1
|
Aksan B, Kenkel AK, Yan J, Sánchez Romero J, Missirlis D, Mauceri D. VEGFD signaling balances stability and activity-dependent structural plasticity of dendrites. Cell Mol Life Sci 2024; 81:354. [PMID: 39158743 PMCID: PMC11335284 DOI: 10.1007/s00018-024-05357-2] [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: 05/24/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024]
Abstract
Mature neurons have stable dendritic architecture, which is essential for the nervous system to operate correctly. The ability to undergo structural plasticity, required to support adaptive processes like memory formation, is still present in mature neurons. It is unclear what molecular and cellular processes control this delicate balance between dendritic structural plasticity and stabilization. Failures in the preservation of optimal dendrite structure due to atrophy or maladaptive plasticity result in abnormal connectivity and are associated with various neurological diseases. Vascular endothelial growth factor D (VEGFD) is critical for the maintenance of mature dendritic trees. Here, we describe how VEGFD affects the neuronal cytoskeleton and demonstrate that VEGFD exerts its effects on dendrite stabilization by influencing the actin cortex and reducing microtubule dynamics. Further, we found that during synaptic activity-induced structural plasticity VEGFD is downregulated. Our findings revealed that VEGFD, acting on its cognate receptor VEGFR3, opposes structural changes by negatively regulating dendrite growth in cultured hippocampal neurons and in vivo in the adult mouse hippocampus with consequences on memory formation. A phosphoproteomic screening identified several regulatory proteins of the cytoskeleton modulated by VEGFD. Among the actin cortex-associated proteins, we found that VEGFD induces dephosphorylation of ezrin at tyrosine 478 via activation of the striatal-enriched protein tyrosine phosphatase (STEP). Activity-triggered structural plasticity of dendrites was impaired by expression of a phospho-deficient mutant ezrin in vitro and in vivo. Thus, VEGFD governs the equilibrium between stabilization and plasticity of dendrites by acting as a molecular brake of structural remodeling.
Collapse
Affiliation(s)
- Bahar Aksan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Ann-Kristin Kenkel
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Jing Yan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Javier Sánchez Romero
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Dimitris Missirlis
- Department of Cellular Biophysics, Max-Planck-Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany.
- Department Molecular and Cellular Neuroscience, Institute of Anatomy and Cell Biology, University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
2
|
Baltar J, Miranda RM, Cabral M, Rebelo S, Grahammer F, Huber TB, Reguenga C, Monteiro FA. Neph1 is required for neurite branching and is negatively regulated by the PRRXL1 homeodomain factor in the developing spinal cord dorsal horn. Neural Dev 2024; 19:13. [PMID: 39049046 PMCID: PMC11271021 DOI: 10.1186/s13064-024-00190-6] [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: 05/16/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
The cell-adhesion molecule NEPH1 is required for maintaining the structural integrity and function of the glomerulus in the kidneys. In the nervous system of Drosophila and C. elegans, it is involved in synaptogenesis and axon branching, which are essential for establishing functional circuits. In the mammalian nervous system, the expression regulation and function of Neph1 has barely been explored. In this study, we provide a spatiotemporal characterization of Neph1 expression in mouse dorsal root ganglia (DRGs) and spinal cord. After the neurogenic phase, Neph1 is broadly expressed in the DRGs and in their putative targets at the dorsal horn of the spinal cord, comprising both GABAergic and glutamatergic neurons. Interestingly, we found that PRRXL1, a homeodomain transcription factor that is required for proper establishment of the DRG-spinal cord circuit, prevents a premature expression of Neph1 in the superficial laminae of the dorsal spinal cord at E14.5, but has no regulatory effect on the DRGs or on either structure at E16.5. By chromatin immunoprecipitation analysis of the dorsal spinal cord, we identified four PRRXL1-bound regions within the Neph1 introns, suggesting that PRRXL1 directly regulates Neph1 transcription. We also showed that Neph1 is required for branching, especially at distal neurites. Together, our work showed that Prrxl1 prevents the early expression of Neph1 in the superficial dorsal horn, suggesting that Neph1 might function as a downstream effector gene for proper assembly of the DRG-spinal nociceptive circuit.
Collapse
Affiliation(s)
- João Baltar
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Rafael Mendes Miranda
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria Cabral
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Sandra Rebelo
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Departamento de Patologia Clínica, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Florian Grahammer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carlos Reguenga
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Filipe Almeida Monteiro
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal.
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal.
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
3
|
Frostadottir D, Welinder C, Perez R, Dahlin LB. Quantitative mass spectrometry analysis of the injured proximal and distal human digital nerve ends. Front Mol Neurosci 2024; 17:1425780. [PMID: 39015129 PMCID: PMC11250671 DOI: 10.3389/fnmol.2024.1425780] [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: 04/30/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Introduction Proteomic analysis of injured human peripheral nerves, particularly focusing on events occurring in the proximal and distal nerve ends, remains relatively underexplored. This study aimed to investigate the molecular patterns underlying a digital nerve injury, focusing on differences in protein expression between the proximal and distal nerve ends. Methods A total of 26 human injured digital nerve samples (24 men; 2 women; median age 47 [30-66] years), harvested during primary nerve repair within 48 h post-injury from proximal and distal nerve ends, were analyzed using mass spectrometry. Results A total of 3,914 proteins were identified, with 127 proteins showing significant differences in abundance between the proximal and the distal nerve ends. The downregulation of proteins in the distal nerve end was associated with synaptic transmission, autophagy, neurotransmitter regulation, cell adhesion and migration. Conversely, proteins upregulated in the distal nerve end were implicated in cellular stress response, neuromuscular junction stability and muscle contraction, neuronal excitability and neurotransmitter release, synaptic vesicle recycling and axon guidance and angiogenesis. Discussion Investigation of proteins, with functional annotations analysis, in proximal and the distal ends of human injured digital nerves, revealed dynamic cellular responses aimed at promoting tissue degeneration and restoration, while suppressing non-essential processes.
Collapse
Affiliation(s)
- Drifa Frostadottir
- Department of Translational Medicine – Hand Surgery, Lund University, Malmö, Sweden
- Department of Hand Surgery, Skåne University Hospital, Malmö, Sweden
| | - Charlotte Welinder
- Faculty of Medicine, Department of Clinical Sciences, Mass Spectrometry, Lund University, Lund, Sweden
| | - Raquel Perez
- Department of Translational Medicine – Hand Surgery, Lund University, Malmö, Sweden
- Unit for Social Epidemiology, Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
| | - Lars B. Dahlin
- Department of Translational Medicine – Hand Surgery, Lund University, Malmö, Sweden
- Department of Hand Surgery, Skåne University Hospital, Malmö, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Johansson Y, Andreassen M, Hartsch M, Wagner S, Forsby A. Attenuated neuronal differentiation caused by acrylamide is not related to oxidative stress in differentiated human neuroblastoma SH-SY5Y cells. Food Chem Toxicol 2024; 187:114623. [PMID: 38554842 DOI: 10.1016/j.fct.2024.114623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Acrylamide (ACR) is a known neurotoxicant and developmental neurotoxicant. As a soft electrophile, ACR reacts with thiol groups in cysteine. One hypothesis of ACR induced neurotoxicity and developmental neurotoxicity (DNT) is conjugation with reduced glutathione (GSH) leading to GSH depletion, increased reactive oxygen species (ROS) production and further oxidative stress and cellular damage. In this regard, we have investigated the effect of ACR on neuronal differentiation, glutathione levels and ROS production in the human neuroblastoma SH-SY5Y cell model. After 9 days of differentiation and exposure, ACR significantly impaired area neurites per cell at non-cytotoxic concentrations (0.33 μM and 10 μM). Furthermore, 10 μM ACR dysregulated 9 mRNA markers important for neuronal development, 5 of them being associated with cytoskeleton organization and axonal guidance. At the non-cytotoxic concentrations that significantly attenuate neuronal differentiation, ACR did neither decrease the level of GSH or total glutathione levels, nor increased ROS production. In addition, the expression of 5 mRNA markers for cellular stress was assessed with no significant altered regulation after ACR exposure up to 320 μM. Thus, ACR-induced DNT is not due to GSH depletion and increased ROS production, neither at non-cytotoxic nor cytotoxic concentrations, in the SH-SH5Y model during differentiation.
Collapse
Affiliation(s)
- Ylva Johansson
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden.
| | - Mathilda Andreassen
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Muriel Hartsch
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Stella Wagner
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| |
Collapse
|
5
|
Rocha Caldas G, do Amaral L, Munhoz Rodrigues D, Mayrink de Miranda A, Aparecida Guinaim Dos Santos N, Machado Rocha L, Tame Parreira RL, Cardozo Dos Santos A, Kenupp Bastos J. Brazilian Green Propolis' Artepillin C and Its Acetylated Derivative Activate the NGF-Signaling Pathways and Induce Neurite Outgrowth in NGF-Deprived PC12 Cells. Chem Biodivers 2023; 20:e202301294. [PMID: 37953436 DOI: 10.1002/cbdv.202301294] [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: 08/25/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/14/2023]
Abstract
Artepillin C is the most studied compound in Brazilian Green Propolis and, along with its acetylated derivative, displays neurotrophic activity on PC12 cells. Specific inhibitors of the trkA receptor (K252a), PI3K/Akt (LY294002), and MAPK/ERK (U0126) signaling pathways were used to investigate the neurotrophic mechanism. The expression of proteins involved in axonal and synaptic plasticity (GAP-43 and Synapsin I) was assessed by western blotting. Additionally, physicochemical properties, pharmacokinetics, and drug-likeness were evaluated by the SwissADME web tool. Both compounds induced neurite outgrowth by activating the NGF-signaling pathways but through different neuronal proteins. Furthermore, in silico analyses showed interesting physicochemical and pharmacokinetic properties of these compounds. Therefore, these compounds could play an important role in axonal and synaptic plasticity and should be further investigated.
Collapse
Affiliation(s)
- Gabriel Rocha Caldas
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Lilian do Amaral
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Débora Munhoz Rodrigues
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Aline Mayrink de Miranda
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Leandro Machado Rocha
- Natural Products Technology Laboratory-Fluminense Federal University, Niterói, RJ, Brazil
| | | | | | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
6
|
Belian S, Korenkova O, Zurzolo C. Actin-based protrusions at a glance. J Cell Sci 2023; 136:jcs261156. [PMID: 37987375 DOI: 10.1242/jcs.261156] [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] [Indexed: 11/22/2023] Open
Abstract
Actin-based protrusions are at the base of many fundamental cellular processes, such as cell adhesion, migration and intercellular communication. In recent decades, the discovery of new types of actin-based protrusions with unique functions has enriched our comprehension of cellular processes. However, as the repertoire of protrusions continues to expand, the rationale behind the classification of newly identified and previously known structures becomes unclear. Although current nomenclature allows good categorization of protrusions based on their functions, it struggles to distinguish them when it comes to structure, composition or formation mechanisms. In this Cell Science at a Glance article, we discuss the different types of actin-based protrusions, focusing on filopodia, cytonemes and tunneling nanotubes, to help better distinguish and categorize them based on their structural and functional differences and similarities.
