1
|
Kong L, Yang J, Yang H, Xu B, Yang T, Liu W. Research advances on CaMKs-mediated neurodevelopmental injury. Arch Toxicol 2024; 98:3933-3947. [PMID: 39292234 DOI: 10.1007/s00204-024-03865-5] [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: 07/13/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Calcium/calmodulin-dependent protein kinases (CaMKs) are important proteins in the calcium signaling cascade response pathway, which can broadly regulate biological functions in vivo. Multifunctional CaMKs play key roles in neural development, including neuronal circuit building, synaptic plasticity establishment, and neurotrophic factor secretion. Currently, four familial proteins, calcium/calmodulin-dependent protein kinase I (CaMKI), calcium/calmodulin-dependent protein kinase II (CaMKII), eukaryotic elongation factor 2 kinase (eEF2K, popularly known as CaMKIII) and calcium/calmodulin-dependent protein kinase IV (CaMKIV), are thought to have been the most extensively studied during neurodevelopment. Although their spatial structures are extremely similar, as well as the initial starting point of activation, both require the activation of calcium and calmodulin (CaM) complexes to be involved in the process, and the phosphorylation sites and modes of each member are different. Furthermore, due to the high structural similarity of CaMKs, their members may play synergistic roles in the regulation of neural development, but different CaMKs also have their own means of regulating neural development. In this review, we first describe the visualized protein structural forms of CaMKI, CaMKII, eEF2K and CaMKIV, and then describe the functions of each kinase in neurodevelopment. After that, we focus on four main mechanisms of neurodevelopmental damage caused by CaMKs: CaMKI/ERK/CREB pathway inhibition leading to dendritic spine structural damage; Ca2+/CaM/CaMKII through induction of mitochondrial kinetic disorders leading to neurodevelopmental damage; CaMKIII/eEF2 hyperphosphorylation affects the establishment of synaptic plasticity; and CaMKIV/JNK/NF-κB through induction of an inflammatory response leading to neurodevelopmental damage. In conclusion, we briefly discuss the pathophysiological significance of aberrant CaMK family expression in neurodevelopmental disorders, as well as the protective effects of conventional CaMKII and CaMKIII antagonists against neurodevelopmental injury.
Collapse
Affiliation(s)
- Lingxu Kong
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Jing Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Huajie Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Bin Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Tianyao Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China.
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China.
| | - Wei Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, Shenyang, China.
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
2
|
Zhang C, Xu H, Tang Q, Duan Y, Xia H, Huang H, Ye D, Bi F. CaMKII suppresses proteotoxicity by phosphorylating BAG3 in response to proteasomal dysfunction. EMBO Rep 2024; 25:4488-4514. [PMID: 39261742 PMCID: PMC11466968 DOI: 10.1038/s44319-024-00248-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024] Open
Abstract
Protein quality control serves as the primary defense mechanism for cells against proteotoxicity induced by proteasome dysfunction. While cells can limit the build-up of ubiquitinated misfolded proteins during proteasome inhibition, the precise mechanism is unclear. Here, we find that protein kinase Ca2+/Calmodulin (CaM)-dependent protein kinase II (CaMKII) maintains proteostasis during proteasome inhibition. We show that proteasome inhibition activates CaMKII, which phosphorylates B-cell lymphoma 2 (Bcl-2)-associated athanogene 3 (BAG3) at residues S173, S377, and S386. Phosphorylated BAG3 activates the heme-regulated inhibitor (HRI)- eukaryotic initiation factor-2α (eIF2α) signaling pathway, suppressing protein synthesis and the production of aggregated ubiquitinated misfolded proteins, ultimately mitigating the proteotoxic crisis. Inhibition of CaMKII exacerbates the accumulation of aggregated misfolded proteins and paraptosis induced by proteasome inhibitors. Based on these findings, we validate that combined targeting of proteasome and CaMKII accelerates tumor cell death and enhances the efficacy of proteasome inhibitors in tumor treatment. Our data unveil a new proteasomal inhibition-induced misfolded protein quality control mechanism and propose a novel therapeutic intervention for proteasome inhibitor-mediated tumor treatment.
Collapse
Affiliation(s)
- Chenliang Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huanji Xu
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiulin Tang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yichun Duan
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hongwei Xia
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huixi Huang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Di Ye
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Feng Bi
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
3
|
Elmorsy EM, Al-Ghafari AB, Al Doghaither HA, Fawzy MS, Shehata SA. Neurotoxic mechanisms of dexamethasone in SH-SY5Y neuroblastoma cells: Insights into bioenergetics, oxidative stress, and apoptosis. Steroids 2024; 212:109514. [PMID: 39303897 DOI: 10.1016/j.steroids.2024.109514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Despite the known therapeutic uses of dexamethasone (DEX), the specific mechanisms underlying its neurotoxic effects in neuronal cells, particularly in undifferentiated human neuroblastoma (SH-SY5Y) cells, remain inadequately understood. This study aims to elucidate these mechanisms, emphasizing bioenergetics, oxidative stress, and apoptosis, thereby providing novel insights into the cellular vulnerabilities induced by chronic DEX exposure. The findings revealed significant reductions in cell viability, altered membrane integrity with LDH leakage, decreased intracellular ATP production, and the electron transport chain complexes I and III activity inhibition. DEX significantly increased the release of the reactive species and peroxidation of lipids, as well as of Nrf2 expression. At the same time, it simultaneously led to a decline in the activities of the antioxidant catalase and superoxide dismutase enzymes, along with a depletion of glutathione reserves. The apoptosis process was exhibited by a significant elevation of caspases 3 and 8 activities with overexpression of mRNA BAX, inhibition of BCL-2, and a significant upregulation of the BAX/BCL-2 ratio. Assessment of neuronal development genes (GAP43, CAMK2A, CAMK2B, TUBB3, and Wnts) by quantitative PCR assay showed increased expression of CAMK2A, CAMK2B, and Wnt3a with a significant reduction in GAP43 mRNA levels. Collectively, this study proved that DEX was cytotoxic to SH-SY5Y via bioenergetic disruption, mitochondrial dysfunction, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Ekramy M Elmorsy
- Pathology Department, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Ayat B Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Huda A Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Manal S Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia; Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Shaimaa A Shehata
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
4
|
Rodríguez-Carrillo A, Verheyen VJ, Van Nuijs ALN, Fernández MF, Remy S. Brain-derived neurotrophic factor (BDNF): an effect biomarker of neurodevelopment in human biomonitoring programs. FRONTIERS IN TOXICOLOGY 2024; 5:1319788. [PMID: 38268968 PMCID: PMC10806109 DOI: 10.3389/ftox.2023.1319788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024] Open
Abstract
The present narrative review summarizes recent findings focusing on the role of brain-derived neurotrophic factor (BDNF) as a biomarker of effect for neurodevelopmental alterations during adolescence, based on health effects of exposure to environmental chemical pollutants. To this end, information was gathered from the PubMed database and the results obtained in the European project Human Biomonitoring for Europe (HBM4EU), in which BDNF was measured at two levels of biological organization: total BDNF protein (serum) and BDNF gene DNA methylation (whole blood) levels. The obtained information is organized as follows. First, human biomonitoring, biomarkers of effect and the current state of the art on neurodevelopmental alterations in the population are presented. Second, BDNF secretion and mechanisms of action are briefly explained. Third, previous studies using BDNF as an effect biomarker were consulted in PubMed database and summarized. Finally, the impact of bisphenol A (BPA), metals, and non-persistent pesticide metabolites on BDNF secretion patterns and its mediation role with behavioral outcomes are addressed and discussed. These findings were obtained from three pilot studies conducted in HBM4EU project. Published findings suggested that exposure to some chemical pollutants such as fine particle matter (PM), PFAS, heavy metals, bisphenols, and non-persistent pesticides may alter circulating BDNF levels in healthy population. Therefore, BDNF could be used as a valuable effect biomarker to investigate developmental neurotoxicity of some chemical pollutants.
