks, pump failure, and significant costs. We therefore used lentiviral vectors to deliver Igf-1, with a particular focus on the novel integration-deficient lentiviral vectors (IDLVs). A neuron-specific promoter, from the human synapsin 1 gene, excellent for gene expression from IDLVs, was additionally used to enhance Igf-1 expression. An investigation of neurotrophic effects on primary rat neuronal cultures demonstrated that neurons transduced with IDLV-Igf-1 vectors had complete protection on withdrawal of exogenous trophic support. Striatal transduction of such vectors into 6-OHDA-lesioned rats, however, provided neither protection of dopaminergic substantia nigra neurons nor improvement of animal behavior.
Collapse
Affiliation(s)
- Ngoc B Lu-Nguyen
- School of Biological Sciences, Royal Holloway, University of London , Egham, United Kingdom
| | - Martin Broadstock
- School of Biological Sciences, Royal Holloway, University of London , Egham, United Kingdom
| | - Rafael J Yáñez-Muñoz
- School of Biological Sciences, Royal Holloway, University of London , Egham, United Kingdom
| |
Collapse
|
21
|
The effects of testosterone and insulin-like growth factor 1 on motor system form and function. Exp Gerontol 2015; 64:81-6. [PMID: 25681641 DOI: 10.1016/j.exger.2015.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/31/2015] [Accepted: 02/10/2015] [Indexed: 12/25/2022]
Abstract
In this perspective article, we review the effects of selected anabolic hormones on the motoric system and speculate on the role these hormones may have on influencing muscle and physical function via their impact on the nervous system. Both muscle strength and anabolic hormone levels decline around middle age into old age over a similar time period, and several animal and human studies indicate that exogenously increasing anabolic hormones (e.g., testosterone and insulin-like growth factor-1 (IGF-1)) in aged subjects is positively associated with improved muscle strength. While most studies in humans have focused on the effects of anabolic hormones on muscle growth, few have considered the impact these hormones have on the motoric system. However, data from animals demonstrate that administering either testosterone or IGF-1 to cells of the central and peripheral motor system can increase cell excitability, attenuate atrophic changes, and improve regenerative capacity of motor neurons. While these studies do not directly indicate that changes in anabolic hormones contribute to reduced human performance in the elderly (e.g., muscle weakness and physical limitations), they do suggest that additional research is warranted along these lines.
Collapse
|
22
|
Vargas-Martínez F, Uvnäs-Moberg K, Petersson M, Olausson HA, Jiménez-Estrada I. Neuropeptides as neuroprotective agents: Oxytocin a forefront developmental player in the mammalian brain. Prog Neurobiol 2014; 123:37-78. [DOI: 10.1016/j.pneurobio.2014.10.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 02/07/2023]
|
23
|
Jing YH, Song YF, Yao YM, Yin J, Wang DG, Gao LP. Retardation of fetal dendritic development induced by gestational hyperglycemia is associated with brain insulin/IGF-I signals. Int J Dev Neurosci 2014; 37:15-20. [PMID: 24953263 DOI: 10.1016/j.ijdevneu.2014.06.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/12/2014] [Accepted: 06/12/2014] [Indexed: 01/26/2023] Open
Abstract
Hyperglycemia is an essential risk factor for mothers and fetuses in gestational diabetes. Clinical observation has indicated that the offspring of mothers with diabetes shows impaired somatosensory function and IQ. However, only a few studies have explored the effects of hyperglycemia on fetal brain development. Neurodevelopment is susceptible to environmental conditions. Thus, this study aims to investigate the effects of maternal hyperglycemia on fetal brain development and to evaluate insulin and insulin-like growth factor-I (IGF-I) signals in fetal brain under hyperglycemia or controlled hyperglycemia. At day 1 of pregnancy, gestational rats were intraperitoneally injected with streptozocin (60 mg/kg). Some of the hyperglycemic gestational rats were injected with insulin (20 IU, two times a day) to control hyperglycemia; the others were injected with saline of equal volume. The gestational rats were sacrificed at days 14, 16, and 18 of embryo development. The dendritic spines of subplate cortex neurons in the fetal brain were detected by Golgi-Cox staining. The mRNA levels of insulin receptors (IRs) and IGF-IR in the fetal brain were measured using qRT-PCR. The protein levels of synaptophysin, IR, and IGF-IR in the fetal brain were detected by western blot. No significant difference in fetal brain formation was observed between the maternal hyperglycemic group and insulin-treated group. By contrast, obvious retardation of dendritic development in the fetus was observed in the maternal hyperglycemic group. Similarly, synaptophysin expression was lower in the fetus of the maternal hyperglycemic group than in that of the insulin-treated group. The mRNA and protein expression levels of IRs in the fetal brain were higher in the hyperglycemic group than in the insulin-treated group. By contrast, the levels of IGF-IR in the brain were lower in the fetus of the maternal hyperglycemic group than in that of the insulin-treated group. These results suggested that maternal hyperglycemia can retard dendritic development in the fetal brain and that these changes partially resulted from abnormal insulin/IGF-I signaling in the fetal brain.
