1
|
Li X, Wang T, Liu N, Cai A, Zhang J, Zhang F, Liu Q, Wang J, Wu Y, Gao K, Jiang YW. Focal cortical dysplasia II caused by brain somatic mutation of IRS-1 is associated with ERK signaling pathway activation. Cereb Cortex 2024; 34:bhae227. [PMID: 38836287 DOI: 10.1093/cercor/bhae227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Somatic mutations have been identified in 10% to 63% of focal cortical dysplasia type II samples, primarily linked to the mTOR pathway. When the causative genetic mutations are not identified, this opens the possibility of discovering new pathogenic genes or pathways that could be contributing to the condition. In our previous study, we identified a novel candidate pathogenic somatic variant of IRS-1 c.1791dupG in the brain tissue of a child with focal cortical dysplasia type II. This study further explored the variant's role in causing type II focal cortical dysplasia through in vitro overexpression in 293T and SH-SY5Y cells and in vivo evaluation via in utero electroporation in fetal brains, assessing effects on neuronal migration, morphology, and network integrity. It was found that the mutant IRS-1 variant led to hyperactivity of p-ERK, increased cell volume, and was predominantly associated with the MAPK signaling pathway. In vivo, the IRS-1 c.1791dupG variant induced abnormal neuron migration, cytomegaly, and network hyperexcitability. Notably, the ERK inhibitor GDC-0994, rather than the mTOR inhibitor rapamycin, effectively rescued the neuronal defects. This study directly highlighted the ERK signaling pathway's role in the pathogenesis of focal cortical dysplasia II and provided a new therapeutic target for cases of focal cortical dysplasia II that are not treatable by rapamycin analogs.
Collapse
Affiliation(s)
- Xiao Li
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Tianshuang Wang
- Department of Neurology, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Minhang District, Shanghai 201102, China
| | - Nana Liu
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Aojie Cai
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Junjiao Zhang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Fan Zhang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Qingzhu Liu
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| | - Yu-Wu Jiang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Children Epilepsy Center, Peking University First Hospital, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, No. 1 Xi'an Men Street, West District, Beijing 100034, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, No. 1 Xi'an Men Street, West District, Beijing 100034, China
| |
Collapse
|
2
|
Wisessaowapak C, Niyomchan A, Visitnonthachai D, Leelaprachakul N, Watcharasit P, Satayavivad J. Arsenic-induced IGF-1 signaling impairment and neurite shortening: The protective roles of IGF-1 through the PI3K/Akt axis. ENVIRONMENTAL TOXICOLOGY 2024; 39:1119-1128. [PMID: 37853848 DOI: 10.1002/tox.23995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/12/2023] [Accepted: 10/07/2023] [Indexed: 10/20/2023]
Abstract
We recently reported that arsenic caused insulin resistance in differentiated human neuroblastoma SH-SY5Y cells. Herein, we further investigated the effects of sodium arsenite on IGF-1 signaling, which shares downstream signaling with insulin. A time-course experiment revealed that sodium arsenite began to decrease IGF-1-stimulated Akt phosphorylation on Day 3 after treatment, indicating that prolonged sodium arsenite treatment disrupted the neuronal IGF-1 response. Additionally, sodium arsenite decreased IGF-1-stimulated tyrosine phosphorylation of the IGF-1 receptor β (IGF-1Rβ) and its downstream target, insulin receptor substrate 1 (IRS1). These results suggested that sodium arsenite impaired the intrinsic tyrosine kinase activity of IGF-1Rβ, ultimately resulting in a reduction in tyrosine-phosphorylated IRS1. Sodium arsenite also reduced IGF-1 stimulated tyrosine phosphorylation of insulin receptor β (IRβ), indicating the potential inhibition of IGF-1R/IR crosstalk by sodium arsenite. Interestingly, sodium arsenite also induced neurite shortening at the same concentrations that caused IGF-1 signaling impairment. A 24-h IGF-1 treatment partially rescued neurite shortening caused by sodium arsenite. Moreover, the reduction in Akt phosphorylation by sodium arsenite was attenuated by IGF-1. Inhibition of PI3K/Akt by LY294002 diminished the protective effects of IGF-1 against sodium arsenite-induced neurite retraction. Together, our findings suggested that sodium arsenite-impaired IGF-1 signaling, leading to neurite shortening through IGF-1/PI3K/Akt.
Collapse
Affiliation(s)
- Churaibhon Wisessaowapak
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| | - Apichaya Niyomchan
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Naphada Leelaprachakul
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, Thailand
| | - Piyajit Watcharasit
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, Thailand
| | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, Thailand
| |
Collapse
|
3
|
Chaker SC, Saad M, Mayes T, Lineaweaver WC. Burn Injury-related Growth Factor Expressions and Their Potential Roles in Burn-related Neuropathies. J Burn Care Res 2024; 45:25-31. [PMID: 37978864 DOI: 10.1093/jbcr/irad184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Indexed: 11/19/2023]
Abstract
In the context of burn injury, growth factors (GFs) play a significant role in mediating the complex local and systematic processes that occur. Among the many systemic complications that arise following a burn injury, peripheral neuropathy remains one of the most common. Despite the broad understanding of the effects GFs have on multiple tissues, their potential implications in both wound healing and neuropathy remain largely unexplored. Therefore, this review aims to investigate the expression patterns of GFs prominent during the burn wound healing process and explore the potential contributions these GFs have on the development of burn-related peripheral neuropathy.
Collapse
Affiliation(s)
- Sara C Chaker
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232USA
| | - Mariam Saad
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232USA
| | - Taylor Mayes
- Middle Tennessee State University, Murfreesboro, TN, 37132USA
| | - William C Lineaweaver
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232USA
| |
Collapse
|
4
|
Proteomic investigation of Cbl and Cbl-b in neuroblastoma cell differentiation highlights roles for SHP-2 and CDK16. iScience 2021; 24:102321. [PMID: 33889818 PMCID: PMC8050387 DOI: 10.1016/j.isci.2021.102321] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/08/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma is a highly heterogeneous embryonal solid tumor of the sympathetic nervous system. As some tumors can be treated to undergo differentiation, investigating this process can guide differentiation-based therapies of neuroblastoma. Here, we studied the role of E3 ubiquitin ligases Cbl and Cbl-b in regulation of long-term signaling responses associated with extracellular signal-regulated kinase phosphorylation and neurite outgrowth, a morphological marker of neuroblastoma cell differentiation. Using quantitative mass spectrometry (MS)-based proteomics, we analyzed how the neuroblastoma cell line proteome, phosphoproteome, and ubiquitylome were affected by Cbl and Cbl-b depletion. To quantitatively assess neurite outgrowth, we developed a high-throughput microscopy assay that was applied in combination with inhibitor studies to pinpoint signaling underlying neurite outgrowth and to functionally validate proteins identified in the MS data sets. Using this combined approach, we identified a role for SHP-2 and CDK16 in Cbl/Cbl-b-dependent regulation of extracellular signal-regulated kinase phosphorylation and neurite outgrowth, highlighting their involvement in neuroblastoma cell differentiation. Multi-layered proteomics captures cellular changes induced by Cbl/Cbl-b depletion SHP-2 and CDK16 protein and phosphorylation levels increase upon Cbl/Cbl-b depletion SHP-2 and CDK16 regulate phospho-ERK and neurite outgrowth in neuroblastoma cells Inhibition of SHP-2 or CDK16 reverts Cbl/Cbl-b knockdown effects on differentiation
Collapse
|
5
|
Lepa C, Hoppe S, Stöber A, Skryabin BV, Sievers LK, Heitplatz B, Ciarimboli G, Neugebauer U, Lindenmeyer MT, Cohen CD, Drexler HC, Boor P, Weide T, Pavenstädt H, George B. TrkC Is Essential for Nephron Function and Trans-Activates Igf1R Signaling. J Am Soc Nephrol 2021; 32:357-374. [PMID: 33380522 PMCID: PMC8054883 DOI: 10.1681/asn.2020040424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 11/03/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Injury to kidney podocytes often results in chronic glomerular disease and consecutive nephron malfunction. For most glomerular diseases, targeted therapies are lacking. Thus, it is important to identify novel signaling pathways contributing to glomerular disease. Neurotrophic tyrosine kinase receptor 3 (TrkC) is expressed in podocytes and the protein transmits signals to the podocyte actin cytoskeleton. METHODS Nephron-specific TrkC knockout (TrkC-KO) and nephron-specific TrkC-overexpressing (TrkC-OE) mice were generated to dissect the role of TrkC in nephron development and maintenance. RESULTS Both TrkC-KO and TrkC-OE mice exhibited enlarged glomeruli, mesangial proliferation, basement membrane thickening, albuminuria, podocyte loss, and aspects of FSGS during aging. Igf1 receptor (Igf1R)-associated gene expression was dysregulated in TrkC-KO mouse glomeruli. Phosphoproteins associated with insulin, erb-b2 receptor tyrosine kinase (Erbb), and Toll-like receptor signaling were enriched in lysates of podocytes treated with the TrkC ligand neurotrophin-3 (Nt-3). Activation of TrkC by Nt-3 resulted in phosphorylation of the Igf1R on activating tyrosine residues in podocytes. Igf1R phosphorylation was increased in TrkC-OE mouse kidneys while it was decreased in TrkC-KO kidneys. Furthermore, TrkC expression was elevated in glomerular tissue of patients with diabetic kidney disease compared with control glomerular tissue. CONCLUSIONS Our results show that TrkC is essential for maintaining glomerular integrity. Furthermore, TrkC modulates Igf-related signaling in podocytes.
