1
|
Hegde S, Modi S, Deihl EW, Glomb OV, Yogev S, Hoerndli FJ, Koushika SP. Axonal mitochondria regulate gentle touch response through control of axonal actin dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607780. [PMID: 39185223 PMCID: PMC11343141 DOI: 10.1101/2024.08.13.607780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Actin in neuronal processes is both stable and dynamic. The origin & functional roles of the different pools of actin is not well understood. We find that mutants that lack mitochondria, ric-7 and mtx-2; miro-1, in neuronal processes also lack dynamic actin. Mitochondria can regulate actin dynamics upto a distance ~80 μm along the neuronal process. Absence of axonal mitochondria and dynamic actin does not markedly alter the Spectrin Membrane Periodic Skeleton (MPS) in touch receptor neurons (TRNs). Restoring mitochondria inTRNs cell autonomously restores dynamic actin in a sod-2 dependent manner. We find that dynamic actin is necessary and sufficient for the localization of gap junction proteins in the TRNs and for the C. elegans gentle touch response. We identify an in vivo mechanism by which axonal mitochondria locally facilitate actin dynamics through reactive oxygen species that we show is necessary for electrical synapses & behaviour.
Collapse
Affiliation(s)
- Sneha Hegde
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| | - Souvik Modi
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| | - Ennis W. Deihl
- Colorado State University, Anatomy and Zoology W309, 1617 Campus Delivery, Fort Collins, 80523 Colorado
| | - Oliver Vinzenz Glomb
- Yale University, Boyer Center for Molecular Medicine, 295 Congress Ave, New Haven, CT 06510
- Current address: Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, 72074 Tübingen, Germany
| | - Shaul Yogev
- Yale University, Boyer Center for Molecular Medicine, 295 Congress Ave, New Haven, CT 06510
| | - Frederic J. Hoerndli
- Colorado State University, Anatomy and Zoology W309, 1617 Campus Delivery, Fort Collins, 80523 Colorado
| | - Sandhya P. Koushika
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| |
Collapse
|
2
|
Gallo G. The Axonal Actin Filament Cytoskeleton: Structure, Function, and Relevance to Injury and Degeneration. Mol Neurobiol 2024; 61:5646-5664. [PMID: 38216856 DOI: 10.1007/s12035-023-03879-7] [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: 10/26/2023] [Accepted: 12/13/2023] [Indexed: 01/14/2024]
Abstract
Early investigations of the neuronal actin filament cytoskeleton gave rise to the notion that, although growth cones exhibit high levels of actin filaments, the axon shaft exhibits low levels of actin filaments. With the development of new tools and imaging techniques, the axonal actin filament cytoskeleton has undergone a renaissance and is now an active field of research. This article reviews the current state of knowledge about the actin cytoskeleton of the axon shaft. The best understood forms of actin filament organization along axons are axonal actin patches and a submembranous system of rings that endow the axon with protrusive competency and structural integrity, respectively. Additional forms of actin filament organization along the axon have also been described and their roles are being elucidated. Extracellular signals regulate the axonal actin filament cytoskeleton and our understanding of the signaling mechanisms involved is being elaborated. Finally, recent years have seen advances in our perspective on how the axonal actin cytoskeleton is impacted by, and contributes to, axon injury and degeneration. The work to date has opened new venues and future research will undoubtedly continue to provide a richer understanding of the axonal actin filament cytoskeleton.
Collapse
Affiliation(s)
- Gianluca Gallo
- Department of Neural Sciences, Shriners Pediatric Research Center, Lewis Katz School of Medicine at Temple University, 3500 North Broad St, Philadelphia, PA, 19140, USA.
| |
Collapse
|
3
|
Wang XH, Zuo ZF, Meng L, Yang Q, Lv P, Zhao LP, Wang XB, Wang YF, Huang Y, Fu C, Liu WQ, Liu XZ, Zheng DY. Neuroprotective effect of salidroside on hippocampal neurons in diabetic mice via PI3K/Akt/GSK-3β signaling pathway. Psychopharmacology (Berl) 2023; 240:1865-1876. [PMID: 37490132 DOI: 10.1007/s00213-023-06373-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 12/08/2022] [Indexed: 07/26/2023]
Abstract
BACKGROUND Diabetic encephalopathy is manifested by cognitive dysfunction. Salidroside, a nature compound isolated from Rhodiola rosea L, has the effects of anti-inflammatory and antioxidant, hypoglycemic and lipid-lowering, improving insulin resistance, inhibiting cell apoptosis, and protecting neurons. However, the mechanism by which salidroside alleviates neuronal degeneration and improves learning and memory impairment in diabetic mice remains unclear. OBJECTIVE To investigate the effects and mechanisms of salidroside on hippocampal neurons in streptozotocin-induced diabetic mice. MATERIALS AND METHODS C57BL/6 mice were randomly divided into 4 groups to receive either sham (control group (CON)), diabetes mellitus (diabetes group (DM)), diabetes mellitus + salidroside (salidroside group (DM + SAL)), and diabetes mellitus + salidroside + phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 (diabetes mellitus + salidroside + LY294002 group (DM + SAL + LY294002)). After 12 weeks of diabetes onset, the cognitive behaviors were tested using Morris water maze. The number of hippocampal neurons was detected by Nissl staining. The expressions of PI3K, p-PI3K, Akt, p-Akt, GSK-3β, p-GSK-3β, cleaved caspase-3, caspase-3, Bax, Bcl-2, MAP2, and SYN in the hippocampus were detected by Western blot. Moreover, the expression of MAP2 and SYN in the hippocampus was further confirmed by immunofluorescence staining. RESULTS Salidroside increased the time of diabetic mice in the