1
|
McGregor ER, Lasky DJ, Rippentrop OJ, Clark JP, Wright S, Jones MV, Anderson RM. Reversal of neuronal tau pathology via adiponectin receptor activation. Commun Biol 2025; 8:8. [PMID: 39755746 DOI: 10.1038/s42003-024-07391-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025] Open
Abstract
Changes in brain mitochondrial metabolism are coincident with functional decline; however, direct links between the two have not been established. Here, we show that mitochondrial targeting via the adiponectin receptor activator AdipoRon (AR) clears neurofibrillary tangles (NFTs) and rescues neuronal tauopathy-associated defects. AR reduced levels of phospho-tau and lowered NFT burden by a mechanism involving the energy-sensing kinase AMPK and the growth-sensing kinase GSK3b. The transcriptional response to AR included broad metabolic and functional pathways. Induction of lysosomal pathways involved activation of LC3 and p62, and restoration of neuronal outgrowth required the stress-responsive kinase JNK. Negative consequences of NFTs on mitochondrial activity, ATP production, and lipid stores were corrected. Defects in electrophysiological measures (e.g., resting potential, resistance, spiking profiles) were also corrected. These findings reveal a network linking mitochondrial function, cellular maintenance processes, and electrical aspects of neuronal function that can be targeted via adiponectin receptor activation.
Collapse
Affiliation(s)
- Eric R McGregor
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Danny J Lasky
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Olivia J Rippentrop
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Josef P Clark
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI, USA
| | - Samantha Wright
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Mathew V Jones
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Rozalyn M Anderson
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- GRECC William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
2
|
Shi J, Chen Q, Lai J, Zhu J, Zhang R, Mazid MA, Li D, Su H, Qin D. Impact of c-JUN deficiency on thalamus development in mice and human neural models. Cell Biosci 2024; 14:149. [PMID: 39707500 DOI: 10.1186/s13578-024-01303-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/04/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND c-Jun is a key regulator of gene expression. Through the formation of homo- or heterodimers, c-JUN binds to DNA and regulates gene transcription. While c-Jun plays a crucial role in embryonic development, its impact on nervous system development in higher mammals, especially for some deep structures, for example, thalamus in diencephalon, remains unclear. METHODS To investigate the influence of c-JUN on early nervous system development, c-Jun knockout (KO) mice and c-JUN KO H1 embryonic stem cells (ESCs)-derived neural progenitor cells (NPCs), cerebral organoids (COs), and thalamus organoids (ThOs) models were used. We detected the dysplasia via histological examination and immunofluorescence staining, omics analysis, and loss/gain of function analysis. RESULTS At embryonic day 14.5, c-Jun knockout (KO) mice exhibited sparseness of fibers in the brain ventricular parenchyma and malformation of the thalamus in the diencephalon. The absence of c-JUN accelerated the induction of NPCs but impaired the extension of fibers in human neuronal cultures. COs lacking c-JUN displayed a robust PAX6+/NESTIN+ exterior layer but lacked a fibers-connected core. Moreover, the subcortex-like areas exhibited defective thalamus characteristics with transcription factor 7 like 2-positive cells. Notably, in guided ThOs, c-JUN KO led to inadequate thalamus patterning with sparse internal nerve fibers. Chromatin accessibility analysis confirmed a less accessible chromatin state in genes related to the thalamus. Overexpression of c-JUN rescued these defects. RNA-seq identified 18 significantly down-regulated genes including RSPO2, WNT8B, MXRA5, HSPG2 and PLAGL1 while 24 genes including MSX1, CYP1B1, LMX1B, NQO1 and COL2A1 were significantly up-regulated. CONCLUSION Our findings from in vivo and in vitro experiments indicate that c-JUN depletion impedes the extension of nerve fibers and renders the thalamus susceptible to dysplasia during early mouse embryonic development and human ThO patterning. Our work provides evidence for the first time that c-JUN is a key transcription regulator that play important roles in the thalamus/diencephalon development.
Collapse
Affiliation(s)
- Jiantao Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qing Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianheng Lai
- Guangdong Engineering Technology Research Center of Biological Targeting Diagnosis, Therapy and Rehabilitation, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jieying Zhu
- CAS Key Laboratory of Regenerative Biology, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ran Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Md Abdul Mazid
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dongwei Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangdong Engineering Technology Research Center of Biological Targeting Diagnosis, Therapy and Rehabilitation, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
- Guangdong Engineering Research Center of Early Clinical Trials of Biotechnology Drugs, The Fifth Affiliated Hospital,, Guangzhou Medical University, Guangzhou, China.
- Bioland Laboratory Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| |
Collapse
|
3
|
Mardi S, Letafati A, Hosseini A, Faraji R, Hosseini P, Mozhgani SH. Analysis of the Main Checkpoints of the JNK-MAPK Pathway in HTLV-1-Associated Leukemia/Lymphoma via Boolean Network Simulation. Biochem Genet 2024:10.1007/s10528-024-10916-0. [PMID: 39320417 DOI: 10.1007/s10528-024-10916-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
The c-Jun N-terminal kinase (JNK) pathway is a signal transduction pathway that plays a critical role in cell growth and survival. Its dysregulation is related to various cancers, including adult T-cell leukemia/lymphoma (ATLL), an aggressive peripheral T-cell malignancy caused by human T-cell lymphotropic virus type 1 (HTLV-1) infection. There is currently no vaccine or definitive treatment for ATLL. This research aimed to identify the JNK-MAPK pathway checkpoints to identify possible therapeutic targets using Boolean network analysis. First, the genes involved in the JNK pathway and their interactions were identified and mapped. Next, a Boolean network analysis was performed using the R programming language, which suggested protein kinase B (AKT) and MAP kinase phosphatase (MKP) for further evaluation. Finally, to confirm the effect of these two genes, a clinical study was conducted among ATLL patients and healthy individuals. The quantitative real time polymerase chain reaction (qRT‒PCR) results revealed a statistically significant decrease in the expression of AKT and MKP in ATLL patients compared to normal controls. This highlights the potential role of these two genes as potential therapeutic targets in ATLL.
Collapse
Affiliation(s)
- Shayan Mardi
- Student Research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Hosseini
- Department of Computer Engineering, Faculty of Engineering, Raja University, Qazvin, Iran
| | - Reza Faraji
- Department of Animal Sciences, College of Agriculture & Natural Resources, University of Tehran, Karaj, Iran
| | - Parastoo Hosseini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed-Hamidreza Mozhgani
- Noncommunicable Disease Research Center, Alborz University of Medical Sciences, Karaj, Iran.
- Department of Microbiology and Virology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
4
|
Castro-Torres RD, Olloquequi J, Parcerisas A, Ureña J, Ettcheto M, Beas-Zarate C, Camins A, Verdaguer E, Auladell C. JNK signaling and its impact on neural cell maturation and differentiation. Life Sci 2024; 350:122750. [PMID: 38801982 DOI: 10.1016/j.lfs.2024.122750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/10/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
C-Jun-N-terminal-kinases (JNKs), members of the mitogen-activated-protein-kinase family, are significantly linked with neurological and neurodegenerative pathologies and cancer progression. However, JNKs serve key roles under physiological conditions, particularly within the central-nervous-system (CNS), where they are critical in governing neural proliferation and differentiation during both embryogenesis and adult stages. These processes control the development of CNS, avoiding neurodevelopment disorders. JNK are key to maintain the proper activity of neural-stem-cells (NSC) and neural-progenitors (NPC) that exist in adults, which keep the convenient brain plasticity and homeostasis. This review underscores how the interaction of JNK with upstream and downstream molecules acts as a regulatory mechanism to manage the self-renewal capacity and differentiation of NSC/NPC during CNS development and in adult neurogenic niches. Evidence suggests that JNK is reliant on non-canonical Wnt components, Fbw7-ubiquitin-ligase, and WDR62-scaffold-protein, regulating substrates such as transcription factors and cytoskeletal proteins. Therefore, understanding which pathways and molecules interact with JNK will bring knowledge on how JNK activation orchestrates neuronal processes that occur in CNS development and brain disorders.
Collapse
Affiliation(s)
- Rubén D Castro-Torres
- Department de Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalonia, Spain; Department of Cell and Molecular Biology, Laboratory of Neurobiotechnology, C.U.C.B.A, Universidad de Guadalajara, Jalisco 44340, Mexico
| | - Jordi Olloquequi
- Department of Biochemistry and Physiology, Physiology Section, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Avda. Diagonal 641, 08028 Barcelona, Catalonia, Spain; Laboratory of Cellular and Molecular Pathology, Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Av. 5 Poniente 1670, 3460000 Talca, Chile
| | - Antoni Parcerisas
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), 08500 Vic, Catalonia, Spain; Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500 Vic, Catalonia, Spain
| | - Jesús Ureña
- Department de Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Miren Ettcheto
- Department de Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Avda. Diagonal 641, E-08028 Barcelona, Catalonia, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Carlos Beas-Zarate
- Department of Cell and Molecular Biology, Laboratory of Neurobiotechnology, C.U.C.B.A, Universidad de Guadalajara, Jalisco 44340, Mexico
| | - Antoni Camins
- Department de Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Avda. Diagonal 641, E-08028 Barcelona, Catalonia, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ester Verdaguer
- Department de Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalonia, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Catalonia, Spain.
| | - Carme Auladell
- Department de Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalonia, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Catalonia, Spain.
| |
Collapse
|
5
|
DeVault L, Mateusiak C, Palucki J, Brent M, Milbrandt J, DiAntonio A. The response of Dual-leucine zipper kinase (DLK) to nocodazole: Evidence for a homeostatic cytoskeletal repair mechanism. PLoS One 2024; 19:e0300539. [PMID: 38574058 PMCID: PMC10994325 DOI: 10.1371/journal.pone.0300539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/28/2024] [Indexed: 04/06/2024] Open
Abstract
Genetic and pharmacological perturbation of the cytoskeleton enhances the regenerative potential of neurons. This response requires Dual-leucine Zipper Kinase (DLK), a neuronal stress sensor that is a central regulator of axon regeneration and degeneration. The damage and repair aspects of this response are reminiscent of other cellular homeostatic systems, suggesting that a cytoskeletal homeostatic response exists. In this study, we propose a framework for understanding DLK mediated neuronal cytoskeletal homeostasis. We demonstrate that low dose nocodazole treatment activates DLK signaling. Activation of DLK signaling results in a DLK-dependent transcriptional signature, which we identify through RNA-seq. This signature includes genes likely to attenuate DLK signaling while simultaneously inducing actin regulating genes. We identify alterations to the cytoskeleton including actin-based morphological changes to the axon. These results are consistent with the model that cytoskeletal disruption in the neuron induces a DLK-dependent homeostatic mechanism, which we term the Cytoskeletal Stress Response (CSR) pathway.
Collapse
Affiliation(s)
- Laura DeVault
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Chase Mateusiak
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Computer Science & Engineering, Washington University, St. Louis, MO, United States of America
| | - John Palucki
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael Brent
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Computer Science & Engineering, Washington University, St. Louis, MO, United States of America
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
6
|
McGregor ER, Lasky DJ, Rippentrop OJ, Clark JP, Wright SLG, Jones MV, Anderson RM. Reversal of neuronal tau pathology, metabolic dysfunction, and electrophysiological defects via adiponectin pathway-dependent AMPK activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579204. [PMID: 38370802 PMCID: PMC10871331 DOI: 10.1101/2024.02.07.579204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Changes in brain mitochondrial metabolism are coincident with functional decline; however, direct links between the two have not been established. Here, we show that mitochondrial targeting via the adiponectin receptor activator AdipoRon (AR) clears neurofibrillary tangles (NFTs) and rescues neuronal tauopathy-associated defects. AR reduced levels of phospho-tau and lowered NFT burden by a mechanism involving the energy-sensing kinase AMPK and the growth-sensing kinase GSK3b. The transcriptional response to AR included broad metabolic and functional pathways. Induction of lysosomal pathways involved activation of LC3 and p62, and restoration of neuronal outgrowth required the stress-responsive kinase JNK. Negative consequences of NFTs on mitochondrial activity, ATP production, and lipid stores were corrected. Defects in electrophysiological measures (e.g., resting potential, resistance, spiking profiles) were also corrected. These findings reveal a network linking mitochondrial function, cellular maintenance processes, and electrical aspects of neuronal function that can be targeted via adiponectin receptor activation.