Collapse
Affiliation(s)
- Carolin Lepa
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Sascha Hoppe
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Antje Stöber
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Boris V. Skryabin
- Medical Faculty, Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), Westfälische-Wilhelms-University, Münster, Germany
| | | | - Barbara Heitplatz
- Gerhard-Domagk Institute for Pathology, University Hospital Münster, Münster, Germany
| | | | - Ute Neugebauer
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Maja T. Lindenmeyer
- III. Medizinische Klinik und Poliklinik, University Hospital Hamburg-Eppendorf, Germany
| | - Clemens D. Cohen
- Klinik für Nieren-, Hochdruck- und Rheumaerkrankungen, München Klinik Harlaching, Germany
| | - Hannes C.A. Drexler
- Mass Spectrometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Thomas Weide
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | | | - Britta George
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| |
Collapse
|
6
|
Soluble SORLA Enhances Neurite Outgrowth and Regeneration through Activation of the EGF Receptor/ERK Signaling Axis. J Neurosci 2020; 40:5908-5921. [PMID: 32601248 DOI: 10.1523/jneurosci.0723-20.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 01/01/2023] Open
Abstract
SORLA is a transmembrane trafficking protein associated with Alzheimer's disease risk. Although SORLA is abundantly expressed in neurons, physiological roles for SORLA remain unclear. Here, we show that cultured transgenic neurons overexpressing SORLA feature longer neurites, and accelerated neurite regeneration with wounding. Enhanced release of a soluble form of SORLA (sSORLA) is observed in transgenic mouse neurons overexpressing human SORLA, while purified sSORLA promotes neurite extension and regeneration. Phosphoproteomic analyses demonstrate enrichment of phosphoproteins related to the epidermal growth factor (EGFR)/ERK pathway in SORLA transgenic mouse hippocampus from both genders. sSORLA coprecipitates with EGFR in vitro, and sSORLA treatment increases EGFR Y1173 phosphorylation, which is involved in ERK activation in cultured neurons. Furthermore, sSORLA triggers ERK activation, whereas pharmacological EGFR or ERK inhibition reverses sSORLA-dependent enhancement of neurite outgrowth. In search for downstream ERK effectors activated by sSORLA, we identified upregulation of Fos expression in hippocampus from male mice overexpressing SORLA by RNAseq analysis. We also found that Fos is upregulated and translocates to the nucleus in an ERK-dependent manner in neurons treated with sSORLA. Together, these results demonstrate that sSORLA is an EGFR-interacting protein that activates EGFR/ERK/Fos signaling to enhance neurite outgrowth and regeneration.SIGNIFICANCE STATEMENT SORLA is a transmembrane trafficking protein previously known to reduce the levels of amyloid-β, which is critical in the pathogenesis of Alzheimer's disease. In addition, SORLA mutations are a risk factor for Alzheimer's disease. Interestingly, the SORLA ectodomain is cleaved into a soluble form, sSORLA, which has been shown to regulate cytoskeletal signaling pathways and cell motility in cells outside the nervous system. We show here that sSORLA binds and activates the EGF receptor to induce downstream signaling through the ERK serine/threonine kinase and the Fos transcription factor, thereby enhancing neurite outgrowth. These findings reveal a novel role for sSORLA in promoting neurite regeneration through the EGF receptor/ERK/Fos pathway, thereby demonstrating a potential neuroprotective mechanism involving SORLA.
Collapse
|
7
|
Littlejohn EL, Scott D, Saatman KE. Insulin-like growth factor-1 overexpression increases long-term survival of posttrauma-born hippocampal neurons while inhibiting ectopic migration following traumatic brain injury. Acta Neuropathol Commun 2020; 8:46. [PMID: 32276671 PMCID: PMC7147070 DOI: 10.1186/s40478-020-00925-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/29/2020] [Indexed: 01/29/2023] Open
Abstract
Cellular damage associated with traumatic brain injury (TBI) manifests in motor and cognitive dysfunction following injury. Experimental models of TBI reveal cell death in the granule cell layer (GCL) of the hippocampal dentate gyrus acutely after injury. Adult-born neurons residing in the neurogenic niche of the GCL, the subgranular zone, are particularly vulnerable. Injury-induced proliferation of neural progenitors in the subgranular zone supports recovery of the immature neuron population, but their development and localization may be altered, potentially affecting long-term survival. Here we show that increasing hippocampal levels of insulin-like growth factor-1 (IGF1) is sufficient to promote end-stage maturity of posttrauma-born neurons and improve cognition following TBI. Mice with conditional overexpression of astrocyte-specific IGF1 and wild-type mice received controlled cortical impact or sham injury and bromo-2'-deoxyuridine injections for 7d after injury to label proliferating cells. IGF1 overexpression increased the number of GCL neurons born acutely after trauma that survived 6 weeks to maturity (NeuN+BrdU+), and enhanced their outward migration into the GCL while significantly reducing the proportion localized ectopically to the hilus and molecular layer. IGF1 selectively affected neurons, without increasing the persistence of posttrauma-proliferated glia in the dentate gyrus. IGF1 overexpressing animals performed better during radial arm water maze reversal testing, a neurogenesis-dependent cognitive test. These findings demonstrate the ability of IGF1 to promote the long-term survival and appropriate localization of granule neurons born acutely after a TBI, and suggest these new neurons contribute to improved cognitive function.
Collapse
Affiliation(s)
- Erica L. Littlejohn
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, B473 Biomedical & Biological Sciences Research Building (BBSRB), 741 South Limestone St, Lexington, KY 40536-0509 USA
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3901 USA
| | - Danielle Scott
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, B473 Biomedical & Biological Sciences Research Building (BBSRB), 741 South Limestone St, Lexington, KY 40536-0509 USA
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, B473 Biomedical & Biological Sciences Research Building (BBSRB), 741 South Limestone St, Lexington, KY 40536-0509 USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536 USA
| |
Collapse
|
8
|
Nampoothiri SS, Rajanikant GK. miR-9 Upregulation Integrates Post-ischemic Neuronal Survival and Regeneration In Vitro. Cell Mol Neurobiol 2019; 39:223-240. [PMID: 30539420 PMCID: PMC11469860 DOI: 10.1007/s10571-018-0642-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/07/2018] [Indexed: 01/02/2023]
Abstract
The irrefutable change in the expression of brain-enriched microRNAs (miRNAs) following ischemic stroke has promoted the development of radical miRNA-based therapeutics encompassing neuroprotection and neuronal restoration. Our previous report on the systems-level prediction of miR-9 in post-stroke-induced neurogenesis served as a premise to experimentally uncover the functional role of miR-9 in post-ischemic neuronal survival and regeneration. The oxygen-glucose deprivation (OGD) in SH-SY5Y cells significantly reduced miR-9 expression, while miR-9 mimic transfection enhanced post-ischemic neuronal cell viability. The next major objective involved the execution of a drug repositioning strategy to augment miR-9 expression via structure-based screening of Food and Drug Administration (FDA)-approved drugs that bind to Histone Deacetylase 4 (HDAC4), a known miR-9 target. Glucosamine emerged as the top hit and its binding potential to HDAC4 was verified by Molecular Dynamics (MD) Simulation, Drug Affinity Responsive Target Stability (DARTS) assay, and MALDI-TOF MS. It was intriguing that the glucosamine treatment 1-h post-OGD was associated with the increased miR-9 level as well as enhanced neuronal viability. miR-9 mimic or post-OGD glucosamine treatment significantly increased the cellular proliferation (BrdU assay), while the neurite outgrowth assay displayed elongated neurites. The enhanced BCL2 and VEGF parallel with the reduced NFκB1, TNF-α, IL-1β, and iNOS mRNA levels in miR-9 mimic or glucosamine-treated cells further substantiated their post-ischemic neuroprotective and regenerative efficacy. Hence, this study unleashes a potential therapeutic approach that integrates neuronal survival and regeneration via small-molecule-based regulation of miR-9 favoring long-term recovery against ischemic stroke.
Collapse
Affiliation(s)
- Sreekala S Nampoothiri
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India
| | - G K Rajanikant
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India.
| |
Collapse
|
9
|
Wepy JA, Galligan JJ, Kingsley PJ, Xu S, Goodman MC, Tallman KA, Rouzer CA, Marnett LJ. Lysophospholipases cooperate to mediate lipid homeostasis and lysophospholipid signaling. J Lipid Res 2018; 60:360-374. [PMID: 30482805 DOI: 10.1194/jlr.m087890] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/05/2018] [Indexed: 12/20/2022] Open
Abstract
Lysophospholipids (LysoPLs) are bioactive lipid species involved in cellular signaling processes and the regulation of cell membrane structure. LysoPLs are metabolized through the action of lysophospholipases, including lysophospholipase A1 (LYPLA1) and lysophospholipase A2 (LYPLA2). A new X-ray crystal structure of LYPLA2 compared with a previously published structure of LYPLA1 demonstrated near-identical folding of the two enzymes; however, LYPLA1 and LYPLA2 have displayed distinct substrate specificities in recombinant enzyme assays. To determine how these in vitro substrate preferences translate into a relevant cellular setting and better understand the enzymes' role in LysoPL metabolism, CRISPR-Cas9 technology was utilized to generate stable KOs of Lypla1 and/or Lypla2 in Neuro2a cells. Using these cellular models in combination with a targeted lipidomics approach, LysoPL levels were quantified and compared between cell lines to determine the effect of losing lysophospholipase activity on lipid metabolism. This work suggests that LYPLA1 and LYPLA2 are each able to account for the loss of the other to maintain lipid homeostasis in cells; however, when both are deleted, LysoPL levels are dramatically increased, causing phenotypic and morphological changes to the cells.
Collapse
Affiliation(s)
- James A Wepy
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Chemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146
| | - James J Galligan
- Departments of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146
| | - Philip J Kingsley
- Departments of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146
| | - Shu Xu
- Departments of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146
| | - Michael C Goodman
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Chemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146
| | - Keri A Tallman
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Chemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146.,Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232-0146
| | - Carol A Rouzer
- Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232-0146.,Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232-0146
| | - Lawrence J Marnett
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Chemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146 .,Departments of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146.,Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232-0146.,Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232-0146.,Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232-0146
| |
Collapse
|
10
|
Farahnak S, McGovern TK, Kim R, O'Sullivan M, Chen B, Lee M, Yoshie H, Wang A, Jang J, Al Heialy S, Lauzon AM, Martin JG. Basic Fibroblast Growth Factor 2 Is a Determinant of CD4 T Cell-Airway Smooth Muscle Cell Communication through Membrane Conduits. THE JOURNAL OF IMMUNOLOGY 2017; 199:3086-3093. [PMID: 28924004 DOI: 10.4049/jimmunol.1700164] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 08/24/2017] [Indexed: 01/23/2023]
Abstract
Activated CD4 T cells connect to airway smooth muscle cells (ASMCs) in vitro via lymphocyte-derived membrane conduits (LMCs) structurally similar to membrane nanotubes with unknown intercellular signals triggering their formation. We examined the structure and function of CD4 T cell-derived LMCs, and we established a role for ASMC-derived basic fibroblast growth factor 2 (FGF2b) and FGF receptor (FGFR)1 in LMC formation. Blocking FGF2b's synthesis and FGFR1 function reduced LMC formation. Mitochondrial flux from ASMCs to T cells was partially FGF2b and FGFR1 dependent. LMC formation by CD4 T cells and mitochondrial transfer from ASMCs was increased in the presence of asthmatic ASMCs that expressed more mRNA for FGF2b compared with normal ASMCs. These observations identify ASMC-derived FGF2b as a factor needed for LMC formation by CD4 T cells, affecting intercellular communication.