Collapse
Affiliation(s)
- Sevan Belian
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
- Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Olga Korenkova
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
- Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Chiara Zurzolo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
| |
Collapse
|
7
|
Bernardes CP, Santos NAG, Costa TR, Menaldo DL, Sisti FM, Amstalden MK, Ribeiro DL, Antunes LMG, Sampaio SV, Santos AC. Effects of C-Terminal-Ethyl-Esterification in a Snake-Venom-Based Peptide Against the Neurotoxicity of Acrolein in PC12 Cells. Int J Pept Res Ther 2023. [DOI: 10.1007/s10989-023-10517-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
8
|
Exosomes induce neurogenesis of pluripotent P19 cells. Stem Cell Rev Rep 2023:10.1007/s12015-023-10512-6. [PMID: 36811747 PMCID: PMC10366297 DOI: 10.1007/s12015-023-10512-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/24/2023]
Abstract
Exosomes play a role in tissue/organ development and differentiation. Retinoic acid induces differentiation of P19 cells (UD-P19) to P19 neurons (P19N) that behave like cortical neurons and express characteristic neuronal genes such as NMDA receptor subunits. Here we report P19N exosome-mediated differentiation of UD-P19 to P19N. Both UD-P19 and P19N released exosomes with characteristic exosome morphology, size, and common protein markers. P19N internalized significantly higher number of Dil-P19N exosomes as compared to UD-P19 with accumulation in the perinuclear region. Continuous exposure of UD-P19 to P19N exosomes for six days induced formation of small-sized embryoid bodies that differentiated into MAP2-/GluN2B-positive neurons recapitulating RA-induction of neurogenesis. Incubation with UD-P19 exosomes for six days did not affect UD-P19. Small RNA-seq identified enrichment of P19N exosomes with pro-neurogenic non-coding RNAs (ncRNAs) such as miR-9, let-7, MALAT1 and depleted with ncRNAs involved in maintenance of stem cell characteristics. UD-P19 exosomes were rich with ncRNAs required for maintenance of stemness. P19N exosomes provide an alternative method to genetic modifications for cellular differentiation of neurons. Our novel findings on exosomes-mediated differentiation of UD-P19 to P19 neurons provide tools to study pathways directing neuron development/differentiation and develop novel therapeutic strategies in neuroscience.
Collapse
|
9
|
Radler MR, Liu X, Peng M, Doyle B, Toyo-Oka K, Spiliotis ET. Pyramidal neuron morphogenesis requires a septin network that stabilizes filopodia and suppresses lamellipodia during neurite initiation. Curr Biol 2023; 33:434-448.e8. [PMID: 36538929 PMCID: PMC9905282 DOI: 10.1016/j.cub.2022.11.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022]
Abstract
Pyramidal neurons are a major cell type of the forebrain, consisting of a pyramidally shaped soma with axonal and apicobasal dendritic processes. It is poorly understood how the neuronal soma develops its pyramidal morphology, while generating neurites of the proper shape and orientation. Here, we discovered that the spherical somata of immature neurite-less neurons possess a circumferential wreath-like network of septin filaments, which promotes neuritogenesis by balancing the protrusive activity of lamellipodia and filopodia. In embryonic rat hippocampal and mouse cortical neurons, the septin wreath network consists of curvilinear filaments that contain septins 5, 7, and 11 (Sept5/7/11). The Sept5/7/11 wreath network demarcates a zone of myosin II enrichment and Arp2/3 diminution at the base of filopodial actin bundles. In Sept7-depleted neurons, cell bodies are enlarged with hyperextended lamellae and abnormally shaped neurites that originate from lamellipodia. This phenotype is accompanied by diminished myosin II and filopodia lifetimes and increased Arp2/3 and lamellipodial activity. Inhibition of Arp2/3 rescues soma and neurite phenotypes, indicating that the septin wreath network suppresses the extension of lamellipodia, facilitating the formation of neurites from the filopodia of a consolidated soma. We show that this septin function is critical for developing a pyramidally shaped soma with properly distributed and oriented dendrites in cultured rat hippocampal neurons and in vivo in mouse perinatal cortical neurons. Therefore, the somatic septin cytoskeleton provides a key morphogenetic mechanism for neuritogenesis and the development of pyramidal neurons.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Megan Peng
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Brenna Doyle
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
c-Abl Tyrosine Kinase Is Required for BDNF-Induced Dendritic Branching and Growth. Int J Mol Sci 2023; 24:ijms24031944. [PMID: 36768268 PMCID: PMC9916151 DOI: 10.3390/ijms24031944] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) induces activation of the TrkB receptor and several downstream pathways (MAPK, PI3K, PLC-γ), leading to neuronal survival, growth, and plasticity. It has been well established that TrkB signaling regulation is required for neurite formation and dendritic arborization, but the specific mechanism is not fully understood. The non-receptor tyrosine kinase c-Abl is a possible candidate regulator of this process, as it has been implicated in tyrosine kinase receptors' signaling and trafficking, as well as regulation of neuronal morphogenesis. To assess the role of c-Abl in BDNF-induced dendritic arborization, wild-type and c-Abl-KO neurons were stimulated with BDNF, and diverse strategies were employed to probe the function of c-Abl, including the use of pharmacological inhibitors, an allosteric c-Abl activator, and shRNA to downregulates c-Abl expression. Surprisingly, BDNF promoted c-Abl activation and interaction with TrkB receptors. Furthermore, pharmacological c-Abl inhibition and genetic ablation abolished BDNF-induced dendritic arborization and increased the availability of TrkB in the cell membrane. Interestingly, inhibition or genetic ablation of c-Abl had no effect on the classic TrkB downstream pathways. Together, our results suggest that BDNF/TrkB-dependent c-Abl activation is a novel and essential mechanism in TrkB signaling.
Collapse
|
11
|
Baar S, Kuragano M, Tokuraku K, Watanabe S. Towards a comprehensive approach for characterizing cell activity in bright-field microscopic images. Sci Rep 2022; 12:16884. [PMID: 36207347 PMCID: PMC9546915 DOI: 10.1038/s41598-022-20598-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/15/2022] [Indexed: 11/23/2022] Open
Abstract
When studying physical cellular response observed by light microscopy, variations in cell behavior are difficult to quantitatively measure and are often only discussed on a subjective level. Hence, cell properties are described qualitatively based on a researcher’s impressions. In this study, we aim to define a comprehensive approach to estimate the physical cell activity based on migration and morphology based on statistical analysis of a cell population within a predefined field of view and timespan. We present quantitative measurements of the influence of drugs such as cytochalasin D and taxol on human neuroblastoma, SH-SY5Y cell populations. Both chemicals are well known to interact with the cytoskeleton and affect the cell morphology and motility. Being able to compute the physical properties of each cell for a given observation time, requires precise localization of each cell even when in an adhesive state, where cells are not visually differentiable. Also, the risk of confusion through contaminants is desired to be minimized. In relation to the cell detection process, we have developed a customized encoder-decoder based deep learning cell detection and tracking procedure. Further, we discuss the accuracy of our approach to quantify cell activity and its viability in regard to the cell detection accuracy.
Collapse
Affiliation(s)
- Stefan Baar
- Graduate School of Engineering, Muroran Institute of Technology, 27-1 Mizumoto-cho, Muroran, Hokkaido, 050-8585, Japan
| | - Masahiro Kuragano
- Graduate School of Engineering, Muroran Institute of Technology, 27-1 Mizumoto-cho, Muroran, Hokkaido, 050-8585, Japan
| | - Kiyotaka Tokuraku
- Graduate School of Engineering, Muroran Institute of Technology, 27-1 Mizumoto-cho, Muroran, Hokkaido, 050-8585, Japan
| | - Shinya Watanabe
- Graduate School of Engineering, Muroran Institute of Technology, 27-1 Mizumoto-cho, Muroran, Hokkaido, 050-8585, Japan.
| |
Collapse
|
12
|
Fuchs J, Bareesel S, Kroon C, Polyzou A, Eickholt BJ, Leondaritis G. Plasma membrane phospholipid phosphatase-related proteins as pleiotropic regulators of neuron growth and excitability. Front Mol Neurosci 2022; 15:984655. [PMID: 36187351 PMCID: PMC9520309 DOI: 10.3389/fnmol.2022.984655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neuronal plasma membrane proteins are essential for integrating cell extrinsic and cell intrinsic signals to orchestrate neuronal differentiation, growth and plasticity in the developing and adult nervous system. Here, we shed light on the family of plasma membrane proteins phospholipid phosphatase-related proteins (PLPPRs) (alternative name, PRGs; plasticity-related genes) that fine-tune neuronal growth and synaptic transmission in the central nervous system. Several studies uncovered essential functions of PLPPRs in filopodia formation, axon guidance and branching during nervous system development and regeneration, as well as in the control of dendritic spine number and excitability. Loss of PLPPR expression in knockout mice increases susceptibility to seizures, and results in defects in sensory information processing, development of psychiatric disorders, stress-related behaviors and abnormal social interaction. However, the exact function of PLPPRs in the context of neurological diseases is largely unclear. Although initially described as active lysophosphatidic acid (LPA) ecto-phosphatases that regulate the levels of this extracellular bioactive lipid, PLPPRs lack catalytic activity against LPA. Nevertheless, they emerge as atypical LPA modulators, by regulating LPA mediated signaling processes. In this review, we summarize the effects of this protein family on cellular morphology, generation and maintenance of cellular protrusions as well as highlight their known neuronal functions and phenotypes of KO mice. We discuss the molecular mechanisms of PLPPRs including the deployment of phospholipids, actin-cytoskeleton and small GTPase signaling pathways, with a focus on identifying gaps in our knowledge to stimulate interest in this understudied protein family.
Collapse
Affiliation(s)
- Joachim Fuchs
- Institute of Molecular Biology and Biochemistry, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Shannon Bareesel
- Institute of Molecular Biology and Biochemistry, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Cristina Kroon
- Institute of Molecular Biology and Biochemistry, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexandra Polyzou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Britta J. Eickholt
- Institute of Molecular Biology and Biochemistry, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- *Correspondence: Britta J. Eickholt,
| | - George Leondaritis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Institute of Biosciences, University Research Center Ioannina, University of Ioannina, Ioannina, Greece
- George Leondaritis,
| |
Collapse
|
13
|
Lee HN, Hyeon SJ, Kim H, Sim KM, Kim Y, Ju J, Lee J, Wang Y, Ryu H, Seong J. Decreased FAK activity and focal adhesion dynamics impair proper neurite formation of medium spiny neurons in Huntington's disease. Acta Neuropathol 2022; 144:521-536. [PMID: 35857122 DOI: 10.1007/s00401-022-02462-z] [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: 03/25/2022] [Revised: 06/11/2022] [Accepted: 06/25/2022] [Indexed: 11/29/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a polyglutamine expansion in the protein huntingtin (HTT) [55]. While the final pathological consequence of HD is the neuronal cell death in the striatum region of the brain, it is still unclear how mutant HTT (mHTT) causes synaptic dysfunctions at the early stage and during the progression of HD. Here, we discovered that the basal activity of focal adhesion kinase (FAK) is severely reduced in a striatal HD cell line, a mouse model of HD, and the human post-mortem brains of HD patients. In addition, we observed with a FRET-based FAK biosensor [59] that neurotransmitter-induced FAK activation is decreased in HD striatal neurons. Total internal reflection fluorescence (TIRF) imaging revealed that the reduced FAK activity causes the impairment of focal adhesion (FA) dynamics, which further leads to the defect in filopodial dynamics causing the abnormally increased number of immature neurites in HD striatal neurons. Therefore, our results suggest that the decreased FAK and FA dynamics in HD impair the proper formation of neurites, which is crucial for normal synaptic functions [52]. We further investigated the molecular mechanism of FAK inhibition in HD and surprisingly discovered that mHTT strongly associates with phosphatidylinositol 4,5-biphosphate, altering its normal distribution at the plasma membrane, which is crucial for FAK activation [14, 60]. Therefore, our results provide a novel molecular mechanism of FAK inhibition in HD along with its pathological mechanism for synaptic dysfunctions during the progression of HD.
