1
|
Tian X, Yan H, Li J, Wu S, Wang J, Fan L. Neurotrophin Promotes Neurite Outgrowth by Inhibiting Rif GTPase Activation Downstream of MAPKs and PI3K Signaling. Int J Mol Sci 2017; 18:E148. [PMID: 28098758 PMCID: PMC5297781 DOI: 10.3390/ijms18010148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/01/2017] [Accepted: 01/06/2017] [Indexed: 12/16/2022] Open
Abstract
Members of the well-known semaphorin family of proteins can induce both repulsive and attractive signaling in neural network formation and their cytoskeletal effects are mediated in part by small guanosine 5'-triphosphatase (GTPases). The aim of this study was to investigate the cellular role of Rif GTPase in the neurotrophin-induced neurite outgrowth. By using PC12 cells which are known to cease dividing and begin to show neurite outgrowth responding to nerve growth factor (NGF), we found that semaphorin 6A was as effective as nerve growth factor at stimulating neurite outgrowth in PC12 cells, and that its neurotrophic effect was transmitted through signaling by mitogen-activated protein kinases (MAPKs) and phosphatidylinositol-3-kinase (PI3K). We further found that neurotrophin-induced neurite formation in PC12 cells could be partially mediated by inhibition of Rif GTPase activity downstream of MAPKs and PI3K signaling. In conclusion, we newly identified Rif as a regulator of the cytoskeletal rearrangement mediated by semaphorins.
Collapse
Affiliation(s)
- Xiaoxia Tian
- School of Life Sciences, Inner Mongolia University, Hohhot 010021, China.
| | - Huijuan Yan
- School of Life Sciences, Inner Mongolia University, Hohhot 010021, China.
| | - Jiayi Li
- School of Life Sciences, Inner Mongolia University, Hohhot 010021, China.
| | - Shuang Wu
- School of Life Sciences, Inner Mongolia University, Hohhot 010021, China.
| | - Junyu Wang
- School of Life Sciences, Inner Mongolia University, Hohhot 010021, China.
| | - Lifei Fan
- School of Life Sciences, Inner Mongolia University, Hohhot 010021, China.
| |
Collapse
|
2
|
Sury MD, McShane E, Hernandez-Miranda LR, Birchmeier C, Selbach M. Quantitative proteomics reveals dynamic interaction of c-Jun N-terminal kinase (JNK) with RNA transport granule proteins splicing factor proline- and glutamine-rich (Sfpq) and non-POU domain-containing octamer-binding protein (Nono) during neuronal differentiation. Mol Cell Proteomics 2014; 14:50-65. [PMID: 25326457 DOI: 10.1074/mcp.m114.039370] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The c-Jun N-terminal kinase (JNK) is an important mediator of physiological and pathophysiological processes in the central nervous system. Importantly, JNK not only is involved in neuronal cell death, but also plays a significant role in neuronal differentiation and regeneration. For example, nerve growth factor induces JNK-dependent neuronal differentiation in several model systems. The mechanism by which JNK mediates neuronal differentiation is not well understood. Here, we employed a proteomic strategy to better characterize the function of JNK during neuronal differentiation. We used SILAC-based quantitative proteomics to identify proteins that interact with JNK in PC12 cells in a nerve growth factor-dependent manner. Intriguingly, we found that JNK interacted with neuronal transport granule proteins such as Sfpq and Nono upon NGF treatment. We validated the specificity of these interactions by showing that they were disrupted by a specific peptide inhibitor that blocks the interaction of JNK with its substrates. Immunoprecipitation and Western blotting experiments confirmed the interaction of JNK1 with Sfpq/Nono and demonstrated that it was RNA dependent. Confocal microscopy indicated that JNK1 associated with neuronal granule proteins in the cytosol of PC12 cells, primary cortical neurons, and P19 neuronal cells. Finally, siRNA experiments confirmed that Sfpq was necessary for neurite outgrowth in PC12 cells and that it most likely acted in the same pathway as JNK. In summary, our data indicate that the interaction of JNK1 with transport granule proteins in the cytosol of differentiating neurons plays an important role during neuronal development.
Collapse
Affiliation(s)
- Matthias D Sury
- From ‡Cell Signaling/Mass Spectrometry, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Erik McShane
- From ‡Cell Signaling/Mass Spectrometry, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Luis Rodrigo Hernandez-Miranda
- §Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Carmen Birchmeier
- §Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Matthias Selbach
- From ‡Cell Signaling/Mass Spectrometry, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany;
| |
Collapse
|
3
|
Triptolide improves nerve regeneration and functional recovery following crush injury to rat sciatic nerve. Neurosci Lett 2014; 561:198-202. [DOI: 10.1016/j.neulet.2013.12.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 12/28/2013] [Accepted: 12/30/2013] [Indexed: 01/24/2023]
|
4
|
Mechanisms of action of brain insulin against neurodegenerative diseases. J Neural Transm (Vienna) 2014; 121:611-26. [PMID: 24398779 DOI: 10.1007/s00702-013-1147-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 12/17/2013] [Indexed: 12/13/2022]
Abstract
Insulin, a pancreatic hormone, is best known for its peripheral effects on the metabolism of glucose, fats and proteins. There is a growing body of evidence linking insulin action in the brain to neurodegenerative diseases. Insulin present in central nervous system is a regulator of central glucose metabolism nevertheless this glucoregulation is not the main function of insulin in the brain. Brain is known to be specifically vulnerable to oxidative products relative to other organs and altered brain insulin signaling may cause or promote neurodegenerative diseases which invalidates and reduces the quality of life. Insulin located within the brain is mostly of pancreatic origin or is produced in the brain itself crosses the blood-brain barrier and enters the brain via a receptor-mediated active transport system. Brain Insulin, insulin receptor and insulin receptor substrate-mediated signaling pathways play important roles in the regulation of peripheral metabolism, feeding behavior, memory and maintenance of neural functions such as neuronal growth and differentiation, neuromodulation and neuroprotection. In the present review, we would like to summarize the novel biological and pathophysiological roles of neuronal insulin in neurodegenerative diseases and describe the main signaling pathways in use for therapeutic strategies in the use of insulin to the cerebral tissues and their biological applications to neurodegenerative diseases.
Collapse
|
5
|
Claulansine F promotes neuritogenesis in PC12 cells via the ERK signaling pathway. Acta Pharmacol Sin 2013; 34:1499-507. [PMID: 24096602 DOI: 10.1038/aps.2013.95] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 06/24/2013] [Indexed: 12/21/2022]
Abstract
AIM To study the effects of Claulansine F (Clau F), a carbazole alkaloid isolated from the stem of Clausena lansium (Lour) Skeels, on neuritogenesis of PC12 cells, and to elucidate the mechanism of action. METHODS Neuritogenesis of PC12 cells was quantified under an inverted microscope. Expression of the neurite outgrowth marker GAP-43 was detected using immunofluorescence. GAP-43 transcription was measured using RT-PCR. Cell viability was evaluated with MTT assay. The levels of phosphor-ERK1/2, phosphor-CREB, phosphor-AKT and acetylate-p53 in the cells were examined using Western blotting analyses. RESULTS Clau F (10-100 μmol/L) significantly increased the percentage of PC12 cells bearing neurites. Clau F markedly increased the expression of GAP-43 in the cells. The efficiency of Clau F (10 μmol/L) in increasing neuritogenesis and GAP-43 expression was comparable to that of nerve growth factor (50 ng/mL). In addition, Clau F completely blocked the proliferation of PC12 cells within 7 d of incubation, whereas it did not cause cell death in cultured rat cortical neurons. Treatment of PC12 cells with Clau F activated both ERK and AKT signaling pathways. Co-treatment of PC12 cells with the specific ERK inhibitor PD98059, but not the specific PI3K inhibitor LY294002, blocked Clau F-induced neuritogenesis and GAP-43 upregulation. CONCLUSION Clau F promotes neuritogenesis in PC12 cells specifically via activation of the ERK signaling pathway.
Collapse
|
6
|
Su Y, Cui L, Piao C, Li B, Zhao LR. The effects of hematopoietic growth factors on neurite outgrowth. PLoS One 2013; 8:e75562. [PMID: 24116056 PMCID: PMC3792965 DOI: 10.1371/journal.pone.0075562] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 08/14/2013] [Indexed: 01/06/2023] Open
Abstract
Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) are initially discovered as the essential hematopoietic growth factors regulating bone marrow stem cell proliferation and differentiation, and SCF in combination with G-CSF (SCF+G-CSF) has synergistic effects on bone marrow stem cell mobilization. In this study we have determined the effect of SCF and G-CSF on neurite outgrowth in rat cortical neurons. Using molecular and cellular biology and live cell imaging approaches, we have revealed that receptors for SCF and G-CSF are expressed on the growth core of cortical neurons, and that SCF+G-CSF synergistically enhances neurite extension through PI3K/AKT and NFκB signaling pathways. Moreover, SCF+G-CSF induces much greater NFκB activation, NFκB transcriptional binding and brain-derived neurotrophic factor (BDNF) production than SCF or G-CSF alone. In addition, we have also observed that BDNF, the target gene of NFκB, is required for SCF+G-CSF-induced neurite outgrowth. These data suggest that SCF+G-CSF has synergistic effects to promote neurite growth. This study provides new insights into the contribution of hematopoietic growth factors in neuronal plasticity.