Collapse
Affiliation(s)
- Soroor Farahnak
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and.,Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Toby K McGovern
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Rachael Kim
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Michael O'Sullivan
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Brian Chen
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Minhyoung Lee
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Haruka Yoshie
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Anna Wang
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Joyce Jang
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Saba Al Heialy
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and.,Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - James G Martin
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; and .,Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
11
|
Hernandez-Martinez JM, Forrest CM, Darlington LG, Smith RA, Stone TW. Quinolinic acid induces neuritogenesis in SH-SY5Y neuroblastoma cells independently of NMDA receptor activation. Eur J Neurosci 2017; 45:700-711. [PMID: 27973747 DOI: 10.1111/ejn.13499] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 12/25/2022]
Abstract
Glutamate and nicotinamide adenine dinucleotide (NAD+ ) have been implicated in neuronal development and several types of cancer. The kynurenine pathway of tryptophan metabolism includes quinolinic acid (QA) which is both a selective agonist at N-methyl-D-aspartate (NMDA) receptors and also a precursor for the formation of NAD+ . The effect of QA on cell survival and differentiation has therefore been examined on SH-SY5Y human neuroblastoma cells. Retinoic acid (RA, 10 μm) induced differentiation of SH-SY5Y cells into a neuronal phenotype showing neurite growth. QA (50-150 nm) also caused a concentration-dependent increase in the neurite/soma ratio, indicating differentiation. Both RA and QA increased expression of the neuronal marker β3-tubulin in whole-cell homogenates and in the neuritic fraction assessed using a neurite outgrowth assay. Expression of the neuronal proliferation marker doublecortin revealed that, unlike RA, QA did not decrease the number of mitotic cells. QA-induced neuritogenesis coincided with an increase in the generation of reactive oxygen species. Neuritogenesis was prevented by diphenylene-iodonium (an inhibitor of NADPH oxidase) and superoxide dismutase, supporting the involvement of reactive oxygen species. NMDA itself did not promote neuritogenesis and the NMDA antagonist dizocilpine (MK-801) did not prevent quinolinate-induced neuritogenesis, indicating that the effects of QA were independent of NMDA receptors. Nicotinamide caused a significant increase in the neurite/soma ratio and the expression of β3-tubulin in the neuritic fraction. Taken together, these results suggest that QA induces neuritogenesis by promoting oxidizing conditions and affecting the availability of NAD+ , independently of NMDA receptors.
Collapse
Affiliation(s)
- Juan-Manuel Hernandez-Martinez
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | - Caroline M Forrest
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | | | - Robert A Smith
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | - Trevor W Stone
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| |
Collapse
|
12
|
IGF-1 Induces GHRH Neuronal Axon Elongation during Early Postnatal Life in Mice. PLoS One 2017; 12:e0170083. [PMID: 28076448 PMCID: PMC5226784 DOI: 10.1371/journal.pone.0170083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/28/2016] [Indexed: 12/21/2022] Open
Abstract
Nutrition during the perinatal period programs body growth. Growth hormone (GH) secretion from the pituitary regulates body growth and is controlled by Growth Hormone Releasing Hormone (GHRH) neurons located in the arcuate nucleus of the hypothalamus. We observed that dietary restriction during the early postnatal period (i.e. lactation) in mice influences postnatal growth by permanently altering the development of the somatotropic axis in the pituitary gland. This alteration may be due to a lack of GHRH signaling during this critical developmental period. Indeed, underfed pups showed decreased insulin-like growth factor I (IGF-I) plasma levels, which are associated with lower innervation of the median eminence by GHRH axons at 10 days of age relative to normally fed pups. IGF-I preferentially stimulated axon elongation of GHRH neurons in in vitro arcuate explant cultures from 7 day-old normally fed pups. This IGF-I stimulating effect was selective since other arcuate neurons visualized concomitantly by neurofilament labeling, or AgRP immunochemistry, did not significantly respond to IGF-I stimulation. Moreover, GHRH neurons in explants from age-matched underfed pups lost the capacity to respond to IGF-I stimulation. Molecular analyses indicated that nutritional restriction was associated with impaired activation of AKT. These results highlight a role for IGF-I in axon elongation that appears to be cell selective and participates in the complex cellular mechanisms that link underfeeding during the early postnatal period with programming of the growth trajectory.
Collapse
|
13
|
Lunn JS, Sakowski SA, McGinley LM, Pacut C, Hazel TG, Johe K, Feldman EL. Autocrine production of IGF-I increases stem cell-mediated neuroprotection. Stem Cells 2016; 33:1480-9. [PMID: 25532472 DOI: 10.1002/stem.1933] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/01/2014] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder resulting in motor neuron (MN) loss. There are currently no effective therapies; however, cellular therapies using neural progenitor cells protect MNs and attenuate disease progression in G93A-SOD1 ALS rats. Recently, we completed a phase I clinical trial examining intraspinal human spinal stem cell (HSSC) transplantation in ALS patients which demonstrated our approach was safe and feasible, supporting the phase II trial currently in progress. In parallel, efforts focused on understanding the mechanisms underlying the preclinical benefit of HSSCs in vitro and in animal models of ALS led us to investigate how insulin-like growth factor-I (IGF-I) production contributes to cellular therapy neuroprotection. IGF-I is a potent growth factor with proven efficacy in preclinical ALS studies, and we contend that autocrine IGF-I production may enhance the salutary effects of HSSCs. By comparing the biological properties of HSSCs to HSSCs expressing sixfold higher levels of IGF-I, we demonstrate that IGF-I production augments the production of glial-derived neurotrophic factor and accelerates neurite outgrowth without adversely affecting HSSC proliferation or terminal differentiation. Furthermore, we demonstrate that increased IGF-I induces more potent MN protection from excitotoxicity via both indirect and direct mechanisms, as demonstrated using hanging inserts with primary MNs or by culturing with organotypic spinal cord slices, respectively. These findings support our theory that combining autocrine growth factor production with HSSC transplantation may offer a novel means to achieve additive neuroprotection in ALS.
Collapse
|
14
|
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
|
15
|
Wang B, Yuan J, Xu J, Xie J, Wang G, Dong P. Neurotrophin expression and laryngeal muscle pathophysiology following recurrent laryngeal nerve transection. Mol Med Rep 2015; 13:1234-42. [PMID: 26677138 PMCID: PMC4732864 DOI: 10.3892/mmr.2015.4684] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 11/19/2015] [Indexed: 11/05/2022] Open
Abstract
Laryngeal palsy often occurs as a result of recurrent laryngeal or vagal nerve injury during oncological surgery of the head and neck, affecting quality of life and increasing economic burden. Reinnervation following recurrent laryngeal nerve (RLN) injury is difficult despite development of techniques, such as neural anastomosis, nerve grafting and creation of a laryngeal muscle pedicle. In the present study, due to the limited availability of human nerve tissue for research, a rat model was used to investigate neurotrophin expression and laryngeal muscle pathophysiology in RLN injury. Twenty-five male Sprague-Dawley rats underwent right RLN transection with the excision of a 5-mm segment. Vocal fold movements, vocalization, histology and immunostaining were evaluated at different time-points (3, 6, 10 and 16 weeks). Although vocalization was restored, movement of the vocal fold failed to return to normal levels following RLN injury. The expression of brain‑derived neurotrophic factor and glial cell line-derived neurotrophic factor differed in the thyroarytenoid (TA) and posterior cricoarytenoid muscles. The number of axons did not increase to baseline levels over time. Furthermore, normal muscle function was unlikely with spontaneous reinnervation. During regeneration following RLN injury, differences in the expression levels of neurotrophic factors may have resulted in preferential reinnervation of the TA muscles. Data from the present study indicated that neurotrophic factors may be applied for restoring the function of the laryngeal nerve following recurrent injury.
Collapse
Affiliation(s)
- Baoxin Wang
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, P.R. China
| | - Junjie Yuan
- Department of Orthopedics, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai 200011, P.R. China
| | - Jiafeng Xu
- School of Economics and Finance, Shanghai International Studies University, Shanghai 200083, P.R. China
| | - Jin Xie
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, P.R. China
| | - Guoliang Wang
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, P.R. China
| | - Pin Dong
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, P.R. China
| |
Collapse
|
16
|
Influence of insulin-like growth factor I on nerve regeneration using allografts: a sciatic nerve model. J Craniofac Surg 2015; 25:1510-4. [PMID: 25006924 DOI: 10.1097/scs.0000000000000783] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Effects of insulin-like growth factor I (IGF I) on peripheral nerve regeneration was studied using allografts in a rat sciatic nerve model. Thirty male white Wistar rats were divided into 3 experimental groups (n = 10) randomly: normal control group (NC), allograft group (ALLO), and IGF I-treated group (ALLO/IGF). In the NC group, the left sciatic nerve was exposed through a gluteal muscle incision and, after homeostasis, the muscle was sutured. In the ALLO group, the left sciatic nerve was exposed through a gluteal muscle incision and transected proximal to the tibioperoneal bifurcation where a 10-mm segment was excised. The same procedure was performed in the ALLO/IGF group. The harvested nerves of the rats of the ALLO group were served as allograft for the ALLO/IGF group and vice versa. The NC and ALLO groups received 10 μL of sterile phosphate buffered saline intraperitoneally once a day for 1 week, and the ALLO/IGF group received 10 μL of IGF I (100 ng/kg per day) intraperitoneally once a day for 1 week. Behavioral testing, sciatic nerve functional study and the gastrocnemius muscle mass showed earlier regeneration of axons in the ALLO/IGF group than in the ALLO group (P < 0.05). Administration of IGF I could accelerate functional recovery after nerve allografting in the sciatic nerve and may have clinical implications for the surgical management of patients after facial nerve transection.