Collapse
Affiliation(s)
- Hae Nim Lee
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Converging Science and Technology, Kyung Hee University, Seoul, 02453, Republic of Korea
| | - Seung Jae Hyeon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Heejung Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Converging Science and Technology, Kyung Hee University, Seoul, 02453, Republic of Korea
| | - Kyoung Mi Sim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yunha Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongmin Ju
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Junghee Lee
- Department of Neurology, Boston University Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Yingxiao Wang
- Department of Bioengineering, University of California, San Diego, CA, 92093, USA
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- Department of Converging Science and Technology, Kyung Hee University, Seoul, 02453, Republic of Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
14
|
Godini R, Fallahi H, Pocock R. The regulatory landscape of neurite development in Caenorhabditis elegans. Front Mol Neurosci 2022; 15:974208. [PMID: 36090252 PMCID: PMC9453034 DOI: 10.3389/fnmol.2022.974208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/26/2022] [Indexed: 11/18/2022] Open
Abstract
Neuronal communication requires precise connectivity of neurite projections (axons and dendrites). Developing neurites express cell-surface receptors that interpret extracellular cues to enable correct guidance toward, and connection with, target cells. Spatiotemporal regulation of neurite guidance molecule expression by transcription factors (TFs) is critical for nervous system development and function. Here, we review how neurite development is regulated by TFs in the Caenorhabditis elegans nervous system. By collecting publicly available transcriptome and ChIP-sequencing data, we reveal gene expression dynamics during neurite development, providing insight into transcriptional mechanisms governing construction of the nervous system architecture.
Collapse
Affiliation(s)
- Rasoul Godini
- Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- *Correspondence: Rasoul Godini,
| | - Hossein Fallahi
- Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Roger Pocock
- Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Roger Pocock,
| |
Collapse
|
15
|
Higgs VE, Das RM. Establishing neuronal polarity: microtubule regulation during neurite initiation. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac007. [PMID: 38596701 PMCID: PMC10913830 DOI: 10.1093/oons/kvac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/25/2022] [Accepted: 05/02/2022] [Indexed: 04/11/2024]
Abstract
The initiation of nascent projections, or neurites, from the neuronal cell body is the first stage in the formation of axons and dendrites, and thus a critical step in the establishment of neuronal architecture and nervous system development. Neurite formation relies on the polarized remodelling of microtubules, which dynamically direct and reinforce cell shape, and provide tracks for cargo transport and force generation. Within neurons, microtubule behaviour and structure are tightly controlled by an array of regulatory factors. Although microtubule regulation in the later stages of axon development is relatively well understood, how microtubules are regulated during neurite initiation is rarely examined. Here, we discuss how factors that direct microtubule growth, remodelling, stability and positioning influence neurite formation. In addition, we consider microtubule organization by the centrosome and modulation by the actin and intermediate filament networks to provide an up-to-date picture of this vital stage in neuronal development.
Collapse
Affiliation(s)
- Victoria E Higgs
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Raman M Das
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
16
|
Hoff KJ, Aiken JE, Gutierrez MA, Franco SJ, Moore JK. Tubulinopathy mutations in TUBA1A that disrupt neuronal morphogenesis and migration override XMAP215/Stu2 regulation of microtubule dynamics. eLife 2022; 11:76189. [PMID: 35511030 PMCID: PMC9236607 DOI: 10.7554/elife.76189] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Heterozygous, missense mutations in α- or β-tubulin genes are associated with a wide range of human brain malformations, known as tubulinopathies. We seek to understand whether a mutation’s impact at the molecular and cellular levels scale with the severity of brain malformation. Here, we focus on two mutations at the valine 409 residue of TUBA1A, V409I, and V409A, identified in patients with pachygyria or lissencephaly, respectively. We find that ectopic expression of TUBA1A-V409I/A mutants disrupt neuronal migration in mice and promote excessive neurite branching and a decrease in the number of neurite retraction events in primary rat neuronal cultures. These neuronal phenotypes are accompanied by increased microtubule acetylation and polymerization rates. To determine the molecular mechanisms, we modeled the V409I/A mutants in budding yeast and found that they promote intrinsically faster microtubule polymerization rates in cells and in reconstitution experiments with purified tubulin. In addition, V409I/A mutants decrease the recruitment of XMAP215/Stu2 to plus ends in budding yeast and ablate tubulin binding to TOG (tumor overexpressed gene) domains. In each assay tested, the TUBA1A-V409I mutant exhibits an intermediate phenotype between wild type and the more severe TUBA1A-V409A, reflecting the severity observed in brain malformations. Together, our data support a model in which the V409I/A mutations disrupt microtubule regulation typically conferred by XMAP215 proteins during neuronal morphogenesis and migration, and this impact on tubulin activity at the molecular level scales with the impact at the cellular and tissue levels. Proteins are molecules made up of long chains of building blocks called amino acids. When a mutation changes one of these amino acids, it can lead to the protein malfunctioning, which can have many effects at the cell and tissue level. Given that human proteins are made up of 20 different amino acids, each building block in a protein could mutate to any of the other 19 amino acids, and each mutations could have different effects. Tubulins are proteins that form microtubules, thin tubes that help give cells their shape and allow them to migrate. These proteins are added or removed to microtubules depending on the cell’s needs, meaning that microtubules can grow or shrink depending on the situation. Mutations in the tubulin proteins have been linked to malformations of varying severities involving the formation of ridges and folds on the surface of the brain, including lissencephaly, pachygyria or polymicrogyria. Hoff et al. wanted to establish links between tubulin mutations and the effects observed at both cell and tissue level in the brain. They focused on two mutations in the tubulin protein TUBA1A that affect the amino acid in position 409 in the protein, which is normally a valine. One of the mutations turns this valine into an amino acid called isoleucine. This mutation is associated with pachygyria, which leads to the brain developing few ridges that are broad and flat. The second mutation turns the valine into an alanine, and is linked to lissencephaly, a more severe condition in which the brain develops no ridges, appearing smooth. Hoff et al. found that both mutations interfere with the development of the brain by stopping neurons from migrating properly, which prevents them from forming the folds in the brain correctly. At the cellular level, the mutations lead to tubulins becoming harder to remove from microtubules, making microtubules more stable than usual. This results in longer microtubules that are harder for the cell to shorten or destroy as needed. Additionally, Hoff et al. showed that the mutant versions of TUBA1A have weaker interactions with a protein called XMAP215, which controls the addition of tubulin to microtubules. This causes the microtubules to grow uncontrollably. Hoff et al. also established that the magnitude of the effects of each mutation on microtubule growth scale with the severity of the disorder they cause. Specifically, cells in which TUBA1A is not mutated have microtubules that grow at a normal rate, and lead to typical brain development. Meanwhile, cells carrying the mutation that turns a valine into an alanine, which is linked to the more severe condition lissencephaly, have microtubules that grow very fast. Finally, cells in which the valine is mutated to an isoleucine – the mutation associated with the less severe malformation pachygyria – have microtubules that grow at an intermediate rate. These findings provide a link between mutations in tubulin proteins and larger effects on cell movement that lead to brain malformations. Additionally, they also link the severity of the malformation to the severity of the microtubule defect caused by each mutation. Further work could examine whether microtubule stabilization is also seen in other similar diseases, which, in the long term, could reveal ways to detect and treat these illnesses.
Collapse
Affiliation(s)
- Katelyn J Hoff
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Jayne E Aiken
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Mark A Gutierrez
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Santos J Franco
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Jeffrey K Moore
- University of Colorado School of Medicine, Aurora, United States
| |
Collapse
|
17
|
Tian T, Quintana-Urzainqui I, Kozić Z, Pratt T, Price DJ. Pax6 loss alters the morphological and electrophysiological development of mouse prethalamic neurons. Development 2022; 149:274738. [PMID: 35224626 PMCID: PMC8977098 DOI: 10.1242/dev.200052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/16/2022] [Indexed: 12/02/2022]
Abstract
Pax6 is a well-known regulator of early neuroepithelial progenitor development. Its constitutive loss has a particularly strong effect on the developing prethalamus, causing it to become extremely hypoplastic. To overcome this difficulty in studying the long-term consequences of Pax6 loss for prethalamic development, we used conditional mutagenesis to delete Pax6 at the onset of neurogenesis and studied the developmental potential of the mutant prethalamic neurons in vitro. We found that Pax6 loss affected their rates of neurite elongation, the location and length of their axon initial segments, and their electrophysiological properties. Our results broaden our understanding of the long-term consequences of Pax6 deletion in the developing mouse forebrain, suggesting that it can have cell-autonomous effects on the structural and functional development of some neurons. Summary: Pax6 impacts neurite extension, axon initial segment properties and the ability to fire normal action potentials in maturing neurons, revealing actions extending beyond those previously characterised in progenitors.
Collapse
Affiliation(s)
- Tian Tian
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Idoia Quintana-Urzainqui
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69012 Heidelberg, Germany
| | - Zrinko Kozić
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Thomas Pratt
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - David J. Price
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
18
|
Vijayalakshmi V, Sadanandan B, Venkataramanaiah Raghu A. Single walled carbon nanotubes in high concentrations is cytotoxic to the human neuronal cell LN18. RESULTS IN CHEMISTRY 2022. [DOI: 10.1016/j.rechem.2022.100484] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
19
|
Mendieta I, Rodríguez-Nieto M, Nuñez-Anita RE, Menchaca-Arredondo JL, García-Alcocer G, Berumen LC. Ultrastructural changes associated to the neuroendocrine transdifferentiation of the lung adenocarcinoma cell line A549. Acta Histochem 2021; 123:151797. [PMID: 34688180 DOI: 10.1016/j.acthis.2021.151797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022]
Abstract
The neuroendocrine transdifferentiation has been found in many cancer cell types, such as prostate, lung and gastrointestinal cells and is accompanied by a lower patient life expectancy. The transdifferentiation process has been induced in vitro by the exposure to different stimuli in human lung adenocarcinoma. The aim of this work was to identify the morphological characteristics of the neuroendocrine phenotype in a human lung cancer cell line, induced by two cAMP elevating agents (IBMX and FSK). Our results showed two phenotypes, one produced by IBMX with higher volume, cell size and increased number of secondary projections, and the other produced by FSK with higher area, roughness of the membrane, cell neurite percentage, number of outgrowths per cell and increased number of primary projections. In conclusion, we describe some morphological and ultrastructural characteristics of the neuroendocrine phenotype in A549 human lung cancer cell line promoted by IBMX and FSK to contribute to the understanding of the autocrine or paracrine signaling within the tumor microenvironment.