Collapse
Affiliation(s)
- Ye Su
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Lili Cui
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Chunshu Piao
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Bin Li
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Li-Ru Zhao
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
7
|
Nakamura T, Yasuda S, Nagai H, Koinuma S, Morishita S, Goto A, Kinashi T, Wada N. Longest neurite-specific activation of Rap1B in hippocampal neurons contributes to polarity formation through RalA and Nore1A in addition to PI3-kinase. Genes Cells 2013; 18:1020-31. [DOI: 10.1111/gtc.12097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Accepted: 08/13/2013] [Indexed: 12/22/2022]
Affiliation(s)
- Takeshi Nakamura
- Division of Biosignaling, Research Institute for Biomedical Sciences; Tokyo University of Science; Noda Chiba 278-0022 Japan
| | - Sayaka Yasuda
- Division of Biosignaling, Research Institute for Biomedical Sciences; Tokyo University of Science; Noda Chiba 278-0022 Japan
| | - Hiroyuki Nagai
- Division of Biosignaling, Research Institute for Biomedical Sciences; Tokyo University of Science; Noda Chiba 278-0022 Japan
| | - Shingo Koinuma
- Division of Biosignaling, Research Institute for Biomedical Sciences; Tokyo University of Science; Noda Chiba 278-0022 Japan
| | - So Morishita
- Division of Biosignaling, Research Institute for Biomedical Sciences; Tokyo University of Science; Noda Chiba 278-0022 Japan
- Department of Applied Biological Science; Tokyo University of Science; Noda Chiba 278-8510 Japan
| | - Akihiro Goto
- Laboratory of Bioimaging and Cell Signaling; Graduate School of Biostudies; Kyoto University; Kyoto 606-8501 Japan
| | - Tatsuo Kinashi
- Department of Molecular Genetics; Kansai Medical University; Osaka 573-1010 Japan
| | - Naoyuki Wada
- Department of Applied Biological Science; Tokyo University of Science; Noda Chiba 278-8510 Japan
| |
Collapse
|
8
|
Quintanilla RA, Godoy JA, Alfaro I, Cabezas D, von Bernhardi R, Bronfman M, Inestrosa NC. Thiazolidinediones promote axonal growth through the activation of the JNK pathway. PLoS One 2013; 8:e65140. [PMID: 23741474 PMCID: PMC3669289 DOI: 10.1371/journal.pone.0065140] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 04/22/2013] [Indexed: 11/18/2022] Open
Abstract
The axon is a neuronal process involved in protein transport, synaptic plasticity, and neural regeneration. It has been suggested that their structure and function are profoundly impaired in neurodegenerative diseases. Previous evidence suggest that Peroxisome Proliferator-Activated Receptors-γ (PPARγ promote neuronal differentiation on various neuronal cell types. In addition, we demonstrated that activation of PPARγby thiazolidinediones (TZDs) drugs that selectively activate PPARγ prevent neurite loss and axonal damage induced by amyloid-β (Aβ). However, the potential role of TZDs in axonal elongation and neuronal polarity has not been explored. We report here that the activation of PPARγ by TZDs promoted axon elongation in primary hippocampal neurons. Treatments with different TZDs significantly increased axonal growth and branching area, but no significant effects were observed in neurite elongation compared to untreated neurons. Treatment with PPARγ antagonist (GW 9662) prevented TZDs-induced axonal growth. Recently, it has been suggested that the c-Jun N-terminal kinase (JNK) plays an important role regulating axonal growth and neuronal polarity. Interestingly, in our studies, treatment with TZDs induced activation of the JNK pathway, and the pharmacological blockage of this pathway prevented axon elongation induced by TZDs. Altogether, these results indicate that activation of JNK induced by PPARγactivators stimulates axonal growth and accelerates neuronal polarity. These novel findings may contribute to the understanding of the effects of PPARγ on neuronal differentiation and validate the use of PPARγ activators as therapeutic agents in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rodrigo A. Quintanilla
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratorio de Neurociencias, Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan A. Godoy
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ivan Alfaro
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Deny Cabezas
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rommy von Bernhardi
- Laboratorio de Neurociencias, Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel Bronfman
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C. Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
9
|
Phospholipase D1 mediates bFGF-induced Bcl-2 expression leading to neurite outgrowth in H19-7 cells. Biochem J 2012; 441:407-16. [PMID: 21916846 DOI: 10.1042/bj20110302] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The purpose of the present study was to investigate the role of PLD (phospholipase D) in bFGF (basic fibroblast growth factor)-induced Bcl-2 expression and to examine whether overexpressed Bcl-2 influences neurite outgrowth in immortalized hippocampal progenitor cells (H19-7 cells). We found that Bcl-2 expression was maximally induced by bFGF within 24 h, and that this effect was reduced by inhibiting PLD1 expression with PLD1 small interfering RNA or by overexpressing DN (dominant-negative)-PLD1, whereas PLD1 overexpression markedly induced Bcl-2 expression. bFGF treatment activated Ras, Src, PI3K (phosphoinositide 3-kinase), PLCγ (phospholipase Cγ) and PKCα (protein kinase Cα). Among these molecules, Src and PKCα were not required for Bcl-2 expression. PLD activity was decreased by Ras, PI3K or PLCγ inhibitor, suggesting that PLD1 activation occurred through Ras, PI3K or PLCγ. We found that Ras was the most upstream molecule among these proteins, followed by the PI3K/PLCγ pathway, indicating that bFGF-induced PLD activation took place through the Ras/PI3K/PLCγ pathway. Furthermore, PLD1 was required for activation of JNK (c-Jun N-terminal kinase), which led to activation of STAT3 (signal transducer and activator of transcription 3) and finally Bcl-2 expression. When Bcl-2 was overexpressed, neurite outgrowth was stimulated along with induction of neurotrophic factors such as brain-derived neurotrophic factor and neurotrophin 4/5. In conclusion, PLD1 acts as a downstream effector of bFGF/Ras/PI3K/PLCγ signalling and regulates Bcl-2 expression through JNK/STAT3, which leads to neurite outgrowth in H19-7 cells.
Collapse
|
10
|
Insulin promotes dendritic spine and synapse formation by the PI3K/Akt/mTOR and Rac1 signaling pathways. Neuropharmacology 2011; 61:867-79. [PMID: 21683721 DOI: 10.1016/j.neuropharm.2011.06.003] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 05/12/2011] [Accepted: 06/02/2011] [Indexed: 12/13/2022]
Abstract
Insulin and its receptor are broadly expressed throughout the brain and have been postulated to play a crucial role in synaptic plasticity. Although structural remodeling of dendritic spines is associated with stable expression of synaptic plasticity, the role of insulin receptor (IR) signaling in the establishment and dynamic changes of dendritic spines remains unclear. Here we report that insulin promotes dendritic spine formation in primary cultures of rat hippocampal neurons. Conversely, downregulation of IR signaling using a blocking antibody or short hairpin RNAs (shRNAs) resulted in a decrease in number of dendritic spines and caused a significant reduction in the frequency of miniature excitatory postsynaptic currents (mEPSCs) without affecting the distribution of their amplitudes. Pharmacological blockade of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway and the small GTPase Rac1 specifically prevented the insulin-induced increase in dendritic spine density. In parallel, genetic ablation of Rac1 expression by lentiviral infection with shRNA abrogated the increase in dendritic spines induced by insulin. More importantly, the increase in dendritic spine density by insulin was accompanied by increasing in presynaptic marker staining density and displayed an increase in mEPSC frequency. Taken together, these results reveal a novel role for IR signaling in the regulation of dendritic spine formation and excitatory synapse development in hippocampal neurons through activation of the PI3K/Akt/mTOR and Rac1 signaling pathways.
Collapse
|
11
|
Haeusgen W, Herdegen T, Waetzig V. MKK7γ1 reverses nerve growth factor signals: proliferation and cell death instead of neuritogenesis and protection. Cell Signal 2011; 23:1281-90. [PMID: 21406225 DOI: 10.1016/j.cellsig.2011.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 03/03/2011] [Indexed: 12/31/2022]
Abstract
c-Jun N-terminal kinases (JNKs) are the exclusive downstream substrates of mitogen-activated protein kinase kinase 7 (MKK7). Recently, we have shown that a single MKK7 splice variant, MKK7γ1, substantially changes the functions of JNKs in naïve PC12 cells. Here we provide evidence that MKK7γ1 blocks NGF-mediated differentiation and sustains proliferation by interfering with the NGF-triggered differentiation programme at several levels: (i) down-regulation of the NGF receptors TrkA and p75; (ii) attenuation of the differentiation-promoting pathways ERK1/2 and AKT; (iii) increase of JNK1 and JNK2, especially the JNK2 54kDa splice variants; (iv) repression of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1), which normally supports NGF-mediated cell cycle arrest; (v) strong induction of the cell cycle promoter CyclinD1, and (vi) profound changes of p53 functions. Moreover, MKK7γ1 substantially changes the responsiveness to stress. Whereas NGF differentiation protects PC12 cells against taxol-induced apoptosis, MKK7γ1 triggers an escape from cell cycle arrest and renders transfected cells sensitive to taxol-induced death. This stress response completely differs from naïve PC12 cells, where MKK7γ1 protects against taxol-induced cell death. These novel aspects on the regulation of JNK signalling emphasise the importance of MKK7γ1 in its ability to reverse basic cellular programmes by simply using JNKs as effectors. Furthermore, our results highlight the necessity for the cells to balance the expression of JNK activators to ensure precise intracellular processes.