platform quadrant and reduced the escape latency of diabetic mice. Salidroside also increased the expression of p-PI3K, p-Akt, p-GSK-3β, MAP2, SYN, Bcl-2, while suppressed the expression of cleaved caspase-3, caspase3, and Bax in the DM + SAL group compared with the DM group (P < 0.05). The Nissl staining showed that the number of hippocampus neurons in the DM + SAL group was increased with the intact, compact, and regular arrangement, compared with the DM groups (P < 0.05). Interestingly, the protective effects of salidroside on diabetic cognitive dysfunction, hippocampal morphological alterations, and protein expressions were abolished by inhibition of PI3K with LY294002. CONCLUSIONS Salidroside exerts neuroprotective properties in diabetic cognitive dysfunction partly via activating the PI3K/Akt/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Xue-Hua Wang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Zhong-Fu Zuo
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- Department of Anatomy, Histology and Embryology, Postdoctoral Research Station, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lu Meng
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Qi Yang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Pan Lv
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Li-Pan Zhao
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Xiao-Bai Wang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Yu-Fei Wang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Ying Huang
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Cong Fu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Wen-Qiang Liu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Xue-Zheng Liu
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
- Liaoning Key Laboratory of Diabetic Cognitive and Perceptive Dysfunction, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| | - De-Yu Zheng
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| |
Collapse
|
4
|
Holland SM, Gallo G. Actin cytoskeletal dynamics do not impose an energy drain on growth cone bioenergetics. J Cell Sci 2023; 136:jcs261356. [PMID: 37534394 PMCID: PMC10445737 DOI: 10.1242/jcs.261356] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023] Open
Abstract
The regulation of the intracellular level of ATP is a fundamental aspect of bioenergetics. Actin cytoskeletal dynamics have been reported to be an energetic drain in developing neurons and platelets. We addressed the role of actin dynamics in primary embryonic chicken neurons using luciferase assays, and by measurement of the ATP/ADP ratio using the ratiometric reporter PercevalHR and the ATP level using the ratiometric reporter mRuby-iATPSnFR. None of the methods revealed an effect of suppressing actin dynamics on the decline in the neuronal ATP level or the ATP/ADP ratio following shutdown of ATP production. Similarly, we find that treatments that elevate or suppress actin dynamics do not alter the ATP/ADP ratio in growth cones, the subcellular domain with the highest actin dynamics in developing neurons. Collectively, the data indicate that actin cytoskeletal dynamics are not a significant energy drain in developing neurons and that the ATP/ADP ratio is maintained when energy utilization varies. Discrepancies between prior work and the current data are discussed with emphasis on methodology and interpretation of the data.
Collapse
Affiliation(s)
- Sabrina M. Holland
- Lewis Katz School of Medicine at Temple University, Department of Neural Sciences, Shriners Pediatric Research Center, 3500 North Broad St, Philadelphia, PA 19140, USA
| | - Gianluca Gallo
- Lewis Katz School of Medicine at Temple University, Department of Neural Sciences, Shriners Pediatric Research Center, 3500 North Broad St, Philadelphia, PA 19140, USA
| |
Collapse
|
5
|
Kouchi Z, Kojima M. A Structural Network Analysis of Neuronal ArhGAP21/23 Interactors by Computational Modeling. ACS OMEGA 2023; 8:19249-19264. [PMID: 37305272 PMCID: PMC10249030 DOI: 10.1021/acsomega.2c08054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023]
Abstract
RhoGTPase-activating proteins (RhoGAPs) play multiple roles in neuronal development; however, details of their substrate recognition system remain elusive. ArhGAP21 and ArhGAP23 are RhoGAPs that contain N-terminal PDZ and pleckstrin homology domains. In the present study, the RhoGAP domain of these ArhGAPs was computationally modeled by template-based methods and the AlphaFold2 software program, and their intrinsic RhoGTPase recognition mechanism was analyzed from the domain structures using the protein docking programs HADDOCK and HDOCK. ArhGAP21 was predicted to preferentially catalyze Cdc42, RhoA, RhoB, RhoC, and RhoG and to downregulate RhoD and Tc10 activities. Regarding ArhGAP23, RhoA and Cdc42 were deduced to be its substrates, whereas RhoD downregulation was predicted to be less efficient. The PDZ domains of ArhGAP21/23 possess the FTLRXXXVY sequence, and similar globular folding consists of antiparalleled β-sheets and two α-helices that are conserved with PDZ domains of MAST-family proteins. A peptide docking analysis revealed the specific interaction of the ArhGAP23 PDZ domain with the PTEN C-terminus. The pleckstrin homology domain structure of ArhGAP23 was also predicted, and the functional selectivity for the interactors regulated by the folding and disordered domains in ArhGAP21 and ArhGAP23 was examined by an in silico analysis. An interaction analysis of these RhoGAPs revealed the existence of mammalian ArhGAP21/23-specific type I and type III Arf- and RhoGTPase-regulated signaling. Multiple recognition systems of RhoGTPase substrates and selective Arf-dependent localization of ArhGAP21/23 may form the basis of the functional core signaling necessary for synaptic homeostasis and axon/dendritic transport regulated by RhoGAP localization and activities.