Collapse
Affiliation(s)
- Eric R McGregor
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Danny J Lasky
- Department. of Neuroscience, Univ. of Wisconsin-Madison, Madison, WI
| | | | - Josef P Clark
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
| | | | - Mathew V Jones
- Department. of Neuroscience, Univ. of Wisconsin-Madison, Madison, WI
| | - Rozalyn M Anderson
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
- GRECC William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
7
|
Becker CJ, Cigliola V, Gillotay P, Rich A, De Simone A, Han Y, Di Talia S, Poss KD. In toto imaging of glial JNK signaling during larval zebrafish spinal cord regeneration. Development 2023; 150:dev202076. [PMID: 37997694 PMCID: PMC10753585 DOI: 10.1242/dev.202076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023]
Abstract
Identification of signaling events that contribute to innate spinal cord regeneration in zebrafish can uncover new targets for modulating injury responses of the mammalian central nervous system. Using a chemical screen, we identify JNK signaling as a necessary regulator of glial cell cycling and tissue bridging during spinal cord regeneration in larval zebrafish. With a kinase translocation reporter, we visualize and quantify JNK signaling dynamics at single-cell resolution in glial cell populations in developing larvae and during injury-induced regeneration. Glial JNK signaling is patterned in time and space during development and regeneration, decreasing globally as the tissue matures and increasing in the rostral cord stump upon transection injury. Thus, dynamic and regional regulation of JNK signaling help to direct glial cell behaviors during innate spinal cord regeneration.
Collapse
Affiliation(s)
- Clayton J. Becker
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Valentina Cigliola
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose, 06100 Nice, France
| | - Pierre Gillotay
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ashley Rich
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alessandro De Simone
- Department of Genetics and Evolution, University of Geneva, 1211 Geneva, Switzerland
| | - Yanchao Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, 215006 Jiangsu, China
| | - Stefano Di Talia
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kenneth D. Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
8
|
DeVault L, Mateusiak C, Palucki J, Brent M, Milbrandt J, DiAntonio A. The response of Dual-Leucine Zipper Kinase (DLK) to nocodazole: evidence for a homeostatic cytoskeletal repair mechanism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.06.561227. [PMID: 37873434 PMCID: PMC10592635 DOI: 10.1101/2023.10.06.561227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Genetic and pharmacological perturbation of the cytoskeleton enhances the regenerative potential of neurons. This response requires Dual-leucine Zipper Kinase (DLK), a neuronal stress sensor that is a central regulator of axon regeneration and degeneration. The damage and repair aspects of this response are reminiscent of other cellular homeostatic systems, suggesting that a cytoskeletal homeostatic response exists. In this study, we propose a framework for understanding DLK mediated neuronal cytoskeletal homeostasis. We demonstrate that a) low dose nocodazole treatment activates DLK signaling and b) DLK signaling mitigates the microtubule damage caused by the cytoskeletal perturbation. We also perform RNA-seq to discover a DLK-dependent transcriptional signature. This signature includes genes likely to attenuate DLK signaling while simultaneously inducing actin regulating genes and promoting actin-based morphological changes to the axon. These results are consistent with the model that cytoskeletal disruption in the neuron induces a DLK-dependent homeostatic mechanism, which we term the Cytoskeletal Stress Response (CSR) pathway.
Collapse
|
9
|
Bu H, Li Z, Lu Y, Zhuang Z, Zhen Y, Zhang L. Deciphering the multifunctional role of dual leucine zipper kinase (DLK) and its therapeutic potential in disease. Eur J Med Chem 2023; 255:115404. [PMID: 37098296 DOI: 10.1016/j.ejmech.2023.115404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 04/27/2023]
Abstract
Dual leucine zipper kinase (DLK, MAP3K12), a serine/threonine protein kinase, plays a key role in neuronal development, as it regulates axon regeneration and degeneration through its downstream kinase. Importantly, DLK is closely related to the pathogenesis of numerous neurodegenerative diseases and the induction of β-cell apoptosis that leads to diabetes. In this review, we summarize the current understanding of DLK function, and then discuss the role of DLK signaling in human diseases. Furthermore, various types of small molecule inhibitors of DLK that have been published so far are described in detail in this paper, providing some strategies for the design of DLK small molecule inhibitors in the future.
Collapse
Affiliation(s)
- Haiqing Bu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yingying Lu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhiyao Zhuang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yongqi Zhen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
10
|
Scharrenberg R, Richter M, Johanns O, Meka DP, Rücker T, Murtaza N, Lindenmaier Z, Ellegood J, Naumann A, Zhao B, Schwanke B, Sedlacik J, Fiehler J, Hanganu-Opatz IL, Lerch JP, Singh KK, de Anda FC. TAOK2 rescues autism-linked developmental deficits in a 16p11.2 microdeletion mouse model. Mol Psychiatry 2022; 27:4707-4721. [PMID: 36123424 PMCID: PMC9734055 DOI: 10.1038/s41380-022-01785-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022]
Abstract
The precise development of the neocortex is a prerequisite for higher cognitive and associative functions. Despite numerous advances that have been made in understanding neuronal differentiation and cortex development, our knowledge regarding the impact of specific genes associated with neurodevelopmental disorders on these processes is still limited. Here, we show that Taok2, which is encoded in humans within the autism spectrum disorder (ASD) susceptibility locus 16p11.2, is essential for neuronal migration. Overexpression of de novo mutations or rare variants from ASD patients disrupts neuronal migration in an isoform-specific manner. The mutated TAOK2α variants but not the TAOK2β variants impaired neuronal migration. Moreover, the TAOK2α isoform colocalizes with microtubules. Consequently, neurons lacking Taok2 have unstable microtubules with reduced levels of acetylated tubulin and phosphorylated JNK1. Mice lacking Taok2 develop gross cortical and cortex layering abnormalities. Moreover, acute Taok2 downregulation or Taok2 knockout delayed the migration of upper-layer cortical neurons in mice, and the expression of a constitutively active form of JNK1 rescued these neuronal migration defects. Finally, we report that the brains of the Taok2 KO and 16p11.2 del Het mouse models show striking anatomical similarities and that the heterozygous 16p11.2 microdeletion mouse model displayed reduced levels of phosphorylated JNK1 and neuronal migration deficits, which were ameliorated upon the introduction of TAOK2α in cortical neurons and in the developing cortex of those mice. These results delineate the critical role of TAOK2 in cortical development and its contribution to neurodevelopmental disorders, including ASD.
Collapse
Affiliation(s)
- Robin Scharrenberg
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Melanie Richter
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| | - Ole Johanns
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Durga Praveen Meka
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Tabitha Rücker
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Nadeem Murtaza
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, L8S 4A9, Canada
| | - Zsuzsa Lindenmaier
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
| | - Anne Naumann
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Bing Zhao
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Birgit Schwanke
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Jan Sedlacik
- Department of Neuroradiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jens Fiehler
- Department of Neuroradiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford, OX3 9DU, UK
| | - Karun K Singh
- Krembil Research Institute, Donald K. Johnson Eye Institute, University Health Network, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| | - Froylan Calderon de Anda
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
11
|
Bales B, Cotero V, Meyer DE, Roberts JC, Rodriguez-Silva M, Siclovan TM, Chambers JW, Rishel MJ. Radiolabeled Aminopyrazoles as Novel Isoform Selective Probes for pJNK3 Quantification. ACS Med Chem Lett 2022; 13:1606-1614. [PMID: 36262398 PMCID: PMC9575163 DOI: 10.1021/acsmedchemlett.2c00278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
The c-Jun N-terminal kinase 3 (JNK3) is a stress-activated kinase primarily expressed in the brain and implicated as an early mediator of neuronal apoptosis. We sought to develop a PET tracer to visualize pathological JNK3 activation. Because regional JNK3 activation precedes apoptosis, such an imaging agent might enable the detection of "at risk" brain regions prior to neuronal death. We prepared a set of 19F-containing compounds on the basis of the reported aminopyrazoles. The candidate, F3, was tritiated and used in autoradiography experiments to demonstrate regional and temporal changes in JNK3 activation in a mouse model of Parkinson's disease. A significant increase in pJNK3 B max versus control animals in multiple brain regions was observed at 8 months, including the ventral midbrain. Pathological activation of JNK3 in these regions preceded statistically significant neuron loss. Analyses of brain concentrations of [18F]-F3 in naïve rats following intravenous injection revealed a small but detectable signal over the background, but was likely not sufficient to support PET imaging.
Collapse
Affiliation(s)
- Brian
C. Bales
- Department
of Biology and Applied Physics, GE Research, One Research Circle, Niskayuna, New York 12309, United States
| | - Victoria Cotero
- Department
of Biology and Applied Physics, GE Research, One Research Circle, Niskayuna, New York 12309, United States
| | - Dan E. Meyer
- Department
of Biology and Applied Physics, GE Research, One Research Circle, Niskayuna, New York 12309, United States
| | - Jeannette C. Roberts
- Department
of Biology and Applied Physics, GE Research, One Research Circle, Niskayuna, New York 12309, United States
| | - Monica Rodriguez-Silva
- Department
of Environmental Health Sciences, Robert Stempel College of Public
Health & Social Work, Florida International
University, Miami, Florida 33199, United States
| | - Tiberiu M. Siclovan
- Department
of Biology and Applied Physics, GE Research, One Research Circle, Niskayuna, New York 12309, United States
| | - Jeremy W. Chambers
- Department
of Environmental Health Sciences, Robert Stempel College of Public
Health & Social Work, Florida International
University, Miami, Florida 33199, United States
| | - Michael J. Rishel
- Department
of Biology and Applied Physics, GE Research, One Research Circle, Niskayuna, New York 12309, United States
| |
Collapse
|
12
|
Petrović A, Ban J, Ivaničić M, Tomljanović I, Mladinic M. The Role of ATF3 in Neuronal Differentiation and Development of Neuronal Networks in Opossum Postnatal Cortical Cultures. Int J Mol Sci 2022; 23:ijms23094964. [PMID: 35563354 PMCID: PMC9100162 DOI: 10.3390/ijms23094964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 12/14/2022] Open
Abstract
Activating transcription factor 3 (ATF3), a member of the ATF/cAMP response element-binding (CREB) family, is upregulated by various intracellular and extracellular signals such as injury and signals related to cell proliferation. ATF3 also belongs to the regeneration-associated genes (RAG) group of transcription factors. RAG and ATF/CREB transcription factors that play an important role in embryonic neuronal development and PNS regeneration may also be involved in postnatal neuronal differentiation and development, as well as in the regeneration of the injured CNS. Here we investigated the effect of ATF3 in differentiation, neural outgrowth, network formation, and regeneration after injury using postnatal dissociated cortical neurons derived from neonatal opossums (Monodelphis domestica). Our results show that RAG and ATF genes are differentially expressed in early differentiated neurons versus undifferentiated neurospheres and that many members of those families, ATF3 in particular, are upregulated in cortical cultures obtained from younger animals that have the ability to fully functionally regenerate spinal cord after injury. In addition, we observed different intracellular localization of ATF3 that shifts from nuclear (in neuronal progenitors) to cytoplasmic (in more mature neurons) during neuronal differentiation. The ATF3 inhibition, pharmacological or by specific antibody, reduced the neurite outgrowth and differentiation and caused increased cell death in early differentiating cortical neuronal cultures, suggesting the importance of ATF3 in the CNS development of neonatal opossums. Finally, we investigated the regeneration capacity of primary cortical cultures after mechanical injury using the scratch assay. Remarkably, neonatal opossum-derived cultures retain their capacity to regenerate for up to 1 month in vitro. Inhibition of ATF3 correlates with reduced neurite outgrowth and regeneration after injury. These results indicate that ATF3, and possibly other members of RAG and ATF/CREB family of transcription factors, have an important role both during cortical postnatal development and in response after injury.