Collapse
Affiliation(s)
- Wiebke Haeusgen
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Hospitalstrasse 4, 24105 Kiel, Germany
| | | | | |
Collapse
|
12
|
Mehan S, Meena H, Sharma D, Sankhla R. JNK: A Stress-Activated Protein Kinase Therapeutic Strategies and Involvement in Alzheimer’s and Various Neurodegenerative Abnormalities. J Mol Neurosci 2010; 43:376-90. [DOI: 10.1007/s12031-010-9454-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 09/16/2010] [Indexed: 01/26/2023]
|
13
|
Yao L, McCaig CD, Zhao M. Electrical signals polarize neuronal organelles, direct neuron migration, and orient cell division. Hippocampus 2009; 19:855-68. [DOI: 10.1002/hipo.20569] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
14
|
Haeusgen W, Boehm R, Zhao Y, Herdegen T, Waetzig V. Specific activities of individual c-Jun N-terminal kinases in the brain. Neuroscience 2009; 161:951-9. [DOI: 10.1016/j.neuroscience.2009.04.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/06/2009] [Accepted: 04/06/2009] [Indexed: 12/31/2022]
|
15
|
Waetzig V, Loose K, Haeusgen W, Herdegen T. c-Jun N-terminal kinases mediate Fas-induced neurite regeneration in PC12 cells. Biochem Pharmacol 2008; 76:1476-84. [DOI: 10.1016/j.bcp.2008.07.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 07/10/2008] [Accepted: 07/11/2008] [Indexed: 12/27/2022]
|
16
|
Tso PH, Morris CJ, Yung LY, Ip NY, Wong YH. Multiple Gi Proteins Participate in Nerve Growth Factor-Induced Activation of c-Jun N-terminal Kinases in PC12 Cells. Neurochem Res 2008; 34:1101-12. [DOI: 10.1007/s11064-008-9880-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2008] [Indexed: 01/21/2023]
|
17
|
Kanai A, Ihara S, Ohdaira T, Shinohara-Kanda A, Iwamatsu A, Fukui Y. Identification of DOCK4 and its splicing variant as PIP3 binding proteins. IUBMB Life 2008; 60:467-72. [PMID: 18459162 DOI: 10.1002/iub.67] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
DOCK4, a member of DOCK180 family proteins, was originally identified as a product of a gene deleted during tumor progression. Although its tumor suppression properties have been reported, the regulation mechanism of this protein has not been fully elucidated. DOCK4 shares two conserved domains called as DHR-1 and DHR-2 domain as other members including DOCK180. Although DHR-1 in DOCK180 is reported to bind to PIP(3), whether that of DOCK4 exhibits similar function has yet not been examined. In a search for novel PIP(3) binding proteins by the PIP(3) analog beads binding assay, we found that DOCK4 and its novel splicing variant, whose exon1 and exon52 are different from the known one, bind to PIP(3). Binding assay using deletion mutants of DOCK4 revealed that the binding region falls into the DHR-1 domain. These results raise the possibility that DOCK4 may be regulated by PIP(3) to exert its function.
Collapse
Affiliation(s)
- Akinori Kanai
- Division of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | |
Collapse
|
18
|
Bernardino L, Agasse F, Silva B, Ferreira R, Grade S, Malva JO. Tumor necrosis factor-alpha modulates survival, proliferation, and neuronal differentiation in neonatal subventricular zone cell cultures. Stem Cells 2008; 26:2361-71. [PMID: 18583543 DOI: 10.1634/stemcells.2007-0914] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor (TNF)-alpha has been reported to modulate brain injury, but remarkably, little is known about its effects on neurogenesis. We report that TNF-alpha strongly influences survival, proliferation, and neuronal differentiation in cultured subventricular zone (SVZ) neural stem/progenitor cells derived from the neonatal P1-3 C57BL/6 mice. By using single-cell calcium imaging, we developed a method, based on cellular response to KCl and/or histamine, that allows the functional evaluation of neuronal differentiation. Exposure of SVZ cultures to 1 and 10 ng/ml mouse or 1 ng/ml human recombinant TNF-alpha resulted in increased differentiation of cells displaying a neuronal-like profile of [Ca2+](i) responses, compared with the predominant profile of immature cells observed in control, nontreated cultures. Moreover, by using neutralizing antibodies for each TNF-alpha receptor, we found that the proneurogenic effect of 1 ng/ml TNF-alpha is mediated via tumor necrosis factor receptor 1 activation. Accordingly, the percentage of neuronal nuclear protein-positive neurons was increased following exposure to mouse TNF-alpha. Interestingly, exposure of SVZ cultures to 1 ng/ml TNF-alpha induced cell proliferation, whereas 10 and 100 ng/ml TNF-alpha induced apoptotic cell death. Moreover, we found that exposure of SVZ cells to TNF-alpha for 15 minutes or 6 hours caused an increase in the phospho-stress-activated protein kinase/c-Jun N-terminal kinase immunoreactivity initially in the nucleus and then in growing axons, colocalizing with tau, consistent with axonogenesis. Taken together, these results show that TNF-alpha induces neurogenesis in neonatal SVZ cell cultures of mice. TNF-alpha, a proinflammatory cytokine and a proneurogenic factor, may play a central role in promoting neurogenesis and brain repair in response to brain injury and infection.
Collapse
Affiliation(s)
- Liliana Bernardino
- Center for Neuroscience and Cell Biology, Institute of Biochemistry, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | | | | | | | | |
Collapse
|
19
|
Atsumi T, Kato K, Uno K, Iijima K, Koike T, Imatani A, Ohara S, Shimosegawa T. Pathophysiological role of the activation of p38 mitogen-activated protein kinases in poorly differentiated gastric cancer. Pathol Int 2007; 57:635-44. [PMID: 17803652 DOI: 10.1111/j.1440-1827.2007.02152.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
p38 mitogen-activated protein kinases (MAPK) contribute to the loss of cell-cell contact and the round cell shape characteristic of poorly differentiated gastric cancer. In the present study it is demonstrated that phospho-p38 MAPK level significantly increased in poorly differentiated gastric cancers in comparison to differentiated cancers and normal gastric mucosa by immunohistochemistry. Next, the pathophysiological roles of p38 MAPK activation were investigated in differentiated gastric cancer cell lines MKN7 and MKN28 and poorly differentiated gastric cancer cell lines KATO-III and MKN45 cells by incubating with specific p38 inhibitor SB203580 or inactivating analog SB202474. The distribution of F-actin on phalloidin staining was identified as fine cytoskeletal filaments in MKN7 and MKN28, but as dense membranous accumulation in KATO-III and MKN45 cells. The treatment with SB203580 but not SB202474 reduced irregular accumulation of F-actin in KATO-III and MKN45 cells. The expression of E-cadherin, ZO-1, occludin and claudin 4 was higher in MKN7 and MKN28 than KATO-III and MKN45 cells. The expression of E-cadherin in KATO-III cells was increased following treatment with SB203580, suggesting the suppression of E-cadherin at the transcriptional level independent of its genetic alterations. Thus, p38 MAPK signaling might contribute to the acquisition of malignant properties in poorly differentiated phenotypes.
Collapse
Affiliation(s)
- Tomoaki Atsumi
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Freund-Michel V, Frossard N. The nerve growth factor and its receptors in airway inflammatory diseases. Pharmacol Ther 2007; 117:52-76. [PMID: 17915332 DOI: 10.1016/j.pharmthera.2007.07.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 07/30/2007] [Indexed: 11/16/2022]
Abstract
The nerve growth factor (NGF) belongs to the neurotrophin family and induces its effects through activation of 2 distinct receptor types: the tropomyosin-related kinase A (TrkA) receptor, carrying an intrinsic tyrosine kinase activity in its intracellular domain, and the receptor p75 for neurotrophins (p75NTR), belonging to the death receptor family. Through activation of its TrkA receptor, NGF activates signalling pathways, including phospholipase Cgamma (PLCgamma), phosphatidyl-inositol 3-kinase (PI3K), the small G protein Ras, and mitogen-activated protein kinases (MAPK). Through its p75NTR receptor, NGF activates proapoptotic signalling pathways including the MAPK c-Jun N-terminal kinase (JNK), ceramides, and the small G protein Rac, but also activates pathways promoting cell survival through the transcription factor nuclear factor-kappaB (NF-kappaB). NGF was first described by Rita Levi-Montalcini and collaborators as an important factor involved in nerve differentiation and survival. Another role for NGF has since been established in inflammation, in particular of the airways, with increased NGF levels in chronic inflammatory diseases. In this review, we will first describe NGF structure and synthesis and NGF receptors and their signalling pathways. We will then provide information about NGF in the airways, describing its expression and regulation, as well as pointing out its potential role in inflammation, hyperresponsiveness, and remodelling process observed in airway inflammatory diseases, in particular in asthma.
Collapse
Affiliation(s)
- V Freund-Michel
- EA 3771 Inflammation and Environment in Asthma, University Louis Pasteur-Strasbourg I, Faculty of Pharmacy, Illkirch, France.
| | | |
Collapse
|
21
|
Newbern J, Taylor A, Robinson M, Lively MO, Milligan CE. c-Jun N-terminal kinase signaling regulates events associated with both health and degeneration in motoneurons. Neuroscience 2007; 147:680-92. [PMID: 17583433 DOI: 10.1016/j.neuroscience.2007.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Revised: 05/02/2007] [Accepted: 05/04/2007] [Indexed: 12/27/2022]
Abstract
The c-Jun N-terminal kinases (JNKs) are activated by various stimuli and are critical for neuronal development as well as for death following a stressful stimulus. Here, we have evaluated JNK activity in both healthy and dying motoneurons from developing chick embryos and found no apparent difference in overall JNK activity between the conditions, suggesting that this pathway maybe critical in both circumstances. Pharmacological inhibition of JNK in healthy motoneurons supplied with trophic support resulted in decreased mitochondrial membrane potential, neurite outgrowth, and phosphorylation of microtubule-associated protein 1B. On the other hand, in motoneurons deprived of trophic support, inhibition of JNK attenuated caspase activation, and nuclear condensation. We also examined the role of JNK's downstream substrate c-Jun in mediating these events. While c-Jun expression and phosphorylation were greater in cells supplied with trophic support as compared with those deprived, inhibition of c-Jun had no effect on nuclear condensation in dying cells or neurite outgrowth in healthy cells, suggesting that JNK's role in these events is independent of c-Jun. Together, our data underscore the dualistic nature of JNK signaling that is critical for both survival and degenerative changes in motoneurons.