Collapse
Affiliation(s)
- Zen Kouchi
- Department
of Genetics, Institute for Developmental
Research, Aichi Developmental Disability Center, 713-8 Kamiya-cho, Kasugai-city 480-0392 Aichi, Japan
| | - Masaki Kojima
- Laboratory
of Bioinformatics, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji 192-0392, Japan
| |
Collapse
|
6
|
Antagonistic Activities of Fmn2 and ADF Regulate Axonal F-Actin Patch Dynamics and the Initiation of Collateral Branching. J Neurosci 2022; 42:7355-7369. [PMID: 36481742 PMCID: PMC9525169 DOI: 10.1523/jneurosci.3107-20.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 12/15/2022] Open
Abstract
Interstitial collateral branching of axons is a critical component in the development of functional neural circuits. Axon collateral branches are established through a series of cellular processes initiated by the development of a specialized, focal F-actin network in axons. The formation, maintenance and remodeling of this F-actin patch is critical for the initiation of axonal protrusions that are subsequently consolidated to form a collateral branch. However, the mechanisms regulating F-actin patch dynamics are poorly understood. Fmn2 is a formin family member implicated in multiple neurodevelopmental disorders. We find that Fmn2 regulates the initiation of axon collateral protrusions in chick spinal neurons and in zebrafish motor neurons. Fmn2 localizes to the protrusion-initiating axonal F-actin patches and regulates the lifetime and size of these F-actin networks. The F-actin nucleation activity of Fmn2 is necessary for F-actin patch stability but not for initiating patch formation. We show that Fmn2 insulates the F-actin patches from disassembly by the actin-depolymerizing factor, ADF, and promotes long-lived, larger patches that are competent to initiate axonal protrusions. The regulation of axonal branching can contribute to the neurodevelopmental pathologies associated with Fmn2 and the dynamic antagonism between Fmn2 and ADF may represent a general mechanism of formin-dependent protection of Arp2/3-initiated F-actin networks from disassembly.SIGNIFICANCE STATEMENT Axonal branching is a key process in the development of functional circuits and neural plasticity. Axon collateral branching is initiated by the elaboration of F-actin filaments from discrete axonal F-actin networks. We show that the neurodevelopmental disorder-associated formin, Fmn2, is a critical regulator of axon collateral branching. Fmn2 localizes to the collateral branch-inducing F-actin patches in axons and regulates the stability of these actin networks. The F-actin nucleation activity of Fmn2 protects the patches from ADF-mediated disassembly. Opposing activities of Fmn2 and ADF exert a dynamic regulatory control on axon collateral branch initiation and may underly the neurodevelopmental defects associated with Fmn2.
Collapse
|
7
|
Ketschek A, Holland SM, Gallo G. SARM1 Suppresses Axon Branching Through Attenuation of Axonal Cytoskeletal Dynamics. Front Mol Neurosci 2022; 15:726962. [PMID: 35264929 PMCID: PMC8899016 DOI: 10.3389/fnmol.2022.726962] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 01/20/2022] [Indexed: 12/18/2022] Open
Abstract
Axon branching is a fundamental aspect of neuronal morphogenesis, neuronal circuit formation, and response of the nervous system to injury. Sterile alpha and TIR motif containing 1 (SARM1) was initially identified as promoting Wallerian degeneration of axons. We now report a novel function of SARM1 in postnatal sensory neurons; the suppression of axon branching. Axon collateral branches develop from axonal filopodia precursors through the coordination of the actin and microtubule cytoskeleton. In vitro analysis revealed that cultured P0-2 dorsal root ganglion sensory neurons from a SARM1 knockout (KO) mouse exhibit increased numbers of collateral branches and axonal filopodia relative to wild-type neurons. In SARM1 KO mice, cutaneous sensory endings exhibit increased branching in the skin in vivo with normal density of innervation. Transient axonal actin patches serve as cytoskeletal platforms from which axonal filopodia emerge. Live imaging analysis of axonal actin dynamics showed that SARM1 KO neurons exhibit increased rates of axonal actin patch formation and increased probability that individual patches will give rise to a filopodium before dissipating. SARM1 KO axons contain elevated levels of drebrin and cortactin, two actin regulatory proteins that are positive regulators of actin patches, filopodia formation, and branching. Live imaging of microtubule plus tip dynamics revealed an increase in the rate of formation and velocity of polymerizing tips along the axons of SARM1 KO neurons. Stationary mitochondria define sites along the axon where branches may arise, and the axons of SARM1 KO sensory neurons exhibit an increase in stationary mitochondria. These data reveal SARM1 to be a negative regulator of axonal cytoskeletal dynamics and collateral branching.