Collapse
Affiliation(s)
- Irasema Mendieta
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, Cerro de las Campanas 76010, Querétaro, Mexico
| | - Maricela Rodríguez-Nieto
- Instituto de Física y Matemáticas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
| | - Rosa Elvira Nuñez-Anita
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás Hidalgo, Tarímbaro Municipio de Morelia 58920, Michoacán, Mexico
| | - Jorge Luis Menchaca-Arredondo
- Facultad de Ciencias Físico Matemáticas, Universidad Autónoma de Nuevo León, Centro de Investigación en Ciencias Físico Matemáticas, San Nicolás de los Garza 66455, Nuevo León, Mexico
| | - Guadalupe García-Alcocer
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, Cerro de las Campanas 76010, Querétaro, Mexico
| | - Laura Cristina Berumen
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, Cerro de las Campanas 76010, Querétaro, Mexico.
| |
Collapse
|
20
|
Tsutsui K, Kim HS, Yoshikata C, Kimura K, Kubota Y, Shibata Y, Tian C, Liu J, Nishiwaki K. Repulsive guidance molecule acts in axon branching in Caenorhabditis elegans. Sci Rep 2021; 11:22370. [PMID: 34785759 PMCID: PMC8595726 DOI: 10.1038/s41598-021-01853-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/03/2021] [Indexed: 11/25/2022] Open
Abstract
Repulsive guidance molecules (RGMs) are evolutionarily conserved proteins implicated in repulsive axon guidance. Here we report the function of the Caenorhabditis elegans ortholog DRAG-1 in axon branching. The axons of hermaphrodite-specific neurons (HSNs) extend dorsal branches at the region abutting the vulval muscles. The drag-1 mutants exhibited defects in HSN axon branching in addition to a small body size phenotype. DRAG-1 expression in the hypodermal cells was required for the branching of the axons. Although DRAG-1 is normally expressed in the ventral hypodermis excepting the vulval region, its ectopic expression in vulval precursor cells was sufficient to induce the branching. The C-terminal glycosylphosphatidylinositol anchor of DRAG-1 was important for its function, suggesting that DRAG-1 should be anchored to the cell surface. Genetic analyses suggested that the membrane receptor UNC-40 acts in the same pathway with DRAG-1 in HSN branching. We propose that DRAG-1 expressed in the ventral hypodermis signals via the UNC-40 receptor expressed in HSNs to elicit branching activity of HSN axons.
Collapse
Affiliation(s)
- Kaname Tsutsui
- Department of Bioscience, Kwansei Gakuin University, 2-1 Gakuen, Sanda, 669-1337, Japan
| | - Hon-Song Kim
- Department of Bioscience, Kwansei Gakuin University, 2-1 Gakuen, Sanda, 669-1337, Japan
| | - Chizu Yoshikata
- Department of Bioscience, Kwansei Gakuin University, 2-1 Gakuen, Sanda, 669-1337, Japan
| | - Kenji Kimura
- Department of Bioscience, Kwansei Gakuin University, 2-1 Gakuen, Sanda, 669-1337, Japan
| | - Yukihiko Kubota
- Department of Bioscience, Kwansei Gakuin University, 2-1 Gakuen, Sanda, 669-1337, Japan
| | - Yukimasa Shibata
- Department of Bioscience, Kwansei Gakuin University, 2-1 Gakuen, Sanda, 669-1337, Japan
| | - Chenxi Tian
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Kiyoji Nishiwaki
- Department of Bioscience, Kwansei Gakuin University, 2-1 Gakuen, Sanda, 669-1337, Japan.
| |
Collapse
|
21
|
Chakraborty A, Upadhya R, Usman TA, Shetty AK, Rutkowski JM. Chronic VEGFR-3 signaling preserves dendritic arborization and sensitization under stress. Brain Behav Immun 2021; 98:219-233. [PMID: 34389489 PMCID: PMC8511130 DOI: 10.1016/j.bbi.2021.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/15/2021] [Accepted: 08/05/2021] [Indexed: 11/15/2022] Open
Abstract
Dendritic arborization is critical for the establishment and maintenance of precise neural circuits. Vascular endothelial growth factor D (VEGF-D), well-characterized as a "lymphangiogenic" growth factor, reportedly maintains dendritic arborization and synaptic strength in the hippocampus of adult mice through VEGF receptor (VEGFR-3) signaling. Here, we investigated the effect of chronic VEGFR-3-specific activation on adipose arbor morphometry using the Adipo-VD mouse, a model of inducible, adipose-specific VEGF-D overexpression. We examined whether adipose tissue innervation was preserved or functionally different in Adipo-VD mice during stress in vivo and if VEGFR-3 signaling afforded neuroprotection to challenged neurons in vitro. Chronic VEGFR-3 signaling in Adipo-VD subcutaneous adipose tissue resulted in a reduction in the dendrite length, dendritic terminal branches (filament length), and dendritic terminal branch volume (filament volume), but increased dendrite branching. We also identified reduced stimulus-evoked excitatory sympathetic nerve activity in Adipo-VD mice. Following 6-hydroxydopamine (6-OHDA) denervation, Adipo-VD dendritic arbors were preserved, including improved dendritic branch volume, length, and dendritic branches than in wildtype tissues. In vitro, we found that chronic elevation of VEGFR-3 signaling in developing mVC neurons changes the dendritic arbor complexity and improves stress-induced structure remodeling. Developing neurons are conferred neuroprotection against stress, potentially by upregulation of proteolytic conversion of pro-BDNF to mature BDNF. Mature neurons, however, display improved dendritic arbor complexity, and unaltered dendritic structural remodeling and improved resistance to stress with VEGFR-3 signaling. Overall, chronically increasing VEGFR-3 signaling in neurons has a synergistic impact on neurosensitization and neuroprotection during stress.
Collapse
Affiliation(s)
- Adri Chakraborty
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Timaj A. Usman
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Joseph M. Rutkowski
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA,Correspondence: Joseph M Rutkowski, Texas A&M University College of Medicine, 8447 Riverside Parkway, Bryan, TX 77807 USA, Ph: 979-436-0576,
| |
Collapse
|
22
|
Slováčková J, Slavík J, Kulich P, Večeřa J, Kováč O, Paculová H, Straková N, Fedr R, Silva JP, Carvalho F, Machala M, Procházková J. Polychlorinated environmental toxicants affect sphingolipid metabolism during neurogenesis in vitro. Toxicology 2021; 463:152986. [PMID: 34627992 DOI: 10.1016/j.tox.2021.152986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/17/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022]
Abstract
Sphingolipids (SLs) are important signaling molecules and functional components of cellular membranes. Although SLs are known as crucial regulators of neural cell physiology and differentiation, modulations of SLs by environmental neurotoxicants in neural cells and their neuronal progeny have not yet been explored. In this study, we used in vitro models of differentiated neuron-like cells, which were repeatedly exposed during differentiation to model environmental toxicants, and we analyzed changes in sphingolipidome, cellular morphology and gene expression related to SL metabolism or neuronal differentiation. We compared these data with the results obtained in undifferentiated neural cells with progenitor-like features. As model polychlorinated organic pollutants, we used 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 3,3'-dichlorobiphenyl (PCB11) and 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153). PCB153 revealed itself as the most prominent deregulator of SL metabolism and as potent toxicant during early phases of in vitro neurogenesis. TCDD exerted only minor changes in the levels of analysed lipid species, however, it significantly changed the rate of pro-neuronal differentiation and deregulated expression of neuronal markers during neurogenesis. PCB11 acted as a potent disruptor of in vitro neurogenesis, which induced significant alterations in SL metabolism and cellular morphology in both differentiated neuron-like models (differentiated NE4C and NG108-15 cells). We identified ceramide-1-phosphate, lactosylceramides and several glycosphingolipids to be the most sensitive SL species to exposure to polychlorinated pollutants. Additionally, we identified deregulation of several genes related to SL metabolism, which may be explored in future as potential markers of developmental neurotoxicity.
Collapse
Affiliation(s)
- Jana Slováčková
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic
| | - Josef Slavík
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic
| | - Pavel Kulich
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic
| | - Josef Večeřa
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic
| | - Ondrej Kováč
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic
| | - Hana Paculová
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic
| | - Nicol Straková
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic
| | - Radek Fedr
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 61265, Brno, Czech Republic
| | - João Pedro Silva
- Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | - Félix Carvalho
- Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | - Miroslav Machala
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic.
| | - Jiřina Procházková
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100, Brno, Czech Republic; Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 61265, Brno, Czech Republic.
| |
Collapse
|
23
|
Bodakuntla S, Nedozralova H, Basnet N, Mizuno N. Cytoskeleton and Membrane Organization at Axon Branches. Front Cell Dev Biol 2021; 9:707486. [PMID: 34540830 PMCID: PMC8440873 DOI: 10.3389/fcell.2021.707486] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Axon branching is a critical process ensuring a high degree of interconnectivity for neural network formation. As branching occurs at sites distant from the soma, it is necessary that axons have a local system to dynamically control and regulate axonal growth. This machinery depends on the orchestration of cellular functions such as cytoskeleton, subcellular transport, energy production, protein- and membrane synthesis that are adapted for branch formation. Compared to the axon shaft, branching sites show a distinct and dynamic arrangement of cytoskeleton components, endoplasmic reticulum and mitochondria. This review discusses the regulation of axon branching in the context of cytoskeleton and membrane remodeling.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hana Nedozralova
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nirakar Basnet
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States.,National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
24
|
Sensory re-innervation of human skin by human neural stem cell-derived peripheral neurons ex vivo. J Invest Dermatol 2021; 142:257-261.e5. [PMID: 34293348 DOI: 10.1016/j.jid.2021.05.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 04/15/2021] [Accepted: 05/17/2021] [Indexed: 11/20/2022]
|
25
|
Blazejewski SM, Bennison SA, Liu X, Toyo-Oka K. High-throughput kinase inhibitor screening reveals roles for Aurora and Nuak kinases in neurite initiation and dendritic branching. Sci Rep 2021; 11:8156. [PMID: 33854138 PMCID: PMC8047044 DOI: 10.1038/s41598-021-87521-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/31/2021] [Indexed: 12/25/2022] Open
Abstract
Kinases are essential regulators of a variety of cellular signaling processes, including neurite formation—a foundational step in neurodevelopment. Aberrant axonal sprouting and failed regeneration of injured axons are associated with conditions like traumatic injury, neurodegenerative disease, and seizures. Investigating the mechanisms underlying neurite formation will allow for identification of potential therapeutics. We used a kinase inhibitor library to screen 493 kinase inhibitors and observed that 45% impacted neuritogenesis in Neuro2a (N-2a) cells. Based on the screening, we further investigated the roles of Aurora kinases A, B, and C and Nuak kinases 1 and 2. The roles of Aurora and Nuak kinases have not been thoroughly studied in the nervous system. Inhibition or overexpression of Aurora and Nuak kinases in primary cortical neurons resulted in various neuromorphological defects, with Aurora A regulating neurite initiation, Aurora B and C regulating neurite initiation and elongation, all Aurora kinases regulating arborization, and all Nuak kinases regulating neurite initiation and elongation and arborization. Our high-throughput screening and analysis of Aurora and Nuak kinases revealed their functions and may contribute to the identification of therapeutics.