Collapse
Affiliation(s)
- J Newbern
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|
22
|
Aoki K, Nakamura T, Inoue T, Meyer T, Matsuda M. An essential role for the SHIP2-dependent negative feedback loop in neuritogenesis of nerve growth factor-stimulated PC12 cells. ACTA ACUST UNITED AC 2007; 177:817-27. [PMID: 17535963 PMCID: PMC2064282 DOI: 10.1083/jcb.200609017] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The local accumulation of phosphatidylinositol (3,4,5) trisphosphate (PIP(3)) and resulting activation of Rac1/Cdc42 play a critical role in nerve growth factor (NGF)-induced neurite outgrowth. To further explore the mechanism, we visualized PIP(3), phosphatidylinositol (3,4) bisphosphate, and Rac1/Cdc42 activities by fluorescence resonance energy transfer (FRET) imaging in NGF-stimulated PC12 cells. Based on the obtained FRET images, and with the help of in silico kinetic reaction model, we predicted that PI-5-phosphatase negatively regulates PIP(3) upon NGF stimulation. In agreement with this model, depletion of Src homology 2 domain-containing inositol polyphosphate 5-phosphatase 2 (SHIP2) markedly potentiated NGF-induced Rac1/Cdc42 activation and PIP(3) accumulation and considerably increased the number and the length of neurites in phosphate and tensin homologue-depleted PC12 cells. Further refinement of the computational model predicted Rac1 regulation of PI3-kinase and SHIP2, which was also validated experimentally. We propose that the SHIP2-mediated negative feedback on PIP(3) coordinately works with the PI3-kinase-mediated positive feedback to form an initial protrusive pattern and, later, to punctuate the PIP(3) accumulation to maintain proper neurite outgrowth.
Collapse
Affiliation(s)
- Kazuhiro Aoki
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | |
Collapse
|
23
|
Lee WT, Chang WH, Huang CH, Wu KJ. NBS1, the Nijmegen breakage syndrome gene product, regulates neuronal proliferation and differentiation. J Neurochem 2007; 102:141-52. [PMID: 17442057 DOI: 10.1111/j.1471-4159.2007.04477.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Nijmegen breakage syndrome (NBS) is an autosomal recessive disorder, characterized by progressive microcephaly, growth retardation, immunodeficiency, and pre-disposition to tumor formation. To investigate the functions of the NBS gene product, NBS1, on neurons, PC12 cells overexpressing NBS1 and related mutants and primary cortical neuronal culture were used in the present study. Small interfering RNA (siRNA) was applied to repress the expression of endogenous Nbs1 in PC12 cells and primary cortical neurons. We demonstrated that overexpression of NBS1 increases cellular proliferation and decreases the apoptosis of PC12 cells in serum withdrawal and ionizing irradiation, through the activation of phosphatidylinositol 3-kinase (PI 3-kinase)/Akt pathway. Overexpression of NBS1 also decreases neurite elongation on PC12 cells under nerve growth factor stimulation. Transfection of NBS1-overexpressing PC12 cells with a dominant negative Akt mutant attenuates the neuroprotection and cellular proliferation effects of NBS1 while having no effect on neurite elongation. PC12 cells overexpressing NBS657del5 and NBS653 mutants, in which the major NBS1 protein in cells are truncated proteins, have decreased cellular proliferation, increased cell death, and decreased neurite elongation compared with those of control PC12 cells. Repression of Nbs1 by siRNA decreases the PI 3-kinase activity and Akt phosphorylation levels, and induces neurite elongation in PC12 cells even without nerve growth factor stimulation. Repression of Nbs1 by siRNA in primary cortical neurons also increased neurite elongation, but increased neuronal death. We conclude that NBS1 can regulate neuronal proliferation and neuroprotection via PI 3-kinase/Akt pathway while regulating neuronal differentiation in a different pathway. Excessive accumulation of truncated protein secondary to 657del5 mutation may be detrimental to neurons, leading to defective neuronal proliferation and differentiation.
Collapse
Affiliation(s)
- Wang-Tso Lee
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| | | | | | | |
Collapse
|
24
|
Cosker KE, Eickholt BJ. Phosphoinositide 3-kinase signalling events controlling axonal morphogenesis. Biochem Soc Trans 2007; 35:207-10. [PMID: 17371239 DOI: 10.1042/bst0350207] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The establishment of neuronal morphology is essential for the formation of the nervous system. In general, neurons undergo a developmental programme during which their immature processes are specified into one axon and several dendrites. Extension of axons and dendrites is then critical for the establishment of appropriate connectivity. A body of work implicates the PI3K (phosphoinositide 3-kinase) signalling pathway to be crucial during the various events leading to the formation of neuronal circuit. In this review, we will focus specifically on the function of PI3K and downstream signalling cascades that control the establishment of axonal specification and elongation.
Collapse
Affiliation(s)
- K E Cosker
- MRC Centre for Developmental Neurobiology, King's College London, London, UK
| | | |
Collapse
|
25
|
Fukui Y, Tanaka T, Tachikawa H, Ihara S. SWAP-70 is required for oncogenic transformation by v-Src in mouse embryo fibroblasts. Biochem Biophys Res Commun 2007; 356:512-6. [PMID: 17367752 DOI: 10.1016/j.bbrc.2007.03.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 03/01/2007] [Indexed: 11/23/2022]
Abstract
SWAP-70 is a phosphatidylinositol trisphosphate (PtdIns(3,4,5)P(3)) binding protein, which acts in F-actin rearrangement. The role of SWAP-70 in oncogenic transformation of mouse embryo fibroblasts (MEFs) by v-Src was examined by use of MEFs defective in SWAP-70. v-Src morphologically transformed MEFs lacking SWAP-70, but growth of the transformed cells in culture was slower than that of cells supplemented with exogenous SWAP-70. The v-Src-transformed MEFs deficient in SWAP-70 were unable to grow in soft agar while those expressing SWAP70 readily formed colonies, suggesting that SWAP-70 is required for anchorage independent growth of v-Src transformed MEFs. When transplanted in nude mice, tumors formed by the v-Src transformed SWAP-70(-/-) MEFs were smaller than those formed by cells expressing exogenous SWAP-70. These results suggest that SWAP-70 may be required for oncogenic transformation and contributes to cell growth in MEFs transformed by v-Src.
Collapse
Affiliation(s)
- Yasuhisa Fukui
- Division of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan.
| | | | | | | |
Collapse
|
26
|
Hajdo-Milasinović A, Ellenbroek SIJ, van Es S, van der Vaart B, Collard JG. Rac1 and Rac3 have opposing functions in cell adhesion and differentiation of neuronal cells. J Cell Sci 2007; 120:555-66. [PMID: 17244648 DOI: 10.1242/jcs.03364] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rac1 and Rac3 are highly homologous members of the Rho small GTPase family. Rac1 is ubiquitously expressed and regulates cell adhesion, migration and differentiation in various cell types. Rac3 is primarily expressed in brain and may therefore have a specific function in neuronal cells. We found that depletion of Rac1 by short interference RNA leads to decreased cell-matrix adhesions and cell rounding in neuronal N1E-115 cells. By contrast, depletion of Rac3 induces stronger cell adhesions and dramatically increases the outgrowth of neurite-like protrusions, suggesting opposite functions for Rac1 and Rac3 in neuronal cells. Consistent with this, overexpression of Rac1 induces cell spreading, whereas overexpression of Rac3 results in a contractile round morphology. Rac1 is mainly found at the plasma membrane, whereas Rac3 is predominantly localized in the perinuclear region. Residues 185-187, present in the variable polybasic rich region at the carboxyl terminus are responsible for the difference in phenotype induced by Rac1 and Rac3 as well as for their different intracellular localization. The Rac1-opposing function of Rac3 is not mediated by or dependent on components of the RhoA signaling pathway. It rather seems that Rac3 exerts its function through negatively affecting integrin-mediated cell-matrix adhesions. Together, our data reveal that Rac3 opposes Rac1 in the regulation of cell adhesion and differentiation of neuronal cells.
Collapse
Affiliation(s)
- Amra Hajdo-Milasinović
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
27
|
von Philipsborn A, Bastmeyer M. Mechanisms of Gradient Detection: A Comparison of Axon Pathfinding with Eukaryotic Cell Migration. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 263:1-62. [PMID: 17725964 DOI: 10.1016/s0074-7696(07)63001-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The detection of gradients of chemotactic cues is a common task for migrating cells and outgrowing axons. Eukaryotic gradient detection employs a spatial mechanism, meaning that the external gradient has to be translated into an intracellular signaling gradient, which affects cell polarization and directional movement. The sensitivity of gradient detection is governed by signal amplification and adaptation mechanisms. Comparison of the major signal transduction pathways underlying gradient detection in three exemplary chemotaxing cell types, Dictyostelium, neutrophils, and fibroblasts and in neuronal growth cones, reveals conserved mechanisms such as localized PI3 kinase/PIP3 signaling and a common output, the regulation of the cytoskeleton by Rho GTPases. Local protein translation plays a role in directional movement of both fibroblasts and neuronal growth cones. Ca(2+) signaling is prominently involved in growth cone gradient detection. The diversity of signaling between different cell types and its functional implications make sense in the biological context.