Collapse
Affiliation(s)
- Yu-Hong Jing
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, PR China.
| | - Yan-Feng Song
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Ya-Ming Yao
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Jie Yin
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - De-Gui Wang
- Institute of Anatomy and Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China
| |
Collapse
|
24
|
Nikolić M, Gardner H, Tucker K. Postnatal neuronal apoptosis in the cerebral cortex: Physiological and pathophysiological mechanisms. Neuroscience 2013; 254:369-78. [DOI: 10.1016/j.neuroscience.2013.09.035] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 09/14/2013] [Accepted: 09/17/2013] [Indexed: 12/15/2022]
|
25
|
Yang X, Wei A, Liu Y, He G, Zhou Z, Yu Z. IGF-1 protects retinal ganglion cells from hypoxia-induced apoptosis by activating the Erk-1/2 and Akt pathways. Mol Vis 2013; 19:1901-12. [PMID: 24049436 PMCID: PMC3774573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/09/2013] [Indexed: 11/01/2022] Open
Abstract
PURPOSE Hypoxia-induced retinal ganglion cell (RGC) apoptosis has been implicated in many optic neuropathies. Insulin-like growth factor-1 (IGF-1) is important in maintaining neuronal survival, proliferation, and differentiation. The purpose of this study is to explore whether IGF-1 can protect RGCs from hypoxia-induced apoptosis and to determine the precise mechanisms that regulate this process. METHODS Purified RGC cultures were obtained from the retinas of neonatal Sprague Dawley (SD) rats using a two-step panning method. Primary cultured RGCs were cultured in a closed hypoxic chamber (5% O2, 5% CO2, and 90% N2) for 12 h with or without IGF-1. The degree of apoptosis in the RGCs was detected by caspase-3 expression and TUNEL and JC-1 staining assays. The expression and phosphorylation of protein kinase B (Akt), p44/42 mitogen-activated protein kinase (MAPK) (extracellular signal-regulated kinase-1/2 [Erk-1/2]), Bad, and caspase-3 was investigated with immunoblot analysis. RESULTS Hypoxia induces apoptosis in primary Sprague Dawley rat RGCs, as detected by caspase-3 expression and TUNEL and JC-1 staining assays, and that IGF-1 treatment could significantly reduce this effect in RGCs. Interestingly, pretreatment of RGCs with AG1024 (an IGF-1 inhibitor), U0126 (an Erk-1/2 inhibitor), and LY294002 (an Akt inhibitor) markedly attenuated the effects of IGF-1 treatment. Furthermore, western blot analysis suggested that the Erk-1/2 and Akt signaling pathways play a role in the protective effects of IGF-1 on RGCs exposed to hypoxia. CONCLUSIONS These data indicate that IGF-1 can protect primary cultured RGCs against hypoxia-induced apoptosis via the Erk-1/2 and Akt signaling pathways, suggesting that IGF-1 treatment is a potential therapeutic approach for treating hypoxia-induced neurodegeneration in the retina.