Collapse
Affiliation(s)
- Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
26
|
TRPV2 interacts with actin and reorganizes submembranous actin cytoskeleton. Biosci Rep 2021; 40:226528. [PMID: 32985655 PMCID: PMC7560523 DOI: 10.1042/bsr20200118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 11/17/2022] Open
Abstract
The understanding of molecules and their role in neurite initiation and/or extension is not only helpful to prevent different neurodegenerative diseases but also can be important in neuronal damage repair. In this work, we explored the role of transient receptor potential vanilloid 2 (TRPV2), a non-selective cation channel in the context of neurite functions. We confirm that functional TRPV2 is endogenously present in F11 cell line, a model system mimicking peripheral neuron. In F11 cells, TRPV2 localizes in specific subcellular regions enriched with filamentous actin, such as in growth cone, filopodia, lamellipodia and in neurites. TRPV2 regulates actin cytoskeleton and also interacts with soluble actin. Ectopic expression of TRPV2-GFP in F11 cell induces more primary and secondary neurites, confirming its role in neurite initiation, extension and branching events. TRPV2-mediated neuritogenesis is dependent on wildtype TRPV2 as cells expressing TRPV2 mutants reveal no neuritogenesis. These findings are relevant to understand the sprouting of new neurites, neuroregeneration and neuronal plasticity at the cellular, subcellular and molecular levels. Such understanding may have further implications in neurodegeneration and peripheral neuropathy.
Collapse
|
27
|
Sisti FM, Dos Santos NAG, do Amaral L, Dos Santos AC. The Neurotrophic-Like Effect of Carvacrol: Perspective for Axonal and Synaptic Regeneration. Neurotox Res 2021; 39:886-896. [PMID: 33666886 DOI: 10.1007/s12640-021-00341-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
Carvacrol (CARV) is a phytochemical widely used as flavoring, preservative, and fragrance in food and cosmetic industries. CARV is able to cross the blood-brain barrier (BBB) and has demonstrated protective potential against neurodegenerative diseases by several mechanisms, including antioxidant, anti-inflammatory, anticholinesterase, and antiapoptotic effects. However, it is not known whether CARV is able to modulate axonal and synaptic plasticity, crucial events in cognition, memory, and learning. Abnormalities in axonal and synaptic plasticity, low levels of neurotrophins, and bioenergetic failure have been associated with the pathogenesis of neurodegenerative diseases, including Parkinson's (PD) and Alzheimer's diseases (ADs). Small lipophilic molecules with neurotrophic activity might be able to restore the axonal and synaptic networks that are lost in neurodegenerative processes. Therefore, this study investigated the neurotrophic potential of CARV in PC12 cell-based neuronal model. Carvacrol induced neurite outgrowth by activating the NGF high-affinity trkA receptor and the downstream PI3K-AKT and MAPK-ERK pathways, without depending on NGF. In addition, CARV increased the expression of proteins involved in neuronal plasticity (β-tubulin III, F-actin, 200-kDa neurofilament, GAP-43 and synapsin-I) and improved bioenergetics (AMPKα, p-AMPKα, and ATP). Our study showed, for the first time, a promising neurotrophic mechanism of CARV that could be beneficial in neurodegenerative and neurological diseases.
Collapse
Affiliation(s)
- Flávia Malvestio Sisti
- Departamento de Análises Clínicas, Toxicológicas E Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - Universidade de São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Neife Aparecida Guinaim Dos Santos
- Departamento de Análises Clínicas, Toxicológicas E Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - Universidade de São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Lilian do Amaral
- Departamento de Análises Clínicas, Toxicológicas E Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - Universidade de São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Antonio Cardozo Dos Santos
- Departamento de Análises Clínicas, Toxicológicas E Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - Universidade de São Paulo, Av do Café s/n, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
28
|
Arruda GLM, Vigerelli H, Bufalo MC, Longato GB, Veloso RV, Zambelli VO, Picolo G, Cury Y, Morandini AC, Marques AC, Sciani JM. Box Jellyfish (Cnidaria, Cubozoa) Extract Increases Neuron's Connection: A Possible Neuroprotector Effect. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8855248. [PMID: 33748281 PMCID: PMC7954621 DOI: 10.1155/2021/8855248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/14/2021] [Accepted: 02/23/2021] [Indexed: 01/29/2023]
Abstract
Neurodegenerative diseases are one of the major causes of death worldwide, characterized by neurite atrophy, neuron apoptosis, and synapse loss. No effective treatment has been indicated for such diseases so far, and the search for new drugs is being increased in the last years. Animal venoms' secretion/venom can be an alternative for the discovery of new molecules, which could be the prototype for a new treatment. Here, we present the biochemical characterization and activity of the extract from the box jellyfish Chiropsalmus quadrumanus (Cq) on neurites. The Cq methanolic extract was obtained and incubated to human SH-SY5Y neurons, and neurite parameters were evaluated. The extract was tested in other cell types to check its cytotoxicity and was submitted to biochemical analysis by mass spectrometry in order to check its composition. We could verify that the Cq extract increased neurite outgrowth length and branching junctions, amplifying the contact between SH-SY5Y neurons, without affecting cell body and viability. The extract action was selective for neurons, as it did not cause any effects on other cell types, such as tumor line, nontumor line, and red blood cells. Moreover, mass spectrometry analysis revealed that there are no proteins but several low molecular mass compounds and peptides. Three peptides, characterized as cryptides, and 14 low molecular mass compounds were found to be related to cytoskeleton reorganization, cell membrane expansion, and antioxidant/neuroprotective activity, which act together to increase neuritogenesis. After this evaluation, we conclude that the Cq extract is a promising tool for neuronal connection recovery, an essential condition for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Gian Lucas M. Arruda
- Laboratório Multidisciplinar de Pesquisa, Universidade São Francisco, Bragança Paulista 12916-900, Brazil
| | - Hugo Vigerelli
- Laboratório de Genética, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Michelle C. Bufalo
- Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Giovanna B. Longato
- Laboratório de Pesquisa em Farmacologia Molecular e Compostos Bioativos, Universidade São Francisco, Bragança Paulista 12916-900, Brazil
| | - Rodinei V. Veloso
- Laboratório Multidisciplinar de Pesquisa, Universidade São Francisco, Bragança Paulista 12916-900, Brazil
| | - Vanessa O. Zambelli
- Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Gisele Picolo
- Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Yara Cury
- Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo 05503-900, Brazil
| | - André C. Morandini
- Departamento de Zoologia, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
- Centro de Biologia Marinha, Universidade de São Paulo, São Sebastião 11612-109, Brazil
| | - Antonio Carlos Marques
- Departamento de Zoologia, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Juliana Mozer Sciani
- Laboratório Multidisciplinar de Pesquisa, Universidade São Francisco, Bragança Paulista 12916-900, Brazil
| |
Collapse
|
29
|
Lepa C, Hoppe S, Stöber A, Skryabin BV, Sievers LK, Heitplatz B, Ciarimboli G, Neugebauer U, Lindenmeyer MT, Cohen CD, Drexler HC, Boor P, Weide T, Pavenstädt H, George B. TrkC Is Essential for Nephron Function and Trans-Activates Igf1R Signaling. J Am Soc Nephrol 2021; 32:357-374. [PMID: 33380522 PMCID: PMC8054883 DOI: 10.1681/asn.2020040424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 11/03/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Injury to kidney podocytes often results in chronic glomerular disease and consecutive nephron malfunction. For most glomerular diseases, targeted therapies are lacking. Thus, it is important to identify novel signaling pathways contributing to glomerular disease. Neurotrophic tyrosine kinase receptor 3 (TrkC) is expressed in podocytes and the protein transmits signals to the podocyte actin cytoskeleton. METHODS Nephron-specific TrkC knockout (TrkC-KO) and nephron-specific TrkC-overexpressing (TrkC-OE) mice were generated to dissect the role of TrkC in nephron development and maintenance. RESULTS Both TrkC-KO and TrkC-OE mice exhibited enlarged glomeruli, mesangial proliferation, basement membrane thickening, albuminuria, podocyte loss, and aspects of FSGS during aging. Igf1 receptor (Igf1R)-associated gene expression was dysregulated in TrkC-KO mouse glomeruli. Phosphoproteins associated with insulin, erb-b2 receptor tyrosine kinase (Erbb), and Toll-like receptor signaling were enriched in lysates of podocytes treated with the TrkC ligand neurotrophin-3 (Nt-3). Activation of TrkC by Nt-3 resulted in phosphorylation of the Igf1R on activating tyrosine residues in podocytes. Igf1R phosphorylation was increased in TrkC-OE mouse kidneys while it was decreased in TrkC-KO kidneys. Furthermore, TrkC expression was elevated in glomerular tissue of patients with diabetic kidney disease compared with control glomerular tissue. CONCLUSIONS Our results show that TrkC is essential for maintaining glomerular integrity. Furthermore, TrkC modulates Igf-related signaling in podocytes.
Collapse
Affiliation(s)
- Carolin Lepa
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Sascha Hoppe
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Antje Stöber
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Boris V. Skryabin
- Medical Faculty, Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), Westfälische-Wilhelms-University, Münster, Germany
| | | | - Barbara Heitplatz
- Gerhard-Domagk Institute for Pathology, University Hospital Münster, Münster, Germany
| | | | - Ute Neugebauer
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Maja T. Lindenmeyer
- III. Medizinische Klinik und Poliklinik, University Hospital Hamburg-Eppendorf, Germany
| | - Clemens D. Cohen
- Klinik für Nieren-, Hochdruck- und Rheumaerkrankungen, München Klinik Harlaching, Germany
| | - Hannes C.A. Drexler
- Mass Spectrometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Thomas Weide
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | | | - Britta George
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| |
Collapse
|
30
|
Arioka Y, Shishido E, Kushima I, Suzuki T, Saito R, Aiba A, Mori D, Ozaki N. Chromosome 22q11.2 deletion causes PERK-dependent vulnerability in dopaminergic neurons. EBioMedicine 2020; 63:103138. [PMID: 33341442 PMCID: PMC7753137 DOI: 10.1016/j.ebiom.2020.103138] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/23/2020] [Accepted: 11/09/2020] [Indexed: 12/27/2022] Open
Abstract
Background The chromosome 22q11.2 deletion is an extremely high risk genetic factor for various neuropsychiatric disorders; however, the 22q11.2 deletion-related brain pathology in humans at the cellular and molecular levels remains unclear. Methods We generated iPS cells from healthy controls (control group) and patients with 22q11.2 deletion (22DS group), and differentiated them into dopaminergic neurons. Semiquantitative proteomic analysis was performed to compare the two groups. Next, we conducted molecular, cell biological and pharmacological assays. Findings Semiquantitative proteomic analysis identified ‘protein processing in the endoplasmic reticulum (ER)’ as the most altered pathway in the 22DS group. In particular, we found a severe defect in protein kinase R-like endoplasmic reticulum kinase (PERK) expression and its activity in the 22DS group. The decreased PERK expression was also shown in the midbrain of a 22q11.2 deletion mouse model. The 22DS group showed characteristic phenotypes, including poor tolerance to ER stress, abnormal F-actin dynamics, and decrease in protein synthesis. Some of phenotypes were rescued by the pharmacological manipulation of PERK activity and phenocopied in PERK-deficient dopaminergic neurons. We lastly showed that DGCR14 was associated with reduction in PERK expression. Interpretation Our findings led us to conclude that the 22q11.2 deletion causes various vulnerabilities in dopaminergic neurons, dependent on PERK dysfunction. Funding This study was supported by the 10.13039/100010463AMED under grant nos JP20dm0107087, JP20dm0207075, JP20ak0101113, JP20dk0307081, and JP18dm0207004h0005; the MEXT KAKENHI under grant nos. 16K19760, 19K08015, 18H04040, and 18K19511; the 10.13039/100008732Uehara Memorial Foundation under grant no. 201810122; and 2019 iPS Academia Japan Grant.