Collapse
Affiliation(s)
- Anne von Philipsborn
- Department of Cell Biology and Neurobiology, University of Karlsruhe, D-76131 Karlsruhe, Germany
| | | |
Collapse
|
28
|
Chen CH, Chen YJ, Jeng CJ, Yang SH, Tung PY, Wang SM. Role of PKA in the anti-Thy-1 antibody-induced neurite outgrowth of dorsal root ganglionic neurons. J Cell Biochem 2007; 101:566-75. [PMID: 17177293 DOI: 10.1002/jcb.21217] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Thy-1 is highly expressed in the mammalian nervous system. Our previous study showed that addition of anti-Thy-1 antibody to cultured dorsal root ganglionic (DRG) neurons promotes neurite outgrowth. In this study, we identified a novel signaling pathway mediating this event. Treatment with function-blocking anti-Thy-1 antibodies enhanced neurite outgrowth of DRG neurons in terms of total neurite length, longest neurite length, and total neurite branching points. To elucidate the possible signal transduction pathway involved, activation of kinases was evaluated by Western blotting. Transient phosphorylation of protein kinase A (PKA) and mitogen-activated kinase kinase (MEK) was induced after 15 min of anti-Thy-1 antibody treatment. Pretreatment with a PKA inhibitor (PKI) or an MEK inhibitor, PD98059, significantly decreased the neurite outgrowth response triggered by anti-Thy-1 antibody, indicating the involvement of both kinases. In addition, anti-Thy-1 antibody treatment also induced transient phosphorylation of cyclic AMP-response element-binding protein (CREB) and this effect was also blocked by a PKI or PD98059. Furthermore, the fact that PKI abolished anti-Thy-1 antibody-induced MEK phosphorylation showed that PKA acts upstream of the MEK-CREB cascade. In summary, the PKA-MEK-CREB pathway is a new pathway involved in the neurite outgrowth-promoting effect of anti-Thy-1 antibody.
Collapse
Affiliation(s)
- Chien-Hsing Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
29
|
Karkoulias G, Flordellis C. Delayed transactivation of the receptor for nerve growth factor is required for sustained signaling and differentiation by alpha2-adrenergic receptors in transfected PC12 cells. Cell Signal 2006; 19:945-57. [PMID: 17215105 DOI: 10.1016/j.cellsig.2006.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Revised: 11/06/2006] [Accepted: 11/06/2006] [Indexed: 11/26/2022]
Abstract
Alpha2-adrenergic receptors have been reported to induce subtype-specific neuronal differentiation in vitro, but the signaling mechanisms that mediate this effect have not been characterized. In the present study we found that stimulated alpha2-ARs induce delayed transactivation of TrkA in PC12 cells. The transactivation of TrkA was sensitive to the PP1 inhibitor of the Src family kinases and required prior transactivation of the EGF receptor. Moreover, alpha2-adrenergic receptors induced sustained activation of MAPK and Akt. The sustained activation of Akt, but not of MAPK, was subtype-specific and correlated with the neuronal differentiation of PC12 cells, with the order alpha2A<alpha2B<alpha2C. Furthermore, stimulated alpha2-ARs induced an increased over time expression of the cell cycle associated proteins, p21WAF1 and Cyclin D1 and led to cell cycle arrest in a similar subtype-specific manner. Contrary to sustained activation of MAPK, the persistent activation of Akt and of p21WAF1 and Cyclin D1 as well as neurite outgrowth and expression of the neuronal marker peripherin, were all blocked by K252a an inhibitor of TrkA activity. Together these results demonstrate a novel outcome following alpha2-AR-mediated EGFR transactivation, being the consecutive transactivation of TrkA, and that this event may mediate the subtype-specific differentiation of alpha2-AR-expressing PC12 cells.
Collapse
Affiliation(s)
- Georgios Karkoulias
- Department of Pharmacology, School of Medicine, University of Patras, 26110 Rio Patras, Greece
| | | |
Collapse
|
30
|
Waetzig V, Zhao Y, Herdegen T. The bright side of JNKs-Multitalented mediators in neuronal sprouting, brain development and nerve fiber regeneration. Prog Neurobiol 2006; 80:84-97. [PMID: 17045385 DOI: 10.1016/j.pneurobio.2006.08.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 08/25/2006] [Accepted: 08/30/2006] [Indexed: 12/11/2022]
Abstract
The c-Jun N-terminal kinases (JNKs) are important regulators of physiological and pathological processes in the central and peripheral nervous system. In general, JNKs are considered as mediators of neuronal degeneration in response to stress and injury. However, recent data have provided substantial evidence that JNKs are also essential for physiological and regenerative signalling in neurons. This review summarizes the importance of JNKs for neurite formation and outgrowth, brain development, dendritic architecture and regeneration of nerve fibers after injury. We discuss putative mechanisms which control the bipartite actions of individual JNK isoforms for neuronal death and repair after nerve fiber injury with a particular focus on the role of the transcription factor c-Jun.
Collapse
Affiliation(s)
- Vicki Waetzig
- Institute of Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Hospitalstrasse 4, 24105 Kiel, Germany
| | | | | |
Collapse
|
31
|
Shi BH, Nashimoto T, Andoh R, Konishi H, Kobayashi M, Xu Q, Ihara S, Fukui Y. Mutation of the PI3' kinase gene in a human colon carcinoma cell line, HCC2998. DNA Cell Biol 2006; 25:399-405. [PMID: 16848681 DOI: 10.1089/dna.2006.25.399] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
HCC2998 is a highly differentiated human colon carcinoma cell line, which has been shown to be converted to a poorly differentiated one after expression of a constitutively active phosphatidylinositol 3-kinase (PI3' kinase). These cells express aberrant sizes of a regulatory subunit of PI3' kinase, p85alpha, with molecular weights of 50 and 76 kDa at a very low level. To elucidate how these cells express these proteins, we analyzed mutations within the p85alpha gene. DNA sequencing analysis revealed that these mutant proteins were generated by independent point mutations in the two alleles of the p85alpha gene: one in the coding sequence, and the other in the acceptor sequence for splicing. Introduction of wild-type p85alpha into HCC2998 cells induced slight rounding of the cells and enhancement of mucin secretion. At the same time, a membrane receptor, ErbB3, was phosphorylated on tyrosine, which in turn, binds to PI3' kinase. Since ErbB3 is upstream of PI3' kinase, it is likely that there is an autocrine loop in which PI3' kinase is activated by ErbB3, which may contribute to dedifferentiation of the cells.
Collapse
Affiliation(s)
- Bin-Hi Shi
- Laboratory of Biological Chemistry, Division of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Horiguchi K, Hanada T, Fukui Y, Chishti AH. Transport of PIP3 by GAKIN, a kinesin-3 family protein, regulates neuronal cell polarity. ACTA ACUST UNITED AC 2006; 174:425-36. [PMID: 16864656 PMCID: PMC2064238 DOI: 10.1083/jcb.200604031] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Phosphatidylinositol-(3,4,5)-trisphosphate (PIP3), a product of phosphatidylinositol 3-kinase, is an important second messenger implicated in signal transduction and membrane transport. In hippocampal neurons, the accumulation of PIP3 at the tip of neurite initiates the axon specification and neuronal polarity formation. We show that guanylate kinase–associated kinesin (GAKIN), a kinesin-like motor protein, directly interacts with a PIP3-interacting protein, PIP3BP, and mediates the transport of PIP3-containing vesicles. Recombinant GAKIN and PIP3BP form a complex on synthetic liposomes containing PIP3 and support the motility of the liposomes along microtubules in vitro. In PC12 cells and cultured hippocampal neurons, transport activity of GAKIN contributes to the accumulation of PIP3 at the tip of neurites. In hippocampal neurons, altered accumulation of PIP3 by overexpression of GAKIN constructs led to the loss of the axonally differentiated neurites. Together, these results suggest that, in neurons, the GAKIN–PIP3BP complex transports PIP3 to the neurite ends and regulates neuronal polarity formation.
Collapse
Affiliation(s)
- Kaori Horiguchi
- Department of Pharmacology and Cancer Center, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
33
|
Tornieri K, Welshhans K, Geddis MS, Rehder V. Control of neurite outgrowth and growth cone motility by phosphatidylinositol-3-kinase. ACTA ACUST UNITED AC 2006; 63:173-92. [PMID: 16463277 DOI: 10.1002/cm.20115] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Phosphatidylinositol-3-kinase (PI-3K) has been reported to affect neurite outgrowth both in vivo and in vitro. Here we investigated the signaling pathways by which PI-3K affects neurite outgrowth and growth cone motility in identified snail neurons in vitro. Inhibition of PI-3K with wortmannin (2 microM) or LY 294002 (25 microM) resulted in a significant elongation of filopodia and in a slow-down of neurite outgrowth. Experiments using cytochalasin and blebbistatin, drugs that interfere with actin polymerization and myosin II activity, respectively, demonstrated that filopodial elongation resulting from PI-3K inhibition was dependent on actin polymerization. Inhibition of strategic kinases located downstream of PI-3K, such as Akt, ROCK, and MEK, also caused significant filopodial elongation and a slow-down in neurite outgrowth. Another growth cone parameter, filopodial number, was not affected by inhibition of PI-3K, Akt, ROCK, or MEK. A detailed study of growth cone behavior showed that the filopodial elongation induced by inhibiting PI-3K, Akt, ROCK, and MEK was achieved by increasing two motility parameters: the rate with which filopodia extend (extension rate) and the time that filopodia spend elongating. Whereas the inhibition of ROCK or Akt (both activated by the lipid kinase activity of PI-3K) and MEK (activated by the protein kinase activity of PI-3K) had additive effects, simultaneous inhibition of Akt and ROCK showed no additive effect. We further demonstrate that the effects on filopodial dynamics investigated were calcium-independent. Taken together, our results suggest that inhibition of PI-3K signaling results in filopodial elongation and a slow-down of neurite advance, reminiscent of growth cone searching behavior.