Collapse
Affiliation(s)
- Xuesen Yang
- Department of Occupational Health, Third Military Medical University, Chongqing, China,Institute of Tropical Medicine, Third Military Medical University, Chongqing, China
| | - Aimin Wei
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Yong Liu
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University, Chongqing, China
| | - Genlin He
- Institute of Tropical Medicine, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| |
Collapse
|
26
|
Yan Y, Li X, Kover K, Clements M, Ye P. CREB participates in the IGF-I-stimulation cyclin D1 transcription. Dev Neurobiol 2013; 73:559-70. [DOI: 10.1002/dneu.22080] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/04/2013] [Accepted: 02/21/2013] [Indexed: 01/13/2023]
Affiliation(s)
- Yun Yan
- Department of Pediatrics; Division of Endocrinology; University of North Carolina at Chapel Hill; Chapel Hill; North Carolina
| | - Xiaoyu Li
- Department of Pediatrics, Children's Mercy Hospitals & Clinics; University of Missouri-Kansas City; Kansas City; Missouri
| | - Karen Kover
- Department of Pediatrics, Children's Mercy Hospitals & Clinics; University of Missouri-Kansas City; Kansas City; Missouri
| | - Mark Clements
- Department of Pediatrics, Children's Mercy Hospitals & Clinics; University of Missouri-Kansas City; Kansas City; Missouri
| | - Ping Ye
- Department of Pediatrics; Division of Endocrinology; University of North Carolina at Chapel Hill; Chapel Hill; North Carolina
| |
Collapse
|
27
|
Istaphanous GK, Ward CG, Nan X, Hughes EA, McCann JC, McAuliffe JJ, Danzer SC, Loepke AW. Characterization and quantification of isoflurane-induced developmental apoptotic cell death in mouse cerebral cortex. Anesth Analg 2013; 116:845-54. [PMID: 23460572 DOI: 10.1213/ane.0b013e318281e988] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Accumulating evidence indicates that isoflurane and other, similarly acting anesthetics exert neurotoxic effects in neonatal animals. However, neither the identity of dying cortical cells nor the extent of cortical cell loss has been sufficiently characterized. We conducted the present study to immunohistochemically identify the dying cells and to quantify the fraction of cells undergoing apoptotic death in neonatal mouse cortex, a substantially affected brain region. METHODS Seven-day-old littermates (n = 36) were randomly assigned to a 6-hour exposure to either 1.5% isoflurane or fasting in room air. Animals were euthanized immediately after exposure and brain sections were double-stained for activated caspase 3 and one of the following cellular markers: Neuronal Nuclei (NeuN) for neurons, glutamic acid decarboxylase (GAD)65 and GAD67 for GABAergic cells, as well as GFAP (glial fibrillary acidic protein) and S100β for astrocytes. RESULTS In 7-day-old mice, isoflurane exposure led to widespread increases in apoptotic cell death relative to controls, as measured by activated caspase 3 immunolabeling. Confocal analyses of caspase 3-labeled cells in cortical layers II and III revealed that the overwhelming majority of cells were postmitotic neurons, but some were astrocytes. We then quantified isoflurane-induced neuronal apoptosis in visual cortex, an area of substantial injury. In unanesthetized control animals, 0.08% ± 0.001% of NeuN-positive layer II/III cortical neurons were immunoreactive for caspase 3. By contrast, the rate of apoptotic NeuN-positive neurons increased at least 11-fold (lower end of the 95% confidence interval [CI]) to 2.0% ± 0.004% of neurons immediately after isoflurane exposure (P = 0.0017 isoflurane versus control). In isoflurane-treated animals, 2.9% ± 0.02% of all caspase 3-positive neurons in superficial cortex also coexpressed GAD67, indicating that inhibitory neurons may also be affected. Analysis of GABAergic neurons, however, proved unexpectedly complex. In addition to inducing apoptosis among some GAD67-immunoreactive neurons, anesthesia also coincided with a dramatic decrease in both GAD67 (0.98 vs 1.84 ng/mg protein, P < 0.00001, anesthesia versus control) and GAD65 (2.25 ± 0.74 vs 23.03 ± 8.47 ng/mg protein, P = 0.0008, anesthesia versus control) protein levels. CONCLUSIONS Prolonged exposure to isoflurane increased neuronal apoptotic cell death in 7-day-old mice, eliminating approximately 2% of cortical neurons, of which some were identified as GABAergic interneurons. Moreover, isoflurane exposure interfered with the inhibitory nervous system by downregulating the central enzymes GAD65 and GAD67. Conversely, at this age, only a minority of degenerating cells were identified as astrocytes. The clinical relevance of these findings in animals remains to be determined.