Collapse
|
17
|
Shigyo M, Kuboyama T, Sawai Y, Tada-Umezaki M, Tohda C. Extracellular vimentin interacts with insulin-like growth factor 1 receptor to promote axonal growth. Sci Rep 2015; 5:12055. [PMID: 26170015 PMCID: PMC4501001 DOI: 10.1038/srep12055] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 06/16/2015] [Indexed: 12/25/2022] Open
Abstract
Vimentin, an intermediate filament protein, is generally recognised as an intracellular protein. Previously, we reported that vimentin was secreted from astrocytes and promoted axonal growth. The effect of extracellular vimentin in neurons was a new finding, but its signalling pathway was unknown. In this study, we aimed to determine the signalling mechanism of extracellular vimentin that facilitates axonal growth. We first identified insulin-like growth factor 1 receptor (IGF1R) as a receptor that is highly phosphorylated by vimentin stimulation. IGF1R blockades diminished vimentin- or IGF1-induced axonal growth in cultured cortical neurons. IGF1, IGF2 and insulin were not detected in the neuron culture medium after vimentin treatment. The combined drug affinity responsive target stability method and western blotting analysis showed that vimentin and IGF1 interacted with IGF1R directly. In addition, immunoprecipitation and western blotting analyses confirmed that recombinant IGF1R bound to vimentin. The results of a molecular dynamics simulation revealed that C-terminal residues (residue number 330-407) in vimentin are the most appropriate binding sites with IGF1R. Thus, extracellular vimentin may be a novel ligand of IGF1R that promotes axonal growth in a similar manner to IGF1. Our results provide novel findings regarding the role of extracellular vimentin and IGF1R in axonal growth.
Collapse
Affiliation(s)
- Michiko Shigyo
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Tomoharu Kuboyama
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Yusuke Sawai
- Division of Chemo-Bioinformatics, Department of Translational Research, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Masahito Tada-Umezaki
- Division of Chemo-Bioinformatics, Department of Translational Research, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Chihiro Tohda
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
18
|
Vieira M, Gomes JR, Saraiva MJ. Transthyretin Induces Insulin-like Growth Factor I Nuclear Translocation Regulating Its Levels in the Hippocampus. Mol Neurobiol 2014; 51:1468-79. [PMID: 25084758 PMCID: PMC4434863 DOI: 10.1007/s12035-014-8824-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 07/17/2014] [Indexed: 01/02/2023]
Abstract
Transthyretin (TTR) is the carrier protein of thyroxine (T4) and binds to retinol-binding protein (RBP)-retinol complex. It is mainly synthesized by both liver and choroid plexuses of the brain. Besides these properties, it has a neuroprotective role in several contexts such as Alzheimer’s disease (AD) and cerebral ischemia. Activation of insulin-like growth factor receptor I (IGF-IR) pathways and increased levels of TTR are associated with absence of neurodegeneration in an AD mouse model. In the present study, we verified that young/adult TTR null mice had decreased levels of IGF-IR in the hippocampus, but not in choroid plexus when compared with wild-type age-matched controls. Moreover, we could also demonstrate that conditional silencing of peripheral TTR did not have any influence in hippocampal IGF-IR levels, indicating that TTR effect on IGF-IR levels is due to TTR mainly synthesized in the choroid plexus. In vitro cellular studies, using NIH3T3 cell line and primary cultured hippocampal neurons, we showed that TTR upregulates IGF-IR at the transcription and translation levels and that is dependent on receptor internalization. Using a GFP-IGF-IR fusion protein, we also found that TTR triggers IGF-IR nuclear translocation in cultured neurons. We could also see an enrichment of IGF-IR in the nuclear fraction, after TTR stimulation in NIH3T3 cells, indicating that IGF-IR regulation, triggered by TTR is induced by nuclear translocation. In summary, the results provide evidence of a new role of TTR as a transcription inducer of IGF-IR in central nervous system (CNS), unveiling a new role in neuroprotection.
Collapse
Affiliation(s)
- Marta Vieira
- Molecular Neurobiology Unit, IBMC - Instituto de Biologia Molecular e Celular, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| | | | | |
Collapse
|
19
|
Mohammadi R, Mahmoodi H. Improvement of peripheral nerve regeneration following nerve repair by silicone tube filled with curcumin: A preliminary study in the rat model. Int J Surg 2013; 11:819-25. [DOI: 10.1016/j.ijsu.2013.08.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/22/2013] [Accepted: 08/18/2013] [Indexed: 12/11/2022]
|
20
|
Effect of local administration of insulin-like growth factor I combined with inside-out artery graft on peripheral nerve regeneration. Injury 2013; 44:1295-301. [PMID: 23747124 DOI: 10.1016/j.injury.2013.04.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/09/2013] [Accepted: 04/14/2013] [Indexed: 02/02/2023]
Abstract
The objective was to assess the effect of topically administered insulin-like growth factor (IGF I) on peripheral nerve regeneration and functional recovery. Eighty male healthy white Wistar rats were divided into four experimental groups (n=20), randomly: in transected group (TC), the left sciatic nerve was transected and stumps were fixed in the adjacent muscle. In treatment group, defect was bridged using an inside-out artery graft (IOAG/IGF) filled with 10 μL IGF I (100 ng/kg). In artery graft group (IOAG), the graft was filled with phosphate-buffered saline alone. In sham-operated group (SHAM), sciatic nerve was exposed and manipulated. Each group was subdivided into five subgroups of five animals each and regenerated nerve fibres were studied 4, 8, 12 and 16 weeks after surgery. Behavioural testing, sciatic nerve functional study, gastrocnemius muscle mass and morphometric indices confirmed faster recovery of regenerated axons in IOAG/IGF than IOAG group (P<0.05). In immunohistochemistry, location of reactions to S-100 in IOAG/IGF was clearly more positive than that in IOAG group. When loaded in an artery graft, IGF I accelerated and improved functional recovery and morphometric indices of sciatic nerve.
Collapse
|
21
|
Molina DP, Ariwodola OJ, Weiner JL, Brunso-Bechtold JK, Adams MM. Growth hormone and insulin-like growth factor-I alter hippocampal excitatory synaptic transmission in young and old rats. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1575-87. [PMID: 22851280 PMCID: PMC3776110 DOI: 10.1007/s11357-012-9460-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 07/11/2012] [Indexed: 06/01/2023]
Abstract
In rats, as in humans, normal aging is characterized by a decline in hippocampal-dependent learning and memory, as well as in glutamatergic function. Both growth hormone (GH) and insulin-like growth factor-I (IGF-I) levels have been reported to decrease with age, and treatment with either GH or IGF-I can ameliorate age-related cognitive decline. Interestingly, acute GH and IGF-I treatments enhance glutamatergic synaptic transmission in the rat hippocampus of juvenile animals. However, whether this enhancement also occurs in old rats, when cognitive impairment is ameliorated by GH and IGF-I (des-IGF-I), remains to be determined. To address this issue, we used an in vitro CA1 hippocampal slice preparation and extracellular recording techniques to study the effects of acute application of GH and IGF-I on compound field excitatory postsynaptic potentials (fEPSPs), as well as AMPA- and NMDA-dependent fEPSPs, in young adult (10 months) and old (28 months) rats. The results indicated that both GH and IGF-I increased compound-, AMPA-, and NMDA-dependent fEPSPs to a similar extent in slices from both age groups and that this augmentation was likely mediated via a postsynaptic mechanism. Initial characterization of the signaling cascades underlying these effects revealed that the GH-induced enhancement was not mediated by the JAK2 signaling element in either young adult or old rats but that the IGF-I-induced enhancement involved a PI3K-mediated mechanism in old, but not young adults. The present findings are consistent with a role for a GH- or IGF-I-induced enhancement of glutamatergic transmission in mitigating age-related cognitive impairment in old rats.
Collapse
Affiliation(s)
- Doris P. Molina
- />Departments of Neurobiology and Anatomy, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1010 USA
| | - Olusegun J. Ariwodola
- />Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1010 USA
| | - Jeff L. Weiner
- />Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1010 USA
| | - Judy K. Brunso-Bechtold
- />Departments of Neurobiology and Anatomy, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1010 USA
- />Roena Kulynych Center for Memory and Cognition Research, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1010 USA
| | - Michelle M. Adams
- />Departments of Neurobiology and Anatomy, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1010 USA
- />Roena Kulynych Center for Memory and Cognition Research, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1010 USA
- />Department of Psychology, Bilkent University, 06800 Bilkent, Ankara, Turkey
| |
Collapse
|
22
|
Dwane S, Durack E, Kiely PA. Optimising parameters for the differentiation of SH-SY5Y cells to study cell adhesion and cell migration. BMC Res Notes 2013; 6:366. [PMID: 24025096 PMCID: PMC3847106 DOI: 10.1186/1756-0500-6-366] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 09/04/2013] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Cell migration is a fundamental biological process and has an important role in the developing brain by regulating a highly specific pattern of connections between nerve cells. Cell migration is required for axonal guidance and neurite outgrowth and involves a series of highly co-ordinated and overlapping signalling pathways. The non-receptor tyrosine kinase, Focal Adhesion Kinase (FAK) has an essential role in development and is the most highly expressed kinase in the developing CNS. FAK activity is essential for neuronal cell adhesion and migration. RESULTS The objective of this study was to optimise a protocol for the differentiation of the neuroblastoma cell line, SH-SY5Y. We determined the optimal extracellular matrix proteins and growth factor combinations required for the optimal differentiation of SH-SY5Y cells into neuronal-like cells and determined those conditions that induce the expression of FAK. It was confirmed that the cells were morphologically and biochemically differentiated when compared to undifferentiated cells. This is in direct contrast to commonly used differentiation methods that induce morphological differentiation but not biochemical differentiation. CONCLUSIONS We conclude that we have optimised a protocol for the differentiation of SH-SY5Y cells that results in a cell population that is both morphologically and biochemically distinct from undifferentiated SH-SY5Y cells and has a distinct adhesion and spreading pattern and display extensive neurite outgrowth. This protocol will provide a neuronal model system for studying FAK activity during cell adhesion and migration events.