Collapse
Affiliation(s)
- Karine Tornieri
- Department of Biology, Georgia State University, Atlanta, Georgia 30302, USA
| | | | | | | |
Collapse
|
34
|
Celik-Ozenci C, Bayram Z, Akkoyunlu G, Korgun ET, Erdogru T, Seval Y, Ustunel I, Baykara M, Demir R. Localization of NGF and nNOS in varicocele-induced rat testis. Acta Histochem 2006; 107:435-42. [PMID: 16325891 DOI: 10.1016/j.acthis.2005.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2005] [Revised: 10/12/2005] [Accepted: 10/20/2005] [Indexed: 10/25/2022]
Abstract
Nerve growth factor (NGF) is synthesized in male germ cells. The presence of neuronal nitric oxide synthase (nNOS) in Leydig cells is related to its role in the regulation of testosterone release. Varicocele is often characterized by abnormal sperm quality and influences the fertilizing capacity of the haploid gamete. We investigated the localization of NGF and nNOS in testes of adult Wistar rats with experimentally induced varicocele after 9, 11, and 13 weeks, as well as in sham-operated controls by immunohistochemistry and Western blot. In control testis, we detected NGF in nuclei of Sertoli cells and also as small vesicular-like structures in the cytoplasm of primary spermatocytes, and in round and elongating spermatids. Varicocele-induction revealed a slight decrease of NGF at 13 weeks, especially in Sertoli cells. In control tissue, nNOS protein was present mainly in Leydig cells and in Sertoli cell cytoplasm. Additionally, nNOS immunoreactivity was present in the heads of elongated spermatids. Western blot results revealed that the decrease of NGF was not significant in the 13-week varicocele group, moreover, the amount of nNOS was not altered in any of the varicocele groups. In conclusion, NGF and nNOS have important roles for normal gametogenesis and our data for the first time indicates that varicocele induction does not necessarily affect the expression of NGF and nNOS. Thus, these two molecules do not appear to be related to varicocele induction.
Collapse
Affiliation(s)
- Ciler Celik-Ozenci
- Department of Histology, Faculty of Medicine, Akdeniz University, Antalya, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Xiao J, Pradhan A, Liu Y. Functional role of JNK in neuritogenesis of PC12-N1 cells. Neurosci Lett 2006; 392:231-4. [PMID: 16216413 DOI: 10.1016/j.neulet.2005.09.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Accepted: 09/10/2005] [Indexed: 11/23/2022]
Abstract
JNKs, also known as SAPKs, are activated in response to a wide variety of factors including growth factors, cytokines, UV radiation, and heat shock. In the rat pheochromocytoma PC12 cells, the JNK signaling pathway mediates diverse functions such as differentiation and apoptosis. We have previously shown that activated JNK is required for later stages of neuritogeneis induced by NGF in a variant PC12 cell line (N1). Here, the functional role of JNK in N1 cells is further investigated. We show that NGF treatment, which induces extensive neurite branching and cell soma enlargement in the N1 cells, stimulates a biphasic activation of JNK. The first phase of activation is rapid and transient, beginning at 15 min after NGF exposure and lasting approximately 45 min. The second phase of activation is sustained, beginning at 9-12 h of NGF treatment and lasting for at least 24 h. Similar biphasic pattern of JNK activation is also observed in the parental PC12 cells. Using the specific JNK inhibitor SP600125, we show that the biphasic activation is necessary for neurite outgrowth and branching, and that inhibition of either phase suppresses neuritogenesis in the N1 cells. These results suggest that dynamic JNK activation may play a key role in neurite outgrowth during neuronal development.
Collapse
Affiliation(s)
- Jingnan Xiao
- Department of Pathology, University of Oklahoma Health Sciences Center, PO Box 26901, 975 NE 10th St. BRC-256, Oklahoma City, 73190, USA
| | | | | |
Collapse
|
36
|
Bowen RAR, Clandinin MT. Maternal dietary 22 : 6n-3 is more effective than 18 : 3n-3 in increasing the 22 : 6n-3 content in phospholipids of glial cells from neonatal rat brain. Br J Nutr 2005; 93:601-11. [PMID: 15975158 DOI: 10.1079/bjn20041390] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
One of the debates in infant nutrition concerns whether dietary 18 : 3n-3 (linolenic acid) can provide for the accretion of 22 : 6n-3 (docosahexaenoic acid, DHA) in neonatal tissues. The objective of the present study was to determine whether low or high 18 : 3n-3 v. preformed 22 : 6n-3 in the maternal diet enabled a similar 22 : 6n-3 content in the phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylinositol (PI) and phosphatidylserine (PS) of glial cells from whole brain (cerebrum and cerebellum) of 2-week-old rat pups. At parturition, the dams were fed semi-purified diets containing either increasing amounts of 18 : 3n-3 (18 : 2n-6 to 18 : 3n-3 fatty acid ratio of 7.8 : 1, 4.4 : 1 or 1 : 1), preformed DHA, or preformed 20 : 4n-6 (arachidonic acid)+DHA. During the first 2 weeks of life, the rat pups from the respective dams received only their dam's milk. The fatty acid composition of the pups' stomach contents (dam's milk) and phospholipids from glial cells were quantified. The 20 : 4n-6 and 22 : 6n-3 content in the stomach from rat pups at 2 weeks of age reflected the fatty acid composition of the dam's diet. The 20 : 4n-6 content of PE and PS in the glial cells was unaffected by maternal diet treatments. Preformed 22 : 6n-3 in the maternal diet increased the 22 : 6n-3 content of glial cell PE and PS compared with maternal diets providing an 18 : 2n-6 to 18 : 3 n-3 fatty acid ratio of 7.8 : 1, 4.4 : 1 or 1 : 1 (P<0.0001). There was no significant difference in the 20 : 4n-6 and 22 : 6n-3 content of glial cell PC and PI among maternal diet treatments. It was concluded that maternal dietary 22 : 6n-3 is more effective than low or high levels of maternal dietary 18 : 3n-3 at increasing the 22 : 6n-3 content in PE and PS of glial cells from the whole brain of rat pups at 2 weeks of age. The findings from the present study have important implications for human infants fed infant formulas that are devoid of 22 : 6n-3.
Collapse
Affiliation(s)
- Raffick A R Bowen
- Nutrition and Metabolism Research Group, Department of Agricultural, Food and Nutritional Science, Uniersity of Alberta, Edmonton, Canada
| | | |
Collapse
|
37
|
Waetzig V, Herdegen T. MEKK1 controls neurite regrowth after experimental injury by balancing ERK1/2 and JNK2 signaling. Mol Cell Neurosci 2005; 30:67-78. [PMID: 16006144 DOI: 10.1016/j.mcn.2005.06.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Revised: 06/02/2005] [Accepted: 06/06/2005] [Indexed: 10/25/2022] Open
Abstract
After injury, peripheral neuronal cells initiate complex signaling cascades to promote survival and regeneration. In the present study, we have identified the mitogen-activated protein kinase (MAPK) isoforms which are necessary for nerve growth factor (NGF)-induced neurite regrowth after injury of differentiated PC12 cells. Extracellular signal-regulated kinases 1 and 2 (ERK1/2) and the usually pro-apoptotic c-Jun N-terminal kinase 2 (JNK2) are crucial for neurite regrowth, while p38 plays no role in this context. Surprisingly, the MEK1 inhibitors PD 98059 and U 0126 blocked both ERK1/2 and JNK phosphorylation, indicating a novel form of balancing MAPK cascade cross-talk. Results from RNAi experiments excluded direct ERK/JNK interactions. We identified the upstream kinase MEKK1 as an activator of both the ERK1/2 and JNK2 pathways, whereby the ERK1/2 kinase MEK1 and the JNK kinase MKK7 bind to MEKK1 in a competing fashion. Our findings suggest an important role of JNK2 and MAPK pathway cross-talk in neurite regeneration.
Collapse
Affiliation(s)
- Vicki Waetzig
- Institute of Pharmacology, University Hospital Schleswig-Holstein, Hospitalstrasse 4, D-24105 Kiel, Germany.
| | | |
Collapse
|
38
|
Cao D, Xue R, Xu J, Liu Z. Effects of docosahexaenoic acid on the survival and neurite outgrowth of rat cortical neurons in primary cultures. J Nutr Biochem 2005; 16:538-46. [PMID: 16115542 DOI: 10.1016/j.jnutbio.2005.02.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2004] [Indexed: 11/23/2022]
Abstract
Effects of docosahexaenoic acid (DHA) on survival and neurite outgrowth were investigated in primary cultures of rat cortical neurons. Cell cultures were prepared from cortex on embryonic day 18 (E-18) for treatment with a series of DHA concentrations (12.5, 25, 50, 75, 100 and 200 microM). Docosahexaenoic acid (25-50 microM) significantly enhanced neuronal viability, but lower concentration of DHA (12.5 microM) did not show an obvious effect. In contrast, higher concentrations of DHA (100-200 microM) exerted the significant opposite effects by decreasing neuronal viability. Furthermore, treatment with 25 microM DHA significantly prevented the neurons from death after different culture days in vitro (DIV). Moreover, measurements from the cultures exposed to 25 microM DHA immediately after plating showed significant increases in the percentage of cells with neurites, the mean number of neurite branches, the total neuritic length per cell and the length of the longest neurite in each cell after 24 and 48 h in vitro (HIV). The DHA-treated neurons had greater growth-associated protein-43 (GAP-43) immunoactivity and higher phosphatidylserine (PS) and phosphatidylethanolamine (PE) contents, but lower phosphatidylcholine (PC) content than control neurons. The significant increased DHA contents were also observed in both PE and PS in the treated neurons. These findings suggest that optimal DHA (25 microM) may have positive effects on the survival and the neurite outgrowth of the cultured fetal rat cortical neurons, and the effects probably are related to DHA-stimulating neuron-specific protein synthesis and its enhancing the discrete phospholipid (PL) content through enrichment of DHA in the PL species.