Collapse
Affiliation(s)
- Andrea Rodríguez-Carrillo
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
- Toxicological Centre, University of Antwerp, Universiteitsplein, Wilrijk, Belgium
| | - Veerle J. Verheyen
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | | | - Mariana F. Fernández
- Biomedical Research Center and School of Medicine, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), University of Granada, Granada, Spain
| | - Sylvie Remy
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| |
Collapse
|
5
|
Ripoli C, Dagliyan O, Renna P, Pastore F, Paciello F, Sollazzo R, Rinaudo M, Battistoni M, Martini S, Tramutola A, Sattin A, Barone E, Saneyoshi T, Fellin T, Hayashi Y, Grassi C. Engineering memory with an extrinsically disordered kinase. SCIENCE ADVANCES 2023; 9:eadh1110. [PMID: 37967196 PMCID: PMC10651130 DOI: 10.1126/sciadv.adh1110] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/13/2023] [Indexed: 11/17/2023]
Abstract
Synaptic plasticity plays a crucial role in memory formation by regulating the communication between neurons. Although actin polymerization has been linked to synaptic plasticity and dendritic spine stability, the causal link between actin polymerization and memory encoding has not been identified yet. It is not clear whether actin polymerization and structural changes in dendritic spines are a driver or a consequence of learning and memory. Using an extrinsically disordered form of the protein kinase LIMK1, which rapidly and precisely acts on ADF/cofilin, a direct modifier of actin, we induced long-term enlargement of dendritic spines and enhancement of synaptic transmission in the hippocampus on command. The activation of extrinsically disordered LIMK1 in vivo improved memory encoding and slowed cognitive decline in aged mice exhibiting reduced cofilin phosphorylation. The engineered memory by an extrinsically disordered LIMK1 supports a direct causal link between actin-mediated synaptic transmission and memory.
Collapse
Affiliation(s)
- Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Onur Dagliyan
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17165 Stockholm, Sweden
| | - Pietro Renna
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Pastore
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Raimondo Sollazzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Martina Battistoni
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Sara Martini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy
| | - Andrea Sattin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy
| | - Takeo Saneyoshi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
6
|
Urrutia PJ, González-Billault C. A Role for Second Messengers in Axodendritic Neuronal Polarity. J Neurosci 2023; 43:2037-2052. [PMID: 36948585 PMCID: PMC10039749 DOI: 10.1523/jneurosci.1065-19.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 03/24/2023] Open
Abstract
Neuronal polarization is a complex molecular process regulated by intrinsic and extrinsic mechanisms. Nerve cells integrate multiple extracellular cues to generate intracellular messengers that ultimately control cell morphology, metabolism, and gene expression. Therefore, second messengers' local concentration and temporal regulation are crucial elements for acquiring a polarized morphology in neurons. This review article summarizes the main findings and current understanding of how Ca2+, IP3, cAMP, cGMP, and hydrogen peroxide control different aspects of neuronal polarization, and highlights questions that still need to be resolved to fully understand the fascinating cellular processes involved in axodendritic polarization.
Collapse
Affiliation(s)
- Pamela J Urrutia
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile 7800003
- School of Medical Technology, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile 7510157
| | - Christian González-Billault
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile 7800003
- Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile 8380453
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile 7800003
- Buck Institute for Research on Aging, Novato, California 94945
| |
Collapse
|
7
|
Simmons P, Trujillo M, McElroy T, Binz R, Pathak R, Allen AR. Evaluating the effects of low-dose simulated galactic cosmic rays on murine hippocampal-dependent cognitive performance. Front Neurosci 2022; 16:908632. [PMID: 36561122 PMCID: PMC9765097 DOI: 10.3389/fnins.2022.908632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/12/2022] [Indexed: 12/12/2022] Open
Abstract
Space exploration has advanced substantially over recent decades and plans to increase the duration of deep space missions are in preparation. One of the primary health concerns is potential damage to the central nervous system (CNS), resulting in loss of cognitive abilities and function. The majority of ground-based research on space radiation-induced health risks has been conducted using single particle simulations, which do not effectively model real-world scenarios. Thus, to improve the safety of space missions, we must expand our understanding of the effects of simulated galactic cosmic rays (GCRs) on the CNS. To assess the effects of low-dose GCR, we subjected 6-month-old male BALB/c mice to 50 cGy 5-beam simplified GCR spectrum (1H, 28Si, 4He, 16O, and 56Fe) whole-body irradiation at the NASA Space Radiation Laboratory. Animals were tested for cognitive performance with Y-maze and Morris water maze tests 3 months after irradiation. Irradiated animals had impaired short-term memory and lacked spatial memory retention on day 5 of the probe trial. Glial cell analysis by flow cytometry showed no significant changes in oligodendrocytes, astrocytes, microglia or neural precursor cells (NPC's) between the sham group and GCR group. Bone marrow cytogenetic data showed a significant increase in the frequency of chromosomal aberrations after GCR exposure. Finally, tandem mass tag proteomics identified 3,639 proteins, 113 of which were differentially expressed when comparing sham versus GCR exposure (fold change > 1.5; p < 0.05). Our data suggest exposure to low-dose GCR induces cognitive deficits by impairing short-term memory and spatial memory retention.
Collapse
Affiliation(s)
- Pilar Simmons
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Madison Trujillo
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Taylor McElroy
- Department of Aging, University of Florida, Gainesville, FL, United States
| | - Regina Binz
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Rupak Pathak
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States,*Correspondence: Antiño R. Allen,
| |
Collapse
|
8
|
Rodríguez-Carrillo A, D'Cruz SC, Mustieles V, Suárez B, Smagulova F, David A, Peinado F, Artacho-Cordón F, López LC, Arrebola JP, Olea N, Fernández MF, Freire C. Exposure to non-persistent pesticides, BDNF, and behavioral function in adolescent males: Exploring a novel effect biomarker approach. ENVIRONMENTAL RESEARCH 2022; 211:113115. [PMID: 35292247 DOI: 10.1016/j.envres.2022.113115] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 05/22/2023]
Abstract
BACKGROUND Numerous contemporary non-persistent pesticides may elicit neurodevelopmental impairments. Brain-derived neurotrophic factor (BDNF) has been proposed as a novel effect biomarker of neurological function that could help to understand the biological responses of some environmental exposures. OBJECTIVES To investigate the relationship between exposure to various non-persistent pesticides, BDNF, and behavioral functioning among adolescents. METHODS The concentrations of organophosphate (OP) insecticide metabolites 3,5,6-trichloro-2-pyridinol (TCPy), 2-isopropyl-4-methyl-6-hydroxypyrimidine (IMPy), malathion diacid (MDA), and diethyl thiophosphate (DETP); metabolites of pyrethroids 3-phenoxybenzoic acid (3-PBA) and dimethylcyclopropane carboxylic acid (DCCA), the metabolite of insecticide carbaryl 1-naphthol (1-N), and the metabolite of ethylene-bis-dithiocarbamate fungicides ethylene thiourea (ETU) were measured in spot urine samples, as well as serum BDNF protein levels and blood DNA methylation of Exon IV of BDNF gene in 15-17-year-old boys from the INMA-Granada cohort in Spain. Adolescents' behavior was reported by parents using the Child Behavior Check List (CBCL/6-18). This study included 140 adolescents of whom 118 had data on BDNF gene DNA methylation. Multivariable linear regression, weighted quantile sum (WQS) for mixture effects, and mediation models were fit. RESULTS IMPy, MDA, DCCA, and ETU were detected in more than 70% of urine samples, DETP in 53%, and TCPy, 3-PBA, and 1-N in less than 50% of samples. Higher levels of IMPy, TCPy, and ETU were significantly associated with more behavioral problems as social, thought problems, and rule-breaking symptoms. IMPy, MDA, DETP, and 1-N were significantly associated with decreased serum BDNF levels, while MDA, 3-PBA, and ETU were associated with higher DNA methylation percentages at several CpGs. WQS models suggest a mixture effect on more behavioral problems and BDNF DNA methylation at several CpGs. A mediated effect of serum BDNF within IMPy-thought and IMPy-rule breaking associations was suggested. CONCLUSION BDNF biomarkers measured at different levels of biological complexity provided novel information regarding the potential disruption of behavioral function due to contemporary pesticides, highlighting exposure to diazinon (IMPy) and the combined effect of IMPy, MDA, DCCA, and ETU. However, further research is warranted.