Collapse
Affiliation(s)
- George K Istaphanous
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Puche JE, Castilla-Cortázar I. Human conditions of insulin-like growth factor-I (IGF-I) deficiency. J Transl Med 2012; 10:224. [PMID: 23148873 PMCID: PMC3543345 DOI: 10.1186/1479-5876-10-224] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 11/07/2012] [Indexed: 12/13/2022] Open
Abstract
Insulin-like growth factor I (IGF-I) is a polypeptide hormone produced mainly by the liver in response to the endocrine GH stimulus, but it is also secreted by multiple tissues for autocrine/paracrine purposes. IGF-I is partly responsible for systemic GH activities although it possesses a wide number of own properties (anabolic, antioxidant, anti-inflammatory and cytoprotective actions). IGF-I is a closely regulated hormone. Consequently, its logical therapeutical applications seems to be limited to restore physiological circulating levels in order to recover the clinical consequences of IGF-I deficiency, conditions where, despite continuous discrepancies, IGF-I treatment has never been related to oncogenesis. Currently the best characterized conditions of IGF-I deficiency are Laron Syndrome, in children; liver cirrhosis, in adults; aging including age-related-cardiovascular and neurological diseases; and more recently, intrauterine growth restriction. The aim of this review is to summarize the increasing list of roles of IGF-I, both in physiological and pathological conditions, underlying that its potential therapeutical options seem to be limited to those proven states of local or systemic IGF-I deficiency as a replacement treatment, rather than increasing its level upper the normal range.
Collapse
Affiliation(s)
- Juan E Puche
- Applied Molecular Medicine Institute (IMMA), School of Medicine, Department of Medical Physiology, Universidad CEU San Pablo, Madrid, Spain
| | - Inma Castilla-Cortázar
- Applied Molecular Medicine Institute (IMMA), School of Medicine, Department of Medical Physiology, Universidad CEU San Pablo, Madrid, Spain
| |
Collapse
|
29
|
O’Kusky J, Ye P. Neurodevelopmental effects of insulin-like growth factor signaling. Front Neuroendocrinol 2012; 33:230-51. [PMID: 22710100 PMCID: PMC3677055 DOI: 10.1016/j.yfrne.2012.06.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/09/2012] [Accepted: 06/07/2012] [Indexed: 11/28/2022]
Abstract
Insulin-like growth factor (IGF) signaling greatly impacts the development and growth of the central nervous system (CNS). IGF-I and IGF-II, two ligands of the IGF system, exert a wide variety of actions both during development and in adulthood, promoting the survival and proliferation of neural cells. The IGFs also influence the growth and maturation of neural cells, augmenting dendritic growth and spine formation, axon outgrowth, synaptogenesis, and myelination. Specific IGF actions, however, likely depend on cell type, developmental stage, and local microenvironmental milieu within the brain. Emerging research also indicates that alterations in IGF signaling likely contribute to the pathogenesis of some neurological disorders. This review summarizes experimental studies and shed light on the critical roles of IGF signaling, as well as its mechanisms, during CNS development.
Collapse
Affiliation(s)
- John O’Kusky
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada V5Z 1M9
| | - Ping Ye
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
30
|
Lee KY, Miki T, Yokoyama T, Ueki M, Warita K, Suzuki S, Ohta KI, Wang ZY, Jamal M, Yakura T, Liu JQ, Hosomi N, Takeuchi Y. Neonatal repetitive maternal separation causes long-lasting alterations in various neurotrophic factor expression in the cerebral cortex of rats. Life Sci 2012; 90:578-84. [PMID: 22365961 DOI: 10.1016/j.lfs.2012.01.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/13/2012] [Accepted: 01/27/2012] [Indexed: 01/08/2023]
Abstract
AIMS This study was carried out to examine the effects of early postnatal maternal separation stress on the development of the cerebral cortex with respect to time-dependent fluctuations of neurotrophic factor ligand and receptor expression. MAIN METHODS Wistar rats were separated from their mothers for 3h per day during postnatal days (PND) 10 to 15. The cerebral cortex was analyzed by real-time RT-PCR for the evaluation of the expression of mRNA for brain-derived neurotrophic factor (BDNF), TrkB, insulin-like growth factor-1 (IGF-1), and type 1 IGF receptor (IGF-1R) on PND16, 20, 30, and 60. KEY FINDINGS The expression of these neurotrophic factor ligands and receptors in the cerebral cortex was enhanced on PND16 and PND20, and then it returned to baseline levels on PND30. By PND60, however, the expression levels were attenuated. SIGNIFICANCE The important implication of this study is the persistent abnormal fluctuation of neurotrophic factor expression for a prolonged period, triggered even after the brain growth spurt. Given that neurotrophic factors play important roles in brain development, it can be speculated that the altered expression of these factors induced by maternal separation may interrupt normal brain development and ultimately lead to functional disruption. However, the possibility of such changes leading to various functional disruptions and the underlying mechanisms involved require further study.