Collapse
|
13
|
ASK1 is a novel molecular target for preventing aminoglycoside-induced hair cell death. J Mol Med (Berl) 2022; 100:797-813. [PMID: 35471608 PMCID: PMC9110505 DOI: 10.1007/s00109-022-02188-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 02/07/2022] [Accepted: 03/03/2022] [Indexed: 10/31/2022]
Abstract
Aminoglycoside antibiotics are lifesaving medicines, crucial for the treatment of chronic or drug resistant infections. However, aminoglycosides are toxic to the sensory hair cells in the inner ear. As a result, aminoglycoside-treated individuals can develop permanent hearing loss and vestibular impairment. There is considerable evidence that reactive oxygen species (ROS) production and the subsequent phosphorylation of c-Jun N-terminal kinase (JNK) and P38 mitogen-activated protein kinase (P38) drives apoptosis in aminoglycoside-treated hair cells. However, treatment strategies that directly inhibit ROS, JNK, or P38 are limited by the importance of these molecules for normal cellular function. Alternatively, the upstream regulator apoptosis signal-regulating kinase 1 (ASK1/MAP3K5) is a key mediator of ROS-induced JNK and P38 activation under pathologic but not homeostatic conditions. We investigated ASK1 as a mediator of drug-induced hair cell death using cochlear explants from Ask1 knockout mice, demonstrating that Ask1 deficiency attenuates neomycin-induced hair cell death. We then evaluated pharmacological inhibition of ASK1 with GS-444217 as a potential otoprotective therapy. GS-444217 significantly attenuated hair cell death in neomycin-treated explants but did not impact aminoglycoside efficacy against P. aeruginosa in the broth dilution test. Overall, we provide significant pre-clinical evidence that ASK1 inhibition represents a novel strategy for preventing aminoglycoside ototoxicity. KEY MESSAGES: • ASK1 is an upstream, redox-sensitive regulator of P38 and JNK, which are known mediators of hair cell death. • Ask1 knockout does not affect hair cell development in vivo, but significantly reduces aminoglycoside-induced hair cell death in vitro. • A small-molecule inhibitor of ASK1 attenuates neomycin-induced hair cell death, and does not impact antibiotic efficacy in vitro. • ASK1 may be a novel molecular target for preventing aminoglycoside-induced hearing loss.
Collapse
|
14
|
Cunningham JG, Scripter JD, Nti SA, Tucker ES. Early construction of the thalamocortical axon pathway requires c-Jun N-terminal kinase signaling within the ventral forebrain. Dev Dyn 2022; 251:459-480. [PMID: 34494344 PMCID: PMC8891049 DOI: 10.1002/dvdy.416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Thalamocortical connectivity is essential for normal brain function. This important pathway is established during development, when thalamic axons extend a long distance through the forebrain before reaching the cerebral cortex. In this study, we identify a novel role for the c-Jun N-terminal kinase (JNK) signaling pathway in guiding thalamocortical axons through intermediate target territories. RESULTS Complete genetic removal of JNK signaling from the Distal-less 5/6 (Dlx5/6) domain in mice prevents thalamocortical axons from crossing the diencephalon-telencephalon boundary (DTB) and the internal capsule fails to form. Ventral telencephalic cells critical for thalamocortical axon extensions including corridor and guidepost neurons are also disrupted. In addition, corticothalamic, striatonigral, and nigrostriatal axons fail to cross the DTB. Analyses of different JNK mutants demonstrate that thalamocortical axon pathfinding has a non-autonomous requirement for JNK signaling. CONCLUSIONS We conclude that JNK signaling within the Dlx5/6 territory enables the construction of major axonal pathways in the developing forebrain. Further exploration of this intermediate axon guidance territory is needed to uncover mechanisms of axonal pathfinding during normal brain development and to elucidate how this vital process may be compromised in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jessica G. Cunningham
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506,Neuroscience Graduate Program, West Virginia University School of Medicine, Morgantown, WV 26506,Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - James D. Scripter
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506,Neuroscience Graduate Program, West Virginia University School of Medicine, Morgantown, WV 26506,Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Stephany A. Nti
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506,Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Eric S. Tucker
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506,Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506
| |
Collapse
|
15
|
Negi P, Cheke RS, Patil VM. Recent advances in pharmacological diversification of Src family kinase inhibitors. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00172-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Background
Src kinase, a nonreceptor protein-tyrosine kinase is composed of 11 members (in human) and is involved in a wide variety of essential functions required to sustain cellular homeostasis and survival.
Main body of the abstract
Deregulated activity of Src family kinase is related to malignant transformation. In 2001, Food and Drug Administration approved imatinib for the treatment of chronic myeloid leukemia followed by approval of various other inhibitors from this category as effective therapeutics for cancer patients. In the past decade, Src family kinase has been investigated for the treatment of diverse pathologies in addition to cancer. In this regard, we provide a systematic evaluation of Src kinase regarding its mechanistic role in cancer and other diseases. Here we comment on preclinical and clinical success of Src kinase inhibitors in cancer followed by diabetes, hypertension, tuberculosis, and inflammation.
Short conclusion
Studies focusing on the diversified role of Src kinase as potential therapeutical target for the development of medicinally active agents might produce significant advances in the management of not only various types of cancer but also other diseases which are in demand for potent and safe therapeutics.
Collapse
|
16
|
Abstract
Obesity, which has long since reached epidemic proportions worldwide, is associated with long-term stress to a variety of organs and results in diseases including type 2 diabetes. In the brain, overnutrition induces hypothalamic stress associated with the activation of several signalling pathways, together with central insulin and leptin resistance. This central action of nutrient overload appears very rapidly, suggesting that nutrition-induced hypothalamic stress is a major upstream initiator of obesity and associated diseases. The cellular response to nutrient overload includes the activation of the stress-activated c-Jun N-terminal kinases (JNKs) JNK1, JNK2 and JNK3, which are widely expressed in the brain. Here, we review recent findings on the regulation and effects of these kinases, with particular focus on the hypothalamus, a key brain region in the control of energy and glucose homeostasis. JNK1 blocks the hypothalamic-pituitary-thyroid axis, reducing energy expenditure and promoting obesity. Recently, opposing roles have been identified for JNK1 and JNK3 in hypothalamic agouti gene-related protein (AgRP) neurons: while JNK1 activation in AgRP neurons induces feeding and weight gain and impairs insulin and leptin signalling, JNK3 (also known as MAPK10) deletion in the same neuronal population produces very similar effects. The opposing roles of these kinases, and the unknown role of hypothalamic JNK2, reflect the complexity of JNK biology. Future studies should address the specific function of each kinase, not only in different neuronal subsets, but also in non-neuronal cells in the central nervous system. Decoding the puzzle of brain stress kinases will help to define the central stimuli and mechanisms implicated in the control of energy balance. Graphical abstract.
Collapse
Affiliation(s)
- Rubén Nogueiras
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Guadalupe Sabio
- Department of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
17
|
Igarashi M, Honda A, Kawasaki A, Nozumi M. Neuronal Signaling Involved in Neuronal Polarization and Growth: Lipid Rafts and Phosphorylation. Front Mol Neurosci 2020; 13:150. [PMID: 32922262 PMCID: PMC7456915 DOI: 10.3389/fnmol.2020.00150] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
Neuronal polarization and growth are developmental processes that occur during neuronal cell differentiation. The molecular signaling mechanisms involved in these events in in vivo mammalian brain remain unclear. Also, cellular events of the neuronal polarization process within a given neuron are thought to be constituted of many independent intracellular signal transduction pathways (the "tug-of-war" model). However, in vivo results suggest that such pathways should be cooperative with one another among a given group of neurons in a region of the brain. Lipid rafts, specific membrane domains with low fluidity, are candidates for the hotspots of such intracellular signaling. Among the signals reported to be involved in polarization, a number are thought to be present or translocated to the lipid rafts in response to extracellular signals. As part of our analysis, we discuss how such novel molecular mechanisms are combined for effective regulation of neuronal polarization and growth, focusing on the significance of the lipid rafts, including results based on recently introduced methods.
Collapse
Affiliation(s)
- Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Atsuko Honda
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| |
Collapse
|
18
|
Involvement of JNK1 in Neuronal Polarization During Brain Development. Cells 2020; 9:cells9081897. [PMID: 32823764 PMCID: PMC7466125 DOI: 10.3390/cells9081897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/16/2022] Open
Abstract
The c-Jun N-terminal Kinases (JNKs) are a group of regulatory elements responsible for the control of a wide array of functions within the cell. In the central nervous system (CNS), JNKs are involved in neuronal polarization, starting from the cell division of neural stem cells and ending with their final positioning when migrating and maturing. This review will focus mostly on isoform JNK1, the foremost contributor of total JNK activity in the CNS. Throughout the text, research from multiple groups will be summarized and discussed in order to describe the involvement of the JNKs in the different steps of neuronal polarization. The data presented support the idea that isoform JNK1 is highly relevant to the regulation of many of the processes that occur in neuronal development in the CNS.
Collapse
|
19
|
Belrose JL, Prasad A, Sammons MA, Gibbs KM, Szaro BG. Comparative gene expression profiling between optic nerve and spinal cord injury in Xenopus laevis reveals a core set of genes inherent in successful regeneration of vertebrate central nervous system axons. BMC Genomics 2020; 21:540. [PMID: 32758133 PMCID: PMC7430912 DOI: 10.1186/s12864-020-06954-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 07/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The South African claw-toed frog, Xenopus laevis, is uniquely suited for studying differences between regenerative and non-regenerative responses to CNS injury within the same organism, because some CNS neurons (e.g., retinal ganglion cells after optic nerve crush (ONC)) regenerate axons throughout life, whereas others (e.g., hindbrain neurons after spinal cord injury (SCI)) lose this capacity as tadpoles metamorphose into frogs. Tissues from these CNS regions (frog ONC eye, tadpole SCI hindbrain, frog SCI hindbrain) were used in a three-way RNA-seq study of axotomized CNS axons to identify potential core gene expression programs for successful CNS axon regeneration. RESULTS Despite tissue-specific changes in expression dominating the injury responses of each tissue, injury-induced changes in gene expression were nonetheless shared between the two axon-regenerative CNS regions that were not shared with the non-regenerative region. These included similar temporal patterns of gene expression and over 300 injury-responsive genes. Many of these genes and their associated cellular functions had previously been associated with injury responses of multiple tissues, both neural and non-neural, from different species, thereby demonstrating deep phylogenetically conserved commonalities between successful CNS axon regeneration and tissue regeneration in general. Further analyses implicated the KEGG adipocytokine signaling pathway, which links leptin with metabolic and gene regulatory pathways, and a novel gene regulatory network with genes regulating chromatin accessibility at its core, as important hubs in the larger network of injury response genes involved in successful CNS axon regeneration. CONCLUSIONS This study identifies deep, phylogenetically conserved commonalities between CNS axon regeneration and other examples of successful tissue regeneration and provides new targets for studying the molecular underpinnings of successful CNS axon regeneration, as well as a guide for distinguishing pro-regenerative injury-induced changes in gene expression from detrimental ones in mammals.
Collapse
Affiliation(s)
- Jamie L Belrose
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Aparna Prasad
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Kurt M Gibbs
- Department of Biology and Chemistry, Morehead State University, Morehead, KY, 40351, USA
| | - Ben G Szaro
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA.
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA.
| |
Collapse
|
20
|
Teravskis PJ, Ashe KH, Liao D. The Accumulation of Tau in Postsynaptic Structures: A Common Feature in Multiple Neurodegenerative Diseases? Neuroscientist 2020; 26:503-520. [PMID: 32389059 DOI: 10.1177/1073858420916696] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increasingly, research suggests that neurodegenerative diseases and dementias are caused not by unique, solitary cellular mechanisms, but by multiple contributory mechanisms manifesting as heterogeneous clinical presentations. However, diverse neurodegenerative diseases also share common pathological hallmarks and cellular mechanisms. One such mechanism involves the redistribution of the microtubule associated protein tau from the axon into the somatodendritic compartment of neurons, followed by the mislocalization of tau into dendritic spines, resulting in postsynaptic functional deficits. Here we review various signaling pathways that trigger the redistribution of tau to the cell body and dendritic tree, and its mislocalization to dendritic spines. The convergence of multiple pathways in different disease models onto this final common pathway suggests that it may be an attractive pathway to target for developing new treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter J Teravskis
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.,University of Minnesota Medical School, Minneapolis, MN, USA
| | - Karen H Ashe
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.,N. Budd Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA.,Geriatric Research Education and Clinical Center, Veterans Affairs Medical Center, Minneapolis, MN, USA
| | - Dezhi Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
21
|
Schellino R, Boido M, Vercelli A. JNK Signaling Pathway Involvement in Spinal Cord Neuron Development and Death. Cells 2019; 8:E1576. [PMID: 31817379 PMCID: PMC6953032 DOI: 10.3390/cells8121576] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022] Open
Abstract
The c-Jun NH2-terminal protein kinase (JNK) is a Janus-faced kinase, which, in the nervous system, plays important roles in a broad range of physiological and pathological processes. Three genes, encoding for 10 JNK isoforms, have been identified: jnk1, jnk2, and jnk3. In the developing spinal cord, JNK proteins control neuronal polarity, axon growth/pathfinding, and programmed cell death; in adulthood they can drive degeneration and regeneration, after pathological insults. Indeed, recent studies have highlighted a role for JNK in motor neuron (MN) diseases, such as amyotrophic lateral sclerosis and spinal muscular atrophy. In this review we discuss how JNK-dependent signaling regulates apparently contradictory functions in the spinal cord, in both the developmental and adult stages. In addition, we examine the evidence that the specific targeting of JNK signaling pathway may represent a promising therapeutic strategy for the treatment of MN diseases.