Collapse
Affiliation(s)
- Andrea Rodríguez-Carrillo
- University of Granada, Biomedical Research Center (CIBM), Department of Radiology, 18016, Granada, Spain
| | - Shereen C D'Cruz
- Univ Rennes, EHESP, INSERM, IRSET (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Vicente Mustieles
- University of Granada, Biomedical Research Center (CIBM), Department of Radiology, 18016, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Spain
| | - Beatriz Suárez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain
| | - Fátima Smagulova
- Univ Rennes, EHESP, INSERM, IRSET (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Arthur David
- Univ Rennes, EHESP, INSERM, IRSET (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Francisco Peinado
- University of Granada, Biomedical Research Center (CIBM), Department of Radiology, 18016, Granada, Spain
| | - Francisco Artacho-Cordón
- University of Granada, Biomedical Research Center (CIBM), Department of Radiology, 18016, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Spain
| | - Luis C López
- University of Granada, Department of Physiology, 18016, Granada, Spain
| | - Juan P Arrebola
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Spain; University of Granada, Department of Preventive Medicine and Public Health, 18016, Granada, Spain
| | - Nicolás Olea
- University of Granada, Biomedical Research Center (CIBM), Department of Radiology, 18016, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Spain
| | - Mariana F Fernández
- University of Granada, Biomedical Research Center (CIBM), Department of Radiology, 18016, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Spain.
| | - Carmen Freire
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012, Granada, Spain; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Spain
| |
Collapse
|
9
|
Fading memories in aging and neurodegeneration: Is p75 neurotrophin receptor a culprit? Ageing Res Rev 2022; 75:101567. [PMID: 35051645 DOI: 10.1016/j.arr.2022.101567] [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: 09/14/2021] [Revised: 12/12/2021] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
Aging and age-related neurodegenerative diseases have become one of the major concerns in modern times as cognitive abilities tend to decline when we get older. It is well known that the main cause of this age-related cognitive deficit is due to aberrant changes in cellular, molecular circuitry and signaling pathways underlying synaptic plasticity and neuronal connections. The p75 neurotrophin receptor (p75NTR) is one of the important mediators regulating the fate of the neurons in the nervous system. Its importance in neuronal apoptosis is well documented. However, the mechanisms involving the regulation of p75NTR in synaptic plasticity and cognitive function remain obscure, although cognitive impairment has been associated with a higher expression of p75NTR in neurons. In this review, we discuss the current understanding of how neurons are influenced by p75NTR function to maintain normal neuronal synaptic strength and connectivity, particularly to support learning and memory in the hippocampus. We then discuss the age-associated alterations in neurophysiological mechanisms of synaptic plasticity and cognitive function. Furthermore, we also describe current evidence that has begun to elucidate how p75NTR regulates synaptic changes in aging and age-related neurodegenerative diseases, focusing on the hippocampus. Elucidating the role that p75NTR signaling plays in regulating synaptic plasticity will contribute to a better understanding of cognitive processes and pathological conditions. This will in turn provide novel approaches to improve therapies for the treatment of neurological diseases in which p75NTR dysfunction has been demonstrated.
Collapse
|
10
|
Nawalpuri B, Sharma A, Chattarji S, Muddashetty RS. Distinct temporal expression of the GW182 paralog TNRC6A in neurons regulates dendritic arborization. J Cell Sci 2021; 134:271120. [PMID: 34328181 DOI: 10.1242/jcs.258465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/19/2021] [Indexed: 01/11/2023] Open
Abstract
Precise development of the dendritic architecture is a critical determinant of mature neuronal circuitry. MicroRNA (miRNA)-mediated regulation of protein synthesis plays a crucial role in dendritic morphogenesis, but the role of miRNA-induced silencing complex (miRISC) protein components in this process is less studied. Here, we show an important role of a key miRISC protein, the GW182 paralog TNRC6A, in the regulation of dendritic growth. We identified a distinct brain region-specific spatiotemporal expression pattern of GW182 during rat postnatal development. We found that the window of peak GW182 expression coincides with the period of extensive dendritic growth, both in the hippocampus and cerebellum. Perturbation of GW182 function during a specific temporal window resulted in reduced dendritic growth of cultured hippocampal neurons. Mechanistically, we show that GW182 modulates dendritic growth by regulating global somatodendritic translation and actin cytoskeletal dynamics of developing neurons. Furthermore, we found that GW182 affects dendritic architecture by regulating the expression of actin modulator LIMK1. Taken together, our data reveal a previously undescribed neurodevelopmental expression pattern of GW182 and its role in dendritic morphogenesis, which involves both translational control and actin cytoskeletal rearrangement. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Bharti Nawalpuri
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore 560065, India.,School of Chemical and Biotechnology, Shanmugha Arts, Science, and Technology and Research Academy (SASTRA) University, Thanjavur 613401, India.,Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | - Arpita Sharma
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore 560065, India
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Bangalore 560065, India.,Simons Initiative for the Developing Brain and Centre for Discovery Brain Sciences, University of Edinburgh EH8 9XD, Edinburgh, UK
| | - Ravi S Muddashetty
- Centre for Brain Development and Repair, Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore 560065, India.,Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
11
|
Kaltezioti V, Foskolou IP, Lavigne MD, Ninou E, Tsampoula M, Fousteri M, Margarity M, Politis PK. Prox1 inhibits neurite outgrowth during central nervous system development. Cell Mol Life Sci 2021; 78:3443-3465. [PMID: 33247761 PMCID: PMC11072475 DOI: 10.1007/s00018-020-03709-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022]
Abstract
During central nervous system (CNS) development, proper and timely induction of neurite elongation is critical for generating functional, mature neurons, and neuronal networks. Despite the wealth of information on the action of extracellular cues, little is known about the intrinsic gene regulatory factors that control this developmental decision. Here, we report the identification of Prox1, a homeobox transcription factor, as a key player in inhibiting neurite elongation. Although Prox1 promotes acquisition of early neuronal identity and is expressed in nascent post-mitotic neurons, it is heavily down-regulated in the majority of terminally differentiated neurons, indicating a regulatory role in delaying neurite outgrowth in newly formed neurons. Consistently, we show that Prox1 is sufficient to inhibit neurite extension in mouse and human neuroblastoma cell lines. More importantly, Prox1 overexpression suppresses neurite elongation in primary neuronal cultures as well as in the developing mouse brain, while Prox1 knock-down promotes neurite outgrowth. Mechanistically, RNA-Seq analysis reveals that Prox1 affects critical pathways for neuronal maturation and neurite extension. Interestingly, Prox1 strongly inhibits many components of Ca2+ signaling pathway, an important mediator of neurite extension and neuronal maturation. In accordance, Prox1 represses Ca2+ entry upon KCl-mediated depolarization and reduces CREB phosphorylation. These observations suggest that Prox1 acts as a potent suppressor of neurite outgrowth by inhibiting Ca2+ signaling pathway. This action may provide the appropriate time window for nascent neurons to find the correct position in the CNS prior to initiation of neurites and axon elongation.
Collapse
Affiliation(s)
- Valeria Kaltezioti
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Iosifina P Foskolou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matthieu D Lavigne
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Elpinickie Ninou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Maria Fousteri
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Marigoula Margarity
- Laboratory of Human and Animal Physiology, Department of Biology, School of Natural Sciences, University of Patras, 26500, Rio Achaias, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece.
| |
Collapse
|
12
|
Martínez MA, Rodríguez JL, Lopez-Torres B, Martínez M, Martínez-Larrañaga MR, Maximiliano JE, Anadón A, Ares I. Use of human neuroblastoma SH-SY5Y cells to evaluate glyphosate-induced effects on oxidative stress, neuronal development and cell death signaling pathways. ENVIRONMENT INTERNATIONAL 2020; 135:105414. [PMID: 31874349 DOI: 10.1016/j.envint.2019.105414] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 06/10/2023]
Abstract
Glyphosate-containing herbicides are the most used agrochemicals in the world. Their indiscriminate application raises some concerns regarding the possible health and environmental hazards. In this study, we investigated in human neuroblastoma cell line SH-SY5Y if oxidative stress, altered neurodevelopment and cell death pathways are involved in response to glyphosate and its metabolite aminomethylphosphonic acid (AMPA) exposures. MTT and LDH assays were carried out to assess the glyphosate and AMPA cytotoxicity. Lipid peroxides measured as malondialdehyde (MDA), nitric oxide (NO) and reactive oxygen species (ROS) production, and caspase-Glo 3/7 activity were evaluated. The neuroprotective role of melatonin (MEL), Trolox, N-acetylcysteine (NAC) and Sylibin against glyphosate- and AMPA-induced oxidative stress was examined. Glyphosate and AMPA effects on neuronal development related gene transcriptions, and gene expression profiling of cell death pathways by Real-Time PCR array were also investigated. Glyphosate (5 mM) and AMPA (10 mM) induced a significant increase in MDA levels, NO and ROS production and caspase 3/7 activity. Glyphosate exposure induced up-regulation of Wnt3a, Wnt5a, Wnt7a, CAMK2A, CAMK2B and down-regulation of GAP43 and TUBB3 mRNA expression involved in normal neural cell development. In relation to gene expression profiling of cell death pathways, of the 84 genes examined in cells a greater than 2-fold change was observed for APAF1, BAX, BCL2, CASP3, CASP7, CASP9, SYCP2, TNF, TP53, CTSB, NFκB1, PIK3C3, SNCA, SQSTMT, HSPBAP1 and KCNIPI mRNA expression for glyphosate and AMPA exposures. These gene expression data can help to define neurotoxic mechanisms of glyphosate and AMPA. Our results demonstrated that glyphosate and AMPA induced cytotoxic effects on neuronal development, oxidative stress and cell death via apoptotic, autophagy and necrotic pathways and confirmed that glyphosate environmental exposure becomes a concern. This study demonstrates that SH-SY5Y cell line could be considered an in vitro system for pesticide screening.