Collapse
Affiliation(s)
- Andrea Ketschek
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Sabrina M. Holland
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
8
|
Tariq K, Luikart BW. Striking a balance: PIP 2 and PIP 3 signaling in neuronal health and disease. EXPLORATION OF NEUROPROTECTIVE THERAPY 2022; 1:86-100. [PMID: 35098253 PMCID: PMC8797975 DOI: 10.37349/ent.2021.00008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Phosphoinositides are membrane phospholipids involved in a variety of cellular processes like growth, development, metabolism, and transport. This review focuses on the maintenance of cellular homeostasis of phosphatidylinositol 4,5-bisphosphate (PIP2), and phosphatidylinositol 3,4,5-trisphosphate (PIP3). The critical balance of these PIPs is crucial for regulation of neuronal form and function. The activity of PIP2 and PIP3 can be regulated through kinases, phosphatases, phospholipases and cholesterol microdomains. PIP2 and PIP3 carry out their functions either indirectly through their effectors activating integral signaling pathways, or through direct regulation of membrane channels, transporters, and cytoskeletal proteins. Any perturbations to the balance between PIP2 and PIP3 signaling result in neurodevelopmental and neurodegenerative disorders. This review will discuss the upstream modulators and downstream effectors of the PIP2 and PIP3 signaling, in the context of neuronal health and disease.
Collapse
Affiliation(s)
- Kamran Tariq
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
9
|
Fuchs J, Eickholt BJ. Precursor types predict the stability of neuronal branches. J Cell Sci 2021; 134:273430. [PMID: 34766183 PMCID: PMC8714070 DOI: 10.1242/jcs.258983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/03/2021] [Indexed: 11/20/2022] Open
Abstract
Branches are critical for neuron function, generating the morphological complexity required for functional networks. They emerge from different, well-described, cytoskeletal precursor structures that elongate to branches. While branches are thought to be maintained by shared cytoskeletal regulators, our data from mouse hippocampal neurons indicate that the precursor structures trigger alternative branch maintenance mechanisms with differing stabilities. Whereas branches originating from lamellipodia or growth cone splitting events collapse soon after formation, branches emerging from filopodia persist. Furthermore, compared to other developing neurites, axons stabilise all branches and preferentially initiate branches from filopodia. These differences explain the altered stability of branches we observe in neurons lacking the plasma membrane protein phospholipid phosphatase-related protein 3 (PLPPR3, also known as PRG2) and in neurons treated with netrin-1. Rather than altering branch stability directly, PLPPR3 and netrin-1 boost a 'filopodia branch programme' on axons, thereby indirectly initiating more long-lived branches. In summary, we propose that studies on branching should distinguish overall stabilising effects from effects on precursor types, ideally using multifactorial statistical models, as exemplified in this study.
Collapse
Affiliation(s)
- Joachim Fuchs
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Molecular Biology and Biochemistry, Virchowweg 6, 10117 Berlin, Germany
| | - Britta J Eickholt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Molecular Biology and Biochemistry, Virchowweg 6, 10117 Berlin, Germany
| |
Collapse
|
10
|
Sainath R, Gallo G. Bioenergetic Requirements and Spatiotemporal Profile of Nerve Growth Factor Induced PI3K-Akt Signaling Along Sensory Axons. Front Mol Neurosci 2021; 14:726331. [PMID: 34630035 PMCID: PMC8497901 DOI: 10.3389/fnmol.2021.726331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Nerve Growth Factor (NGF) promotes the elaboration of axonal filopodia and branches through PI3K-Akt. NGF activates the TrkA receptor resulting in an initial transient high amplitude burst of PI3K-Akt signaling followed by a maintained lower steady state, hereafter referred to as initiation and steady state phases. Akt initially undergoes phosphorylation at T308 followed by phosphorylation at S473, resulting in maximal kinase activation. We report that during the initiation phase the localization of PI3K signaling, reported by visualizing sites of PIP3 formation, and Akt signaling, reflected by Akt phosphorylation at T308, correlates with the positioning of axonal mitochondria. Mitochondrial oxidative phosphorylation but not glycolysis is required for Akt phosphorylation at T308. In contrast, the phosphorylation of Akt at S473 is not spatially associated with mitochondria and is dependent on both oxidative phosphorylation and glycolysis. Under NGF steady state conditions, maintenance of phosphorylation at T308 shows dual dependence on oxidative phosphorylation and glycolysis. Phosphorylation at S473 is more dependent on glycolysis but also requires oxidative phosphorylation for maintenance over longer time periods. The data indicate that NGF induced PI3K-Akt signaling along axons is preferentially initiated at sites containing mitochondria, in a manner dependent on oxidative phosphorylation. Steady state signaling is discussed in the context of combined contributions by mitochondria and the possibility of glycolysis occurring in association with endocytosed signalosomes.
Collapse
Affiliation(s)
- Rajiv Sainath
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
11
|
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
|
12
|
Gohel D, Singh R. Mitohormesis; Potential implications in neurodegenerative diseases. Mitochondrion 2020; 56:40-46. [PMID: 33220499 DOI: 10.1016/j.mito.2020.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/06/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Mitochondrial dysfunction is known to be associated with neurodegenerative diseases (NDDs), which is a major burden on the society. Therefore, understanding the regulation of mitochondrial dysfunctions and its implication in neurodegeneration has been major goal for exploiting these mechanisms to rescue neuronal death. The crosstalk between mitochondria and nucleus is important for different neuronal functions including axonal branching, energy homeostasis, neuroinflammation and neuronal survival. The decreased mitochondria capacity during progressive neurodegeneration leads to the altered OXPHOS activity and generation of ROS. The ROS levels in narrow physiological range can reprogram nuclear gene expression to enhance the cellular survival by phenomenon called mitohormesis. Here, we have systematically reviewed the existing reports of mitochondrial dysfunctions causing altered ROS levels in NDDs. We further discussed the role of ROS in regulating mitohormesis and emphasized the importance of mitohormesis in neuronal homeostasis. The emerging role of mitohormesis highlights its importance in future studies on intracellular ROS mediated rescue of mitochondrial dysfunction along with other prevailing mechanisms to alleviate neurodegeneration.