Collapse
Affiliation(s)
- Yuko Arioka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan; Institute for Advanced Research, Nagoya University, Nagoya, Japan.
| | - Emiko Shishido
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; National Institute for Physiological Sciences, Okazaki, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan
| | - Toshiaki Suzuki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryo Saito
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsu Aiba
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan; Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan; Brain and Mind Research Center, Nagoya University, Nagoya, Japan.
| |
Collapse
|
31
|
Li M, Knapp SK, Iden S. Mechanisms of melanocyte polarity and differentiation: What can we learn from other neuroectoderm-derived lineages? Curr Opin Cell Biol 2020; 67:99-108. [PMID: 33099084 DOI: 10.1016/j.ceb.2020.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 01/16/2023]
Abstract
Melanocytes are neuroectoderm-derived pigment-producing cells with highly polarized dendritic morphology. They protect the skin against ultraviolet radiation by providing melanin to neighbouring keratinocytes. However, the mechanisms underlying melanocyte polarization and its relevance for diseases remain mostly elusive. Numerous studies have instead revealed roles for polarity regulators in other neuroectoderm-derived lineages including different neuronal cell types. Considering the shared ontogeny and morphological similarities, these lineages may be used as reference models for the exploration of melanocyte polarity, for example, regarding dendrite formation, spine morphogenesis and polarized organelle transport. In this review, we summarize and compare the latest progress in understanding polarity regulation in neuronal cells and melanocytes and project key open questions for future work.
Collapse
Affiliation(s)
- Mengnan Li
- Cell and Developmental Biology, Center for Human and Molecular Biology (ZHMB), Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| | - Sina K Knapp
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Germany
| | - Sandra Iden
- Cell and Developmental Biology, Center for Human and Molecular Biology (ZHMB), Saarland University, Faculty of Medicine, Homburg/Saar, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany.
| |
Collapse
|
32
|
Lyons EL, Leone-Kabler S, Kovach AL, Thomas BF, Howlett AC. Cannabinoid receptor subtype influence on neuritogenesis in human SH-SY5Y cells. Mol Cell Neurosci 2020; 109:103566. [PMID: 33049367 DOI: 10.1016/j.mcn.2020.103566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 10/23/2022] Open
Abstract
Human SH-SY5Y neuroblastoma cells stably expressing exogenous CB1 (CB1XS) or CB2 (CB2XS) receptors were developed to investigate endocannabinoid signaling in the extension of neuronal projections. Expression of cannabinoid receptors did not alter proliferation rate, viability, or apoptosis relative to parental SH-SY5Y. Transcripts for endogenous cannabinoid system enzymes (diacylglycerol lipase, monoacylglycerol lipase, α/β-hydrolase domain containing proteins 6 and 12, N-acyl phosphatidylethanolamine-phospholipase D, and fatty acid amide hydrolase) were not altered by CB1 or CB2 expression. Endocannabinoid ligands 2-arachidonoylglycerol (2-AG) and anandamide were quantitated in SH-SY5Y cells, and diacylglycerol lipase inhibitor tetrahydrolipstatin decreased 2-AG abundance by 90% but did not alter anandamide abundance. M3 muscarinic agonist oxotremorine M, and inhibitors of monoacylglycerol lipase and α/β hydrolase domain containing proteins 6 &12 increased 2-AG abundance. CB1 receptor expression increased lengths of short (<30 μm) and long (>30 μm) projections, and this effect was significantly reduced by tetrahydrolipstatin, indicative of stimulation by endogenously produced 2-AG. Pertussis toxin, Gβγ inhibitor gallein, and β-arrestin inhibitor barbadin did not significantly alter long projection length in CB1XS, but significantly reduced short projections, with gallein having the greatest inhibition. The rho kinase inhibitor Y27632 increased CB1 receptor-mediated long projection extension, indicative of actin cytoskeleton involvement. CB1 receptor expression increased GAP43 and ST8SIA2 mRNA and decreased ITGA1 mRNA, whereas CB2 receptor expression increased NCAM and SYT mRNA. We propose that basal endogenous production of 2-AG provides autocrine stimulation of CB1 receptor signaling through Gi/o, Gβγ, and β-arrestin mechanisms to promote neuritogenesis, and rho kinase influences process extension.
Collapse
Affiliation(s)
- Erica L Lyons
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, One Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | - Sandra Leone-Kabler
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, One Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | - Alexander L Kovach
- Discovery Sciences, RTI International, PO Box 12194, Research Triangle Park, NC 27709, USA.
| | - Brian F Thomas
- Discovery Sciences, RTI International, PO Box 12194, Research Triangle Park, NC 27709, USA.
| | - Allyn C Howlett
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, One Medical Center Blvd., Winston-Salem, NC 27157, USA.
| |
Collapse
|
33
|
Vasudevan A, Koushika SP. Molecular mechanisms governing axonal transport: a C. elegans perspective. J Neurogenet 2020; 34:282-297. [PMID: 33030066 DOI: 10.1080/01677063.2020.1823385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Axonal transport is integral for maintaining neuronal form and function, and defects in axonal transport have been correlated with several neurological diseases, making it a subject of extensive research over the past several years. The anterograde and retrograde transport machineries are crucial for the delivery and distribution of several cytoskeletal elements, growth factors, organelles and other synaptic cargo. Molecular motors and the neuronal cytoskeleton function as effectors for multiple neuronal processes such as axon outgrowth and synapse formation. This review examines the molecular mechanisms governing axonal transport, specifically highlighting the contribution of studies conducted in C. elegans, which has proved to be a tractable model system in which to identify both novel and conserved regulatory mechanisms of axonal transport.
Collapse
Affiliation(s)
- Amruta Vasudevan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
34
|
Wen W, Wang Y, Li H, Xu H, Xu M, Frank JA, Ma M, Luo J. Mesencephalic Astrocyte-Derived Neurotrophic Factor (MANF) Regulates Neurite Outgrowth Through the Activation of Akt/mTOR and Erk/mTOR Signaling Pathways. Front Mol Neurosci 2020; 13:560020. [PMID: 33071755 PMCID: PMC7541815 DOI: 10.3389/fnmol.2020.560020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Neurite outgrowth is essential for brain development and the recovery of brain injury and neurodegenerative diseases. In this study, we examined the role of the neurotrophic factor MANF in regulating neurite outgrowth. We generated MANF knockout (KO) neuro2a (N2a) cell lines using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 and demonstrated that MANF KO N2a cells failed to grow neurites in response to RA stimulation. Using MANF siRNA, this finding was confirmed in human SH-SY5Y neuronal cell line. Nevertheless, MANF overexpression by adenovirus transduction or addition of MANF into culture media facilitated the growth of longer neurites in RA-treated N2a cells. MANF deficiency resulted in inhibition of Akt, Erk, mTOR, and P70S6, and impaired protein synthesis. MANF overexpression on the other hand facilitated the growth of longer neurites by activating Akt, Erk, mTOR, and P70S6. Pharmacological blockade of Akt, Erk or mTOR eliminated the promoting effect of MANF on neurite outgrowth. These findings suggest that MANF positively regulated neurite outgrowth by activating Akt/mTOR and Erk/mTOR signaling pathways.
Collapse
Affiliation(s)
- Wen Wen
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Yongchao Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Hui Li
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Hong Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jacqueline A Frank
- Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Murong Ma
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jia Luo
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
35
|
Nalinratana N, Meksuriyen D, Ongpipattanakul B. Asiaticoside but not its aglycone exhibits neuritogenicity through TrkA receptor signaling: a bridge between ERK1/2-CREB and Akt-GSK3β/RhoA. Neuroreport 2020; 30:1261-1270. [PMID: 31651704 DOI: 10.1097/wnr.0000000000001352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The neuritogenicity and the neuroregenerative potential of asiaticoside (AS) and its aglycone, asiatic acid (AA), has been generally reported. We recently identified the participation of extracellular signal-regulated protein kinases 1/2 (ERK1/2) and protein kinase B (Akt) in the neuritogenic mechanism of AS and AA. In this study, we further investigated the possible upstream target molecule and the associated downstream signaling of both triterpenoids in mouse neuroblastoma Neuro-2a cells. Our immunoblotting and immunofluorescence assays revealed that either AS or AA exerted neurite extension activity through inhibitory effect on glycogen synthase kinase 3β (GSK3β) and Ras homolog gene family member A (RhoA). AS appeared significantly more potent in promoting neurite elongation than AA, and concurrently expressed a higher degree of inhibition on GSK3β and RhoA activations. The mediation of GSK3β and RhoA activities in AS-treated cells involved Akt signaling. Moreover, when using GW441756, a specific tropomyosin receptor kinase A (TrkA) receptor signaling inhibitor, the ERK1/2 and Akt phosphorylation, the inhibitory effects on GSK3β and RhoA and the neurite outgrowth induced by AS, but not AA, were totally suppressed. In conclusion, our findings supported the different upstream regulators of AS and AA in promoting neuritogenicity in Neuro-2a cells. Although both AS and AA could enhance neurite elongation through the suppression of GSK3β and RhoA activities, only AS could modulate the effect through TrkA receptor signaling.
Collapse
Affiliation(s)
- Nonthaneth Nalinratana
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok
| | - Duangdeun Meksuriyen
- Drug and Health Product Research and Development Center, College of Pharmacy, Rangsit University, Pathum Thani
| | - Boonsri Ongpipattanakul
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok.,Chulalongkorn University Drugs and Health Products Innovation and Promotion Center, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
36
|
Theis T, Kumar S, Wei E, Nguyen J, Glynos V, Paranjape N, Askarifirouzjaei H, Khajouienejad L, Berthiaume F, Young W, Schachner M. Myristoylated alanine-rich C-kinase substrate effector domain peptide improves sex-specific recovery and axonal regrowth after spinal cord injury. FASEB J 2020; 34:12677-12690. [PMID: 32729988 DOI: 10.1096/fj.202000026rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 11/11/2022]
Abstract
Myristoylated alanine-rich C-kinase substrate (MARCKS) is an intracellular receptor for polysialic acid. MARCKS supports development, synaptic plasticity, and regeneration after injury. MARCKS binds with its functionally essential effector domain (ED) to polysialic acid. A 25-mer peptide comprising the ED of MARCKS stimulates neuritogenesis of primary hippocampal neurons after addition to the culture. This motivated us to investigate whether ED peptide has similar effects in spinal cord injury. ED peptide supported recovery and regrowth of monoaminergic axons in female, but not in male mice. Sex-specific differences in response to ED peptide application also occurred in cultured neurons. In female but not male neurons, the ED peptide enhanced neurite outgrowth that could be suppressed by inhibitors of the estrogen receptors α and β, fibroblast growth factor receptor-1, protein kinase C, and matrix metalloproteinase 2. In addition, we observed female-specific elevation of phosphorylated MARCKS levels after ED peptide treatment. In male neurons, the ED peptide enhanced neuritogenesis in the presence of an androgen receptor inhibitor to the extent seen in ED peptide-treated female neurons. However, inhibition of androgen receptor did not lead to increased phosphorylation of MARCKS. These results provide insights into the functions of a novel compound contributing to gender-dependent regeneration.