Collapse
Affiliation(s)
- María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José-Luis Rodríguez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Marta Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Jorge-Enrique Maximiliano
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
13
|
Wong LW, Tann JY, Ibanez CF, Sajikumar S. The p75 Neurotrophin Receptor Is an Essential Mediator of Impairments in Hippocampal-Dependent Associative Plasticity and Memory Induced by Sleep Deprivation. J Neurosci 2019; 39:5452-5465. [PMID: 31085607 PMCID: PMC6616296 DOI: 10.1523/jneurosci.2876-18.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/22/2023] Open
Abstract
Sleep deprivation (SD) interferes with hippocampal structural and functional plasticity, formation of long-term memory and cognitive function. The molecular mechanisms underlying these effects are incompletely understood. Here, we show that SD impaired synaptic tagging and capture and behavioral tagging, two major mechanisms of associative learning and memory. Strikingly, mutant male mice lacking the p75 neurotrophin receptor (p75NTR) were resistant to the detrimental effects of SD on hippocampal plasticity at both cellular and behavioral levels. Mechanistically, SD increased p75NTR expression and its interaction with phosphodiesterase. p75NTR deletion preserved hippocampal structural and functional plasticity by preventing SD-mediated effects on hippocampal cAMP-CREB-BDNF, cAMP-PKA-LIMK1-cofilin, and RhoA-ROCK2 pathways. Our study identifies p75NTR as an important mediator of hippocampal structural and functional changes associated with SD, and suggests that targeting p75NTR could be a promising strategy to limit the memory and cognitive deficits that accompany sleep loss.SIGNIFICANCE STATEMENT The lack of sufficient sleep is a major health concern in today's world. Sleep deprivation (SD) affects cognitive functions such as memory. We have investigated how associative memory mechanisms, synaptic tagging and capture (STC), was impaired in SD mice at cellular and behavioral level. Interestingly, mutant male mice that lacked the p75 neurotrophin receptor (p75NTR) were seen to be resistant to the SD-induced impairments in hippocampal synaptic plasticity and STC. Additionally, we elucidated the molecular pathways responsible for this rescue of plasticity in the mutant mice. Our study has thus identified p75NTR as a promising target to limit the cognitive deficits associated with SD.
Collapse
Affiliation(s)
- Lik-Wei Wong
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| | - Jason Y Tann
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| | - Carlos F Ibanez
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm S-17177, Sweden
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore,
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| |
Collapse
|
14
|
Nicole O, Bell DM, Leste-Lasserre T, Doat H, Guillemot F, Pacary E. A novel role for CAMKIIβ in the regulation of cortical neuron migration: implications for neurodevelopmental disorders. Mol Psychiatry 2018; 23:2209-2226. [PMID: 29712998 PMCID: PMC6129389 DOI: 10.1038/s41380-018-0046-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/11/2018] [Accepted: 02/28/2018] [Indexed: 12/19/2022]
Abstract
Perturbation of CaMKIIβ expression has been associated with multiple neuropsychiatric diseases, highlighting CaMKIIβ as a gene of interest. Yet, in contrast to CaMKIIα, the specific functions of CaMKIIβ in the brain remain poorly explored. Here, we reveal a novel function for this CaMKII isoform in vivo during neuronal development. By using in utero electroporation, we show that CaMKIIβ is an important regulator of radial migration of projection neurons during cerebral cortex development. Knockdown of CaMKIIβ causes accelerated migration of nascent pyramidal neurons, whereas overexpression of CaMKIIβ inhibits migration, demonstrating that precise regulation of CaMKIIβ expression is required for correct neuronal migration. More precisely, CaMKIIβ controls the multipolar-bipolar transition in the intermediate zone and locomotion in the cortical plate through its actin-binding and -bundling activities. In addition, our data indicate that a fine-tuned balance between CaMKIIβ and cofilin activities is necessary to ensure proper migration of cortical neurons. Thus, our findings define a novel isoform-specific function for CaMKIIβ, demonstrating that CaMKIIβ has a major biological function in the developing brain.
Collapse
Affiliation(s)
- Olivier Nicole
- CNRS, UMR5293, Institut des Maladies Neurodégénératives, F-33000 Bordeaux, France,Université de Bordeaux, F-33000 Bordeaux, France
| | - Donald M. Bell
- Confocal and Image Analysis Facility, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | | - Hélène Doat
- Transcriptome Facility, INSERM U1215, Neurocentre Magendie, F-33000 Bordeaux, France
| | | | - Emilie Pacary
- Université de Bordeaux, F-33000, Bordeaux, France. .,INSERM U1215, Neurocentre Magendie, F-33000, Bordeaux, France.
| |
Collapse
|
15
|
Nguyen HTN, Kato H, Masuda K, Yamaza H, Hirofuji Y, Sato H, Pham TTM, Takayama F, Sakai Y, Ohga S, Taguchi T, Nonaka K. Impaired neurite development associated with mitochondrial dysfunction in dopaminergic neurons differentiated from exfoliated deciduous tooth-derived pulp stem cells of children with autism spectrum disorder. Biochem Biophys Rep 2018; 16:24-31. [PMID: 30258988 PMCID: PMC6153399 DOI: 10.1016/j.bbrep.2018.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 08/15/2018] [Accepted: 09/14/2018] [Indexed: 01/08/2023] Open
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder characterized by impaired social interactions, restrictive interests, and repetitive stereotypic behaviors. Among the various mechanisms underlying the pathogenesis of ASD, dysfunctions of dopaminergic signaling and mitochondria have been hypothesized to explain the core symptoms of children with ASD. However, only a few studies focusing on the pathological association between dopaminergic neurons (DN) and mitochondria in ASD have been performed using patient-derived stem cells and in vitro differentiated neurons. Stem cells from human exfoliated deciduous teeth (SHED) are neural crest-derived mesenchymal stem cells present in the dental pulp of exfoliated deciduous teeth; these cells can differentiate into dopaminergic neurons (DN) in vitro. This study aimed to investigate the pathological association between development of DN and mitochondria in ASD by using SHED as a disease- or patient-specific cellular model. The SHED obtained from three children with ASD and three typically developing children were differentiated into DN, and the neurobiology of these cells was examined. The DN derived from children with ASD showed impaired neurite outgrowth and branching, associated with decreased mitochondrial membrane potential, ATP production, number of mitochondria within the neurites, amount of mitochondria per cell area and intracellular calcium level. In addition, impaired neurite outgrowth and branching of ASD-derived DN were not improved by brain-derived neurotrophic factor (BDNF), suggesting impairment of the BDNF signaling pathway in ASD. These results imply that intracerebral dopamine production may have decreased in these children. The earliest age at which deciduous teeth spontaneously exfoliate in humans, and SHED can be noninvasively collected, is approximately 6 years. Our results suggest that in vitro analysis of SHED-derived DN obtained from children with ASD provides neurobiological information that may be useful in determining treatment strategies in the early stages of ASD. Dental pulp stem cells of autistic patient differentiate into dopaminergic neurons. These neurons show impaired neurite development compared with those from controls. This impairment is associated with mitochondrial dysfunction. Dental pulp stem cells may help establish treatment strategies against autism.