Collapse
Affiliation(s)
- Susan Dwane
- Department of Life Sciences and Materials and Surface Science Institute, University of Limerick, Limerick, Ireland.
| | | | | |
Collapse
|
23
|
Taniguchi T, Tanaka S, Ishii A, Watanabe M, Fujitani N, Sugeo A, Gotoh S, Ohta T, Hiyoshi M, Matsuzaki H, Sakai N, Konishi H. A brain-specific Grb2-associated regulator of extracellular signal-regulated kinase (Erk)/mitogen-activated protein kinase (MAPK) (GAREM) subtype, GAREM2, contributes to neurite outgrowth of neuroblastoma cells by regulating Erk signaling. J Biol Chem 2013; 288:29934-42. [PMID: 24003223 DOI: 10.1074/jbc.m113.492520] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Grb2-associated regulator of Erk/MAPK1 (GAREM) is an adaptor molecule in the EGF-mediated signaling pathway. GAREM is expressed ubiquitously in human organs and cultured cells. Two GAREM homologues are encoded by the human genome. Therefore, previously identified GAREM is named GAREM1. Here we characterized a new subtype of GAREM, GAREM2, that is specifically expressed in the mouse, rat, and human brain. Three GAREM2 tyrosines (Tyr-102, Tyr-429, and Tyr-551) are phosphorylated upon EGF stimulation and are necessary for binding to Grb2. Furthermore, GAREM2 and Shp2 regulate Erk activity in EGF-stimulated cells. These characteristics are similar to those of GAREM1. GAREM2 is expressed in some neuroblastoma cell lines and is also tyrosine-phosphorylated and bound to Grb2 after treatment with EGF. Eventually, GAREM2 regulates Erk activation in the presence of EGF or insulin like growth factor 1. GAREM2 also regulates insulin-like growth factor 1-induced neuronal differentiation of the SH-SY5Y neuroblastoma cell line. Although the structure and function of both GAREM subtypes are similar, GAREM1 is recruited into the nucleus and GAREM2 is not. Nuclear localization of GAREM1 might be controlled by a GAREM1-specific nuclear localization sequence and 14-3-3ε binding. The N-terminal 20 amino acids of GAREM1 make up its nuclear localization sequence that is also a 14-3-3ε binding site. The GAREM family is a new class of adaptor molecules with subtype-specific biological functions.
Collapse
Affiliation(s)
- Tomonori Taniguchi
- From the Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shobara, Hiroshima 727-0023, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
The elements of peripheral nerve regeneration comprise a complex combination of nerve growth, muscle satellite cells proliferation and differentiation and vessel growth. There is also increasing evidence that growth factors may act at multiple levels in the regenerative response. One such factor affecting multiple cell processes is insulin-like growth factor (IGF-1). As a neurotrophic factor IGF-1 is known to promote nerve elongation and branching. As a myogenic factor, IGF-1 promotes satellite cell proliferation, differentiation and muscle hypertrophy. As an angiogenic factor, IGF-1 is known to promote angiogenesis in regenerating skeletal muscle by activating VEGF and VEGF receptors. Additionally, recent studies show that IGF-1 may also promote the activation of muscle stem cells during the regenerative process. This review will outline the pathways by which IGF-1 affects multiple layers of the regenerative response and how these pathways converge to promote the regeneration of nerves.
Collapse
Affiliation(s)
- Eric D Rabinovsky
- Michael E. DeBakey Department of Surgery, Division of Plastic Surgery, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Role of Physical Exercise for Improving Posttraumatic Nerve Regeneration. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 109:125-49. [DOI: 10.1016/b978-0-12-420045-6.00006-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
26
|
Sakowski SA, Feldman EL. Insulin-like growth factors in the peripheral nervous system. Endocrinol Metab Clin North Am 2012; 41:375-93, vii. [PMID: 22682636 DOI: 10.1016/j.ecl.2012.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin-like growth factors (IGFs) play an integral role in development, growth, and survival. This article details the current understanding of the effects of IGFs in the peripheral nervous system (PNS) during health and disease, and introduces how the IGF system regulates PNS development and impacts growth and survival of PNS cells. Also discussed are implications of IGF signaling in neurodegeneration and the status and prospects of IGF therapies for PNS conditions. There is substantial support for the application of IGF therapies in the treatment of PNS injury and disease.
Collapse
Affiliation(s)
- Stacey A Sakowski
- A. Alfred Taubman Medical Research Institute, University of Michigan, 109 Zina Pitcher Place, 4019 AAT-BSRB, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
27
|
γ-Enolase C-terminal peptide promotes cell survival and neurite outgrowth by activation of the PI3K/Akt and MAPK/ERK signalling pathways. Biochem J 2012; 443:439-50. [PMID: 22257123 DOI: 10.1042/bj20111351] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
γ-Enolase, a glycolytic enzyme, is expressed specifically in neurons. It exerts neurotrophic activity and has been suggested to regulate growth, differentiation, survival and regeneration of neurons. In the present study, we investigated the involvement of γ-enolase in PI3K (phosphoinositide 3-kinase)/Akt and MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) signalling, the two pathways triggered predominantly by neurotrophic factors. Whereas the PI3K/Akt pathway, rather than the MAPK/ERK pathway, is involved in γ-enolase-enhanced cell survival, γ-enolase-stimulated neurite outgrowth requires both pathways, i.e. the activation of both PI3K and ERK1/2, leading to subsequent expression of the growth-cone-specific protein GAP-43 (growth-associated protein of 43 kDa). MEK (MAPK/ERK kinase) and PI3K inhibition blocked or attenuated the neurite outgrowth associated with dynamic remodelling of the actin-based cytoskeleton. We show that γ-enolase-mediated PI3K activation regulates RhoA kinase, a key regulator of actin cytoskeleton organization. Moreover, the inhibition of RhoA downstream effector ROCK (Rho-associated kinase) results in enhanced γ-enolase-induced neurite outgrowth, accompanied by actin polymerization and its redistribution to growth cones. Our results show that γ-enolase controls neuronal survival, differentiation and neurite regeneration by activating the PI3K/Akt and MAPK/ERK signalling pathways, resulting in downstream regulation of the molecular and cellular processes of cytoskeleton reorganization and cell remodelling, activation of transcriptional factors and regulation of the cell cycle.
Collapse
|
28
|
Woo JK, Choi Y, Oh SH, Jeong JH, Choi DH, Seo HS, Kim CW. Mucin 1 enhances the tumor angiogenic response by activation of the AKT signaling pathway. Oncogene 2011; 31:2187-98. [PMID: 21927028 DOI: 10.1038/onc.2011.410] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although the hyper-glycosylated transmembrane protein Mucin 1 (MUC1) is aberrantly overexpressed in human breast carcinoma, the biological significance of MUC1 overexpression is unclear. This study showed that MUC1 expression promoted the synthesis and secretion of vascular endothelial growth factor (VEGF) through the AKT signaling pathway. Increase VEGF production through MUC1 expression had a number of effect. First, MUC1 transfection increased expression of VEGF in breast cancer cells. Second, MUC1-mediated VEGF induction was attenuated by a chemical inhibitor of AKT or MUC1 knock-down by MUC1 siRNA. Third, MUC1 expression led to the activation of insulin-like growth factor-1 receptor, which correlated with VEGF expression. In addition, when MDA-MB-231 human breast cancer cells were directly injected into NOD/SCID mice, MUC1 expression accelerated xenograft tumor growth in vivo. Finally, MUC1 expression enhanced tumor growth and angiogenesis in a PyMT-MMTV/hMUC1 transgenic mouse model. Concurrent with these results, analysis of a human tissue microarray identified a high correlation between MUC1 and VEGF expression in human breast carcinoma. The current report is the first to demonstrate that MUC1 expression promotes angiogenesis in human breast cancer in vivo and in vitro.
Collapse
Affiliation(s)
- J K Woo
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
29
|
In vitro optimization of retinoic acid–induced neuritogenesis and TH endogenous expression in human SH-SY5Y neuroblastoma cells by the antioxidant Trolox. Mol Cell Biochem 2011; 358:325-34. [DOI: 10.1007/s11010-011-0983-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 06/29/2011] [Indexed: 12/20/2022]
|
30
|
Dokas LA, Malone AM, Williams FE, Nauli SM, Messer WS. Multiple protein kinases determine the phosphorylated state of the small heat shock protein, HSP27, in SH-SY5Y neuroblastoma cells. Neuropharmacology 2011; 61:12-24. [PMID: 21338617 PMCID: PMC3105189 DOI: 10.1016/j.neuropharm.2011.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 01/25/2011] [Accepted: 02/11/2011] [Indexed: 01/21/2023]
Abstract
In SH-SY5Y human neuroblastoma cells, the cholinergic agonist, carbachol, stimulates phosphorylation of the small heat shock protein 27 (HSP27). Carbachol increases phosphorylation of both Ser-82 and Ser-78 while the phorbol ester, phorbol-12, 13-dibutyrate (PDB) affects only Ser-82. Muscarinic receptor activation by carbachol was confirmed by sensitivity of Ser-82 phosphorylation to hyoscyamine with no effect of nicotine or bradykinin. This response to carbachol is partially reduced by inhibition of protein kinase C (PKC) with GF 109203X and p38 mitogen-activated protein kinase (MAPK) with SB 203580. In contrast, phosphorylation produced by PDB is completely reversed by GF 109203X or CID 755673, an inhibitor of PKD. Inhibition of phosphatidylinositol 3-kinase or Akt with LY 294002 or Akti-1/2 stimulates HSP27 phosphorylation while rapamycin, which inhibits mTORC1, does not. The stimulatory effect of Akti-1/2 is reversed by SB 203580 and correlates with increased p38 MAPK phosphorylation. SH-SY5Y cells differentiated with a low concentration of PDB and basic fibroblast growth factor to a more neuronal phenotype retain carbachol-, PDB- and Akti-1/2-responsive HSP27 phosphorylation. Immunofluorescence microscopy confirms increased HSP27 phosphorylation in response to carbachol or PDB. At cell margins, PDB causes f-actin to reorganize forming lamellipodial structures from which phospho-HSP27 is segregated. The resultant phenotypic change in cell morphology is dependent upon PKC, but not PKD, activity. The major conclusion from this study is that the phosphorylated state of HSP27 in SH-SY5Y cells results from integrated signaling involving PKC, p38 MAPK and Akt.