Collapse
Affiliation(s)
- Kyoung-Youl Lee
- Department of Health Science, Kongju National University, Chungnam, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
West EL, Pearson RA, Duran Y, Gonzalez-Cordero A, MacLaren RE, Smith AJ, Sowden JC, Ali RR. Manipulation of the recipient retinal environment by ectopic expression of neurotrophic growth factors can improve transplanted photoreceptor integration and survival. Cell Transplant 2012; 21:871-87. [PMID: 22325046 DOI: 10.3727/096368911x623871] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Degeneration of the neural retina is the leading cause of untreatable blindness in the developed world. Stem cell replacement therapy offers a novel strategy for retinal repair. Postmitotic photoreceptor precursors derived from the early postnatal (P) retina are able to migrate and integrate into the adult mouse retina following transplantation into the subretinal space, but it is likely that a large number of these cells would be required to restore vision. The adult recipient retina presents a very different environment to that from which photoreceptor precursor donor cells isolated from the developing postnatal retina are derived. Here we considered the possibility that modulation of the recipient environment by ectopic expression of developmentally regulated growth factors, normally present during photoreceptor development, might enhance the migration and integration of transplanted cells into the adult neural retina. Adeno-associated viral (AAV) vectors were used to introduce three growth factors previously reported to play a role in photoreceptor development, IGF1, FGF2, and CNTF, into the adult retina, prior to transplantation of P4 cells derived from the Nrl.GFP(+ve) neural retina. At 3 weeks posttransplantation the number of integrated, differentiated photoreceptor cells present in AAV-mediated neurotrophic factor-treated eyes was assessed and compared to control treated contralateral eyes. We show, firstly, that it is possible to manipulate the recipient retinal microenvironment via rAAV-mediated gene transfer with respect to these developmentally relevant growth factors. Moreover, when combined with cell transplantation, AAV-mediated expression of IGF1 led to significantly increased levels of cell integration, while overexpression of FGF2 had no significant effect on integrated cell number. Conversely, expression of CNTF led to a significant decrease in cell integration and an exacerbated glial response that led to glial scarring. Together, these findings demonstrate the importance of the extrinsic environment of the recipient retina for photoreceptor cell transplantation and show for the first time that it is possible to manipulate this environment using viral vectors to influence photoreceptor transplantation efficiency.
Collapse
Affiliation(s)
- E L West
- Department of Genetics, University College London Institute of Ophthalmology, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Lunn JS, Pacut C, Backus C, Hong Y, Johe K, Hefferan M, Marsala M, Feldman EL. The pleotrophic effects of insulin-like growth factor-I on human spinal cord neural progenitor cells. Stem Cells Dev 2010; 19:1983-93. [PMID: 20406098 DOI: 10.1089/scd.2010.0003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Most stem cell therapies involve direct, intraparachymal placement of neural progenitor cells. These cells provide physical support to the endogenous neuronal population and may be engineered to provide in situ growth factor support. Insulin-like growth factor-I (IGF-I) has potent neurotrophic and neuroprotective properties and is expressed by human neural stem cells (hNSCs). IGF-I is implicated in multiple aspects of cell behavior, including proliferation, differentiation, and survival. Enhancing hNSC function through IGF-I overexpression may increase the benefits of stem cell therapy. As a first step to that goal, we examined the direct effects of IGF-I on hNSC behavior in vitro. We demonstrate that IGF-I treatment enhances both the number and length of hNSC neurites. This is correlated with a decrease in proliferation, suggesting that IGF-I promotes neurite outgrowth but not proliferation. While IGF-I activates both AKT and MAPK signaling in hNSCs, we demonstrate that IGF-I-mediated neurite outgrowth is dependent only on AKT signaling. Finally, we demonstrate that IGF-I is neuroprotective after glutamate exposure in a model of excitotoxic cell death.