Collapse
Affiliation(s)
- Roberta Schellino
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy
| | - Marina Boido
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy
- National Institute of Neuroscience (INN), 10125 Turin, Italy
| | - Alessandro Vercelli
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano (TO), Italy
- National Institute of Neuroscience (INN), 10125 Turin, Italy
| |
Collapse
|
22
|
Horgusluoglu-Moloch E, Risacher SL, Crane PK, Hibar D, Thompson PM, Saykin AJ, Nho K. Genome-wide association analysis of hippocampal volume identifies enrichment of neurogenesis-related pathways. Sci Rep 2019; 9:14498. [PMID: 31601890 PMCID: PMC6787090 DOI: 10.1038/s41598-019-50507-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 09/09/2019] [Indexed: 01/04/2023] Open
Abstract
Adult neurogenesis occurs in the dentate gyrus of the hippocampus during adulthood and contributes to sustaining the hippocampal formation. To investigate whether neurogenesis-related pathways are associated with hippocampal volume, we performed gene-set enrichment analysis using summary statistics from a large-scale genome-wide association study (N = 13,163) of hippocampal volume from the Enhancing Neuro Imaging Genetics through Meta-Analysis (ENIGMA) Consortium and two year hippocampal volume changes from baseline in cognitively normal individuals from Alzheimer's Disease Neuroimaging Initiative Cohort (ADNI). Gene-set enrichment analysis of hippocampal volume identified 44 significantly enriched biological pathways (FDR corrected p-value < 0.05), of which 38 pathways were related to neurogenesis-related processes including neurogenesis, generation of new neurons, neuronal development, and neuronal migration and differentiation. For genes highly represented in the significantly enriched neurogenesis-related pathways, gene-based association analysis identified TESC, ACVR1, MSRB3, and DPP4 as significantly associated with hippocampal volume. Furthermore, co-expression network-based functional analysis of gene expression data in the hippocampal subfields, CA1 and CA3, from 32 normal controls showed that distinct co-expression modules were mostly enriched in neurogenesis related pathways. Our results suggest that neurogenesis-related pathways may be enriched for hippocampal volume and that hippocampal volume may serve as a potential phenotype for the investigation of human adult neurogenesis.
Collapse
Grants
- UL1 TR001108 NCATS NIH HHS
- R01 CA129769 NCI NIH HHS
- R35 CA197289 NCI NIH HHS
- P50 GM115318 NIGMS NIH HHS
- R01 AG019771 NIA NIH HHS
- P30 AG010133 NIA NIH HHS
- R03 AG054936 NIA NIH HHS
- U01 AG024904 NIA NIH HHS
- UL1 TR002369 NCATS NIH HHS
- R01 LM011360 NLM NIH HHS
- U54 EB020403 NIBIB NIH HHS
- K01 AG049050 NIA NIH HHS
- R01 LM012535 NLM NIH HHS
- CIHR
- NLM R01 LM012535, NIA R03 AG054936, NIA R01 AG19771, NIA P30 AG10133, NLM R01 LM011360, NSF IIS-1117335, DOD W81XWH-14-2-0151, NCAA 14132004, NIGMS P50GM115318, NCATS UL1 TR001108, NIA K01 AG049050, the Alzheimer’s Association, the Indiana Clinical and Translational Science Institute, and the IU Health-IU School of Medicine Strategic Neuroscience Research Initiative.
- ENIGMA was supported in part by a Consortium grant (U54EB020403 to PMT) from the NIH Institutes contributing to the Big Data to Knowledge (BD2K) Initiative, including the NIBIB and NCI.
- Data collection and sharing for this project was funded by the Alzheimer's Disease Neuroimaging Initiative (ADNI) (National Institutes of Health Grant U01 AG024904) and DOD ADNI (Department of Defense award number W81XWH-12-2-0012). ADNI is funded by the National Institute on Aging, the National Institute of Biomedical Imaging and Bioengineering, and through generous contributions from the following: AbbVie, Alzheimer’s Association; Alzheimer’s Drug Discovery Foundation; Araclon Biotech; BioClinica, Inc.; Biogen; Bristol-Myers Squibb Company; CereSpir, Inc.; Cogstate; Eisai Inc.; Elan Pharmaceuticals, Inc.; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.; Fujirebio; GE Healthcare; IXICO Ltd.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Pharmaceutical Research & Development LLC.; Lumosity; Lundbeck; Merck & Co., Inc.; Meso Scale Diagnostics, LLC.; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer Inc.; Piramal Imaging; Servier; Takeda Pharmaceutical Company; and Transition Therapeutics. The Canadian Institutes of Health Research is providing funds to support ADNI clinical sites in Canada. Private sector contributions are facilitated by the Foundation for the National Institutes of Health (www.fnih.org). The grantee organization is the Northern California Institute for Research and Education, and the study is coordinated by the Alzheimer’s Therapeutic Research Institute at the University of Southern California. ADNI data are disseminated by the Laboratory for Neuro Imaging at the University of Southern California. Additional support for data analysis was provided by NLM R01 LM012535, NIA R03 AG054936, NIA R01 AG19771, NIA P30 AG10133, NLM R01 LM011360, NSF IIS-1117335, DOD W81XWH-14-2-0151, NCAA 14132004, NIGMS P50GM115318, NCATS UL1 TR001108, NIA K01 AG049050, the Alzheimer’s Association, the Indiana Clinical and Translational Science Institute, and the IU Health-IU School of Medicine Strategic Neuroscience Research Initiative.
Collapse
Affiliation(s)
- Emrin Horgusluoglu-Moloch
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, School of Medicine, Seattle, WA, USA
| | - Derrek Hibar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Biomarkers, Janssen Research and Development, LLC, San Diego, CA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew J Saykin
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Kwangsik Nho
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
23
|
Igarashi M, Okuda S. Evolutionary analysis of proline-directed phosphorylation sites in the mammalian growth cone identified using phosphoproteomics. Mol Brain 2019; 12:53. [PMID: 31151465 PMCID: PMC6545026 DOI: 10.1186/s13041-019-0476-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/27/2019] [Indexed: 12/15/2022] Open
Abstract
The growth cone is essential for nerve growth and axon regeneration, which directly form and rearrange the neural network. Recently, to clarify the molecular signaling pathways in the growth cone that utilize protein phosphorylation, we performed a phosphoproteomics study of mammalian growth cone membranes derived from the developing rodent brain and identified > 30,000 phosphopeptides from ~ 1200 proteins. We found that the phosphorylation sites were highly proline directed and primarily mitogen-activated protein kinase (MAPK) dependent, due to particular activation of c-jun N-terminal protein kinase (JNK), a member of the MAPK family. Because the MAPK/JNK pathway is also involved in axon regeneration of invertebrate model organisms such Caenorhabditis elegans and Drosophila, we performed evolutionary bioinformatics analysis of the mammalian growth cone phosphorylation sites. Although these sites were generally conserved within vertebrates, they were not necessarily conserved in these invertebrate model organisms. In particular, high-frequency phosphorylation sites (> 20 times) were less conserved than low-frequency sites. Taken together, the mammalian growth cones contain a large number of vertebrate-specific phosphorylation sites and stronger dependence upon MAPK/JNK than C. elegans or Drosophila. We conclude that axon growth/regeneration likely involves many vertebrate-specific phosphorylation sites.
Collapse
Affiliation(s)
- Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi, Chuo-ku, Niigata, 951-8510, Japan.
| | - Shujiro Okuda
- Laboratory of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
24
|
Kinase Inhibitors with Antiepileptic Properties Identified with a Novel in Vitro Screening Platform. Int J Mol Sci 2019; 20:ijms20102502. [PMID: 31117204 PMCID: PMC6566965 DOI: 10.3390/ijms20102502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
Kinase signaling plays an important role in acquired epilepsy, but only a small percentage of the total kinome has been investigated in this context. A major roadblock that prevents the systematic investigation of the contributions of kinase signaling networks is the slow speed of experiments designed to test the chronic effects of target inhibition in epilepsy models. We developed a novel in vitro screening platform based on microwire recordings from an organotypic hippocampal culture model of acquired epilepsy. This platform enables the direct, parallel determination of the effects of compounds on spontaneous epileptiform activity. The platform also enables repeated recordings from the same culture over two-week long experiments. We screened 45 kinase inhibitors and quantified their effects on seizure duration, the frequency of paroxysmal activity, and electrographic load. We identified several inhibitors with previously unknown antiepileptic properties. We also used kinase inhibition profile cross-referencing to identify kinases that are inhibited by seizure-suppressing compounds, but not by compounds that had no effect on seizures.
Collapse
|
25
|
Lin C, Chen P, Chan H, Huang Y, Chang NW. Peroxisome proliferator‐activated receptor alpha accelerates neuronal differentiation and this might involve the mitogen‐activated protein kinase pathway. Int J Dev Neurosci 2018; 71:46-51. [DOI: 10.1016/j.ijdevneu.2018.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023] Open
Affiliation(s)
- Chingju Lin
- Department of PhysiologyCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Pei‐Yi Chen
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Hsu‐Chin Chan
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Yi‐Ping Huang
- Department of PhysiologyCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| | - Nai Wen Chang
- Department of BiochemistryCollege of Medicine, China Medical UniversityTaichungTaiwan, ROC
| |
Collapse
|
26
|
Hapak SM, Rothlin CV, Ghosh S. PAR3-PAR6-atypical PKC polarity complex proteins in neuronal polarization. Cell Mol Life Sci 2018; 75:2735-2761. [PMID: 29696344 PMCID: PMC11105418 DOI: 10.1007/s00018-018-2828-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/03/2018] [Accepted: 04/23/2018] [Indexed: 01/01/2023]
Abstract
Polarity is a fundamental feature of cells. Protein complexes, including the PAR3-PAR6-aPKC complex, have conserved roles in establishing polarity across a number of eukaryotic cell types. In neurons, polarity is evident as distinct axonal versus dendritic domains. The PAR3, PAR6, and aPKC proteins also play important roles in neuronal polarization. During this process, either aPKC kinase activity, the assembly of the PAR3-PAR6-aPKC complex or the localization of these proteins is regulated downstream of a number of signaling pathways. In turn, the PAR3, PAR6, and aPKC proteins control various effector molecules to establish neuronal polarity. Herein, we discuss the many signaling mechanisms and effector functions that have been linked to PAR3, PAR6, and aPKC during the establishment of neuronal polarity.