Collapse
Affiliation(s)
- Linda A. Dokas
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Amy M. Malone
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Frederick E. Williams
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Surya M. Nauli
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
- Department of Medicinal & Biological Chemistry, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - William S. Messer
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
- Department of Medicinal & Biological Chemistry, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| |
Collapse
|
31
|
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
|
32
|
Giachello CNG, Fiumara F, Giacomini C, Corradi A, Milanese C, Ghirardi M, Benfenati F, Montarolo PG. MAPK/Erk-dependent phosphorylation of synapsin mediates formation of functional synapses and short-term homosynaptic plasticity. J Cell Sci 2010; 123:881-93. [PMID: 20159961 DOI: 10.1242/jcs.056846] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
MAPK/Erk is a protein kinase activated by neurotrophic factors involved in synapse formation and plasticity, which acts at both the nuclear and cytoplasmic level. Synapsin proteins are synaptic-vesicle-associated proteins that are well known to be MAPK/Erk substrates at phylogenetically conserved sites. However, the physiological role of MAPK/Erk-dependent synapsin phosphorylation in regulating synaptic formation and function is poorly understood. Here, we examined whether synapsin acts as a physiological effector of MAPK/Erk in synaptogenesis and plasticity. To this aim, we developed an in vitro model of soma-to-soma paired Helix B2 neurons, that establish bidirectional excitatory synapses. We found that the formation and activity-dependent short-term plasticity of these synapses is dependent on the MAPK/Erk pathway. To address the role of synapsin in this pathway, we generated non-phosphorylatable and pseudo-phosphorylated Helix synapsin mutants at the MAPK/Erk sites. Overexpression experiments revealed that both mutants interfere with presynaptic differentiation, synapsin clustering, and severely impair post-tetanic potentiation, a form of short-term homosynaptic plasticity. Our findings show that MAPK/Erk-dependent synapsin phosphorylation has a dual role both in the establishment of functional synaptic connections and their short-term plasticity, indicating that some of the multiple extranuclear functions of MAPK/Erk in neurons can be mediated by the same multifunctional presynaptic target.
Collapse
|
33
|
Harrill JA, Freudenrich TM, Machacek DW, Stice SL, Mundy WR. Quantitative assessment of neurite outgrowth in human embryonic stem cell-derived hN2 cells using automated high-content image analysis. Neurotoxicology 2010; 31:277-90. [PMID: 20188755 DOI: 10.1016/j.neuro.2010.02.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 02/09/2010] [Accepted: 02/17/2010] [Indexed: 02/04/2023]
Abstract
Throughout development neurons undergo a number of morphological changes including neurite outgrowth from the cell body. Exposure to neurotoxic chemicals that interfere with this process may result in permanent deficits in nervous system function. Traditionally, rodent primary neural cultures and immortalized human and non-human clonal cell lines have been used to investigate the molecular mechanisms controlling neurite outgrowth and examine chemical effects on this process. The present study characterizes the molecular phenotype of hN2 human embryonic stem cell (hESC)-derived neural cells and uses automated high-content image analysis to measure neurite outgrowth in vitro. At 24h post-plating hN2 cells express a number of protein markers indicative of a neuronal phenotype, including: nestin, beta(III)-tubulin, microtubule-associated protein 2 (MAP2) and phosphorylated neurofilaments. Neurite outgrowth in hN2 cells proceeded rapidly, with a majority of cells extending one to three neurites by 48h in culture. In addition, concentration-dependent decreases in neurite outgrowth and ATP-content were observed following treatment of hN2 cells with either bisindolylmaleimide I, U0126, lithium chloride, sodium orthovanadate and brefeldin A, all of which have previously been shown to inhibit neurite outgrowth in primary rodent neural cultures. Overall, the molecular phenotype, rate of neurite outgrowth and sensitivity of hN2 cells to neurite outgrowth inhibitors were comparable to other in vitro models previously characterized in the literature. hN2 cells provide a model in which to investigate chemical effects on neurite outgrowth in a non-transformed human-derived cells and provide an alternative to the use of primary rodent neural cultures or immortalized clonal cell lines.
Collapse
Affiliation(s)
- Joshua A Harrill
- Systems Biology Branch, Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, NC 27711, United States
| | | | | | | | | |
Collapse
|
34
|
Edwards JL, Quattrini A, Lentz SI, Figueroa-Romero C, Cerri F, Backus C, Hong Y, Feldman EL. Diabetes regulates mitochondrial biogenesis and fission in mouse neurons. Diabetologia 2010; 53:160-9. [PMID: 19847394 PMCID: PMC4011390 DOI: 10.1007/s00125-009-1553-y] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS Normal mitochondrial activity is a critical component of neuronal metabolism and function. Disruption of mitochondrial activity by altered mitochondrial fission and fusion is the root cause of both neurodegenerative disorders and Charcot-Marie-Tooth type 2A inherited neuropathy. This study addressed the role of mitochondrial fission in the pathogenesis of diabetic neuropathy. METHODS Mitochondrial biogenesis and fission were assayed in both in vivo and in vitro models of diabetic neuropathy. Gene, protein, mitochondrial DNA and ultrastructural analyses were used to assess mitochondrial biogenesis and fission. RESULTS There was greater mitochondrial biogenesis in dorsal root ganglion neurons from diabetic compared with non-diabetic mice. An essential step in mitochondrial biogenesis is mitochondrial fission, regulated by the mitochondrial fission protein dynamin-related protein 1 (DRP1). Evaluation of diabetic neurons in vivo indicated small, fragmented mitochondria, suggesting increased fission. In vitro studies revealed that short-term hyperglycaemic exposure increased levels of DRP1 protein. The influence of hyperglycaemia-mediated mitochondrial fission on cell viability was evaluated by knockdown of Drp1 (also known as Dnm1l). Knockdown of Drp1 resulted in decreased susceptibility to hyperglycaemic damage. CONCLUSIONS/INTERPRETATION We propose that: (1) mitochondria undergo biogenesis in response to hyperglycaemia, but the increased biogenesis is insufficient to accommodate the metabolic load; (2) hyperglycaemia causes an excess of mitochondrial fission, creating small, damaged mitochondria; and (3) reduction of aberrant mitochondrial fission increases neuronal survival and indicates an important role for the fission-fusion equilibrium in the pathogenesis of diabetic neuropathy.
Collapse
Affiliation(s)
- J L Edwards
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Sakowski SA, Heavener SB, Lunn JS, Fung K, Oh SS, Spratt SK, Hogikyan ND, Feldman EL. Neuroprotection using gene therapy to induce vascular endothelial growth factor-A expression. Gene Ther 2009; 16:1292-9. [PMID: 19727131 PMCID: PMC4215171 DOI: 10.1038/gt.2009.111] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 07/31/2009] [Accepted: 08/01/2009] [Indexed: 01/19/2023]
Abstract
Engineered zinc-finger protein (ZFP) transcription factors induce the expression of endogenous genes and can be remotely delivered using adenoviral vectors. One such factor, Ad-32Ep65-Flag (Ad-p65), targets and induces expression of vascular endothelial growth factor (VEGF; also called VEGF-A) splice variants in their normal biological stoichiometry. We show that Ad-p65 transfection of primary motor neurons results in VEGF variant expression and a significant increase in axon outgrowth in these cells. Given the neuroprotective effects of VEGF and its ability to increase neurite outgrowth, we examined the efficacy of Ad-p65 to enhance motor neuron regeneration in vivo using rats that have undergone recurrent laryngeal nerve (RLN)-crush injury. Injection of Ad-p65 after RLN crush accelerated the return of vocal fold mobility and the percentage of nerve-endplate contacts in the thyroarytenoid muscle. Overall, adenoviral delivery of an engineered ZFP transcription factor inducing VEGF-A splice variant expression enhances nerve regeneration. ZFP transcription factor gene therapy to increase expression of the full complement of VEGF-A splice variants is a promising avenue for the treatment of nerve injury and neurodegeneration.
Collapse
Affiliation(s)
- Stacey A. Sakowski
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - S. Brett Heavener
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - J. Simon Lunn
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Kevin Fung
- Department of Otolaryngology, University of Western Ontario, London, Ontario, Canada
| | - Sang Su Oh
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | | | - Norman D. Hogikyan
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Lunn JS, Sakowski SA, Kim B, Rosenberg AA, Feldman EL. Vascular endothelial growth factor prevents G93A-SOD1-induced motor neuron degeneration. Dev Neurobiol 2009; 69:871-84. [PMID: 19672955 PMCID: PMC2853013 DOI: 10.1002/dneu.20747] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disorder characterized by selective loss of motor neurons (MNs). Twenty percent of familial ALS cases are associated with mutations in Cu(2+)/Zn(2+) superoxide dismutase (SOD1). To specifically understand the cellular mechanisms underlying mutant SOD1 toxicity, we have established an in vitro model of ALS using rat primary MN cultures transfected with an adenoviral vector encoding a mutant SOD1, G93A-SOD1. Transfected cells undergo axonal degeneration and alterations in biochemical responses characteristic of cell death such as activation of caspase-3. Vascular endothelial growth factor (VEGF) is an angiogenic and neuroprotective growth factor that can increase axonal outgrowth, block neuronal apoptosis, and promote neurogenesis. Decreased VEGF gene expression in mice results in a phenotype similar to that seen in patients with ALS, thus linking loss of VEGF to the pathogenesis of MN degeneration. Decreased neurotrophic signals prior to and during disease progression may increase MN susceptibility to mutant SOD1-induced toxicity. In this study, we demonstrate a decrease in VEGF and VEGFR2 levels in the spinal cord of G93A-SOD1 ALS mice. Furthermore, in isolated MN cultures, VEGF alleviates the effects of G93A-SOD1 toxicity and neuroprotection involves phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling. Overall, these studies validate the usefulness of VEGF as a potential therapeutic factor for the treatment of ALS and give valuable insight into the responsible signaling pathways and mechanisms involved.