Collapse
Affiliation(s)
- J Simon Lunn
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Prostaglandin E2-induced synaptic plasticity in neocortical networks of organotypic slice cultures. J Neurosci 2010; 30:11678-87. [PMID: 20810888 DOI: 10.1523/jneurosci.4665-09.2010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of epilepsy, yet the mechanisms underlying the progression from TBI to epilepsy are unknown. TBI induces the expression of COX-2 (cyclooxygenase-2) and increases levels of prostaglandin E2 (PGE2). Here, we demonstrate that acutely applied PGE2 (2 mum) decreases neocortical network activity by postsynaptically reducing excitatory synaptic transmission in acute and organotypic neocortical slices of mice. In contrast, long-term exposure to PGE2 (2 mum; 48 h) presynaptically increases excitatory synaptic transmission, leading to a hyperexcitable network state that is characterized by the generation of paroxysmal depolarization shifts (PDSs). PDSs were also evoked as a result of depriving organotypic slices of activity by treating them with tetrodotoxin (TTX, 1 mum; 48 h). This treatment predominantly increased postsynaptically excitatory synaptic transmission. The network and cellular effects of PGE2 and TTX treatments reversed within 1 week. Differences in the underlying mechanisms (presynaptic vs postsynaptic) as well as occlusion experiments in which slices were exposed to TTX plus PGE2 suggest that the two substances evoke distinct forms of homeostatic plasticity, both of which result in a hyperexcitable network state. PGE2 and TTX (alone or together with PGE2) also increased levels of apoptotic cell death in organotypic slices. Thus, we hypothesize that the increase in excitability and apoptosis may constitute the first steps in a cascade of events that eventually lead to epileptogenesis triggered by TBI.
Collapse
|
34
|
Ye P, Hu Q, Liu H, Yan Y, D'ercole AJ. beta-catenin mediates insulin-like growth factor-I actions to promote cyclin D1 mRNA expression, cell proliferation and survival in oligodendroglial cultures. Glia 2010; 58:1031-41. [PMID: 20235220 PMCID: PMC2917840 DOI: 10.1002/glia.20984] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
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.
Collapse
Affiliation(s)
- Ping Ye
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7039, USA.
| | | | | | | | | |
Collapse
|
35
|
Sanno H, Shen X, Kuru N, Bormuth I, Bobsin K, Gardner HAR, Komljenovic D, Tarabykin V, Erzurumlu RS, Tucker KL. Control of postnatal apoptosis in the neocortex by RhoA-subfamily GTPases determines neuronal density. J Neurosci 2010; 30:4221-31. [PMID: 20335457 PMCID: PMC2852171 DOI: 10.1523/jneurosci.3318-09.2010] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2009] [Revised: 01/25/2010] [Accepted: 01/30/2010] [Indexed: 12/26/2022] Open
Abstract
Apoptosis of neurons in the maturing neocortex has been recorded in a wide variety of mammals, but very little is known about its effects on cortical differentiation. Recent research has implicated the RhoA GTPase subfamily in the control of apoptosis in the developing nervous system and in other tissue types. Rho GTPases are important components of the signaling pathways linking extracellular signals to the cytoskeleton. To investigate the role of the RhoA GTPase subfamily in neocortical apoptosis and differentiation, we have engineered a mouse line in which a dominant-negative RhoA mutant (N19-RhoA) is expressed from the Mapt locus, such that all neurons of the developing nervous system are expressing the N19-RhoA inhibitor. Postnatal expression of N19-RhoA led to no major changes in neocortical anatomy. Six layers of the neocortex developed and barrels (whisker-related neural modules) formed in layer IV. However, the density and absolute number of neurons in the somatosensory cortex increased by 12-26% compared with wild-type littermates. This was not explained by a change in the migration of neurons during the formation of cortical layers but rather by a large decrease in the amount of neuronal apoptosis at postnatal day 5, the developmental maximum of cortical apoptosis. In addition, overexpression of RhoA in cortical neurons was seen to cause high levels of apoptosis. These results demonstrate that RhoA-subfamily members play a major role in developmental apoptosis in postnatal neocortex of the mouse but that decreased apoptosis does not alter cortical cytoarchitecture and patterning.