Collapse
Affiliation(s)
- Sophie M Hapak
- Department of Medicine, School of Medicine, University of Minnesota, 401 East River Parkway, Minneapolis, MN, 55455, USA.
| | - Carla V Rothlin
- Department of Immunobiology, School of Medicine, Yale University, 300 Cedar Street, New Haven, CT, 06520, USA
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Sourav Ghosh
- Department of Neurology, School of Medicine, Yale University, 300 George Street, New Haven, CT, 06511, USA
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| |
Collapse
|
27
|
Kawasaki A, Okada M, Tamada A, Okuda S, Nozumi M, Ito Y, Kobayashi D, Yamasaki T, Yokoyama R, Shibata T, Nishina H, Yoshida Y, Fujii Y, Takeuchi K, Igarashi M. Growth Cone Phosphoproteomics Reveals that GAP-43 Phosphorylated by JNK Is a Marker of Axon Growth and Regeneration. iScience 2018; 4:190-203. [PMID: 30240740 PMCID: PMC6147025 DOI: 10.1016/j.isci.2018.05.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/05/2018] [Accepted: 05/25/2018] [Indexed: 12/20/2022] Open
Abstract
Neuronal growth cones are essential for nerve growth and regeneration, as well as for the formation and rearrangement of the neural network. To elucidate phosphorylation-dependent signaling pathways and establish useful molecular markers for axon growth and regeneration, we performed a phosphoproteomics study of mammalian growth cones, which identified >30,000 phosphopeptides of ∼1,200 proteins. The phosphorylation sites were highly proline directed and primarily MAPK dependent, owing to the activation of JNK, suggesting that proteins that undergo proline-directed phosphorylation mediate nerve growth in the mammalian brain. Bioinformatics analysis revealed that phosphoproteins were enriched in microtubules and the cortical cytoskeleton. The most frequently phosphorylated site was S96 of GAP-43 (growth-associated protein 43-kDa), a vertebrate-specific protein involved in axon growth. This previously uncharacterized phosphorylation site was JNK dependent. S96 phosphorylation was specifically detected in growing and regenerating axons as the most frequent target of JNK signaling; thus it represents a promising new molecular marker for mammalian axonal growth and regeneration. Phosphoproteomics of mammalian growth cone membranes reveals activation of MAPK JNK is the activated MAPK in growth cones and phosphorylates S96 of GAP-43 pS96 of GAP-43, the most frequent site, is observed in growing axons pS96 is biochemically detected in the regenerating axons of the peripheral nerves
Collapse
Affiliation(s)
- Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Masayasu Okada
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan; Department of Neurosurgery, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Atsushi Tamada
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan; Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Shujiro Okuda
- Laboratory of Bioinformatics, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Yasuyuki Ito
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan
| | - Daiki Kobayashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan
| | - Tokiwa Yamasaki
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryo Yokoyama
- K.K. Sciex Japan, Shinagawa-ku, Tokyo 140-0001, Japan
| | | | - Hiroshi Nishina
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yutaka Yoshida
- Center for Coordination of Research, Institute for Research Promotion, Niigata University, Ikarashi, Niigata 951-2181, Japan
| | - Yukihiko Fujii
- Department of Neurosurgery, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Kosei Takeuchi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan; Department of Medical Cell Biology, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi, Chuo-ku, Niigata 951-8510, Japan; Center for Trans-disciplinary Research, Institute for Research Promotion, Niigata University, Chuo-ku, Niigata 951-8510, Japan.
| |
Collapse
|
28
|
JNK1 controls adult hippocampal neurogenesis and imposes cell-autonomous control of anxiety behaviour from the neurogenic niche. Mol Psychiatry 2018; 23:362-374. [PMID: 27843149 PMCID: PMC5794884 DOI: 10.1038/mp.2016.203] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 10/03/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022]
Abstract
Promoting adult hippocampal neurogenesis is expected to induce neuroplastic changes that improve mood and alleviate anxiety. However, the underlying mechanisms remain largely unknown and the hypothesis itself is controversial. Here we show that mice lacking Jnk1, or c-Jun N-terminal kinase (JNK) inhibitor-treated mice, display increased neurogenesis in adult hippocampus characterized by enhanced cell proliferation and survival, and increased maturation in the ventral region. Correspondingly, anxiety behaviour is reduced in a battery of tests, except when neurogenesis is prevented by AraC treatment. Using engineered retroviruses, we show that exclusive inhibition of JNK in adult-born granule cells alleviates anxiety and reduces depressive-like behaviour. These data validate the neurogenesis hypothesis of anxiety. Moreover, they establish a causal role for JNK in the hippocampal neurogenic niche and anxiety behaviour, and advocate targeting of JNK as an avenue for novel therapies against affective disorders.
Collapse
|
29
|
Ji Y, Teng L, Zhang R, Sun J, Guo Y. NRG-1β exerts neuroprotective effects against ischemia reperfusion-induced injury in rats through the JNK signaling pathway. Neuroscience 2017; 362:13-24. [PMID: 28843994 DOI: 10.1016/j.neuroscience.2017.08.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/15/2017] [Accepted: 08/15/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND Neuregulin-1β (NRG-1β) has great potential to be developed into therapeutics for neuroprotection. The aim of the current study was to analyze the effects and possible signaling pathway of NRG-1β on brain tissues in a rat model of middle cerebral artery occlusion/reperfusion (MCAO/R). METHODS In order to observe the protective effect of NRG-1β on MCAO/R, the neurological deficit and infarct volume were measured using a modified neurological severity score (mNSS) test and by triphenyl tetrazolium chloride (TTC) staining. In order to detect the antagonistic effect of NRG-1β on nerve cells and the blood-brain barrier (BBB), the morphology and structure of cortical brain tissues were observed by Evans Blue (EB) staining, hematoxylin-eosin (H&E) and Nissl staining, in situ cell death detection kit, and transmission electron microscopy (TEM). In order to investigate whether NRG-1β exhibited a significant neuroprotective effect via the JNK signaling pathway, the activity of JNK and the levels of phospho-MKK4, phospho-JNK, pan-JNK and phospho-c-Jun were tested using a JNK activity screening kit, immunofluorescent labeling, and western blot analysis, respectively. RESULTS In the NRG-1β treatment group, accompanied with a decrease in JNK activity, the protein levels of phospho-JNK, phospho-MKK4 and phospho-c-Jun decreased, the ischemia-induced apoptosis decreased, the abnormal morphological structures of nerve cells were ameliorated, the integrity of the BBB was restored, and infarct volume was reduced. At the same time, neurological function was significantly recovered. CONCLUSION NRG-1β exerts a neuroprotective effect through the JNK signaling pathway in MCAO/R rats.
Collapse
Affiliation(s)
- Yaqing Ji
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| | - Lei Teng
- Department of Biology, Qingdao University, Qingdao 266021, Shandong, China.
| | - Rui Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| | - Jinping Sun
- Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| | - Yunliang Guo
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| |
Collapse
|
30
|
Kon E, Cossard A, Jossin Y. Neuronal Polarity in the Embryonic Mammalian Cerebral Cortex. Front Cell Neurosci 2017; 11:163. [PMID: 28670267 PMCID: PMC5472699 DOI: 10.3389/fncel.2017.00163] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/26/2017] [Indexed: 11/13/2022] Open
Abstract
The cerebral cortex is composed of billions of neurons that can grossly be subdivided into two broad classes: inhibitory GABAergic interneurons and excitatory glutamatergic neurons. The majority of cortical neurons in mammals are the excitatory type and they are the main focus of this review article. Like many of the cells in multicellular organisms, fully differentiated neurons are both morphologically and functionally polarized. However, they go through several changes in polarity before reaching this final mature differentiated state. Neurons are derived from polarized neuronal progenitor/stem cells and their commitment to neuronal fate is decided by cellular and molecular asymmetry during their last division in the neurogenic zone. They migrate from their birthplace using so-called multipolar migration, during which they switch direction of movement several times, and repolarize for bipolar migration when the axon is specified. Therefore, neurons have to break their previous symmetry, change their morphology and adequately respond to polarizing signals during migration in order to reach the correct position in the cortex and start making connections. Finally, the dendritic tree is elaborated and the axon/dendrite morphological polarity is set. Here we will describe the function, establishment and maintenance of polarity during the different developmental steps starting from neural stem cell (NSC) division, neuronal migration and axon specification at embryonic developmental stages.
Collapse
Affiliation(s)
- Elif Kon
- Mammalian Development and Cell Biology Unit, Institute of Neuroscience, Université catholique de LouvainBrussels, Belgium
| | - Alexia Cossard
- Mammalian Development and Cell Biology Unit, Institute of Neuroscience, Université catholique de LouvainBrussels, Belgium
| | - Yves Jossin
- Mammalian Development and Cell Biology Unit, Institute of Neuroscience, Université catholique de LouvainBrussels, Belgium
| |
Collapse
|
31
|
Jang Y, Lee MJ, Han J, Kim SJ, Ryu I, Ju X, Ryu MJ, Chung W, Oh E, Kweon GR, Heo JY. A High-fat Diet Induces a Loss of Midbrain Dopaminergic Neuronal Function That Underlies Motor Abnormalities. Exp Neurobiol 2017; 26:104-112. [PMID: 28442947 PMCID: PMC5403908 DOI: 10.5607/en.2017.26.2.104] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Movement defects in obesity are associated with peripheral muscle defects, arthritis, and dysfunction of motor control by the brain. Although movement functionality is negatively correlated with obesity, the brain regions and downstream signaling pathways associated with movement defects in obesity are unclear. A dopaminergic neuronal pathway from the substantia nigra (SN) to the striatum is responsible for regulating grip strength and motor initiation through tyrosine hydroxylase (TH) activity-dependent dopamine release. We found that mice fed a high-fat diet exhibited decreased movement in open-field tests and an increase in missteps in a vertical grid test compared with normally fed mice. This motor abnormality was associated with a significant reduction of TH in the SN and striatum. We further found that phosphorylation of c-Jun N-terminal kinase (JNK), which modulates TH expression in the SN and striatum, was decreased under excess-energy conditions. Our findings suggest that high calorie intake impairs motor function through JNK-dependent dysregulation of TH in the SN and striatum.
Collapse
Affiliation(s)
- Yunseon Jang
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Jeongsu Han
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Soo Jeong Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Ilhwan Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Xianshu Ju
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Woosuk Chung
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Gi Ryang Kweon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.,Brain Research Institute, Chungnam National University School of Medicine, Daejeon 35015, Korea
| |
Collapse
|
32
|
Muñoz-Llancao P, de Gregorio C, Las Heras M, Meinohl C, Noorman K, Boddeke E, Cheng X, Lezoualc'h F, Schmidt M, Gonzalez-Billault C. Microtubule-regulating proteins and cAMP-dependent signaling in neuroblastoma differentiation. Cytoskeleton (Hoboken) 2017; 74:143-158. [PMID: 28164467 DOI: 10.1002/cm.21355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 01/15/2023]
Abstract
Neurons are highly differentiated cells responsible for the conduction and transmission of information in the nervous system. The proper function of a neuron relies on the compartmentalization of their intracellular domains. Differentiated neuroblastoma cells have been extensively used to study and understand the physiology and cell biology of neuronal cells. Here, we show that differentiation of N1E-115 neuroblastoma cells is more pronounced upon exposure of a chemical analog of cyclic AMP (cAMP), db-cAMP. We next analysed the expression of key microtubule-regulating proteins in differentiated cells and the expression and activation of key cAMP players such as EPAC, PKA and AKAP79/150. Most of the microtubule-promoting factors were up regulated during differentiation of N1E-115 cells, while microtubule-destabilizing proteins were down regulated. We observed an increase in tubulin post-translational modifications related to microtubule stability. As expected, db-cAMP increased PKA- and EPAC-dependent signalling. Consistently, pharmacological modulation of EPAC activity instructed cell differentiation, number of neurites, and neurite length in N1E-115 cells. Moreover, disruption of the PKA-AKAP interaction reduced these morphometric parameters. Interestingly, PKA and EPAC act synergistically to induce neuronal differentiation in N1E-115. Altogether these results show that the changes observed in the differentiation of N1E-115 cells proceed by regulating several microtubule-stabilizing factors, and the acquisition of a neuronal phenotype is a process involving concerted although independent functions of EPAC and PKA.