Collapse
Affiliation(s)
- J. Simon Lunn
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stacey A. Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
37
|
Kim B, Feldman EL. Insulin receptor substrate (IRS)-2, not IRS-1, protects human neuroblastoma cells against apoptosis. Apoptosis 2009; 14:665-73. [PMID: 19259821 DOI: 10.1007/s10495-009-0331-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Insulin receptor substrates (IRS)-1 and -2 are major substrates of insulin and type I insulin-like growth factor (IGF-I) receptor (IGF-IR) signaling. In this study, SH-EP human neuroblastoma cells are used as a model system to examine the differential roles of IRS-1 and IRS-2 on glucose-mediated apoptosis. In the presence of high glucose, IRS-1 underwent caspase-mediated degradation, followed by focal adhesion kinase (FAK) and Akt degradation and apoptosis. IRS-2 expression blocked all these changes whereas IRS-1 overexpression had no effect. In parallel, IRS-2, but not IRS-1, overexpression enhanced IGF-I-mediated Akt activation without affecting extracellular regulated kinase signaling. While IRS-1 was readily degraded by caspases, hyperglycemia-mediated IRS-2 degradation was unaffected by caspase inhibitors but blocked by proteasome and calpain inhibitors. Our data suggest that the differential degradation of IRS-1 and IRS-2 contributes to their distinct modes of action and the increased neuroprotective effects of IRS-2 in this report are due, in part, to its resistance to caspase-mediated degradation.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of Neurology, University of Michigan, 5371 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA.
| | | |
Collapse
|
38
|
Feliciano DM, Edelman AM. Repression of Ca2+/calmodulin-dependent protein kinase IV signaling accelerates retinoic acid-induced differentiation of human neuroblastoma cells. J Biol Chem 2009; 284:26466-81. [PMID: 19633294 DOI: 10.1074/jbc.m109.027680] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuroblastoma cells having stem cell-like qualities are widely employed models for the study of neural stem/progenitor cell proliferation and differentiation. We find that human BE(2)C neuroblastoma cells possess a signaling cascade initiated by Ca(2+) influx via voltage-dependent calcium channels and the N-methyl-D-aspartate (NMDA) receptor and culminating in nuclear calmodulin-dependent protein kinase IV (CaMKIV)-mediated phosphorylation and activation of the transcription factors Ca(2+)/cyclic AMP-response element-binding protein (CREB) and ATF1 (activating transcription factor-1). This pathway functions to maintain BE(2)C cells in an undifferentiated, proliferative state. Parallel to this Ca(2+)-dependent pathway is a hormone-responsive program by which retinoic acid (RA) initiates the differentiation of BE(2)C cells toward a neuronal lineage. This is evidenced by RA-dependent induction of the cell cycle inhibitor p21/Cip1 (Cdk-interacting protein 1) and cell cycle arrest, induction of the neuroblastic marker doublecortin and of the neuron-specific intermediate filament protein, peripherin, and by RA-stimulated extension of neuritic processes. During neuronal differentiation there is a complex antagonistic interplay between these two major signaling pathways. RA down-regulates expression of CaMKIV and one of its upstream activators, CaMKK1 (calmodulin-dependent protein kinase kinase 1). This is accompanied by RA-induced suppression of activating phosphorylation of CREB with a time course paralleling that of CaMKIV down-regulation. RA-induced repression of the Ca(2+)/calmodulin-dependent protein kinase kinase/CaMKIV/CREB pathway appears to be involved in regulating the timing of neuronal differentiation, as shown by the effect of RNA interference of CaMKIV to markedly accelerate RA-dependent up-regulation of p21/Cip1 and doublecortin expression and RA-promoted neurite outgrowth. RA-induced repression of the CaMKIV signaling pathway may represent an early event in retinoid-dependent neuronal differentiation.
Collapse
Affiliation(s)
- David M Feliciano
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214, USA
| | | |
Collapse
|
39
|
Gokce O, Runne H, Kuhn A, Luthi-Carter R. Short-term striatal gene expression responses to brain-derived neurotrophic factor are dependent on MEK and ERK activation. PLoS One 2009; 4:e5292. [PMID: 19390590 PMCID: PMC2669182 DOI: 10.1371/journal.pone.0005292] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Accepted: 03/26/2009] [Indexed: 12/25/2022] Open
Abstract
Background Brain-derived neurotrophic factor (BDNF) is believed to be an important regulator of striatal neuron survival, differentiation, and plasticity. Moreover, reduction of BDNF delivery to the striatum has been implicated in the pathophysiology of Huntington's disease. Nevertheless, many essential aspects of BDNF responses in striatal neurons remain to be elucidated. Methodology/Principal Findings In this study, we assessed the relative contributions of multipartite intracellular signaling pathways to the short-term induction of striatal gene expression by BDNF. To identify genes regulated by BDNF in these GABAergic cells, we first used DNA microarrays to quantify their transcriptomic responses following 3 h of BDNF exposure. The signal transduction pathways underlying gene induction were subsequently dissected using pharmacological agents and quantitative real-time PCR. Gene expression responses to BDNF were abolished by inhibitors of TrkB (K252a) and calcium (chelator BAPTA-AM and transient receptor potential cation channel [TRPC] antagonist SKF-96365). Interestingly, inhibitors of mitogen-activated protein kinase kinases 1 and 2 (MEK1/2) and extracellular signal-regulated kinase ERK also blocked the BDNF-mediated induction of all tested BDNF-responsive genes. In contrast, inhibitors of nitric oxide synthase (NOS), phosphotidylinositol-3-kinase (PI3K), and CAMK exhibited less prevalent, gene-specific effects on BDNF-induced RNA expression. At the nuclear level, the activation of both Elk-1 and CREB showed MEK dependence. Importantly, MEK-dependent activation of transcription was shown to be required for BDNF-induced striatal neurite outgrowth, providing evidence for its contribution to striatal neuron plasticity. Conclusions These results show that the MEK/ERK pathway is a major mediator of neuronal plasticity and other important BDNF-dependent striatal functions that are fulfilled through the positive regulation of gene expression.
Collapse
Affiliation(s)
- Ozgun Gokce
- Laboratory of Functional Neurogenomics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Heike Runne
- Laboratory of Functional Neurogenomics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alexandre Kuhn
- Laboratory of Functional Neurogenomics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ruth Luthi-Carter
- Laboratory of Functional Neurogenomics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
40
|
Yamaguchi S, Asanoma K, Takao T, Kato K, Wake N. Homeobox gene HOPX is epigenetically silenced in human uterine endometrial cancer and suppresses estrogen-stimulated proliferation of cancer cells by inhibiting serum response factor. Int J Cancer 2009; 124:2577-88. [PMID: 19173292 DOI: 10.1002/ijc.24217] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
HOPX (homeodomain only protein X) is a newly identified homeobox gene whose loss of expression has been reported for several types of neoplasm. Although we found most human uterine endometrial cancers (HEC) defective in HOPX expression, genetic mutations in the HOPX gene were undetectable. As is the case with several tumor suppressor genes, the promoter region of HOPX is densely methylated in HEC tissue samples obtained by laser capture microdissection. HOPX mRNA and protein levels were reduced in the majority of samples, and this correlated with hypermethylation of the HOPX promoter. Forced expression of HOPX resulted in a partial block in cell proliferation, in vivo tumorigenicity and c-fos gene expression in HEC and MCF7 cells in response to 17beta-estradiol (E(2)) stimulation. Analysis of the serum response element (SRE) of c-fos gene promoter showed that the effect of HOPX expression is associated with inhibition of E(2)-induced c-fos activation through the serum response factor (SRF) motif. Knockdown of HOPX in immortalized human endometrial cells resulted in accelerated proliferation. Our study indicates that transcriptional silencing of HOPX results from hypermethylation of the HOPpromoter, which leads to HEC development.
Collapse
Affiliation(s)
- Shinichiro Yamaguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
41
|
Wu H, Ichikawa S, Tani C, Zhu B, Tada M, Shimoishi Y, Murata Y, Nakamura Y. Docosahexaenoic acid induces ERK1/2 activation and neuritogenesis via intracellular reactive oxygen species production in human neuroblastoma SH-SY5Y cells. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:8-16. [DOI: 10.1016/j.bbalip.2008.10.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 08/19/2008] [Accepted: 10/03/2008] [Indexed: 01/21/2023]
|
42
|
Langhorst MF, Jaeger FA, Mueller S, Sven Hartmann L, Luxenhofer G, Stuermer CA. Reggies/flotillins regulate cytoskeletal remodeling during neuronal differentiation via CAP/ponsin and Rho GTPases. Eur J Cell Biol 2008; 87:921-31. [DOI: 10.1016/j.ejcb.2008.07.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2008] [Revised: 06/29/2008] [Accepted: 07/03/2008] [Indexed: 10/21/2022] Open
|
43
|
Abstract
IGF-I and -II are potent neuronal mitogens and survival factors. The actions of IGF-I and -II are mediated via the type I IGF receptor (IGF-IR) and IGF binding proteins regulate the bioavailability of the IGFs. Cell viability correlates with IGF-IR expression and intact IGF-I/IGF-IR signaling pathways, including activation of MAPK/phosphatidylinositol-3 kinase. The expression of IGF-I and -II, IGF-IR, and IGF binding proteins are developmentally regulated in the central and peripheral nervous system. IGF-I therapy demonstrates mixed therapeutic results in the treatment of peripheral nerve injury, neuropathy, and motor neuron diseases such as amyotrophic lateral sclerosis. In this review we discuss the role of IGFs during peripheral nervous system development and the IGF signaling system as the potential therapeutic target for the treatment of nerve injury and motor neuron diseases.
Collapse
Affiliation(s)
- Kelli A Sullivan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | | | | |
Collapse
|
44
|
Wilczak N, De Keyser J, Chesik D. Targeting Insulin-Like Growth Factor-1 Signaling into the Central Nervous System for Promoting Myelin Repair. Drug Target Insights 2008. [DOI: 10.4137/dti.s362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Nadine Wilczak
- Department of Neurology, University Medical Center Groningen, the Netherlands
| | - Jacques De Keyser
- Department of Neurology, University Medical Center Groningen, the Netherlands
| | - Daniel Chesik
- Department of Neurology, University Medical Center Groningen, the Netherlands
| |
Collapse
|
45
|
Meyer GE, Chesler L, Liu D, Gable K, Maddux BA, Goldenberg DD, Youngren JF, Goldfine ID, Weiss WA, Matthay KK, Rosenthal SM. Nordihydroguaiaretic acid inhibits insulin-like growth factor signaling, growth, and survival in human neuroblastoma cells. J Cell Biochem 2007; 102:1529-41. [PMID: 17486636 PMCID: PMC3001106 DOI: 10.1002/jcb.21373] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Neuroblastoma is a common pediatric malignancy that metastasizes to the liver, bone, and other organs. Children with metastatic disease have a less than 50% chance of survival with current treatments. Insulin-like growth factors (IGFs) stimulate neuroblastoma growth, survival, and motility, and are expressed by neuroblastoma cells and the tissues they invade. Thus, therapies that disrupt the effects of IGFs on neuroblastoma tumorigenesis may slow disease progression. We show that NVP-AEW541, a specific inhibitor of the IGF-I receptor (IGF-IR), potently inhibits neuroblastoma growth in vitro. Nordihydroguaiaretic acid (NDGA), a phenolic compound isolated from the creosote bush (Larrea divaricata), has anti-tumor properties against a number of malignancies, has been shown to inhibit the phosphorylation and activation of the IGF-IR in breast cancer cells, and is currently in Phase I trials for prostate cancer. In the present study in neuroblastoma, NDGA inhibits IGF-I-mediated activation of the IGF-IR and disrupts activation of ERK and Akt signaling pathways induced by IGF-I. NDGA inhibits growth of neuroblastoma cells and induces apoptosis at higher doses, causing IGF-I-resistant activation of caspase-3 and a large increase in the fraction of sub-G0 cells. In addition, NDGA inhibits the growth of xenografted human neuroblastoma tumors in nude mice. These results indicate that NDGA may be useful in the treatment of neuroblastoma and may function in part via disruption of IGF-IR signaling.