Collapse
Affiliation(s)
- Dehua Cao
- Department of Biology, School of Life Sciences, Nanjing University, Jiangsu, Nanjing 210093, P. R. China
| | | | | | | |
Collapse
|
39
|
Laurino L, Wang XX, de la Houssaye BA, Sosa L, Dupraz S, Cáceres A, Pfenninger KH, Quiroga S. PI3K activation by IGF-1 is essential for the regulation of membrane expansion at the nerve growth cone. J Cell Sci 2005; 118:3653-62. [PMID: 16046480 DOI: 10.1242/jcs.02490] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Exocytotic incorporation of plasmalemmal precursor vesicles (PPVs) into the cell surface is necessary for axonal outgrowth and is known to occur mainly at the nerve growth cone. We have demonstrated recently that plasmalemmal expansion is regulated at the growth cone by IGF-1, but not by BDNF, in a manner that is quasi independent of the neuron's perikaryon. To begin elucidating the signaling pathway by which exocytosis of the plasmalemmal precursor is regulated, we studied activation of the IRS/PI3K/Akt pathway in isolated growth cones and hippocampal neurons in culture stimulated with IGF-1 or BDNF. Our results show that IGF-1, but not BDNF, significantly and rapidly stimulates IRS/PI3K/Akt and membrane expansion. Inhibition of PI3K with Wortmannin or LY294002 blocked IGF-1-stimulated plasmalemmal expansion at the growth cones of cultured neurons. Finally, our results show that, upon stimulation with IGF-1, most active PI3K becomes associated with distal microtubules in the proximal or central domain of the growth cone. Taken together, our results suggest a critical role for IGF-1 and the IRS/PI3K/Akt pathway in the process of membrane assembly at the axonal growth cone.
Collapse
Affiliation(s)
- Lisandro Laurino
- Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba y CIQUIBIC, CONICET, Córdoba 5000, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Dey N, Howell BW, De PK, Durden DL. CSK negatively regulates nerve growth factor induced neural differentiation and augments AKT kinase activity. Exp Cell Res 2005; 307:1-14. [PMID: 15890337 DOI: 10.1016/j.yexcr.2005.02.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2004] [Revised: 01/12/2005] [Accepted: 02/10/2005] [Indexed: 11/26/2022]
Abstract
Src family kinases are involved in transducing growth factor signals for cellular differentiation and proliferation in a variety of cell types. The activity of all Src family kinases (SFKs) is controlled by phosphorylation at their C-terminal 527-tyrosine residue by C-terminal SRC kinase, CSK. There is a paucity of information regarding the role of CSK and/or specific Src family kinases in neuronal differentiation. Pretreatment of PC12 cells with the Src family kinase inhibitor, PP1, blocked NGF-induced activation of SFKs and obliterated neurite outgrowth. To confirm a role for CSK and specific isoforms of SFKs in neuronal differentiation, we overexpressed active and catalytically dead CSK in the rat pheochromocytoma cell line, PC12. CSK overexpression caused a profound inhibition of NGF-induced activation of FYN, YES, RAS, and ERK and inhibited neurite outgrowth, NGF-stimulated integrin-directed migration and blocked the NGF-induced conversion of GDP-RAC to its GTP-bound active state. CSK overexpression markedly augmented the activation state of AKT following NGF stimulation. In contrast, kinase-dead CSK augmented the activation of FYN, RAS, and ERK and increased neurite outgrowth. These data suggest a distinct requirement for CSK in the regulation of NGF/TrkA activation of RAS, RAC, ERK, and AKT via the differential control of SFKs in the orchestration of neuronal differentiation.
Collapse
Affiliation(s)
- Nandini Dey
- Section of Pediatric Hematology/Oncology, Department of Pediatrics, AFLAC Cancer Center and Blood Disorders Services, Emory University School of Medicine, Atlanta, GA 30022, USA
| | | | | | | |
Collapse
|
41
|
Andres RH, Ducray AD, Huber AW, Pérez-Bouza A, Krebs SH, Schlattner U, Seiler RW, Wallimann T, Widmer HR. Effects of creatine treatment on survival and differentiation of GABA-ergic neurons in cultured striatal tissue. J Neurochem 2005; 95:33-45. [PMID: 16045451 DOI: 10.1111/j.1471-4159.2005.03337.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder, characterized by a prominent loss of GABA-ergic medium-sized spiny neurons in the caudate putamen. There is evidence that impaired energy metabolism contributes to neuronal death in HD. Creatine is an endogenous substrate for creatine kinases and thereby supports cellular ATP levels. This study investigated the effects of creatine supplementation (5 mm) on cell survival and neuronal differentiation in striatal cultures. Chronic creatine treatment resulted in significant increased densities of GABA-immunoreactive (-ir) neurons, although total neuronal cell number and general viability were not affected. Similar effects were seen after short-term treatment, suggesting that creatine acted as a differentiation factor. Inhibitors of transcription or translation did not abolish the creatine-mediated effects, nor did omission of extracellular calcium, whereas inhibition of mitogen-activated protein kinase and phosphatidylinositol-3-kinase significantly attenuated the creatine induced increase in GABA-ir cell densities. Creatine exhibited significant neuroprotection against toxicity instigated either by glucose- and serum deprivation or addition of 3-nitropropionic acid. In sum, the neuroprotective properties in combination with promotion of neuronal differentiation suggest that creatine has potential as a therapeutic drug in the treatment of neurodegenerative diseases, like HD.
Collapse
Affiliation(s)
- R H Andres
- Department of Neurosurgery, University Hospital, Berne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pan J, Kao YL, Joshi S, Jeetendran S, Dipette D, Singh US. Activation of Rac1 by phosphatidylinositol 3‐kinasein vivo: role in activation of mitogen‐activated protein kinase (MAPK) pathways and retinoic acid‐induced neuronal differentiation of SH‐SY5Y cells. J Neurochem 2005; 93:571-83. [PMID: 15836616 DOI: 10.1111/j.1471-4159.2005.03106.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rho GTPases such as RhoA, Rac1 and Cdc42 are crucial players in the regulation of signal transduction pathways required for neuronal differentiation. Using an in vitro cell culture model of neuroblastoma SH-SY5Y cells, we demonstrated previously that RhoA is an in vivo substrate of tissue transglutaminase (TGase) and retinoic acid (RA) promoted activation of RhoA by transamidation. Although activation of RhoA promoted cytoskeletal rearrangement in SH-SY5Y cells, it was not involved in induction of neurite outgrowth. Here, we demonstrate that RA promotes activation of Rac1 in SH-SY5Y cells in a transamidation-independent manner. RA-induced activation of Rac1 is mediated by phosphatidylinositol 3-kinase (PI3K), probably because of phosphorylation of the p85 regulatory subunit by Src kinases. Over-expression of constitutively active PI3K or Rac1-V12 induces neurite outgrowth, activation of mitogen activated protein kinases (MAPKs), and expression of neuronal markers. The PI3K inhibitor LY294002, or over-expression of dominant negative Rac1-N17, blocks RA-induced neurite outgrowth, activation of MAPKs, and expression of neuronal markers, suggesting that activation of PI3K/Rac1 signaling represents a potential mechanism for regulation of neuronal differentiation in SH-SY5Y cells.
Collapse
Affiliation(s)
- Jing Pan
- Cardiovascular Research Institute, Department of Internal Medicine, The Texas A&M University System Health Science Center, College of Medicine, Temple, Texas 76504, USA
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Our brain serves as a center for cognitive function and neurons within the brain relay and store information about our surroundings and experiences. Modulation of this complex neuronal circuitry allows us to process that information and respond appropriately. Proper development of neurons is therefore vital to the mental health of an individual, and perturbations in their signaling or morphology are likely to result in cognitive impairment. The development of a neuron requires a series of steps that begins with migration from its birth place and initiation of process outgrowth, and ultimately leads to differentiation and the formation of connections that allow it to communicate with appropriate targets. Over the past several years, it has become clear that the Rho family of GTPases and related molecules play an important role in various aspects of neuronal development, including neurite outgrowth and differentiation, axon pathfinding, and dendritic spine formation and maintenance. Given the importance of these molecules in these processes, it is therefore not surprising that mutations in genes encoding a number of regulators and effectors of the Rho GTPases have been associated with human neurological diseases. This review will focus on the role of the Rho GTPases and their associated signaling molecules throughout neuronal development and discuss how perturbations in Rho GTPase signaling may lead to cognitive disorders.
Collapse
Affiliation(s)
- Eve-Ellen Govek
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | | | | |
Collapse
|
44
|
Riese U, Ziegler E, Hamburger M. Militarinone A induces differentiation in PC12 cells via MAP and Akt kinase signal transduction pathways. FEBS Lett 2005; 577:455-9. [PMID: 15556627 DOI: 10.1016/j.febslet.2004.10.045] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Revised: 09/23/2004] [Accepted: 10/11/2004] [Indexed: 10/26/2022]
Abstract
The fungal metabolite militarinone A (MILI A) promotes neurite outgrowth in PC12 cells. This study was conducted to investigate the signaling pathways involved in the cellular differentiation processes induced by the compound, with a focus on cascades implicated with nerve growth factor (NGF)-mediated neuritogenesis. MILI A possessed pronounced amphiphilic properties. The compound rapidly accumulated in the cell membrane and was slowly released into the cytoplasma. In primed PC12 cells, an early activation of protein kinase B (Akt), representing a downstream target of phosphoinositol 3 (PI3) kinase, and a delayed phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), and of transcription factor cAMP responsive element binding protein (CREB) was found. The NGF-dependent activation of c-Jun amino terminal kinase (SAPK/JNK1) was potentiated. Morphological differentiation of cells and the phosphorylation of specific signal molecules were blocked by the MAP kinase (MEK1) inhibitor PD098059, the PI3-kinase (PI3K) inhibitor wortmannin and the adenylyl cyclase inhibitor 9-cyclopentyladenine.