Collapse
Affiliation(s)
- Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Elena Wei
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Jennifer Nguyen
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Vicci Glynos
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Nikita Paranjape
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Hadi Askarifirouzjaei
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Leila Khajouienejad
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Wise Young
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
37
|
Srivastava AK, Yadav SS, Mishra S, Yadav SK, Parmar D, Yadav S. A combined microRNA and proteome profiling to investigate the effect of ZnO nanoparticles on neuronal cells. Nanotoxicology 2020; 14:757-773. [DOI: 10.1080/17435390.2020.1759726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Ankur Kumar Srivastava
- Developmental Toxicology Laboratory, Systems Toxicology, and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Smriti Singh Yadav
- Developmental Toxicology Laboratory, Systems Toxicology, and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India
| | - Saumya Mishra
- Developmental Toxicology Laboratory, Systems Toxicology, and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanjeev Kumar Yadav
- Developmental Toxicology Laboratory, Systems Toxicology, and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Devendra Parmar
- Developmental Toxicology Laboratory, Systems Toxicology, and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanjay Yadav
- Developmental Toxicology Laboratory, Systems Toxicology, and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- All India Institute of Medical Sciences (AIIMS), Raebareli, Uttar Pradesh, India
| |
Collapse
|
38
|
Burger CA, Alevy J, Casasent AK, Jiang D, Albrecht NE, Liang JH, Hirano AA, Brecha NC, Samuel MA. LKB1 coordinates neurite remodeling to drive synapse layer emergence in the outer retina. eLife 2020; 9:e56931. [PMID: 32378514 PMCID: PMC7237215 DOI: 10.7554/elife.56931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 04/11/2020] [Indexed: 12/04/2022] Open
Abstract
Structural changes in pre and postsynaptic neurons that accompany synapse formation often temporally and spatially overlap. Thus, it has been difficult to resolve which processes drive patterned connectivity. To overcome this, we use the laminated outer murine retina. We identify the serine/threonine kinase LKB1 as a key driver of synapse layer emergence. The absence of LKB1 in the retina caused a marked mislocalization and delay in synapse layer formation. In parallel, LKB1 modulated postsynaptic horizontal cell refinement and presynaptic photoreceptor axon growth. Mislocalized horizontal cell processes contacted aberrant cone axons in LKB1 mutants. These defects coincided with altered synapse protein organization, and horizontal cell neurites were misdirected to ectopic synapse protein regions. Together, these data suggest that LKB1 instructs the timing and location of connectivity in the outer retina via coordinate regulation of pre and postsynaptic neuron structure and the localization of synapse-associated proteins.
Collapse
Affiliation(s)
- Courtney A Burger
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Jonathan Alevy
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Anna K Casasent
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Danye Jiang
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Nicholas E Albrecht
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Justine H Liang
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Arlene A Hirano
- Department of Neurobiology, David Geffen School of Medicine at UCLALos AngelesUnited States
- United States Veterans Administration Greater Los Angeles Healthcare SystemLos AngelesUnited States
| | - Nicholas C Brecha
- Department of Neurobiology, David Geffen School of Medicine at UCLALos AngelesUnited States
- United States Veterans Administration Greater Los Angeles Healthcare SystemLos AngelesUnited States
| | - Melanie A Samuel
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
39
|
Omelchenko A, Menon H, Donofrio SG, Kumar G, Chapman HM, Roshal J, Martinez-Montes ER, Wang TL, Spaller MR, Firestein BL. Interaction Between CRIPT and PSD-95 Is Required for Proper Dendritic Arborization in Hippocampal Neurons. Mol Neurobiol 2020; 57:2479-2493. [PMID: 32157575 PMCID: PMC7176523 DOI: 10.1007/s12035-020-01895-5] [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: 10/09/2019] [Accepted: 02/24/2020] [Indexed: 01/10/2023]
Abstract
CRIPT, the cysteine-rich PDZ-binding protein, binds to the third PDZ domain of PSD-95 (postsynaptic density protein 95) family proteins and directly binds microtubules, linking PSD-95 family proteins to the neuronal cytoskeleton. Here, we show that overexpression of a full-length CRIPT leads to a modest decrease, and knockdown of CRIPT leads to an increase in dendritic branching in cultured rat hippocampal neurons. Overexpression of truncated CRIPT lacking the PDZ domain-binding motif, which does not bind to PSD-95, significantly decreases dendritic arborization. Conversely, overexpression of a full-length CRIPT significantly increases the number of immature and mature dendritic spines, and this effect is not observed when CRIPT∆PDZ is overexpressed. Competitive inhibition of CRIPT binding to the third PDZ domain of PSD-95 with PDZ3-binding peptides resulted in differential effects on dendritic arborization based on the origin of respective peptide sequence. These results highlight multifunctional roles of CRIPT during development and underscore the significance of the interaction between CRIPT and the third PDZ domain of PSD-95.
Collapse
Affiliation(s)
- Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
- Neuroscience Graduate Program, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Harita Menon
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Sarah G Donofrio
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Gaurav Kumar
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Heidi M Chapman
- Geisel School of Medicine, Department of Medical Education and Norris Cotton Cancer Center, Dartmouth College, Lebanon, NH, 03756, USA
| | - Joshua Roshal
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Eduardo R Martinez-Montes
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Tiffany L Wang
- Geisel School of Medicine, Department of Medical Education and Norris Cotton Cancer Center, Dartmouth College, Lebanon, NH, 03756, USA
| | - Mark R Spaller
- Geisel School of Medicine, Department of Medical Education and Norris Cotton Cancer Center, Dartmouth College, Lebanon, NH, 03756, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA.
| |
Collapse
|
40
|
Genetic predispositions of Parkinson's disease revealed in patient-derived brain cells. NPJ PARKINSONS DISEASE 2020; 6:8. [PMID: 32352027 PMCID: PMC7181694 DOI: 10.1038/s41531-020-0110-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent neurological disorder and has been the focus of intense investigations to understand its etiology and progression, but it still lacks a cure. Modeling diseases of the central nervous system in vitro with human induced pluripotent stem cells (hiPSC) is still in its infancy but has the potential to expedite the discovery and validation of new treatments. Here, we discuss the interplay between genetic predispositions and midbrain neuronal impairments in people living with PD. We first summarize the prevalence of causal Parkinson's genes and risk factors reported in 74 epidemiological and genomic studies. We then present a meta-analysis of 385 hiPSC-derived neuronal lines from 67 recent independent original research articles, which point towards specific impairments in neurons from Parkinson's patients, within the context of genetic predispositions. Despite the heterogeneous nature of the disease, current iPSC models reveal converging molecular pathways underlying neurodegeneration in a range of familial and sporadic forms of Parkinson's disease. Altogether, consolidating our understanding of robust cellular phenotypes across genetic cohorts of Parkinson's patients may guide future personalized drug screens in preclinical research.
Collapse
|
41
|
Bennison SA, Blazejewski SM, Smith TH, Toyo-Oka K. Protein kinases: master regulators of neuritogenesis and therapeutic targets for axon regeneration. Cell Mol Life Sci 2020; 77:1511-1530. [PMID: 31659414 PMCID: PMC7166181 DOI: 10.1007/s00018-019-03336-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/16/2019] [Accepted: 10/08/2019] [Indexed: 12/25/2022]
Abstract
Proper neurite formation is essential for appropriate neuronal morphology to develop and defects at this early foundational stage have serious implications for overall neuronal function. Neuritogenesis is tightly regulated by various signaling mechanisms that control the timing and placement of neurite initiation, as well as the various processes necessary for neurite elongation to occur. Kinases are integral components of these regulatory pathways that control the activation and inactivation of their targets. This review provides a comprehensive summary of the kinases that are notably involved in regulating neurite formation, which is a complex process that involves cytoskeletal rearrangements, addition of plasma membrane to increase neuronal surface area, coupling of cytoskeleton/plasma membrane, metabolic regulation, and regulation of neuronal differentiation. Since kinases are key regulators of these functions during neuromorphogenesis, they have high potential for use as therapeutic targets for axon regeneration after injury or disease where neurite formation is disrupted.
Collapse
Affiliation(s)
- Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Trevor H Smith
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
42
|
Wlodarczyk-Li SA, Vassoler FM, Byrnes EM, Schonhoff CM. Oxycodone Decreases Dendritic Complexity in Female but not Male Rat Striatal Neurons In Vitro. Neurosci Lett 2020; 722:134856. [PMID: 32088199 DOI: 10.1016/j.neulet.2020.134856] [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: 12/16/2019] [Revised: 02/14/2020] [Accepted: 02/19/2020] [Indexed: 10/25/2022]
Abstract
The use of oxycodone in the past two decades has dramatically risen, yet the amount of research regarding how it impacts neuronal health is lacking. As prescription use and misuse in women of reproductive age increases there has been a corresponding increase in the number of infants who have been exposed to oxycodone in utero. Given the critical role of the striatum in motor control and reward regulation, the aim of the current study was to examine the effects of oxycodone on developing rat striatal neurons. Sex-specific effects of oxycodone on neuronal cytoarchitecture were examined in cultured rat striatal neurons with a primary focus on dendritic arborization. Neurons were extracted from either male or female embryonic day 18 rat striata and cultured and exposed to varying concentrations of oxycodone over a ten-day period. Dendritic complexity of the neurons was measured using Sholl analysis. Results indicate that oxycodone inhibits dendritic complexity in a dose-dependent manner in female but not male striatal neurons. Additional analysis indicated the number of non-primary dendrites in female striatal neurons significantly decreased with increasing concentrations of oxycodone, while the number of primary dendrites as well as the length of primary and non-primary dendrites was unaffected by oxycodone treatment in both sexes. These in vitro findings demonstrate sex-specific effects of oxycodone on the development of striatal dendritic architecture which may be important for understanding the effects of oxycodone exposure in utero.
Collapse
Affiliation(s)
- Sara A Wlodarczyk-Li
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, 01536, United States
| | - Fair M Vassoler
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, 01536, United States
| | - Elizabeth M Byrnes
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, 01536, United States
| | - Christopher M Schonhoff
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, 01536, United States.
| |
Collapse
|
43
|
Regulation of dopaminergic neuronal phenotypes by the estrogen-related receptor gamma ligand GSK4716 via the activation of CREB signaling. Neurochem Int 2020; 136:104726. [PMID: 32173553 DOI: 10.1016/j.neuint.2020.104726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 02/13/2020] [Accepted: 03/10/2020] [Indexed: 01/09/2023]
Abstract
Midbrain dopaminergic (DAergic) neurotransmission plays a crucial role in regulating motor, cognitive, and emotional functions. The orphan nuclear receptor estrogen-related receptor gamma (ERRγ) is highly expressed in the adult brain and in the developing fetal brain. Our previous study showed the relevance of ERRγ in the regulation of the DAergic neuronal phenotype with the upregulation of dopamine synthesizing tyrosine hydroxylase (TH) and dopamine transporter (DAT) and the possibility that ERRγ could be a novel target for regulating DAergic neuronal differentiation. In this study, we examined whether ERRγ ligands could be small molecule regulators of DAergic phenotypes. The ERRγ agonist GSK4716 increased DAT and TH expression, and the ERRγ inverse agonist GSK5182 attenuated the retinoic acid-induced upregulation of DAT and TH in differentiated SH-SY5Y cells. We found that biphasic activation of the protein kinase A/cyclic AMP response element-binding (CREB) protein signaling pathway was involved in the GSK4716-induced increase in the DAergic phenotype in SH-SY5Y cells. CREB signaling activated as early as 3 h after GSK4716 treatment in an ERRγ-independent manner, but increased following ERRγ activation after 3 days. Protein kinase A inhibitor H-89 attenuated GSK4716-induced DAT and TH upregulation. In primary cultured DAergic neurons, GSK4716 increased neurite length and the number of DAT and TH-double-positive (DAT + TH+) neurons compared to that in control cells. These findings suggest that ERRγ ligands could serve as useful chemical tools for obtaining a better understanding of the regulation of DAergic phenotypes and might facilitate the development of small molecule therapeutics to treat DA-related neurological diseases.