Collapse
Affiliation(s)
- Huong Thi Nguyen Nguyen
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Hiroki Kato
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
- Corresponding authors.
| | - Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
- Corresponding authors.
| | - Haruyoshi Yamaza
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Yuta Hirofuji
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Hiroshi Sato
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Thanh Thi Mai Pham
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Fumiko Takayama
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Kazuaki Nonaka
- Section of Oral Medicine for Children, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka 812-8582, Japan
| |
Collapse
|
16
|
Vogt J, Kirischuk S, Unichenko P, Schlüter L, Pelosi A, Endle H, Yang JW, Schmarowski N, Cheng J, Thalman C, Strauss U, Prokudin A, Bharati BS, Aoki J, Chun J, Lutz B, Luhmann HJ, Nitsch R. Synaptic Phospholipid Signaling Modulates Axon Outgrowth via Glutamate-dependent Ca2+-mediated Molecular Pathways. Cereb Cortex 2018; 27:131-145. [PMID: 27909001 PMCID: PMC5939201 DOI: 10.1093/cercor/bhw370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Indexed: 12/28/2022] Open
Abstract
Altered synaptic bioactive lipid signaling has been recently shown to augment neuronal excitation in the hippocampus of adult animals by activation of presynaptic LPA2-receptors leading to increased presynaptic glutamate release. Here, we show that this results in higher postsynaptic Ca2+ levels and in premature onset of spontaneous neuronal activity in the developing entorhinal cortex. Interestingly, increased synchronized neuronal activity led to reduced axon growth velocity of entorhinal neurons which project via the perforant path to the hippocampus. This was due to Ca2+-dependent molecular signaling to the axon affecting stabilization of the actin cytoskeleton. The spontaneous activity affected the entire entorhinal cortical network and thus led to reduced overall axon fiber numbers in the mature perforant path that is known to be important for specific memory functions. Our data show that precise regulation of early cortical activity by bioactive lipids is of critical importance for proper circuit formation.
Collapse
Affiliation(s)
- Johannes Vogt
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Sergei Kirischuk
- Institute of Physiology, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Petr Unichenko
- Institute of Physiology, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Leslie Schlüter
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Assunta Pelosi
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Heiko Endle
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Jenq-Wei Yang
- Institute of Physiology, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Nikolai Schmarowski
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Jin Cheng
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Carine Thalman
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Ulf Strauss
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin, 10119 Berlin, Germany
| | - Alexey Prokudin
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - B Suman Bharati
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Jerold Chun
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Robert Nitsch
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center, Johannes Gutenberg-University, 55131 Mainz, Germany
| |
Collapse
|
17
|
Woolfrey KM, O'Leary H, Goodell DJ, Robertson HR, Horne EA, Coultrap SJ, Dell'Acqua ML, Bayer KU. CaMKII regulates the depalmitoylation and synaptic removal of the scaffold protein AKAP79/150 to mediate structural long-term depression. J Biol Chem 2017; 293:1551-1567. [PMID: 29196604 DOI: 10.1074/jbc.m117.813808] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/21/2017] [Indexed: 11/06/2022] Open
Abstract
Both long-term potentiation (LTP) and depression (LTD) of excitatory synapse strength require the Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) and its autonomous activity generated by Thr-286 autophosphorylation. Additionally, LTP and LTD are correlated with dendritic spine enlargement and shrinkage that are accompanied by the synaptic accumulation or removal, respectively, of the AMPA-receptor regulatory scaffold protein A-kinase anchoring protein (AKAP) 79/150. We show here that the spine shrinkage associated with LTD indeed requires synaptic AKAP79/150 removal, which in turn requires CaMKII activity. In contrast to normal CaMKII substrates, the substrate sites within the AKAP79/150 N-terminal polybasic membrane-cytoskeletal targeting domain were phosphorylated more efficiently by autonomous compared with Ca2+/CaM-stimulated CaMKII activity. This unusual regulation was mediated by Ca2+/CaM binding to the substrate sites resulting in protection from phosphorylation in the presence of Ca2+/CaM, a mechanism that favors phosphorylation by prolonged, weak LTD stimuli versus brief, strong LTP stimuli. Phosphorylation by CaMKII inhibited AKAP79/150 association with F-actin; it also facilitated AKAP79/150 removal from spines but was not required for it. By contrast, LTD-induced spine removal of AKAP79/150 required its depalmitoylation on two Cys residues within the N-terminal targeting domain. Notably, such LTD-induced depalmitoylation was also blocked by CaMKII inhibition. These results provide a mechanism how CaMKII can indeed mediate not only LTP but also LTD through regulated substrate selection; however, in the case of AKAP79/150, indirect CaMKII effects on palmitoylation are more important than the effects of direct phosphorylation. Additionally, our results provide the first direct evidence for a function of the well-described AKAP79/150 trafficking in regulating LTD-induced spine shrinkage.
Collapse
Affiliation(s)
- Kevin M Woolfrey
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Heather O'Leary
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Dayton J Goodell
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Holly R Robertson
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Eric A Horne
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Steven J Coultrap
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Mark L Dell'Acqua
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - K Ulrich Bayer
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| |
Collapse
|
18
|
Dombert B, Balk S, Lüningschrör P, Moradi M, Sivadasan R, Saal-Bauernschubert L, Jablonka S. BDNF/trkB Induction of Calcium Transients through Ca v2.2 Calcium Channels in Motoneurons Corresponds to F-actin Assembly and Growth Cone Formation on β2-Chain Laminin (221). Front Mol Neurosci 2017; 10:346. [PMID: 29163025 PMCID: PMC5670157 DOI: 10.3389/fnmol.2017.00346] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/12/2017] [Indexed: 12/11/2022] Open
Abstract
Spontaneous Ca2+ transients and actin dynamics in primary motoneurons correspond to cellular differentiation such as axon elongation and growth cone formation. Brain-derived neurotrophic factor (BDNF) and its receptor trkB support both motoneuron survival and synaptic differentiation. However, in motoneurons effects of BDNF/trkB signaling on spontaneous Ca2+ influx and actin dynamics at axonal growth cones are not fully unraveled. In our study we addressed the question how neurotrophic factor signaling corresponds to cell autonomous excitability and growth cone formation. Primary motoneurons from mouse embryos were cultured on the synapse specific, β2-chain containing laminin isoform (221) regulating axon elongation through spontaneous Ca2+ transients that are in turn induced by enhanced clustering of N-type specific voltage-gated Ca2+ channels (Cav2.2) in axonal growth cones. TrkB-deficient (trkBTK-/-) mouse motoneurons which express no full-length trkB receptor and wildtype motoneurons cultured without BDNF exhibited reduced spontaneous Ca2+ transients that corresponded to altered axon elongation and defects in growth cone morphology which was accompanied by changes in the local actin cytoskeleton. Vice versa, the acute application of BDNF resulted in the induction of spontaneous Ca2+ transients and Cav2.2 clustering in motor growth cones, as well as the activation of trkB downstream signaling cascades which promoted the stabilization of β-actin via the LIM kinase pathway and phosphorylation of profilin at Tyr129. Finally, we identified a mutual regulation of neuronal excitability and actin dynamics in axonal growth cones of embryonic motoneurons cultured on laminin-221/211. Impaired excitability resulted in dysregulated axon extension and local actin cytoskeleton, whereas upon β-actin knockdown Cav2.2 clustering was affected. We conclude from our data that in embryonic motoneurons BDNF/trkB signaling contributes to axon elongation and growth cone formation through changes in the local actin cytoskeleton accompanied by increased Cav2.2 clustering and local calcium transients. These findings may help to explore cellular mechanisms which might be dysregulated during maturation of embryonic motoneurons leading to motoneuron disease.