Collapse
Affiliation(s)
- Ulrike Riese
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Semmelweisstrasse 10, D-07743 Jena, Germany
| | | | | |
Collapse
|
45
|
Okamoto T, Namikawa K, Asano T, Takaoka K, Kiyama H. Differential regulation of the regulatory subunits for phosphatidylinositol 3-kinase in response to motor nerve injury. ACTA ACUST UNITED AC 2004; 131:119-25. [PMID: 15530660 DOI: 10.1016/j.molbrainres.2004.08.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2004] [Indexed: 01/22/2023]
Abstract
Type Ia phosphatidylinositol 3-kinase (PI3K) generates lipid products that operate as one of major second messengers following activation of tyrosine kinase receptors. PI3K is a heterodimer composed of a 110-kDa catalytic subunit and a regulatory subunit. In this study, we determined the expression of mRNA for the regulatory subunits after injury of rat hypoglossal nerves. In situ hybridization histochemistry revealed that the expression of PI3K regulatory subunit alpha isoforms (p85alpha, p55alpha, and p50alpha) was significantly enhanced in injured motor neurons, whereas other regulatory subunits such as p85beta or p55gamma were not detected. Of the alpha isoforms, the greatest increase was observed in p55alpha mRNA levels, while there were smaller increases in p85alpha and p50alpha mRNA expression. These results were confirmed by RT-PCR analysis. Further immunohistochemical analysis also confirmed the increased level of p55alpha protein in injured motor neurons. Taken together with the previously reported induction of the p110alpha catalytic subunit in injured neurons, these results suggest that PI3K, consisting of p55alpha and p110alpha, plays a crucial role in the process of nerve regeneration.
Collapse
Affiliation(s)
- Takashi Okamoto
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | | | | | | | | |
Collapse
|
46
|
Gambarotta G, Garzotto D, Destro E, Mautino B, Giampietro C, Cutrupi S, Dati C, Cattaneo E, Fasolo A, Perroteau I. ErbB4 Expression in Neural Progenitor Cells (ST14A) Is Necessary to Mediate Neuregulin-1β1-induced Migration. J Biol Chem 2004; 279:48808-16. [PMID: 15355992 DOI: 10.1074/jbc.m408374200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Activation of the receptor tyrosine kinase ErbB4 leads to various cellular responses such as proliferation, survival, differentiation, and chemotaxis. Two pairs of naturally occurring ErbB4 isoforms differing in their juxtamembrane (JMa/JMb) and C termini (cyt1/cyt2) have been described. To examine the role of ErbB4 in neuron migration, we cloned and stably transfected each of the four ErbB4 isoforms in ST14A cells (a neural progenitor cell line derived from the striatum of embryonic day 14 rats) endogenously expressing the other members of the ErbB family: ErbB1, ErbB2, and ErbB3. Using immunoprecipitation assays, we showed that the neuregulin-1beta1 (NRG1beta1) stimulus induced ErbB4 tyrosine phosphorylation and phosphatidylinositol 3-kinase (PI3K) recruitment and activation (as demonstrated by Akt phosphorylation) either directly (ErbB4 cyt1 isoform) or indirectly (ErbB4 cyt2 isoform). We examined the ability of the four ErbB4 isoforms to induce chemotaxis and cell proliferation in response to NRG1beta1 stimulation. Using migration assays, we observed that only ErbB4-expressing cells stimulated with NRG1beta1 showed a significant increase in migration, whereas the growth rate remained unchanged. Additional assays showed that inhibition of PI3K (but not of phospholipase Cgamma) dramatically reduced migratory activity. Our data show that ErbB4 signaling via PI3K activation plays a fundamental role in controlling NRG1beta1-induced migration.
Collapse
Affiliation(s)
- Giovanna Gambarotta
- Department of Human and Animal Biology, University of Torino, Torino 10123, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Alleaume C, Eychène A, Harnois T, Bourmeyster N, Constantin B, Caigneaux E, Muller JM, Philippe M. Vasoactive intestinal peptide-induced neurite remodeling in human neuroblastoma SH-SY5Y cells implicates the Cdc42 GTPase and is independent of Ras-ERK pathway. Exp Cell Res 2004; 299:511-24. [PMID: 15350548 DOI: 10.1016/j.yexcr.2004.06.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2004] [Revised: 06/15/2004] [Indexed: 11/28/2022]
Abstract
Vasoactive intestinal peptide (VIP) is known to regulate proliferation or differentiation in normal and tumoral cells. SH-SY5Y is a differentiated cell subclone derived from the SK-N-SH human neuroblastoma cell line and possess all the components for an autocrine action of VIP. In the present study, we investigated the morphological changes and intracellular signaling pathways occurring upon VIP treatment of SH-SY5Y cells. VIP induced an early remodeling of cell projections: a branched neurite network spread out and prominent varicosities developed along neurites. Although activated by VIP, the Ras/ERK pathway was not required for the remodeling process. In contrast, pull-down experiments revealed a strong Cdc42 activation by VIP while expression of a dominant-negative Cdc42 prevented the VIP-induced neurite changes, suggesting an important role for this small GTPase in the process. These data provide the first evidence for a regulation of the activity of Rho family GTPases by VIP and bring new insights in the signaling pathways implicated in neurite remodeling process induced by VIP in neuroblastoma cells.
Collapse
Affiliation(s)
- Céline Alleaume
- Equipe Neuropeptides, Institut de Physiologie et Biologie Cellulaires, CNRS UMR 6187, Université de Poitiers-Pôle Biologie Santé, 86022 Poitiers Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Laurent CE, Smithgall TE. The c-Fes tyrosine kinase cooperates with the breakpoint cluster region protein (Bcr) to induce neurite extension in a Rac- and Cdc42-dependent manner. Exp Cell Res 2004; 299:188-98. [PMID: 15302586 DOI: 10.1016/j.yexcr.2004.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2004] [Revised: 05/10/2004] [Indexed: 10/26/2022]
Abstract
The c-fes locus encodes a cytoplasmic protein-tyrosine kinase (Fes) previously shown to accelerate nerve growth factor (NGF)-induced neurite outgrowth in rat PC12 cells. Here, we investigated the role of the Rho family small GTPases Rac1 and Cdc42 in Fes-mediated neuritogenesis, which have been implicated in neuronal differentiation in other systems. Fes-induced acceleration of neurite outgrowth in response to NGF treatment was completely blocked by the expression of dominant-negative Rac1 or Cdc42. Expression of a kinase-active mutant of Fes induced constitutive relocalization of endogenous Rac1 to the cell periphery in the absence of NGF, and led to dramatic actin reorganization and spontaneous neurite extension. We also investigated the breakpoint cluster region protein (Bcr), which possesses the Dbl and PH domains characteristic of guanine nucleotide exchange factors for Rho family GTPases, as a possible link between Fes, Rac/Cdc42 activation, and neuritogenesis. Coexpression of a GFP-Bcr fusion protein containing the Fes binding and tyrosine phosphorylation sites (amino acids 162-413) completely suppressed neurite outgrowth triggered by Fes. Conversely, coexpression of full-length Bcr with wild-type Fes in PC12 cells induced NGF-independent neurite formation. Taken together, these data suggest that Fes and Bcr cooperate to activate Rho family GTPases as part of a novel pathway regulating neurite extension in PC12 cells, and provide more evidence for an emerging role for Fes in neuronal differentiation.
Collapse
Affiliation(s)
- Charles E Laurent
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
49
|
Li P, Yamakuni T, Matsunaga K, Kondo S, Ohizumi Y. Nardosinone enhances nerve growth factor-induced neurite outgrowth in a mitogen-activated protein kinase- and protein kinase C-dependent manner in PC12D cells. J Pharmacol Sci 2004; 93:122-5. [PMID: 14501162 DOI: 10.1254/jphs.93.122] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The mechanism to enhance nerve growth factor (NGF, 2 ng/ml)-induced neurite outgrowth from PC12D cells by nardosinone isolated from Nardostachys chinensis was examined. It was shown that the potentiation of the NGF-induced neurite outgrowth by nardosinone was mitogen-activated protein (MAP) kinase-dependent, but was not accompanied by stimulation of NGF-induced increase in MAP kinase phosphorylation. Furthermore, this augmentation of NGF-induced neurite outgrowth was abolished by GF109203X, a protein kinase C (PKC) inhibitor. These results suggest that the enhancement of NGF-induced neurite outgrowth from PC12D cells by nardosinone involves activation of a down-stream step of the MAP kinase-dependent cascade of NGF coupled with PKC.
Collapse
Affiliation(s)
- Ping Li
- Department of Pharmaceutical Molecular Biology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | | | | | | | | |
Collapse
|
50
|
Abstract
Recent experiments in various cell types such as mammalian neutrophils and Dictyostelium discoideum amoebae point to a key role for the lipid product of PI 3-kinase, PIP(3), in determining internal polarity. In neurons, as a consequence of the elongation of one neurite, the axon is specified and the cell acquires its polarity. To test the hypothesis that PI 3-kinase and PIP(3) may play a role in neuronal polarity, and especially in axon specification, we observed localization of PIP(3) visualized by Akt-PH-GFP in developing hippocampal neurons. We found that PIP(3) accumulates in the tip of the growing processes. This accumulation is inhibited by addition of PI 3-kinase inhibitors. Those inhibitors, consistently with a role of PIP(3) in process formation and elongation, delay the transition from stage 1 neurons to stage 3 neurons, and both axon formation and elongation. Moreover, when the immature neurite contacts a bead coated with laminin, a substrate known to induce axon specification, PIP(3) accumulates in its growth cone followed by a rapid elongation of the neurite. In such conditions, the addition of PI 3-kinase inhibitors inhibits both PIP(3) accumulation and future axon elongation. These results suggest that PIP(3) is involved in axon specification, possibly by stimulating neurite outgrowth. In addition, when a second neurite contacted the beads, this neurite rapidly elongates whereas the elongation of the first laminin-contacting neurite stops, consistently with the hypothesis of a negative feedback mechanism from the growing future axon to the other neurites.
Collapse
Affiliation(s)
- Céline Ménager
- Department of Cell Pharmacology, Nagoya University, Graduate School of Medicine, Showa, Nagoya, Aichi, Japan
| | | | | | | |
Collapse
|