Collapse
Affiliation(s)
- Pablo Muñoz-Llancao
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile.,Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Cristian de Gregorio
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Macarena Las Heras
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Christopher Meinohl
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Kevin Noorman
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Erik Boddeke
- Department of Medical Physiology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, USA
| | - Frank Lezoualc'h
- Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France.,Université de Toulouse III, Paul Sabatier, Toulouse, France
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Christian Gonzalez-Billault
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile.,The Buck Institute for Research on Aging, Novato, USA
| |
Collapse
|
33
|
Vuong TA, Leem YE, Kim BG, Cho H, Lee SJ, Bae GU, Kang JS. A Sonic hedgehog coreceptor, BOC regulates neuronal differentiation and neurite outgrowth via interaction with ABL and JNK activation. Cell Signal 2016; 30:30-40. [PMID: 27871935 DOI: 10.1016/j.cellsig.2016.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022]
Abstract
Neurite outgrowth is a critical step for neurogenesis and remodeling synaptic circuitry during neuronal development and regeneration. An immunoglobulin superfamily member, BOC functions as Sonic hedgehog (Shh) coreceptor in canonical and noncanonical Shh signaling in neuronal development and axon outgrowth/guidance. However signaling mechanisms responsible for BOC action during these processes remain unknown. In our previous studies, a multiprotein complex containing BOC and a closely related protein CDO promotes myogenic differentiation through activation of multiple signaling pathways, including non-receptor tyrosine kinase ABL. Given that ABL and Jun. N-terminal kinase (JNK) are implicated in actin cytoskeletal dynamics required for neurogenesis, we investigated the relationship between BOC, ABL and JNK during neuronal differentiation. Here, we demonstrate that BOC and ABL are induced in P19 embryonal carcinoma (EC) cells and cortical neural progenitor cells (NPCs) during neuronal differentiation. BOC-depleted EC cells or Boc-/- NPCs exhibit impaired neuronal differentiation with shorter neurite formation. BOC interacts with ABL through its putative SH2 binding domain and seems to be phosphorylated in an ABL activity-dependent manner. Unlike wildtype BOC, ABL-binding defective BOC mutants exhibit impaired JNK activation and neuronal differentiation. Finally, Shh treatment enhances JNK activation which is diminished by BOC depletion. These data suggest that BOC interacts with ABL and activates JNK thereby promoting neuronal differentiation and neurite outgrowth.
Collapse
Affiliation(s)
- Tuan Anh Vuong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Bok-Geon Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Sang-Jin Lee
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Gyu-Un Bae
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea.
| |
Collapse
|
34
|
Oksdath M, Guil AFN, Grassi D, Sosa LJ, Quiroga S. The Motor KIF5C Links the Requirements of Stable Microtubules and IGF-1 Receptor Membrane Insertion for Neuronal Polarization. Mol Neurobiol 2016; 54:6085-6096. [DOI: 10.1007/s12035-016-0144-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 09/19/2016] [Indexed: 11/24/2022]
|
35
|
Leucine Zipper-bearing Kinase promotes axon growth in mammalian central nervous system neurons. Sci Rep 2016; 6:31482. [PMID: 27511108 PMCID: PMC4980599 DOI: 10.1038/srep31482] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/04/2016] [Indexed: 11/11/2022] Open
Abstract
Leucine Zipper-bearing Kinase (LZK/MAP3K13) is a member of the mixed lineage kinase family with high sequence identity to Dual Leucine Zipper Kinase (DLK/MAP3K12). While DLK is established as a key regulator of axonal responses to injury, the role of LZK in mammalian neurons is poorly understood. By gain- and loss-of-function analyses in neuronal cultures, we identify LZK as a novel positive regulator of axon growth. LZK signals specifically through MKK4 and JNKs among MAP2Ks and MAPKs respectively in neuronal cells, with JNK activity positively regulating LZK protein levels. Neuronal maturation or activity deprivation activates the LZK-MKK4-JNK pathway. LZK and DLK share commonalities in signaling, regulation, and effects on axon extension. Furthermore, LZK-dependent regulation of DLK protein expression and the lack of additive effects on axon growth upon co-manipulation suggest complex functional interaction and cross-regulation between these two kinases. Together, our data support the possibility for two structurally related MAP3Ks to work in concert to mediate axonal responses to external insult or injury in mammalian CNS neurons.
Collapse
|
36
|
JNK Signaling: Regulation and Functions Based on Complex Protein-Protein Partnerships. Microbiol Mol Biol Rev 2016; 80:793-835. [PMID: 27466283 DOI: 10.1128/mmbr.00043-14] [Citation(s) in RCA: 355] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs), as members of the mitogen-activated protein kinase (MAPK) family, mediate eukaryotic cell responses to a wide range of abiotic and biotic stress insults. JNKs also regulate important physiological processes, including neuronal functions, immunological actions, and embryonic development, via their impact on gene expression, cytoskeletal protein dynamics, and cell death/survival pathways. Although the JNK pathway has been under study for >20 years, its complexity is still perplexing, with multiple protein partners of JNKs underlying the diversity of actions. Here we review the current knowledge of JNK structure and isoforms as well as the partnerships of JNKs with a range of intracellular proteins. Many of these proteins are direct substrates of the JNKs. We analyzed almost 100 of these target proteins in detail within a framework of their classification based on their regulation by JNKs. Examples of these JNK substrates include a diverse assortment of nuclear transcription factors (Jun, ATF2, Myc, Elk1), cytoplasmic proteins involved in cytoskeleton regulation (DCX, Tau, WDR62) or vesicular transport (JIP1, JIP3), cell membrane receptors (BMPR2), and mitochondrial proteins (Mcl1, Bim). In addition, because upstream signaling components impact JNK activity, we critically assessed the involvement of signaling scaffolds and the roles of feedback mechanisms in the JNK pathway. Despite a clarification of many regulatory events in JNK-dependent signaling during the past decade, many other structural and mechanistic insights are just beginning to be revealed. These advances open new opportunities to understand the role of JNK signaling in diverse physiological and pathophysiological states.
Collapse
|
37
|
Saraiva C, Paiva J, Santos T, Ferreira L, Bernardino L. MicroRNA-124 loaded nanoparticles enhance brain repair in Parkinson's disease. J Control Release 2016; 235:291-305. [PMID: 27269730 DOI: 10.1016/j.jconrel.2016.06.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/29/2016] [Accepted: 06/02/2016] [Indexed: 01/17/2023]
Abstract
Modulation of the subventricular zone (SVZ) neurogenic niche can enhance brain repair in several disorders including Parkinson's disease (PD). Herein, we used biocompatible and traceable polymeric nanoparticles (NPs) containing perfluoro-1,5-crown ether (PFCE) and coated with protamine sulfate to complex microRNA-124 (miR-124), a neuronal fate determinant. The ability of NPs to efficiently deliver miR-124 and prompt SVZ neurogenesis and brain repair in PD was evaluated. In vitro, miR-124 NPs were efficiently internalized by neural stem/progenitors cells and neuroblasts and promoted their neuronal commitment and maturation. The expression of Sox9 and Jagged1, two miR-124 targets and stemness-related genes, were also decreased upon miR-124 NP treatment. In vivo, the intracerebral administration of miR-124 NPs increased the number of migrating neuroblasts that reached the granule cell layer of the olfactory bulb, both in healthy and in a 6-hydroxydopamine (6-OHDA) mouse model for PD. MiR-124 NPs were also able to induce migration of neurons into the lesioned striatum of 6-OHDA-treated mice. Most importantly, miR-124 NPs proved to ameliorate motor symptoms of 6-OHDA mice, monitored by the apomorphine-induced rotation test. Altogether, we provide clear evidences to support the use of miR-124 NPs as a new therapeutic approach to boost endogenous brain repair mechanisms in a setting of neurodegeneration.
Collapse
Affiliation(s)
- C Saraiva
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - J Paiva
- CNC-Center for Neuroscience and Cell Biology, 3004-504 Coimbra, Portugal; Biocant - Center of Innovation in Biotechnology, 3060-197 Cantanhede, Portugal
| | - T Santos
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - L Ferreira
- CNC-Center for Neuroscience and Cell Biology, 3004-504 Coimbra, Portugal; Biocant - Center of Innovation in Biotechnology, 3060-197 Cantanhede, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-789 Coimbra, Portugal
| | - L Bernardino
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6201-506 Covilhã, Portugal.
| |
Collapse
|
38
|
Padzik A, Deshpande P, Hollos P, Franker M, Rannikko EH, Cai D, Prus P, Mågård M, Westerlund N, Verhey KJ, James P, Hoogenraad CC, Coffey ET. KIF5C S176 Phosphorylation Regulates Microtubule Binding and Transport Efficiency in Mammalian Neurons. Front Cell Neurosci 2016; 10:57. [PMID: 27013971 PMCID: PMC4791394 DOI: 10.3389/fncel.2016.00057] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/24/2016] [Indexed: 12/15/2022] Open
Abstract
Increased phosphorylation of the KIF5 anterograde motor is associated with impaired axonal transport and neurodegeneration, but paradoxically also with normal transport, though the details are not fully defined. JNK phosphorylates KIF5C on S176 in the motor domain; a site that we show is phosphorylated in brain. Microtubule pelleting assays demonstrate that phosphomimetic KIF5C(1-560)(S176D) associates weakly with microtubules compared to KIF5C(1-560)(WT). Consistent with this, 50% of KIF5C(1-560)(S176D) shows diffuse movement in neurons. However, the remaining 50% remains microtubule bound and displays decreased pausing and increased bidirectional movement. The same directionality switching is observed with KIF5C(1-560)(WT) in the presence of an active JNK chimera, MKK7-JNK. Yet, in cargo trafficking assays where peroxisome cargo is bound, KIF5C(1-560)(S176D)-GFP-FRB transports normally to microtubule plus ends. We also find that JNK increases the ATP hydrolysis of KIF5C in vitro. These data suggest that phosphorylation of KIF5C-S176 primes the motor to either disengage entirely from microtubule tracks as previously observed in response to stress, or to display improved efficiency. The final outcome may depend on cargo load and motor ensembles.