Collapse
Affiliation(s)
- Dhruv Gohel
- Department of Biochemistry, Faculty of Science, The M.S. University of Baroda, Vadodara 390002, Gujarat, India
| | - Rajesh Singh
- Department of Biochemistry, Faculty of Science, The M.S. University of Baroda, Vadodara 390002, Gujarat, India.
| |
Collapse
|
13
|
Gallo G. The bioenergetics of neuronal morphogenesis and regeneration: Frontiers beyond the mitochondrion. Dev Neurobiol 2020; 80:263-276. [PMID: 32750228 DOI: 10.1002/dneu.22776] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/27/2022]
Abstract
The formation of axons and dendrites during development, and their regeneration following injury, are energy intensive processes. The underlying assembly and dynamics of the cytoskeleton, axonal transport mechanisms, and extensive signaling networks all rely on ATP and GTP consumption. Cellular ATP is generated through oxidative phosphorylation (OxP) in mitochondria, glycolysis and "regenerative" kinase systems. Recent investigations have focused on the role of the mitochondrion in axonal development and regeneration emphasizing the importance of this organelle and OxP in axon development and regeneration. In contrast, the understanding of alternative sources of ATP in neuronal morphogenesis and regeneration remains largely unexplored. This review focuses on the current state of the field of neuronal bioenergetics underlying morphogenesis and regeneration and considers the literature on the bioenergetics of non-neuronal cell motility to emphasize the potential contributions of non-mitochondrial energy sources.
Collapse
Affiliation(s)
- Gianluca Gallo
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, PA, USA
| |
Collapse
|
14
|
Brosig A, Fuchs J, Ipek F, Kroon C, Schrötter S, Vadhvani M, Polyzou A, Ledderose J, van Diepen M, Holzhütter HG, Trimbuch T, Gimber N, Schmoranzer J, Lieberam I, Rosenmund C, Spahn C, Scheerer P, Szczepek M, Leondaritis G, Eickholt BJ. The Axonal Membrane Protein PRG2 Inhibits PTEN and Directs Growth to Branches. Cell Rep 2020; 29:2028-2040.e8. [PMID: 31722215 PMCID: PMC6856728 DOI: 10.1016/j.celrep.2019.10.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 08/09/2019] [Accepted: 10/09/2019] [Indexed: 01/03/2023] Open
Abstract
In developing neurons, phosphoinositide 3-kinases (PI3Ks) control axon growth and branching by positively regulating PI3K/PI(3,4,5)P3, but how neurons are able to generate sufficient PI(3,4,5)P3 in the presence of high levels of the antagonizing phosphatase PTEN is difficult to reconcile. We find that normal axon morphogenesis involves homeostasis of elongation and branch growth controlled by accumulation of PI(3,4,5)P3 through PTEN inhibition. We identify a plasma membrane-localized protein-protein interaction of PTEN with plasticity-related gene 2 (PRG2). PRG2 stabilizes membrane PI(3,4,5)P3 by inhibiting PTEN and localizes in nanoclusters along axon membranes when neurons initiate their complex branching behavior. We demonstrate that PRG2 is both sufficient and necessary to account for the ability of neurons to generate axon filopodia and branches in dependence on PI3K/PI(3,4,5)P3 and PTEN. Our data indicate that PRG2 is part of a neuronal growth program that induces collateral branch growth in axons by conferring local inhibition of PTEN. Neuronal axon growth and branching is globally regulated by PI3K/PTEN signaling PRG2 inhibits PTEN and stabilizes PIP3 and F-actin PRG2 localizes to nanoclusters on the axonal membrane and coincides with branching PRG2 promotes axonal filopodia and branching dependent on PI3K/PTEN
Collapse
Affiliation(s)
- Annika Brosig
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Joachim Fuchs
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Fatih Ipek
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Group Protein X-ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Cristina Kroon
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Sandra Schrötter
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Department of Genetics and Complex Diseases, T.H. Chan Harvard School of Public Health, Boston, MA 02120, USA
| | - Mayur Vadhvani
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Alexandra Polyzou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Julia Ledderose
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michiel van Diepen
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Hermann-Georg Holzhütter
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Niclas Gimber
- Advanced Medical Bioimaging Core Facility (AMBIO), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jan Schmoranzer
- Advanced Medical Bioimaging Core Facility (AMBIO), Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ivo Lieberam
- Centre for Stem Cells and Regenerative Medicine and Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College, London, UK
| | - Christian Rosenmund
- NeuroCure-Cluster of Excellence, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Christian Spahn
- NeuroCure-Cluster of Excellence, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Patrick Scheerer
- Group Protein X-ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michal Szczepek
- Group Protein X-ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - George Leondaritis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece.