Collapse
Affiliation(s)
- Hitomi Sanno
- Interdisciplinary Center for Neurosciences and
- Institute of Anatomy, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Xiao Shen
- Interdisciplinary Center for Neurosciences and
- Institute of Anatomy, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Nilgün Kuru
- Department of Biology, Faculty of Education, Cumhuriyet University, TR-58140 Sivas, Turkey
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Ingo Bormuth
- Max Planck Institute for Experimental Medicine, D-37075 Göttingen, Germany
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Campus Mitte, D-10098 Berlin, Germany, and
| | - Kristin Bobsin
- Interdisciplinary Center for Neurosciences and
- Institute of Anatomy, University of Heidelberg, D-69120 Heidelberg, Germany
| | | | - Dorde Komljenovic
- Institute of Anatomy, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Victor Tarabykin
- Max Planck Institute for Experimental Medicine, D-37075 Göttingen, Germany
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Campus Mitte, D-10098 Berlin, Germany, and
| | - Reha S. Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Kerry L. Tucker
- Interdisciplinary Center for Neurosciences and
- Institute of Anatomy, University of Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
36
|
Liu W, Ye P, O'Kusky JR, D'Ercole AJ. Type 1 insulin-like growth factor receptor signaling is essential for the development of the hippocampal formation and dentate gyrus. J Neurosci Res 2010; 87:2821-32. [PMID: 19437543 DOI: 10.1002/jnr.22129] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
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.
Collapse
Affiliation(s)
- Wen Liu
- Department of Pediatrics, Division of Endocrinology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
37
|
Beresewicz M, Majewska M, Makarewicz D, Vayro S, Zabłocka B, Górecki DC. Changes in the expression of insulin-like growth factor 1 variants in the postnatal brain development and in neonatal hypoxia-ischaemia. Int J Dev Neurosci 2009; 28:91-7. [PMID: 19766709 DOI: 10.1016/j.ijdevneu.2009.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 09/07/2009] [Accepted: 09/13/2009] [Indexed: 01/08/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is a multifunctional peptide of which numerous isoforms exist. The predominant form, IGF-1Ea is involved in physiological processes while IGF-1Ec (mechano-growth factor, MGF) is expressed in response to a different set of stimuli. We have identified specific changes in the expression patterns of these IGF-1 variants in brain development in normal rats and following neonatal hypoxia-ischaemia (HI). Both IGF-1Ea and IGF-1Ec are expressed during normal postnatal brain development, albeit with highly specific temporal distributions. In contrast, HI produced increased and prolonged expression of the IGF-1Ec isoform only. Importantly, hypoxia alone stimulated the expression of IGF-1Ec as well. Thus, IGF-1Ec may play a role in HI pathology. Neonatal hypoxia-ischaemia occurs in approximately 1:4000-1:10,000 newborns and causes neurological deficits in approximately 75% of those affected. Unfortunately, no specific treatment is available. IGF-1 is known to have neuroprotective activity and its IGF-1Ec variant appears to be an endogenous protective factor in hypoxia-ischaemia. Therefore, IGF-1Ec could potentially be developed into a therapeutic modality for the attenuation or prevention of neuronal damage in this and related disorders.
Collapse
Affiliation(s)
- Małgorzata Beresewicz
- Molecular Biology Unit, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
38
|
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.
Collapse
Affiliation(s)
- A Joseph D'Ercole
- Department of Pediatrics, CB 7039, University of North Carolina, Chapel Hill, North Carolina 27599-7039, USA.
| | | |
Collapse
|
39
|
Sciara AA, Somoza GM, Arranz SE. Insulin-like growth factor-I of pejerrey, Odontesthes bonariensis: cDNA characterization, tissue distribution and expression profiles after growth hormone administration. ACTA ACUST UNITED AC 2008; 309:407-18. [PMID: 18521839 DOI: 10.1002/jez.466] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The liver production of the insulin-like growth factor-I (IGF-I) is a key factor in the endocrine control of body growth by a growth hormone. As pejerrey Odontesthes bonariensis has been reported as a fish with low growth rates in captivity, basic research on this respect is needed in order to understand it. In this context, the pejerrey IGF-I cDNA was cloned and its hepatic expression was examined in fish after recombinant pejerrey growth hormone (pjGHr) administration. The full length of IGF-I transcript showed a high sequence similarity to other teleost sequences. The tissue distribution analysis by reverse transcriptase polymerase chain reaction in adult fish revealed that IGF-I expressed ubiquitously with the highest mRNA levels in the liver, posterior intestine and brain. No alternative IGF-I mRNA was found in the liver, as it was reported for other teleosts. IGF-I transcript was measured in the liver after pjGHr in vivo stimulation by means of quantitative real-time polymerase chain reaction assays. A dose-dependent response of IGF-I mRNA was observed after pjGHr administration, and reached a six-fold IGF-I maximum increase over control group when 2.5 microg pjGH/g-body weight (bw) was injected. Temporal analysis of hepatic IGF-I mRNA level showed that administration of a single dose of pjGHr into juvenile pejerrey resulted in a significant increase (P <0.02) 9 hours post-injection (hpi). These results add novel information on the nucleotide sequence of IGF-I in Atheriniformes and demonstrate that pjGHr could promote a dramatic response in liver, increasing the IGF-I mRNA level.