Collapse
Affiliation(s)
- Artur Padzik
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Prasannakumar Deshpande
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Patrik Hollos
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Mariella Franker
- Cell Biology, Faculty of Science, Utrecht University Utrecht, Netherlands
| | - Emmy H Rannikko
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Dawen Cai
- Department of Cell and Developmental Biology, University of Michigan Ann Arbor, MI, USA
| | - Piotr Prus
- Department of Biochemistry, University of Oulu Oulu, Finland
| | - Mats Mågård
- Department of Immunotechnology, Lund University Medicon, Lund, Sweden
| | - Nina Westerlund
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Kristen J Verhey
- Department of Cell and Developmental Biology, University of Michigan Ann Arbor, MI, USA
| | - Peter James
- Department of Immunotechnology, Lund University Medicon, Lund, Sweden
| | | | - Eleanor T Coffey
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| |
Collapse
|
39
|
Vernia S, Morel C, Madara JC, Cavanagh-Kyros J, Barrett T, Chase K, Kennedy NJ, Jung DY, Kim JK, Aronin N, Flavell RA, Lowell BB, Davis RJ. Excitatory transmission onto AgRP neurons is regulated by cJun NH2-terminal kinase 3 in response to metabolic stress. eLife 2016; 5:e10031. [PMID: 26910012 PMCID: PMC4798947 DOI: 10.7554/elife.10031] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 02/22/2016] [Indexed: 11/13/2022] Open
Abstract
The cJun NH2-terminal kinase (JNK) signaling pathway is implicated in the response to metabolic stress. Indeed, it is established that the ubiquitously expressed JNK1 and JNK2 isoforms regulate energy expenditure and insulin resistance. However, the role of the neuron-specific isoform JNK3 is unclear. Here we demonstrate that JNK3 deficiency causes hyperphagia selectively in high fat diet (HFD)-fed mice. JNK3 deficiency in neurons that express the leptin receptor LEPRb was sufficient to cause HFD-dependent hyperphagia. Studies of sub-groups of leptin-responsive neurons demonstrated that JNK3 deficiency in AgRP neurons, but not POMC neurons, was sufficient to cause the hyperphagic response. These effects of JNK3 deficiency were associated with enhanced excitatory signaling by AgRP neurons in HFD-fed mice. JNK3 therefore provides a mechanism that contributes to homeostatic regulation of energy balance in response to metabolic stress. DOI:http://dx.doi.org/10.7554/eLife.10031.001 Consuming the right amount of food is important for health. Eating too little for a long time causes damage to organs, and overeating can cause harm as well, in the form of conditions such as obesity and type 2 diabetes. Several signaling molecules and brain regions are linked to controlling food consumption and ensuring the body receives the correct amount of nutrients to fuel its activities. Previous studies have found that two proteins called JNK1 and JNK2, which are found in most tissues of the body, can reduce how much energy cells use. This can trigger insulin resistance and fat accumulation, and so suggests that blocking the activity of these proteins may help to treat type 2 diabetes and obesity. However, the role of another JNK protein – JNK3, which is mostly found in the brain – was not known. Now, Vernia, Morel et al. have investigated the role of JNK3 in metabolism. It was found that JNK3 reduced the amount of food consumed by mice provided with a cafeteria (high fat) diet. Mice that lacked JNK3 ate far more food and gained more weight on a high fat diet than normal mice. However, JNK3 played no role in food consumption when mice were fed a standard chow diet. Treating normal mice with leptin – an appetite-suppressing hormone – caused them to lose weight, but did not affect mice that lacked JNK3. Examining the brains of the mice revealed that in normal mice, JNK3 in a specific sub-population of neurons decreases the production of proteins that promote eating. However, the proteins continued to be produced in mice that lacked JNK3, encouraging overeating. Overall, the results suggest that blocking the activity of all the JNK proteins will not help treat obesity and diabetes as shutting down JNK3 could encourage overeating. Therefore, future investigation into treatments for these conditions should focus on drugs that specifically target JNK1 and JNK2, and not JNK3. DOI:http://dx.doi.org/10.7554/eLife.10031.002
Collapse
Affiliation(s)
- Santiago Vernia
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Caroline Morel
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Joseph C Madara
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States.,Harvard Medical School, Boston, United States
| | - Julie Cavanagh-Kyros
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, United States
| | - Tamera Barrett
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, United States
| | - Kathryn Chase
- Department of Medicine, Division of Endocrinology, University of Massachusetts Medical School, Worcester, United States
| | - Norman J Kennedy
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Dae Young Jung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Department of Medicine, Division of Endocrinology, University of Massachusetts Medical School, Worcester, United States
| | - Neil Aronin
- Department of Medicine, Division of Endocrinology, University of Massachusetts Medical School, Worcester, United States
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, United States.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, United States
| | - Bradford B Lowell
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States.,Harvard Medical School, Boston, United States
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
40
|
Cai C, Lin J, Sun S, He Y. JNK Inhibition Inhibits Lateral Line Neuromast Hair Cell Development. Front Cell Neurosci 2016; 10:19. [PMID: 26903805 PMCID: PMC4742541 DOI: 10.3389/fncel.2016.00019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 01/18/2016] [Indexed: 12/21/2022] Open
Abstract
JNK signaling is known to play a role in regulating cell behaviors such as cell cycle progression, cell proliferation, and apoptosis, and recent studies have suggested important roles for JNK signaling in embryonic development. However, the precise function of JNK signaling in hair cell development remains poorly studied. In this study, we used the small molecule JNK inhibitor SP600125 to examine the effect of JNK signaling abrogation on the development of hair cells in the zebrafish lateral line neuromast. Our results showed that SP600125 reduced the numbers of both hair cells and supporting cells in neuromasts during larval development in a dose-dependent manner. Additionally, JNK inhibition strongly inhibited the proliferation of neuromast cells, which likely explains the decrease in the number of differentiated hair cells in inhibitor-treated larvae. Furthermore, western blot and in situ analysis showed that JNK inhibition induced cell cycle arrest through induction of p21 expression. We also showed that SP600125 induced cell death in developing neuromasts as measured by cleaved caspase-3 immunohistochemistry, and this was accompanied with an induction of p53 gene expression. Together these results indicate that JNK might be an important regulator in the development of hair cells in the lateral line in zebrafish by controlling both cell cycle progression and apoptosis.
Collapse
Affiliation(s)
- Chengfu Cai
- Department of Otolaryngology, Affiliated Eye and ENT Hospital of Fudan UniversityShanghai, China; Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital, Xiamen UniversityXiamen, Fujian, China
| | - Jinchao Lin
- Department of Otolaryngology-Head and Neck Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University Quanzhou, Fujian, China
| | - Shaoyang Sun
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Medical Sciences, School of Basic Medical Sciences, Fudan University Shanghai, China
| | - Yingzi He
- Department of Otolaryngology, Affiliated Eye and ENT Hospital of Fudan UniversityShanghai, China; Research Center, Affiliated Eye and ENT Hospital of Fudan UniversityShanghai, China; Key Laboratory of Hearing Medicine, Ministry of Health, Affiliated Eye and ENT Hospital of Fudan UniversityShanghai, China
| |
Collapse
|
41
|
Yagi H, Ohkawara B, Nakashima H, Ito K, Tsushima M, Ishii H, Noto K, Ohta K, Masuda A, Imagama S, Ishiguro N, Ohno K. Zonisamide Enhances Neurite Elongation of Primary Motor Neurons and Facilitates Peripheral Nerve Regeneration In Vitro and in a Mouse Model. PLoS One 2015; 10:e0142786. [PMID: 26571146 PMCID: PMC4646494 DOI: 10.1371/journal.pone.0142786] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 10/27/2015] [Indexed: 12/25/2022] Open
Abstract
No clinically applicable drug is currently available to enhance neurite elongation after nerve injury. To identify a clinically applicable drug, we screened pre-approved drugs for neurite elongation in the motor neuron-like NSC34 cells. We found that zonisamide, an anti-epileptic and anti-Parkinson’s disease drug, promoted neurite elongation in cultured primary motor neurons and NSC34 cells in a concentration-dependent manner. The neurite-scratch assay revealed that zonisamide enhanced neurite regeneration. Zonisamide was also protective against oxidative stress-induced cell death of primary motor neurons. Zonisamide induced mRNA expression of nerve growth factors (BDNF, NGF, and neurotrophin-4/5), and their receptors (tropomyosin receptor kinase A and B). In a mouse model of sciatic nerve autograft, intragastric administration of zonisamide for 1 week increased the size of axons distal to the transected site 3.9-fold. Zonisamide also improved the sciatic function index, a marker for motor function of hindlimbs after sciatic nerve autograft, from 6 weeks after surgery. At 8 weeks after surgery, zonisamide was protective against denervation-induced muscle degeneration in tibialis anterior, and increased gene expression of Chrne, Colq, and Rapsn, which are specifically expressed at the neuromuscular junction. We propose that zonisamide is a potential therapeutic agent for peripheral nerve injuries as well as for neuropathies due to other etiologies.
Collapse
Affiliation(s)
- Hideki Yagi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Bisei Ohkawara
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Nakashima
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenyu Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikito Tsushima
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hisao Ishii
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Hand Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kimitoshi Noto
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Hand Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kyotaro Ohta
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akio Masuda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoki Ishiguro
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail:
| |
Collapse
|
42
|
Exchange Protein Directly Activated by cAMP (EPAC) Regulates Neuronal Polarization through Rap1B. J Neurosci 2015; 35:11315-29. [PMID: 26269639 DOI: 10.1523/jneurosci.3645-14.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Acquisition of neuronal polarity is a complex process involving cellular and molecular events. The second messenger cAMP is involved in axonal specification through activation of protein kinase A. However, an alternative cAMP-dependent mechanism involves the exchange protein directly activated by cAMP (EPAC), which also responds to physiological changes in cAMP concentration, promoting activation of the small Rap GTPases. Here, we present evidence that EPAC signaling contributes to axon specification and elongation. In primary rat hippocampal neurons, EPAC isoforms were expressed differentially during axon specification. Furthermore, 8-pCPT, an EPAC pharmacological activator, and genetic manipulations of EPAC in neurons induced supernumerary axons indicative of Rap1b activation. Moreover, 8-pCPT-treated neurons expressed ankyrin G and other markers of mature axons such as synaptophysin and axonal accumulation of vGLUT1. In contrast, pharmacological inhibition of EPAC delayed neuronal polarity. Genetic manipulations to inactivate EPAC1 using either shRNA or neurons derived from EPAC1 knock-out (KO) mice led to axon elongation and polarization defects. Interestingly, multiaxonic neurons generated by 8-pCPT treatments in wild-type neurons were not found in EPAC1 KO mice neurons. Altogether, these results propose that EPAC signaling is an alternative and complementary mechanism for cAMP-dependent axon determination. SIGNIFICANCE STATEMENT This study identifies the guanine exchange factor responsible for Rap1b activation during neuronal polarization and provides an alternate explanation for cAMP-dependent acquisition of neuronal polarity.
Collapse
|
43
|
Namba T, Funahashi Y, Nakamuta S, Xu C, Takano T, Kaibuchi K. Extracellular and Intracellular Signaling for Neuronal Polarity. Physiol Rev 2015; 95:995-1024. [PMID: 26133936 DOI: 10.1152/physrev.00025.2014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neurons are one of the highly polarized cells in the body. One of the fundamental issues in neuroscience is how neurons establish their polarity; therefore, this issue fascinates many scientists. Cultured neurons are useful tools for analyzing the mechanisms of neuronal polarization, and indeed, most of the molecules important in their polarization were identified using culture systems. However, we now know that the process of neuronal polarization in vivo differs in some respects from that in cultured neurons. One of the major differences is their surrounding microenvironment; neurons in vivo can be influenced by extrinsic factors from the microenvironment. Therefore, a major question remains: How are neurons polarized in vivo? Here, we begin by reviewing the process of neuronal polarization in culture conditions and in vivo. We also survey the molecular mechanisms underlying neuronal polarization. Finally, we introduce the theoretical basis of neuronal polarization and the possible involvement of neuronal polarity in disease and traumatic brain injury.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chundi Xu
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
44
|
Sahu SK, Garding A, Tiwari N, Thakurela S, Toedling J, Gebhard S, Ortega F, Schmarowski N, Berninger B, Nitsch R, Schmidt M, Tiwari VK. JNK-dependent gene regulatory circuitry governs mesenchymal fate. EMBO J 2015; 34:2162-81. [PMID: 26157010 PMCID: PMC4557668 DOI: 10.15252/embj.201490693] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 06/05/2015] [Indexed: 12/14/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a biological process in which cells lose cell–cell contacts and become motile. EMT is used during development, for example, in triggering neural crest migration, and in cancer metastasis. Despite progress, the dynamics of JNK signaling, its role in genomewide transcriptional reprogramming, and involved downstream effectors during EMT remain largely unknown. Here, we show that JNK is not required for initiation, but progression of phenotypic changes associated with EMT. Such dependency resulted from JNK-driven transcriptional reprogramming of critical EMT genes and involved changes in their chromatin state. Furthermore, we identified eight novel JNK-induced transcription factors that were required for proper EMT. Three of these factors were also highly expressed in invasive cancer cells where they function in gene regulation to maintain mesenchymal identity. These factors were also induced during neuronal development and function in neuronal migration in vivo. These comprehensive findings uncovered a kinetically distinct role for the JNK pathway in defining the transcriptome that underlies mesenchymal identity and revealed novel transcription factors that mediate these responses during development and disease.
Collapse
Affiliation(s)
| | | | - Neha Tiwari
- Institute of Physiological Chemistry University Medical Center Johannes Gutenberg University, Mainz, Germany
| | | | | | - Susanne Gebhard
- Department of Obstetrics and Gynecology, Johannes Gutenberg University, Mainz, Germany
| | - Felipe Ortega
- Institute of Physiological Chemistry University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Nikolai Schmarowski
- Institute for Microscopic Anatomy and Neurobiology University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Benedikt Berninger
- Institute of Physiological Chemistry University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Robert Nitsch
- Institute for Microscopic Anatomy and Neurobiology University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Marcus Schmidt
- Department of Obstetrics and Gynecology, Johannes Gutenberg University, Mainz, Germany
| | | |
Collapse
|
45
|
Fröhlich D, Kuo WP, Frühbeis C, Sun JJ, Zehendner CM, Luhmann HJ, Pinto S, Toedling J, Trotter J, Krämer-Albers EM. Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0510. [PMID: 25135971 DOI: 10.1098/rstb.2013.0510] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Exosomes are small membranous vesicles of endocytic origin that are released by almost every cell type. They exert versatile functions in intercellular communication important for many physiological and pathological processes. Recently, exosomes attracted interest with regard to their role in cell-cell communication in the nervous system. We have shown that exosomes released from oligodendrocytes upon stimulation with the neurotransmitter glutamate are internalized by neurons and enhance the neuronal stress tolerance. Here, we demonstrate that oligodendroglial exosomes also promote neuronal survival during oxygen-glucose deprivation, a model of cerebral ischaemia. We show the transfer from oligodendrocytes to neurons of superoxide dismutase and catalase, enzymes which are known to help cells to resist oxidative stress. Additionally, we identify various effects of oligodendroglial exosomes on neuronal physiology. Electrophysiological analysis using in vitro multi-electrode arrays revealed an increased firing rate of neurons exposed to oligodendroglial exosomes. Moreover, gene expression analysis and phosphorylation arrays uncovered differentially expressed genes and altered signal transduction pathways in neurons after exosome treatment. Our study thus provides new insight into the broad spectrum of action of oligodendroglial exosomes and their effects on neuronal physiology. The exchange of extracellular vesicles between neural cells may exhibit remarkable potential to impact brain performance.