Collapse
|
44
|
Swenarchuk LE. Nerve, Muscle, and Synaptogenesis. Cells 2019; 8:cells8111448. [PMID: 31744142 PMCID: PMC6912269 DOI: 10.3390/cells8111448] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/06/2019] [Accepted: 11/06/2019] [Indexed: 12/21/2022] Open
Abstract
The vertebrate skeletal neuromuscular junction (NMJ) has long served as a model system for studying synapse structure, function, and development. Over the last several decades, a neuron-specific isoform of agrin, a heparan sulfate proteoglycan, has been identified as playing a central role in synapse formation at all vertebrate skeletal neuromuscular synapses. While agrin was initially postulated to be the inductive molecule that initiates synaptogenesis, this model has been modified in response to work showing that postsynaptic differentiation can develop in the absence of innervation, and that synapses can form in transgenic mice in which the agrin gene is ablated. In place of a unitary mechanism for neuromuscular synapse formation, studies in both mice and zebrafish have led to the proposal that two mechanisms mediate synaptogenesis, with some synapses being induced by nerve contact while others involve the incorporation of prepatterned postsynaptic structures. Moreover, the current model also proposes that agrin can serve two functions, to induce synaptogenesis and to stabilize new synapses, once these are formed. This review examines the evidence for these propositions, and concludes that it remains possible that a single molecular mechanism mediates synaptogenesis at all NMJs, and that agrin acts as a stabilizer, while its role as inducer is open to question. Moreover, if agrin does not act to initiate synaptogenesis, it follows that as yet uncharacterized molecular interactions are required to play this essential inductive role. Several alternatives to agrin for this function are suggested, including focal pericellular proteolysis and integrin signaling, but all require experimental validation.
Collapse
|
45
|
A Synthetic Snake-Venom-Based Tripeptide Protects PC12 Cells from the Neurotoxicity of Acrolein by Improving Axonal Plasticity and Bioenergetics. Neurotox Res 2019; 37:227-237. [DOI: 10.1007/s12640-019-00111-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
|
46
|
Yang G, Shcheglovitov A. Probing disrupted neurodevelopment in autism using human stem cell-derived neurons and organoids: An outlook into future diagnostics and drug development. Dev Dyn 2019; 249:6-33. [PMID: 31398277 DOI: 10.1002/dvdy.100] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorders (ASDs) represent a spectrum of neurodevelopmental disorders characterized by impaired social interaction, repetitive or restrictive behaviors, and problems with speech. According to a recent report by the Centers for Disease Control and Prevention, one in 68 children in the US is diagnosed with ASDs. Although ASD-related diagnostics and the knowledge of ASD-associated genetic abnormalities have improved in recent years, our understanding of the cellular and molecular pathways disrupted in ASD remains very limited. As a result, no specific therapies or medications are available for individuals with ASDs. In this review, we describe the neurodevelopmental processes that are likely affected in the brains of individuals with ASDs and discuss how patient-specific stem cell-derived neurons and organoids can be used for investigating these processes at the cellular and molecular levels. Finally, we propose a discovery pipeline to be used in the future for identifying the cellular and molecular deficits and developing novel personalized therapies for individuals with idiopathic ASDs.
Collapse
Affiliation(s)
- Guang Yang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah.,Neuroscience Graduate Program, University of Utah, Salt Lake City, Utah
| | - Alex Shcheglovitov
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah.,Neuroscience Graduate Program, University of Utah, Salt Lake City, Utah
| |
Collapse
|
47
|
Taylor KL, Taylor RJ, Richters KE, Huynh B, Carrington J, McDermott ME, Wilson RL, Dent EW. Opposing functions of F-BAR proteins in neuronal membrane protrusion, tubule formation, and neurite outgrowth. Life Sci Alliance 2019; 2:2/3/e201800288. [PMID: 31160379 PMCID: PMC6549137 DOI: 10.26508/lsa.201800288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 01/08/2023] Open
Abstract
Neurite formation is a fundamental antecedent to axon and dendrite formation, but the mechanisms that underlie this important process are poorly characterized. Here, we demonstrate that two F-BAR proteins, CIP4 and FBP17, have opposing functions in early cortical neuron development. The F-BAR family of proteins play important roles in many cellular processes by regulating both membrane and actin dynamics. The CIP4 family of F-BAR proteins is widely recognized to function in endocytosis by elongating endocytosing vesicles. However, in primary cortical neurons, CIP4 concentrates at the tips of extending lamellipodia and filopodia and inhibits neurite outgrowth. Here, we report that the highly homologous CIP4 family member, FBP17, induces tubular structures in primary cortical neurons and results in precocious neurite formation. Through domain swapping and deletion experiments, we demonstrate that a novel polybasic region between the F-BAR and HR1 domains is required for membrane bending. Moreover, the presence of a poly-PxxP region in longer splice isoforms of CIP4 and FBP17 largely reverses the localization and function of these proteins. Thus, CIP4 and FBP17 function as an antagonistic pair to fine-tune membrane protrusion, endocytosis, and neurite formation during early neuronal development.
Collapse
Affiliation(s)
- Kendra L Taylor
- University of Wisconsin-Madison, Neuroscience Training Program, Madison, WI, USA
| | - Russell J Taylor
- University of Wisconsin-Madison, Neuroscience Training Program, Madison, WI, USA
| | - Karl E Richters
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Brandon Huynh
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Justin Carrington
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Maeve E McDermott
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Rebecca L Wilson
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Erik W Dent
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| |
Collapse
|
48
|
Ferreira RS, Dos Santos NAG, Bernardes CP, Sisti FM, Amaral L, Fontana ACK, Dos Santos AC. Caffeic Acid Phenethyl Ester (CAPE) Protects PC12 Cells Against Cisplatin-Induced Neurotoxicity by Activating the AMPK/SIRT1, MAPK/Erk, and PI3k/Akt Signaling Pathways. Neurotox Res 2019; 36:175-192. [PMID: 31016689 DOI: 10.1007/s12640-019-00042-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 01/01/2023]
Abstract
Peripheral sensory neuropathy (PSN) is a well-known side effect of cisplatin characterized by axonal damage. In the early stage of neurotoxicity, cisplatin affects proteins that modulate neurite outgrowth and neuroplasticity, without inducing mitochondrial damage or apoptosis. There are no preventive therapies for cisplatin-induced peripheral neuropathy; therefore, measures to improve axonal growth and connectivity would be beneficial. Caffeic acid phenethyl ester (CAPE) is a bioactive component of propolis with neurotrophic and neuroprotective activities. We have recently showed that CAPE protects against cisplatin-induced neurotoxicity by activating NGF high-affinity receptors (trkA) and inducing neuroplasticity. We have now assessed other potential early targets of cisplatin and additional mechanisms involved in the neuroprotection of CAPE. Cisplatin reduced axonal cytoskeletal proteins (F-actin and β-III-tubulin) without inducing oxidative damage in PC12 cells. It also reduced energy-related proteins (AMPK α, p-AMPK α, and SIRT1) and glucose uptake. At this stage of neurotoxicity, glutamate excitotoxicity is not involved in the toxicity of cisplatin. CAPE attenuated the downregulation of the cytoskeleton and energy-related markers as well as SIRT1 and phosphorylated AMPK α. Moreover, the neuroprotective mechanism of CAPE also involves the activation of the neurotrophic signaling pathways MAPK/Erk and PI3k/Akt. The PI3K/Akt pathway is involved in the upregulation of SIRT1 induced by CAPE, but not in the upregulation of cytoskeletal proteins. Altogether, these findings suggest that the neuroprotective effect of CAPE against cisplatin-induced neurotoxicity involves both (a) a neurotrophic mechanism that mimics the mechanism triggered by the NGF itself and (b) a non-neurotrophic mechanism that upregulates the cytoskeletal proteins.
Collapse
Affiliation(s)
- Rafaela Scalco Ferreira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Neife Aparecida Guinaim Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Carolina P Bernardes
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Flávia Malvestio Sisti
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Lilian Amaral
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Andreia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Antonio Cardozo Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
49
|
Pergu R, Dagar S, Kumar H, Kumar R, Bhattacharya J, Mylavarapu SVS. The chaperone ERp29 is required for tunneling nanotube formation by stabilizing MSec. J Biol Chem 2019; 294:7177-7193. [PMID: 30877198 DOI: 10.1074/jbc.ra118.005659] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/14/2019] [Indexed: 01/23/2023] Open
Abstract
Tunneling nanotubes (TNTs) are membrane conduits that mediate long-distance intercellular cross-talk in several organisms and play vital roles during development, pathogenic transmission, and cancer metastasis. However, the molecular mechanisms of TNT formation and function remain poorly understood. The protein MSec (also known as TNFα-induced protein 2 (TNFAIP2) and B94) is essential for TNT formation in multiple cell types. Here, using affinity protein purification, mass spectrometric identification, and confocal immunofluorescence microscopy assays, we found that MSec interacts with the endoplasmic reticulum (ER) chaperone ERp29. siRNA-mediated ERp29 depletion in mammalian cells significantly reduces TNT formation, whereas its overexpression induces TNT formation, but in a strictly MSec-dependent manner. ERp29 stabilized MSec protein levels, but not its mRNA levels, and the chaperone activity of ERp29 was required for maintaining MSec protein stability. Subcellular ER fractionation and subsequent limited proteolytic treatment suggested that MSec is associated with the outer surface of the ER. The ERp29-MSec interaction appeared to require the presence of other bridging protein(s), perhaps triggered by post-translational modification of ERp29. Our study implicates MSec as a target of ERp29 and reveals an indispensable role for the ER in TNT formation, suggesting new modalities for regulating TNT numbers in cells and tissues.
Collapse
Affiliation(s)
- Rajaiah Pergu
- From the Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, and.,the Manipal Academy of Higher Education, Manipal Karnataka 576104, and
| | - Sunayana Dagar
- From the Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, and.,the Kalinga Institute of Industrial Technology, Bhubaneswar Odisha 751024, India
| | - Harsh Kumar
- From the Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, and.,the Manipal Academy of Higher Education, Manipal Karnataka 576104, and
| | - Rajesh Kumar
- the HIV Vaccine Translational Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad Haryana 121001
| | - Jayanta Bhattacharya
- the HIV Vaccine Translational Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad Haryana 121001
| | - Sivaram V S Mylavarapu
- From the Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, and .,the Manipal Academy of Higher Education, Manipal Karnataka 576104, and.,the Kalinga Institute of Industrial Technology, Bhubaneswar Odisha 751024, India
| |
Collapse
|
50
|
Wang JL, Lin YC, Young TH, Chen MH. Far-infrared ray radiation promotes neurite outgrowth of neuron-like PC12 cells through AKT1 signaling. J Formos Med Assoc 2019; 118:600-610. [DOI: 10.1016/j.jfma.2018.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/14/2022] Open
|