Collapse
Affiliation(s)
- Andrés A Sciara
- Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Provincia de Santa Fe, Argentina
| | | | | |
Collapse
|
40
|
Sanz A, Carrero P, Pernía O, Garcia-Segura LM. Pubertal maturation modifies the regulation of insulin-like growth factor-I receptor signaling by estradiol in the rat prefrontal cortex. Dev Neurobiol 2008; 68:1018-28. [PMID: 18446778 DOI: 10.1002/dneu.20641] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The transition from adolescence to adulthood is accompanied by substantial plastic modifications in the cerebral cortex, including changes in the growth and retraction of neuronal processes and in the rate of synaptic formation and neuronal loss. Some of these plastic changes are prevented in female rats by prepubertal ovariectomy. The ovarian hormone estradiol modulates neuronal differentiation and survival and these effects are in part mediated by the interaction with insulin-like growth factor-I (IGF-I). In this study, we have explored whether the activation by estradiol of some components of IGF-I receptor signaling is altered in the prefrontal cortex during puberty. Estradiol administration to rats ovariectomized after puberty resulted, 24 h after the hormonal administration, in a sustained phosphorylation of Akt and glycogen synthase kinase 3 beta in the prefrontal cortex. However, this hormonal effect was not observed in animals ovariectomized before puberty. These findings suggest that during pubertal maturation there is a programming by ovarian hormones of the future regulatory actions of estradiol on IGF-I receptor signaling in the prefrontal cortex. The modification in the regulation of IGF-I receptor signaling by estradiol during pubertal maturation may have implications for the developmental changes occurring in the prefrontal cortex in the transition from adolescence to adulthood.
Collapse
Affiliation(s)
- Amaya Sanz
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, E-28002 Madrid, Spain
| | | | | | | |
Collapse
|
41
|
Ning H, Lin G, Fandel T, Banie L, Lue TF, Lin CS. Insulin growth factor signaling mediates neuron-like differentiation of adipose-tissue-derived stem cells. Differentiation 2008; 76:488-94. [PMID: 18021255 PMCID: PMC2908590 DOI: 10.1111/j.1432-0436.2007.00240.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Our previous study showed that adipose tissue-derived stem cells (ADSC) could be induced by isobutylmethylxanthine (IBMX) to differentiate into neuron-like cells. In the present study, ADSC were treated with IBMX in the presence or in the absence of each of eight specific inhibitors of different signaling pathways (JAK/STAT, PKA, PI3K, MEK, Wnt/Frizzled, ERK/MAPK, TGF-beta, and insulin growth factor [IGF]-I). PPP, a specific inhibitor of IGF-I signaling, was the only inhibitor that showed significant inhibition of IBMX-induced ADSC neuronal differentiation, as determined by changes in cell morphology in the initial screening. Further examination by immunofluorescence staining showed that the neuronal marker, beta-III-tubulin, was highly induced in IBMX-treated ADSC, and the induction was significantly suppressed by PPP. Western blotting, followed by densitometry showed that PPP suppressed IBMX-induced beta-III-tubulin expression by 43%, 88%, and 84% when used to treat the cells for 1, 3, and 24 hr, respectively. Treatment of ADSC with IBMX also led to the phosphorylation of IGF-I receptor at tyrosine 1136 (Y1136), as determined by immunofluorescence staining with an antibody that reacts specifically with Y1136. This effect was also abrogated by PPP. Thus, the IBMX-induced neuron-like differentiation of ADSC is mediated by IGF signaling through the phosphorylation of IGF-IR at Y1136.
Collapse
Affiliation(s)
- Hongxiu Ning
- Knuppe Molecular Urology Laboratory Department of Urology, School of Medicine, University of California San Francisco, CA 94143-0738, USA
| | | | | | | | | | | |
Collapse
|
42
|
Erickson RI, Paucar AA, Jackson RL, Visnyei K, Kornblum H. Roles of insulin and transferrin in neural progenitor survival and proliferation. J Neurosci Res 2008; 86:1884-94. [DOI: 10.1002/jnr.21631] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|