| | - Britta J Eickholt
- Institute of Biochemistry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; NeuroCure-Cluster of Excellence, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
15
|
Kim J, Cheong JH. Role of Mitochondria-Cytoskeleton Interactions in the Regulation of Mitochondrial Structure and Function in Cancer Stem Cells. Cells 2020; 9:cells9071691. [PMID: 32674438 PMCID: PMC7407978 DOI: 10.3390/cells9071691] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 12/16/2022] Open
Abstract
Despite the promise of cancer medicine, major challenges currently confronting the treatment of cancer patients include chemoresistance and recurrence. The existence of subpopulations of cancer cells, known as cancer stem cells (CSCs), contributes to the failure of cancer therapies and is associated with poor clinical outcomes. Of note, one of the recently characterized features of CSCs is augmented mitochondrial function. The cytoskeleton network is essential in regulating mitochondrial morphology and rearrangement, which are inextricably linked to its functions, such as oxidative phosphorylation (OXPHOS). The interaction between the cytoskeleton and mitochondria can enable CSCs to adapt to challenging conditions, such as a lack of energy sources, and to maintain their stemness. Cytoskeleton-mediated mitochondrial trafficking and relocating to the high energy requirement region are crucial steps in epithelial-to-mesenchymal transition (EMT). In addition, the cytoskeleton itself interplays with and blocks the voltage-dependent anion channel (VDAC) to directly regulate bioenergetics. In this review, we describe the regulation of cellular bioenergetics in CSCs, focusing on the cytoskeleton-mediated dynamic control of mitochondrial structure and function.
Collapse
Affiliation(s)
- Jungmin Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Jae-Ho Cheong
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea;
- Department of Surgery, Yonsei University Health System, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-2-2228-2094; Fax: +82-2-313-8289
| |
Collapse
|
16
|
Gao P, Mu M, Chen Y, He J, Tao X, Song C. Yes-associated protein upregulates filopodia formation to promote alveolar epithelial-cell phagocytosis. Immunol Lett 2020; 225:44-49. [PMID: 32554050 DOI: 10.1016/j.imlet.2020.06.009] [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: 03/24/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 02/08/2023]
Abstract
Cells engulf particles larger than 0.5 μm in diameter by phagocytosis, which is driven by cytoskeletal rearrangements. Phagocytosis by alveolar epithelial cells (AECs) helps to maintain the alveolar homeostasis. Yes-associated protein (YAP), a transcriptional coactivator of the Hippo pathway, affects proliferation, differentiation, and cytoskeletal rearrangement of AECs, but it is not clear whether YAP regulates phagocytosis. In this study, interference with YAP expression inhibited phagocytosis in MLE-12 cells and in primary cultures of AEC. Filopodia formation promoted phagocytosis in AECs, and YAP enhanced filopodia formation in AECs. Blocking PI3K signaling resulted in reduced YAP protein expression and inhibition of phagocytosis. The results indicate that YAP expression was regulated by PI3K signaling and promoted phagocytosis in AECs by upregulating filopodia formation.
Collapse
Affiliation(s)
- Peiyu Gao
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, PR China; Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Mimi Mu
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, PR China; Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Yan Chen
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, PR China; Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Jing He
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, PR China; Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Xiangnan Tao
- Clinical Laboratory, The Second Affiliated Hospital of Bengbu Medical College, Anhui, 233004, PR China
| | - Chuanwang Song
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, PR China; Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, PR China.
| |
Collapse
|
17
|
Gallop J. Filopodia and their links with membrane traffic and cell adhesion. Semin Cell Dev Biol 2020; 102:81-89. [DOI: 10.1016/j.semcdb.2019.11.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 01/24/2023]
|
18
|
Skelton PD, Stan RV, Luikart BW. The Role of PTEN in Neurodevelopment. MOLECULAR NEUROPSYCHIATRY 2020; 5:60-71. [PMID: 32399470 PMCID: PMC7206585 DOI: 10.1159/000504782] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
Abstract
PTEN is a lipid and protein phosphatase that regulates cell growth and survival. Mutations to PTEN are highly penetrant for autism spectrum disorder (ASD). Here, we briefly review the evidence linking PTEN mutations to ASD and the mouse models that have been used to study the role of PTEN in neurodevelopment. We then focus on the cellular phenotypes associated with PTEN loss in neurons, highlighting the role PTEN plays in neuronal proliferation, migration, survival, morphology, and plasticity.
Collapse
Affiliation(s)
- Patrick D. Skelton
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Radu V. Stan
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Bryan W. Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
19
|
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
|
20
|
Armijo-Weingart L, Ketschek A, Sainath R, Pacheco A, Smith GM, Gallo G. Neurotrophins induce fission of mitochondria along embryonic sensory axons. eLife 2019; 8:e49494. [PMID: 31789589 PMCID: PMC6887118 DOI: 10.7554/elife.49494] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/26/2019] [Indexed: 12/30/2022] Open
Abstract
Neurotrophins are growth factors that have a multitude of roles in the nervous system. We report that neurotrophins induce the fission of mitochondria along embryonic chick sensory axons driven by combined PI3K and Mek-Erk signaling. Following an initial burst of fission, a new steady state of neurotrophin-dependent mitochondria length is established. Mek-Erk controls the activity of the fission mediator Drp1 GTPase, while PI3K may contribute to the actin-dependent aspect of fission. Drp1-mediated fission is required for nerve growth factor (NGF)-induced collateral branching in vitro and expression of dominant negative Drp1 impairs the branching of axons in the developing spinal cord in vivo. Fission is also required for NGF-induced mitochondria-dependent intra-axonal translation of the actin regulatory protein cortactin, a previously determined component of NGF-induced branching. Collectively, these observations unveil a novel biological function of neurotrophins; the regulation of mitochondrial fission and steady state mitochondrial length and density in axons.