Collapse
Affiliation(s)
- Gary E. Meyer
- Department of Pediatrics, University of California, San Francisco, California
| | - Louis Chesler
- Department of Pediatrics, University of California, San Francisco, California
| | - Dandan Liu
- Department of Pediatrics, University of California, San Francisco, California
| | - Karissa Gable
- Diabetes and Endocrine Research, University of California, San Francisco/Mt. Zion Medical Center, San Francisco, California
| | - Betty A. Maddux
- Diabetes and Endocrine Research, University of California, San Francisco/Mt. Zion Medical Center, San Francisco, California
| | - David D. Goldenberg
- Department of Neurology, University of California, San Francisco, California
| | - Jack F. Youngren
- Diabetes and Endocrine Research, University of California, San Francisco/Mt. Zion Medical Center, San Francisco, California
| | - Ira D. Goldfine
- Diabetes and Endocrine Research, University of California, San Francisco/Mt. Zion Medical Center, San Francisco, California
| | - William A. Weiss
- Department of Neurology, University of California, San Francisco, California
| | | | | |
Collapse
|
46
|
Schwab TS, Madison BB, Grauman AR, Feldman EL. Insulin-like growth factor-I induces the phosphorylation and nuclear exclusion of forkhead transcription factors in human neuroblastoma cells. Apoptosis 2007; 10:831-40. [PMID: 16133873 DOI: 10.1007/s10495-005-0429-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Akt-mediated phosphorylation of forkhead transcription factors is linked to growth factor-stimulated cell survival. We investigated whether the survival activity of insulin-like growth factor-I (IGF-I) in SH-SY5Y human neuroblastoma (NBL) cells is associated with phosphorylation and/or localization changes in forkhead proteins. IGF-I induced phosphorylation of Erks (p42/p44), FKHR (FOXO1a) (Ser 253), FKHRL1 (FOXO3a) (Ser 256), and Akt (Ser 473). PI3-K inhibitor, LY294002, reduced IGF-I-stimulated phosphorylation of FKHR, FKHRL1, and Akt, but did not affect Erk phosphorylation. Using a GFP-FKHR construct, FKHR imported into the nucleus during growth factor withdrawal-induced apoptosis. In addition, IGF-I rescue from serum withdrawal-induced apoptosis is associated with a rapid export of GFP-FKHR into the cytoplasm. Leptomycin B, an inhibitor of Crm1-mediated nuclear export, decreased the level of FKHRL1 phosphorylation in the presence of IGF-I in vector and FKHR overexpressing cells, but had no effect on the phosphorylation status of FKHR. In addition, leptomycin B prevented IGF-I stimulated nuclear export of GFP-FKHR. These studies show IGF-I phosphorylation of FKHR and FKHRL1 via a PI3-K-dependent pathway in NBL cells.
Collapse
Affiliation(s)
- T S Schwab
- Department of Neurology, University of Michigan, 4414 Kresge III, 200 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
47
|
Delaune A, Corbière C, Benjelloun FD, Legrand E, Vannier JP, Ripoll C, Vasse M. Promyelocytic leukemia-nuclear body formation is an early event leading to retinoic acid-induced differentiation of neuroblastoma cells. J Neurochem 2007; 104:89-99. [PMID: 17986232 DOI: 10.1111/j.1471-4159.2007.05019.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neuroblastoma is one of the most common cancers in children. Neuroblastoma differentiation is linked to the presence of the promyelocytic leukemia (PML) protein. Retinoic acid, a powerful differentiation-inducer in vitro, is a potent agent for the treatment of neuroblastoma. Using two different human neuroblastoma cell lines, SH-SY5Y and LA-N-5, we show here that PML protein leads to the formation of nuclear bodies (PML-NB) after only 1 h of retinoic acid treatment and that this formation is mediated by the extracellular signal-regulated kinase (ERK) pathway. Inhibition of protein kinase C also leads to formation of PML-NB via the ERK pathway. Both sumoylation and phosphorylation of PML in an ERK-dependent pathway are also required for formation of PML-NB. Finally, we show that PML-NB formation in neuroblastoma cells is associated with neurite outgrowth. These results support the proposal that the formation of PML-NB is correlated with the differentiation of neuroblastoma cells.
Collapse
Affiliation(s)
- A Delaune
- Groupe de recherche MERCI & IHURBM, Faculté de médecine et pharmacie, Rouen, France
| | | | | | | | | | | | | |
Collapse
|
48
|
Grotzer MA, Guerreiro AS, Bourquin JP, Arcaro A. IGF signaling as a therapeutic target in pediatric solid tumors of the central and peripheral nervous system. Expert Rev Endocrinol Metab 2007; 2:677-688. [PMID: 30736130 DOI: 10.1586/17446651.2.5.677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Similar to many other growth factor systems, the IGF system consists of more than a single ligand interacting with a single receptor. There are three ligands (IGF-I, IGF-II and insulin) that interact with at least four receptors. In addition, the IGF system also involves six well-characterized binding proteins that regulate IGF action. Type I IGF receptor-mediated signaling plays a fundamental role in cell growth and malignant transformation and is an important mediator of anti-apoptotic signals. This review describes the roles of IGF signaling in childhood tumors of the CNS and PNS, including neuroblastoma, medulloblastoma, atypical teratoid/rhabdoid tumors and craniopharyngioma. Moreover, it describes strategies to disrupt the IGF signaling as a potential cancer therapy.
Collapse
Affiliation(s)
- Michael A Grotzer
- a University Children's Hospital of Zurich, Division of Oncology, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland.
| | - Ana S Guerreiro
- b University Children's Hospital of Zurich, Division of Clinical Chemistry and Biochemistry, Zurich, Switzerland
| | - Jean-Pierre Bourquin
- c University Children's Hospital of Zurich, Division of Oncology, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland
| | - Alexandre Arcaro
- b University Children's Hospital of Zurich, Division of Clinical Chemistry and Biochemistry, Zurich, Switzerland
| |
Collapse
|
49
|
Huang CW, Huang CC, Cheng JT, Tsai JJ, Wu SN. Glucose and hippocampal neuronal excitability: role of ATP-sensitive potassium channels. J Neurosci Res 2007; 85:1468-77. [PMID: 17410601 DOI: 10.1002/jnr.21284] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Hyperglycemia-related neuronal excitability and epileptic seizures are not uncommon in clinical practice. However, their underlying mechanism remains elusive. ATP-sensitive K(+) (K(ATP)) channels are found in many excitable cells, including cardiac myocytes, pancreatic beta cells, and neurons. These channels provide a link between the electrical activity of cell membranes and cellular metabolism. We investigated the effects of higher extracellular glucose on hippocampal K(ATP) channel activities and neuronal excitability. The cell-attached patch-clamp configuration on cultured hippocampal cells and a novel multielectrode recording system on hippocampal slices were employed. In addition, a simulation modeling hippocampal CA3 pyramidal neurons (Pinsky-Rinzel model) was analyzed to investigate the role of K(ATP) channels in the firing of simulated action potentials. We found that incremental extracellular glucose could attenuate the activities of hippocampal K(ATP) channels. The effect was concentration dependent and involved mainly in open probabilities, not single-channel conductance. Additionally, higher levels of extracellular glucose could enhance neuropropagation; this could be attenuated by diazoxide, a K(ATP) channel agonist. In simulations, high levels of intracellular ATP, used to mimic increased extracellular glucose or reduced conductance of K(ATP) channels, enhanced the firing of action potentials in model neurons. The stochastic increases in intracellular ATP levels also demonstrated an irregular and clustered neuronal firing pattern. This phenomenon of K(ATP) channel attenuation could be one of the underlying mechanisms of glucose-related neuronal hyperexcitability and propagation.
Collapse
Affiliation(s)
- Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Medical Center, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
50
|
Huang CW, Huang CC, Wu SN. Activation by zonisamide, a newer antiepileptic drug, of large-conductance calcium-activated potassium channel in differentiated hippocampal neuron-derived H19-7 cells. J Pharmacol Exp Ther 2007; 321:98-106. [PMID: 17255467 DOI: 10.1124/jpet.106.116954] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Zonisamide (ZNS; 3-sulfamoylmethyl-1,2-benzisoxazole), as one of the newer antiepileptic drugs, has been demonstrated its broad-spectrum clinical efficacy on various neuropsychiatric disorders. However, little is known regarding the mechanism of ZNS actions on ion currents in neurons. We thus investigated its effect on ion currents in differentiated hippocampal 19-7 cells. In whole-cell configuration of patch-clamp technology, the ZNS (30 microM) reversibly increased the amplitude of K+ outward currents, and paxilline (1 microM) was effective in suppressing the ZNS-induced increase of K+ outward currents. In inside-out configuration, ZNS (30 microM) applied to the intracellular face of the membrane did not alter single-channel conductance; however, it did enhance the activity of large-conductance Ca2+-activated K+ (BK(Ca)) channels primarily by decreasing mean closed time. In addition, the EC50 value for ZNS-stimulated BK(Ca) channels was 34 microM. This drug caused a left shift in the activation curve of BK(Ca) channels, with no change in the gating charge of these channels. Moreover, ZNS at a concentration greater than 100 microM also reduced the amplitude of A-type K+ current in these cells. A simulation modeling based on hippocampal CA3 pyramidal neurons (Pinsky-Rinzel model) was also analyzed to investigate the inhibitory effect of ZNS on the firing of simulated action potentials. Taken together, this study suggests that, in hippocampal neurons during the exposure to ZNS, the ZNS-mediated effects on BK(Ca) channels and A-type K+ current could be potential mechanisms through which it affects neuronal excitability.
Collapse
Affiliation(s)
- Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Medical College, 1 University Road, Tainan 70101, Taiwan
| | | | | |
Collapse
|