Collapse
Affiliation(s)
- Dominik Fröhlich
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany
| | - Wen Ping Kuo
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| | - Carsten Frühbeis
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany
| | - Jyh-Jang Sun
- Institute of Physiology, University Medical Center, 55128 Mainz, Germany Neuro-Electronics Research Flanders, 3001 Leuven, Belgium
| | - Christoph M Zehendner
- Institute of Physiology, University Medical Center, 55128 Mainz, Germany ZIM III, Department of Cardiology, Goethe University Frankfurt, 60389 Frankfurt, Germany
| | - Heiko J Luhmann
- Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany Institute of Physiology, University Medical Center, 55128 Mainz, Germany
| | - Sheena Pinto
- Division of Developmental Immunology, DKFZ Heidelberg, 69120 Heidelberg, Germany
| | - Joern Toedling
- Institute of Molecular Biology gGmbH, 55128 Mainz, Germany
| | - Jacqueline Trotter
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| | - Eva-Maria Krämer-Albers
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| |
Collapse
|
46
|
Margevicius DR, Bastian C, Fan Q, Davis RJ, Pimplikar SW. JNK-interacting protein 1 mediates Alzheimer's-like pathological features in AICD-transgenic mice. Neurobiol Aging 2015; 36:2370-9. [PMID: 26022769 DOI: 10.1016/j.neurobiolaging.2015.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 12/20/2022]
Abstract
Amyloid precursor protein, which generates amyloid beta peptides, is intimately associated with Alzheimer's disease (AD) pathogenesis. We previously showed that transgenic mice overexpressing amyloid precursor protein intracellular domain (AICD), a peptide generated simultaneously with amyloid beta, develop AD-like pathologies, including hyperphosphorylated tau, loss of synapses, and memory impairments. AICD is known to bind c-Jun N-terminal kinase (JNK)-interacting protein 1 (JIP1), a scaffold protein that associates with and activates JNK. The aim of this study was to examine the role of JIP1 in AICD-induced AD-like pathologies in vivo, since the JNK pathway is aberrantly activated in AD brains and contributes to AD pathologies. We generated AICD-Tg mice lacking the JIP1 gene (AICD; JIP1(-/-)) and found that although AICD; JIP1(-/-) mice exhibit increased AICD, the absence of JIP1 results in decreased levels of hyperphosphorylated tau and activated JNK. AICD; JIP1(-/-) mice are also protected from synaptic loss and show improved performance in behavioral tests. These results suggest that JIP1 mediates AD-like pathologies in AICD-Tg mice and that JNK signaling may contribute to amyloid-independent mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Daniel R Margevicius
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA.
| | - Chinthasagar Bastian
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School and Howard Hughes Medical Institute, Worcestor, MA, USA
| | - Sanjay W Pimplikar
- Department of Neurosciences, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
47
|
Frias B, Santos J, Morgado M, Sousa MM, Gray SMY, McCloskey KD, Allen S, Cruz F, Cruz CD. The role of brain-derived neurotrophic factor (BDNF) in the development of neurogenic detrusor overactivity (NDO). J Neurosci 2015; 35:2146-60. [PMID: 25653370 PMCID: PMC4315839 DOI: 10.1523/jneurosci.0373-14.2015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 11/21/2014] [Accepted: 11/27/2014] [Indexed: 12/24/2022] Open
Abstract
Neurogenic detrusor overactivity (NDO) is a well known consequence of spinal cord injury (SCI), recognizable after spinal shock, during which the bladder is areflexic. NDO emergence and maintenance depend on profound plastic changes of the spinal neuronal pathways regulating bladder function. It is well known that neurotrophins (NTs) are major regulators of such changes. NGF is the best-studied NT in the bladder and its role in NDO has already been established. Another very abundant neurotrophin is BDNF. Despite being shown that, acting at the spinal cord level, BDNF is a key mediator of bladder dysfunction and pain during cystitis, it is presently unclear if it is also important for NDO. This study aimed to clarify this issue. Results obtained pinpoint BDNF as an important regulator of NDO appearance and maintenance. Spinal BDNF expression increased in a time-dependent manner together with NDO emergence. In chronic SCI rats, BDNF sequestration improved bladder function, indicating that, at later stages, BDNF contributes NDO maintenance. During spinal shock, BDNF sequestration resulted in early development of bladder hyperactivity, accompanied by increased axonal growth of calcitonin gene-related peptide-labeled fibers in the dorsal horn. Chronic BDNF administration inhibited the emergence of NDO, together with reduction of axonal growth, suggesting that BDNF may have a crucial role in bladder function after SCI via inhibition of neuronal sprouting. These findings highlight the role of BDNF in NDO and may provide a significant contribution to create more efficient therapies to manage SCI patients.
Collapse
Affiliation(s)
- Bárbara Frias
- Department of Experimental Biology, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal, Translational NeuroUrology and
| | - João Santos
- Department of Experimental Biology, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal
| | - Marlene Morgado
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal
| | - Mónica Mendes Sousa
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal
| | - Susannah M Y Gray
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT7 1 NN Belfast, United Kingdom
| | - Karen D McCloskey
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, BT7 1 NN Belfast, United Kingdom
| | - Shelley Allen
- Molecular Neurobiology Unit, University of Bristol, School of Clinical Sciences, BS10 5NB Bristol, United Kingdom
| | - Francisco Cruz
- Translational NeuroUrology and Department of Urology, Hospital de S. João, 4200-319 Porto, Portugal, and
| | - Célia Duarte Cruz
- Department of Experimental Biology, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal, Translational NeuroUrology and
| |
Collapse
|
48
|
Oyanagi K, Negishi T, Tashiro T. Action of thyroxine on the survival and neurite maintenance of cerebellar granule neurons in culture. J Neurosci Res 2014; 93:592-603. [PMID: 25447738 DOI: 10.1002/jnr.23519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 09/27/2014] [Accepted: 10/17/2014] [Indexed: 01/28/2023]
Abstract
Developmental hypothyroidism causes severe impairments in the cerebellum. To understand the role of thyroid hormones (THs) in cerebellar development, we examined the effect of three different THs, thyroxine (T4), 3,5,3'-triidothyronine (T3), and 3,3',5'-triiodothyronine (reverse T3; rT3), on the survival and morphology of cerebellar granule neurons (CGNs) in culture and found novel actions specific to T4. Rat CGNs obtained at postnatal day 6 were first cultured for 2 days in serum-containing medium with 25 mM K(+) (K25), then switched to serum-free medium with physiological 5 mM K(+) (K5) or with K25 and cultured for an additional 2 or 4 days. CGNs underwent apoptosis in K5 but survived in K25. Addition of T4 at concentrations of 100-200 nM but not T3 or rT3 rescued CGNs from cell death in K5 in a dose-dependent manner. Furthermore, 200 nM T4 was also effective in maintaining the neurites of CGNs in K5. In K5, T4 suppressed tau phosphorylation at two developmentally regulated sites as well as phosphorylation of c-jun N-terminal kinase (JNK) necessary for its activation and localization to axons. These results suggest that, during cerebellar development, T4 exerts its activity in cell survival and neurite maintenance in a manner distinct from the other two thyroid hormones through regulating the activity and localization of JNK.
Collapse
Affiliation(s)
- Koshi Oyanagi
- Department of Chemistry and Biological Science, School of Science and Engineering, Aoyama Gakuin University, Kanagawa, Japan
| | | | | |
Collapse
|
49
|
Komulainen E, Zdrojewska J, Freemantle E, Mohammad H, Kulesskaya N, Deshpande P, Marchisella F, Mysore R, Hollos P, Michelsen KA, Mågard M, Rauvala H, James P, Coffey ET. JNK1 controls dendritic field size in L2/3 and L5 of the motor cortex, constrains soma size, and influences fine motor coordination. Front Cell Neurosci 2014; 8:272. [PMID: 25309320 PMCID: PMC4162472 DOI: 10.3389/fncel.2014.00272] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/20/2014] [Indexed: 11/23/2022] Open
Abstract
Genetic anomalies on the JNK pathway confer susceptibility to autism spectrum disorders, schizophrenia, and intellectual disability. The mechanism whereby a gain or loss of function in JNK signaling predisposes to these prevalent dendrite disorders, with associated motor dysfunction, remains unclear. Here we find that JNK1 regulates the dendritic field of L2/3 and L5 pyramidal neurons of the mouse motor cortex (M1), the main excitatory pathway controlling voluntary movement. In Jnk1-/- mice, basal dendrite branching of L5 pyramidal neurons is increased in M1, as is cell soma size, whereas in L2/3, dendritic arborization is decreased. We show that JNK1 phosphorylates rat HMW-MAP2 on T1619, T1622, and T1625 (Uniprot P15146) corresponding to mouse T1617, T1620, T1623, to create a binding motif, that is critical for MAP2 interaction with and stabilization of microtubules, and dendrite growth control. Targeted expression in M1 of GFP-HMW-MAP2 that is pseudo-phosphorylated on T1619, T1622, and T1625 increases dendrite complexity in L2/3 indicating that JNK1 phosphorylation of HMW-MAP2 regulates the dendritic field. Consistent with the morphological changes observed in L2/3 and L5, Jnk1-/- mice exhibit deficits in limb placement and motor coordination, while stride length is reduced in older animals. In summary, JNK1 phosphorylates HMW-MAP2 to increase its stabilization of microtubules while at the same time controlling dendritic fields in the main excitatory pathway of M1. Moreover, JNK1 contributes to normal functioning of fine motor coordination. We report for the first time, a quantitative Sholl analysis of dendrite architecture, and of motor behavior in Jnk1-/- mice. Our results illustrate the molecular and behavioral consequences of interrupted JNK1 signaling and provide new ground for mechanistic understanding of those prevalent neuropyschiatric disorders where genetic disruption of the JNK pathway is central.
Collapse
Affiliation(s)
- Emilia Komulainen
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Justyna Zdrojewska
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Erika Freemantle
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Hasan Mohammad
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | | | - Prasannakumar Deshpande
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Francesca Marchisella
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Raghavendra Mysore
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | - Patrik Hollos
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| | | | - Mats Mågard
- Institute for Immune Technology, Medicon Village, University of Lund Lund, Sweden
| | - Heikki Rauvala
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Peter James
- Institute for Immune Technology, Medicon Village, University of Lund Lund, Sweden
| | - Eleanor T Coffey
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| |
Collapse
|
50
|
Soares L, Parisi M, Bonini NM. Axon injury and regeneration in the adult Drosophila. Sci Rep 2014; 4:6199. [PMID: 25160612 PMCID: PMC4145289 DOI: 10.1038/srep06199] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/04/2014] [Indexed: 01/09/2023] Open
Abstract
Neural regeneration is a fascinating process with profound impact on human health, such that defining biological and genetic pathways is of interest. Here we describe an in vivo preparation for neuronal regeneration in the adult Drosophila. The nerve along the anterior margin of the wing is comprised of ~225 neurons that send projections into the central neuropil (thorax). Precise ablation can be induced with a pulsed laser to sever the entire axonal tract. The animal can be recovered, and response to injury assessed over time. Upon ablation, there is local loss of axons near the injury site, scar formation, a rapid impact on the cytoskeleton, and stimulation of hemocytes. By 7d, ~50% of animals show nerve regrowth, with axons from the nerve cells extending down towards the injury or re-routing. Inhibition of JNK signaling promotes regrowth through the injury site, enabling regeneration of the axonal tract.
Collapse
Affiliation(s)
- Lorena Soares
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael Parisi
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|