Collapse
Affiliation(s)
- Benjamin Dombert
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Stefanie Balk
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Mehri Moradi
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Rajeeve Sivadasan
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Sibylle Jablonka
- Institute of Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
19
|
The functions of Reelin in membrane trafficking and cytoskeletal dynamics: implications for neuronal migration, polarization and differentiation. Biochem J 2017; 474:3137-3165. [PMID: 28887403 DOI: 10.1042/bcj20160628] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023]
Abstract
Reelin is a large extracellular matrix protein with relevant roles in mammalian central nervous system including neurogenesis, neuronal polarization and migration during development; and synaptic plasticity with its implications in learning and memory, in the adult. Dysfunctions in reelin signaling are associated with brain lamination defects such as lissencephaly, but also with neuropsychiatric diseases like autism, schizophrenia and depression as well with neurodegeneration. Reelin signaling involves a core pathway that activates upon reelin binding to its receptors, particularly ApoER2 (apolipoprotein E receptor 2)/LRP8 (low-density lipoprotein receptor-related protein 8) and very low-density lipoprotein receptor, followed by Src/Fyn-mediated phosphorylation of the adaptor protein Dab1 (Disabled-1). Phosphorylated Dab1 (pDab1) is a hub in the signaling cascade, from which several other downstream pathways diverge reflecting the different roles of reelin. Many of these pathways affect the dynamics of the actin and microtubular cytoskeleton, as well as membrane trafficking through the regulation of the activity of small GTPases, including the Rho and Rap families and molecules involved in cell polarity. The complexity of reelin functions is reflected by the fact that, even now, the precise mode of action of this signaling cascade in vivo at the cellular and molecular levels remains unclear. This review addresses and discusses in detail the participation of reelin in the processes underlying neurogenesis, neuronal migration in the cerebral cortex and the hippocampus; and the polarization, differentiation and maturation processes that neurons experiment in order to be functional in the adult brain. In vivo and in vitro evidence is presented in order to facilitate a better understanding of this fascinating system.
Collapse
|
20
|
Shi C, Cai Y, Li Y, Li Y, Hu N, Ma S, Hu S, Zhu P, Wang W, Zhou H. Yap promotes hepatocellular carcinoma metastasis and mobilization via governing cofilin/F-actin/lamellipodium axis by regulation of JNK/Bnip3/SERCA/CaMKII pathways. Redox Biol 2017; 14:59-71. [PMID: 28869833 PMCID: PMC5582718 DOI: 10.1016/j.redox.2017.08.013] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/08/2017] [Accepted: 08/21/2017] [Indexed: 12/16/2022] Open
Abstract
Despite the increasingly important role of Hippo-Yap in hepatocellular carcinoma (HCC) development and progression, little insight is available at the time regarding the specifics interaction of Yap and cancer cells migration. Here, we identified the mechanism by which tumor-intrinsic Yap deletion resulted in HCC migratory inhibition. Yap was greatly upregulated in HCC and its expression promoted the cells migration. Functional studies found that knockdown of Yap induced JNK phosphorylation which closely bound to the Bnip3 promoter and contributed to Bnip3 expression. Higher Bnip3 employed excessive mitophagy leading to mitochondrial dysfunction and ATP shortage. The insufficient ATP inactivated SERCA and consequently triggered intracellular calcium overload. As the consequence of calcium oscillation, Ca/calmodulin-dependent protein kinases II (CaMKII) was signaled and subsequently inhibited cofilin activity via phosphorylated modification. The phosphorylated cofilin failed to manipulate F-actin polymerization and lamellipodium formation, resulting into the impairment of lamellipodium-based migration. Collectively, our results identified Hippo-Yap as the tumor promoter in hepatocellular carcinoma that mediated via activation of cofilin/F-actin/lamellipodium axis by limiting JNK-Bnip3-SERCA-CaMKII pathways, with potential application to HCC therapy involving cancer metastasis. Yap is upregulated in the hepatocellular carcinoma and promotes cancer cell migration. Loss of Yap impairs cell mobility via inhibiting cofilin/F-actin/lamellipodium by activation of JNK-Bnip3-SERCA-CaMKII. Loss of Yap enhances JNK phosphorylation which triggers Bnip3-required mitophagy. Excessive mitophagy induces mitochondrial energy disorder which blunts SERCA and causes calcium overload. The calcium overload drives CaMKII which inactivates cofilin, leading to F-actin degradation and lamellipodium collapse.
Collapse
Affiliation(s)
- Chen Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yong Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yongheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ye Li
- Department of Oncology, PLA General Hospital Cancer Center, Beijing, China
| | - Nan Hu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Sai Ma
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Weihu Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China.
| | - Hao Zhou
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; Department of Cardiology, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
21
|
In silico analyses and global transcriptional profiling reveal novel putative targets for Pea3 transcription factor related to its function in neurons. PLoS One 2017; 12:e0170585. [PMID: 28158215 PMCID: PMC5291419 DOI: 10.1371/journal.pone.0170585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 01/08/2017] [Indexed: 01/05/2023] Open
Abstract
Pea3 transcription factor belongs to the PEA3 subfamily within the ETS domain transcription factor superfamily, and has been largely studied in relation to its role in breast cancer metastasis. Nonetheless, Pea3 plays a role not only in breast tumor, but also in other tissues with branching morphogenesis, including kidneys, blood vasculature, bronchi and the developing nervous system. Identification of Pea3 target promoters in these systems are important for a thorough understanding of how Pea3 functions. Present study particularly focuses on the identification of novel neuronal targets of Pea3 in a combinatorial approach, through curation, computational analysis and microarray studies in a neuronal model system, SH-SY5Y neuroblastoma cells. We not only show that quite a number of genes in cancer, immune system and cell cycle pathways, among many others, are either up- or down-regulated by Pea3, but also identify novel targets including ephrins and ephrin receptors, semaphorins, cell adhesion molecules, as well as metalloproteases such as kallikreins, to be among potential target promoters in neuronal systems. Our overall results indicate that rather than early stages of neurite extension and axonal guidance, Pea3 is more involved in target identification and synaptic maturation.
Collapse
|
22
|
Yan X, Liu J, Ye Z, Huang J, He F, Xiao W, Hu X, Luo Z. CaMKII-Mediated CREB Phosphorylation Is Involved in Ca2+-Induced BDNF mRNA Transcription and Neurite Outgrowth Promoted by Electrical Stimulation. PLoS One 2016; 11:e0162784. [PMID: 27611779 PMCID: PMC5017744 DOI: 10.1371/journal.pone.0162784] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 08/29/2016] [Indexed: 11/19/2022] Open
Abstract
Electrical stimulation (ES)-triggered up-regulation of brain-derived neurotrophic factor (BDNF) and neurite outgrowth in cultured rat postnatal dorsal root ganglion neurons (DRGNs) is calcium (Ca2+)-dependent. The effects of increased Ca2+ on BDNF up-regulation and neurite outgrowth remain unclear. We showed here that ES increased phosphorylation of the cAMP-response element binding protein (CREB). Blockade of Ca2+ suppressed CREB phosphorylation and neurite outgrowth. Down-regulation of phosphorylated (p)-CREB reduced BDNF transcription and neurite outgrowth triggered by ES. Furthermore, blockade of calmodulin-dependent protein kinase II (CaMKII) using the inhibitors KN93 or KN62 reduced p-CREB, and specific knockdown of the CaMKIIα or CaMKIIβ subunit was sufficient to suppress p-CREB. Recombinant BDNF or hyperforin reversed the effects of Ca2+ blockade and CaMKII knockdown. Taken together, these data establish a potential signaling pathway of Ca2+-CaMKII-CREB in neuronal activation. To our knowledge, this is the first report of the mechanisms of Ca2+-dependent BDNF transcription and neurite outgrowth triggered by ES. These findings might help further investigation of complex molecular signaling networks in ES-triggered nerve regeneration in vivo.
Collapse
Affiliation(s)
- Xiaodong Yan
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China
| | - Juanfang Liu
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China
| | - Zhengxu Ye
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Fei He
- Department of Hereditary and Development, Basic Unit, Fourth Military Medical University, Xi’an 710032, China
| | - Wei Xiao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Xueyu Hu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- * E-mail: (ZL); (XH)
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- * E-mail: (ZL); (XH)
| |
Collapse
|
23
|
Jianru YI, MeiLe LI, Yang Y, Zheng W, Yu LI, Zhao Z. Static compression regulates OPG expression in periodontal ligament cells via the CAMK II pathway. J Appl Oral Sci 2016; 23:549-54. [PMID: 26814456 PMCID: PMC4716692 DOI: 10.1590/1678-775720150156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/01/2015] [Indexed: 01/17/2023] Open
Abstract
Objective This study aimed to investigate the potential role of CAMK II pathway in the compression-regulated OPG expression in periodontal ligament cells (PDLCs). Material and Methods The PDL tissue model was developed by 3-D culturing human PDLCs in a thin sheet of poly lactic-co-glycolic acid (PLGA) scaffolds, which was subjected to static compression of 25 g/cm2 for 3, 6 and 12 h, with or without treatment of KN-93. After that, the expression of OPG, RANKL and NFATC2 was investigated through real-time PCR and western blot analysis. Results After static compression, the NFATC2 and RANKL expression was significantly up-regulated, while partially suppressed by KN-93 for 6 and 12 h respectively. The OPG expression was significantly down-regulated by compression in 3 h, started to elevate in 6 h, and significantly up-regulated in 12 h. The up-regulation after 12 h was significantly suppressed by KN-93. Conclusions Long-term static compression increases OPG expression in PDLCs, at least partially, via the CAMK II pathway.