Collapse
Affiliation(s)
- Lorena Armijo-Weingart
- Department of Anatomy and Cell Biology, Shriner Hospitals Pediatric Research CenterTemple University Lewis Katz School of MedicinePhiladelphiaUnited States
| | - Andrea Ketschek
- Department of Anatomy and Cell Biology, Shriner Hospitals Pediatric Research CenterTemple University Lewis Katz School of MedicinePhiladelphiaUnited States
| | - Rajiv Sainath
- Department of Anatomy and Cell Biology, Shriner Hospitals Pediatric Research CenterTemple University Lewis Katz School of MedicinePhiladelphiaUnited States
| | - Almudena Pacheco
- Department of Anatomy and Cell Biology, Shriner Hospitals Pediatric Research CenterTemple University Lewis Katz School of MedicinePhiladelphiaUnited States
| | - George M Smith
- Department of Neuroscience, Shriner Hospitals Pediatric Research CenterTemple University Lewis Katz School of MedicinePhiladelphiaUnited States
| | - Gianluca Gallo
- Department of Anatomy and Cell Biology, Shriner Hospitals Pediatric Research CenterTemple University Lewis Katz School of MedicinePhiladelphiaUnited States
| |
Collapse
|
21
|
Pacheco A, Merianda TT, Twiss JL, Gallo G. Mechanism and role of the intra-axonal Calreticulin translation in response to axonal injury. Exp Neurol 2019; 323:113072. [PMID: 31669485 DOI: 10.1016/j.expneurol.2019.113072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/17/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022]
Abstract
Following injury, sensory axons locally translate mRNAs that encode proteins needed for the response to injury, locally and through retrograde signaling, and for regeneration. In this study, we addressed the mechanism and role of axotomy-induced intra-axonal translation of the ER chaperone Calreticulin. In vivo peripheral nerve injury increased Calreticulin levels in sensory axons. Using an in vitro model system of sensory neurons amenable to mechanistic dissection we provide evidence that axotomy induces local translation of Calreticulin through PERK (protein kinase RNA-like endoplasmic reticulum kinase) mediated phosphorylation of eIF2α by a mechanism that requires both 5' and 3'UTRs (untranslated regions) elements in Calreticulin mRNA. ShRNA mediated depletion of Calreticulin or inhibition of PERK signaling increased axon retraction following axotomy. In contrast, expression of axonally targeted, but not somatically restricted, Calreticulin mRNA decreased retraction and promoted axon regeneration following axotomy in vitro. Collectively, these data indicate that the intra-axonal translation of Calreticulin in response to axotomy serves to minimize the ensuing retraction, and overexpression of axonally targeted Calreticulin mRNA promotes axon regeneration.
Collapse
Affiliation(s)
- Almudena Pacheco
- Temple University School of Medicine, Shriners Pediatric Research Center, Department of Anatomy and Cell Biology, 3500 North Broad St, Philadelphia, PA 19140, United States of America.
| | - Tanuja T Merianda
- Drexel University, Department of Biology, Philadelphia, PA 19104, United States of America
| | - Jeffery L Twiss
- University of South Carolina, Department of Biological Sciences, Columbia 29208, SC, United States of America.
| | - Gianluca Gallo
- Temple University School of Medicine, Shriners Pediatric Research Center, Department of Anatomy and Cell Biology, 3500 North Broad St, Philadelphia, PA 19140, United States of America.
| |
Collapse
|
22
|
Dong L, Li R, Li D, Wang B, Lu Y, Li P, Yu F, Jin Y, Ni X, Wu Y, Yang S, Lv G, Li X, Xiao J, Wang J. FGF10 Enhances Peripheral Nerve Regeneration via the Preactivation of the PI3K/Akt Signaling-Mediated Antioxidant Response. Front Pharmacol 2019; 10:1224. [PMID: 31680984 PMCID: PMC6805699 DOI: 10.3389/fphar.2019.01224] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022] Open
Abstract
The process of axonal regeneration after peripheral nerve injury (PNI) is slow and mostly incomplete. Previous studies have investigated the neuroprotective effects of fibroblast growth factor 10 (FGF10) against spinal cord injury and cerebral ischemia brain injury. However, the role of FGF10 in peripheral nerve regeneration remains unknown. In this study, we aimed to investigate the underlying therapeutic effects of FGF10 on nerve regeneration and functional recovery after PNI and to explore the associated mechanism. Our results showed that FGF10 administration promoted axonal regeneration and functional recovery after nerve damage. Moreover, exogenous FGF10 treatment also prevented SCs from excessive oxidative stress-induced apoptosis, which was probably related to the activation of phosphatidylinositol-3 kinase/protein kinase B (PI3K/Akt) signaling. The inhibition of the PI3K/Akt pathway by the specific inhibitor LY294002 partially reversed the therapeutic effects of FGF10 both in vivo and in vitro. Thus, from our perspective, FGF10 may be a promising therapeutic drug for repairing sciatic nerve damage through countering excessive oxidative stress-induced SC apoptosis.