Collapse
Affiliation(s)
- Y I Jianru
- State Key Laboratory of Oral Diseases, West China School and Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L I MeiLe
- State Key Laboratory of Oral Diseases, West China School and Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Yang
- State Key Laboratory of Oral Diseases, West China School and Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Zheng
- State Key Laboratory of Oral Diseases, West China School and Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L I Yu
- State Key Laboratory of Oral Diseases, West China School and Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, West China School and Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Gonzalez A, Moya-Alvarado G, Gonzalez-Billaut C, Bronfman FC. Cellular and molecular mechanisms regulating neuronal growth by brain-derived neurotrophic factor. Cytoskeleton (Hoboken) 2016; 73:612-628. [PMID: 27223597 DOI: 10.1002/cm.21312] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 12/31/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptors TrkB and p75 regulate dendritic and axonal growth during development and maintenance of the mature nervous system; however, the cellular and molecular mechanisms underlying this process are not fully understood. In recent years, several advances have shed new light on the processes behind the regulation of BDNF-mediated structural plasticity including control of neuronal transcription, local translation of proteins, and regulation of cytoskeleton and membrane dynamics. In this review, we summarize recent advances in the field of BDNF signaling in neurons to induce neuronal growth. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Andres Gonzalez
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Guillermo Moya-Alvarado
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian Gonzalez-Billaut
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile and Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Francisca C Bronfman
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
25
|
Coullery RP, Ferrari ME, Rosso SB. Neuronal development and axon growth are altered by glyphosate through a WNT non-canonical signaling pathway. Neurotoxicology 2016; 52:150-61. [PMID: 26688330 DOI: 10.1016/j.neuro.2015.12.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 01/25/2023]
Abstract
The growth and morphological differentiation of neurons are critical events in the establishment of proper neuronal connectivity and functioning. The developing nervous system is highly susceptible to damage caused by exposure to environmental contaminants. Glyphosate-containing herbicides are the most used agrochemicals in the world, particularly on genetically modified plants. Previous studies have demonstrated that glyphosate induces neurotoxicity in mammals. Therefore, its action mechanism on the nervous system needs to be determined. In this study, we report about impaired neuronal development caused by glyphosate exposure. Particularly, we observed that the initial axonal differentiation and growth of cultured neurons is affected by glyphosate since most treated cells remained undifferentiated after 1 day in culture. Although they polarized at 2 days in vitro, they elicited shorter and unbranched axons and they also developed less complex dendritic arbors compared to controls. To go further, we attempted to identify the cellular mechanism by which glyphosate affected neuronal morphology. Biochemical approaches revealed that glyphosate led to a decrease in Wnt5a level, a key factor for the initial neurite development and maturation, as well as inducing a down-regulation of CaMKII activity. This data suggests that the morphological defects would likely be a consequence of the decrease in both Wnt5a expression and CaMKII activity induced by glyphosate. Additionally, these changes might be reflected in a subsequent neuronal dysfunction. Therefore, our findings highlight the importance of establishing rigorous control on the use of glyphosate-based herbicides in order to protect mammals' health.
Collapse
Affiliation(s)
- Romina P Coullery
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - María E Ferrari
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Silvana B Rosso
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina.
| |
Collapse
|
26
|
Kim K, Lakhanpal G, Lu HE, Khan M, Suzuki A, Hayashi MK, Narayanan R, Luyben TT, Matsuda T, Nagai T, Blanpied TA, Hayashi Y, Okamoto K. A Temporary Gating of Actin Remodeling during Synaptic Plasticity Consists of the Interplay between the Kinase and Structural Functions of CaMKII. Neuron 2015; 87:813-26. [PMID: 26291163 DOI: 10.1016/j.neuron.2015.07.023] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 06/11/2015] [Accepted: 07/17/2015] [Indexed: 12/20/2022]
Abstract
The structural modification of dendritic spines plays a critical role in synaptic plasticity. CaMKII is a pivotal molecule involved in this process through both kinase-dependent and independent structural functions, but the respective contributions of these two functions to the synaptic plasticity remain unclear. We demonstrate that the transient interplay between the kinase and structural functions of CaMKII during the induction of synaptic plasticity temporally gates the activity-dependent modification of the actin cytoskeleton. Inactive CaMKII binds F-actin, thereby limiting access of actin-regulating proteins to F-actin and stabilizing spine structure. CaMKII-activating stimuli trigger dissociation of CaMKII from F-actin through specific autophosphorylation reactions within the F-actin binding region and permits F-actin remodeling by regulatory proteins followed by reassociation and restabilization. Blocking the autophosphorylation impairs both functional and structural plasticity without affecting kinase activity. These results underpin the importance of the interplay between the kinase and structural functions of CaMKII in defining a time window permissive for synaptic plasticity.
Collapse
Affiliation(s)
- Karam Kim
- Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | - Gurpreet Lakhanpal
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Hsiangmin E Lu
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mustafa Khan
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Akio Suzuki
- Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | - Mariko Kato Hayashi
- RIKEN-MIT Neuroscience Research Center, The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Radhakrishnan Narayanan
- RIKEN-MIT Neuroscience Research Center, The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Thomas T Luyben
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada; Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Tomoki Matsuda
- The Institute of Scientific and Industrial Research, Osaka University, Osaka 567-0047, Japan
| | - Takeharu Nagai
- The Institute of Scientific and Industrial Research, Osaka University, Osaka 567-0047, Japan
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yasunori Hayashi
- Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan; RIKEN-MIT Neuroscience Research Center, The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Brain and Body System Science Institute, Saitama University, Saitama 338-8570, Japan; School of Life Science, South China Normal University, Guangzhou 510631, China.
| | - Kenichi Okamoto
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada; RIKEN-MIT Neuroscience Research Center, The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
27
|
Hernandez-Chan NG, Bannon MJ, Orozco-Barrios CE, Escobedo L, Zamudio S, De la Cruz F, Gongora-Alfaro JL, Armendáriz-Borunda J, Reyes-Corona D, Espadas-Alvarez AJ, Flores-Martínez YM, Ayala-Davila J, Hernandez-Gutierrez ME, Pavón L, García-Villegas R, Nadella R, Martinez-Fong D. Neurotensin-polyplex-mediated brain-derived neurotrophic factor gene delivery into nigral dopamine neurons prevents nigrostriatal degeneration in a rat model of early Parkinson's disease. J Biomed Sci 2015; 22:59. [PMID: 26198255 PMCID: PMC4511027 DOI: 10.1186/s12929-015-0166-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 07/10/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The neurotrophin Brain-Derived Neurotrophic Factor (BDNF) influences nigral dopaminergic neurons via autocrine and paracrine mechanisms. The reduction of BDNF expression in Parkinson's disease substantia nigra (SN) might contribute to the death of dopaminergic neurons because inhibiting BDNF expression in the SN causes parkinsonism in the rat. This study aimed to demonstrate that increasing BDNF expression in dopaminergic neurons of rats with one week of 6-hydroxydopamine lesion recovers from parkinsonism. The plasmids phDAT-BDNF-flag and phDAT-EGFP, coding for enhanced green fluorescent protein, were transfected using neurotensin (NTS)-polyplex, which enables delivery of genes into the dopaminergic neurons via neurotensin-receptor type 1 (NTSR1) internalization. RESULTS Two weeks after transfections, RT-PCR and immunofluorescence techniques showed that the residual dopaminergic neurons retain NTSR1 expression and susceptibility to be transfected by the NTS-polyplex. phDAT-BDNF-flag transfection did not increase dopaminergic neurons, but caused 7-fold increase in dopamine fibers within the SN and 5-fold increase in innervation and dopamine levels in the striatum. These neurotrophic effects were accompanied by a significant improvement in motor behavior. CONCLUSIONS NTS-polyplex-mediated BDNF overexpression in dopaminergic neurons has proven to be effective to remit hemiparkinsonism in the rat. This BDNF gene therapy might be helpful in the early stage of Parkinson's disease.
Collapse
Affiliation(s)
- Nancy G Hernandez-Chan
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Michael J Bannon
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield Ave, 3355 Scott Hall, Detroit, MI, 48201, USA.
| | - Carlos E Orozco-Barrios
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Lourdes Escobedo
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Sergio Zamudio
- Escuela Nacional de Ciencias Biológicas, IPN, Wilfrido Massieu s/n, México D.F, 07738, Mexico.
| | - Fidel De la Cruz
- Escuela Nacional de Ciencias Biológicas, IPN, Wilfrido Massieu s/n, México D.F, 07738, Mexico.
| | - Jose L Gongora-Alfaro
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad de Yucatán, Av. Itzaes # 490 x 59-A, Mérida, Yucatán, 97000, Mexico.
| | - Juan Armendáriz-Borunda
- Institute for Molecular Biology and Gene Therapy, Department of Molecular Biology and Genomics, University of Guadalajara, Sierra Mojada # 950, Guadalajara, Jalisco, 44281, Mexico.
| | - David Reyes-Corona
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Armando J Espadas-Alvarez
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Yazmin M Flores-Martínez
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Jose Ayala-Davila
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Maria E Hernandez-Gutierrez
- Department of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México Xochimilco # 101, México D.F, 14370, Mexico.
| | - Lenin Pavón
- Department of Psychoimmunology, National Institute of Psychiatry "Ramón de la Fuente", Calzada México Xochimilco # 101, México D.F, 14370, Mexico.
| | - Refugio García-Villegas
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Rasajna Nadella
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
- Program de doctorado en Nanociencias and Nanotecnología, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| | - Daniel Martinez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
- Program de doctorado en Nanociencias and Nanotecnología, CINVESTAV, Av. Instituto Politécnico Nacional # 2508, México D.F, 07360, Mexico.
| |
Collapse
|
28
|
Gao L, Li C, Yang RY, Lian WW, Fang JS, Pang XC, Qin XM, Liu AL, Du GH. Ameliorative effects of baicalein in MPTP-induced mouse model of Parkinson's disease: A microarray study. Pharmacol Biochem Behav 2015; 133:155-63. [PMID: 25895692 DOI: 10.1016/j.pbb.2015.04.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 04/02/2015] [Accepted: 04/12/2015] [Indexed: 01/17/2023]
Abstract
Baicalein, a flavonoid from Scutellaria baicalensis Georgi, has been shown to possess neuroprotective properties. The purpose of this study was to explore the effects of baicalein on motor behavioral deficits and gene expression in N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice model of Parkinson's disease (PD). The behavioral results showed that baicalein significantly improves the abnormal behaviors in MPTP-induced mice model of PD, as manifested by shortening the total time for climbing down the pole, prolonging the latent periods of rotarod, and increasing the vertical movements. Using cDNA microarray and subsequent bioinformatic analyses, it was found that baicalein significantly promotes the biological processes including neurogenesis, neuroblast proliferation, neurotrophin signaling pathway, walking and locomotor behaviors, and inhibits dopamine metabolic process through regulation of gene expressions. Based on analysis of gene co-expression networks, the results indicated that the regulation of genes such as LIMK1, SNCA and GLRA1 by baicalein might play central roles in the network. Our results provide experimental evidence for the potential use of baicalein in the treatment of PD, and revealed gene expression profiles, biological processes and pathways influenced by baicalein in MPTP-treated mice.
Collapse
Affiliation(s)
- Li Gao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan 030006, PR China
| | - Chao Li
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Ran-Yao Yang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Wen-Wen Lian
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Jian-Song Fang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Xiao-Cong Pang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan 030006, PR China
| | - Ai-Lin Liu
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; Beijing Key Laboratory of Drug Target Research and Drug Screening, Beijing 100050, PR China.
| | - Guan-Hua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, PR China.
| |
Collapse
|
29
|
Ohashi K. Roles of cofilin in development and its mechanisms of regulation. Dev Growth Differ 2015; 57:275-90. [DOI: 10.1111/dgd.12213] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Kazumasa Ohashi
- Department of Biomolecular Sciences; Graduate School of Life Sciences; Tohoku University; Sendai Miyagi 980-8578 Japan
| |
Collapse
|
30
|
Koch JC, Tönges L, Barski E, Michel U, Bähr M, Lingor P. ROCK2 is a major regulator of axonal degeneration, neuronal death and axonal regeneration in the CNS. Cell Death Dis 2014; 5:e1225. [PMID: 24832597 PMCID: PMC4047920 DOI: 10.1038/cddis.2014.191] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 03/26/2014] [Accepted: 03/28/2014] [Indexed: 12/19/2022]
Abstract
The Rho/ROCK/LIMK pathway is central for the mediation of repulsive environmental signals in the central nervous system. Several studies using pharmacological Rho-associated protein kinase (ROCK) inhibitors have shown positive effects on neurite regeneration and suggest additional pro-survival effects in neurons. However, as none of these drugs is completely target specific, it remains unclear how these effects are mediated and whether ROCK is really the most relevant target of the pathway. To answer these questions, we generated adeno-associated viral vectors to specifically downregulate ROCK2 and LIM domain kinase (LIMK)-1 in rat retinal ganglion cells (RGCs) in vitro and in vivo. We show here that specific knockdown of ROCK2 and LIMK1 equally enhanced neurite outgrowth of RGCs on inhibitory substrates and both induced substantial neuronal regeneration over distances of more than 5 mm after rat optic nerve crush (ONC) in vivo. However, only knockdown of ROCK2 but not LIMK1 increased survival of RGCs after optic nerve axotomy. Moreover, knockdown of ROCK2 attenuated axonal degeneration of the proximal axon after ONC assessed by in vivo live imaging. Mechanistically, we demonstrate here that knockdown of ROCK2 resulted in decreased intraneuronal activity of calpain and caspase 3, whereas levels of pAkt and collapsin response mediator protein 2 and autophagic flux were increased. Taken together, our data characterize ROCK2 as a specific therapeutic target in neurodegenerative diseases and demonstrate new downstream effects of ROCK2 including axonal degeneration, apoptosis and autophagy.
Collapse
Affiliation(s)
- J C Koch
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - L Tönges
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - E Barski
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - U Michel
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - M Bähr
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - P Lingor
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| |
Collapse
|
31
|
Ohashi K, Sampei K, Nakagawa M, Uchiumi N, Amanuma T, Aiba S, Oikawa M, Mizuno K. Damnacanthal, an effective inhibitor of LIM-kinase, inhibits cell migration and invasion. Mol Biol Cell 2014; 25:828-40. [PMID: 24478456 PMCID: PMC3952852 DOI: 10.1091/mbc.e13-09-0540] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Damnacanthal is identified as an effective inhibitor of LIM-kinase. It inhibits chemotaxis of T-cells and migration and invasion of breast carcinoma cells in culture and hapten-induced migration of epidermal Langerhans cells in mouse ears. Damnacanthal is a useful tool for investigating the cellular and physiological functions of LIM-kinase. LIM-kinases (LIMKs) play crucial roles in various cell activities, including migration, division, and morphogenesis, by phosphorylating and inactivating cofilin. Using a bimolecular fluorescence complementation assay to detect the actin–cofilin interaction, we screened LIMK1 inhibitors and identified two effective inhibitors, damnacanthal (Dam) and MO-26 (a pyrazolopyrimidine derivative). These compounds have already been shown to inhibit Lck, a Src family tyrosine kinase. However, in vitro kinase assays revealed that Dam inhibited LIMK1 more effectively than Lck. Dam suppressed LIMK1-induced cofilin phosphorylation and deceleration of actin retrograde flow in lamellipodia in N1E-115 cells. Dam impaired CXCL12-induced chemotactic migration of Jurkat T lymphocytes and Jurkat-derived, Lck-deficient JCaM1.6 cells and also inhibited serum-induced migration and invasion of MDA-MB-231 breast carcinoma cells. These results suggest that Dam has the potential to suppress cell migration and invasion primarily through the inhibition of LIMK kinase activity. Topical application of Dam also suppressed hapten-induced migration of epidermal Langerhans cells in mouse ears. Dam provides a useful tool for investigating cellular and physiological functions of LIMKs and holds promise for the development of agents against LIMK-related diseases. The bimolecular fluorescence complementation assay system used in this study will provide a useful method to screen for inhibitors of various protein kinases.
Collapse
Affiliation(s)
- Kazumasa Ohashi
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8574, Japan Graduate School of Nanobioscience, Yokohama City University, Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | | | | | | | | | | | | | | |
Collapse
|