Collapse
Affiliation(s)
- Lvpeng Dong
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Rui Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China.,School of Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Duohui Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Beini Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Yingfeng Lu
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peifeng Li
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangzheng Yu
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yonglong Jin
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao Ni
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanqing Wu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Shengnan Yang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Guanxi Lv
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jian Wang
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Fricano-Kugler CJ, Getz SA, Williams MR, Zurawel AA, DeSpenza T, Frazel PW, Li M, O’Malley AJ, Moen EL, Luikart BW. Nuclear Excluded Autism-Associated Phosphatase and Tensin Homolog Mutations Dysregulate Neuronal Growth. Biol Psychiatry 2018; 84:265-277. [PMID: 29373119 PMCID: PMC5984669 DOI: 10.1016/j.biopsych.2017.11.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/01/2017] [Accepted: 11/16/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND Phosphatase and tensin homolog (PTEN) negatively regulates downstream protein kinase B signaling, resulting in decreased cellular growth and proliferation. PTEN is mutated in a subset of children with autism spectrum disorder (ASD); however, the mechanism by which specific point mutations alter PTEN function is largely unknown. Here, we assessed how ASD-associated single-nucleotide variations in PTEN (ASD-PTEN) affect function. METHODS We used viral-mediated molecular substitution of human PTEN into Pten knockout mouse neurons and assessed neuronal morphology to determine the functional impact of ASD-PTEN. We employed molecular cloning to examine how PTEN's stability, subcellular localization, and catalytic activity affect neuronal growth. RESULTS We identified a set of ASD-PTEN mutations displaying altered lipid phosphatase function and subcellular localization. We demonstrated that wild-type PTEN can rescue the neuronal hypertrophy, while PTEN H93R, F241S, D252G, W274L, N276S, and D326N failed to rescue this hypertrophy. A subset of these mutations lacked nuclear localization, prompting us to examine the role of nuclear PTEN in regulating neuronal growth. We found that nuclear PTEN alone is sufficient to regulate soma size. Furthermore, forced localization of the D252G and W274L mutations into the nucleus partially restores regulation of soma size. CONCLUSIONS ASD-PTEN mutations display decreased stability, catalytic activity, and/or altered subcellular localization. Mutations lacking nuclear localization uncover a novel mechanism whereby lipid phosphatase activity in the nucleus can regulate mammalian target of rapamycin signaling and neuronal growth.
Collapse
Affiliation(s)
- Catherine J. Fricano-Kugler
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756
| | - Stephanie A. Getz
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756
| | - Michael R. Williams
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756
| | - Ashley A. Zurawel
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756
| | - Tyrone DeSpenza
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756
| | - Paul W. Frazel
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756
| | - Meijie Li
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756
| | - Alistair J. O’Malley
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA,Department of Biomedical Data Science, The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Erika L. Moen
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Bryan W. Luikart
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire 03756,Corresponding Author-
| |
Collapse
|
24
|
Abstract
SIGNIFICANCE Numerous studies have demonstrated the actions of reactive oxygen species (ROS) as regulators of several physiological processes. In this study, we discuss how redox signaling mechanisms operate to control different processes such as neuronal differentiation, oligodendrocyte differentiation, dendritic growth, and axonal growth. Recent Advances: Redox homeostasis regulates the physiology of neural stem cells (NSCs). Notably, the neuronal differentiation process of NSCs is determined by a change toward oxidative metabolism, increased levels of mitochondrial ROS, increased activity of NADPH oxidase (NOX) enzymes, decreased levels of Nrf2, and differential regulation of different redoxins. Furthermore, during the neuronal maturation processes, NOX and MICAL produce ROS to regulate cytoskeletal dynamics, which control the dendritic and axonal growth, as well as the axonal guidance. CRITICAL ISSUES The redox homeostasis changes are, in part, attributed to cell metabolism and compartmentalized production of ROS, which is regulated, sensed, and transduced by different molecules such as thioredoxins, glutaredoxins, peroxiredoxins, and nucleoredoxin to control different signaling pathways in different subcellular regions. The study of how these elements cooperatively act is essential for the understanding of nervous system development, as well as the application of regenerative therapies that recapitulate these processes. FUTURE DIRECTIONS The information about these topics in the last two decades leads us to the conclusion that the role of ROS signaling in development of the nervous system is more important than it was previously believed and makes clear the importance of exploring in more detail the mechanisms of redox signaling. Antioxid. Redox Signal. 28, 1603-1625.
Collapse
Affiliation(s)
- Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
25
|
Papandréou MJ, Leterrier C. The functional architecture of axonal actin. Mol Cell Neurosci 2018; 91:151-159. [PMID: 29758267 DOI: 10.1016/j.mcn.2018.05.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/10/2018] [Accepted: 05/11/2018] [Indexed: 11/17/2022] Open
Abstract
The cytoskeleton builds and supports the complex architecture of neurons. It orchestrates the specification, growth, and compartmentation of the axon: axon initial segment, axonal shaft, presynapses. The cytoskeleton must then maintain this intricate architecture for the whole life of its host, but also drive its adaptation to new network demands and changing physiological conditions. Microtubules are readily visible inside axon shafts by electron microscopy, whereas axonal actin study has long been focused on dynamic structures of the axon such as growth cones. Super-resolution microscopy and live-cell imaging have recently revealed new actin-based structures in mature axons: rings, hotspots and trails. This has caused renewed interest for axonal actin, with efforts underway to understand the precise organization and cellular functions of these assemblies. Actin is also present in presynapses, where its arrangement is still poorly defined, and its functions vigorously debated. Here we review the organization of axonal actin, focusing on recent advances and current questions in this rejuvenated field.
Collapse
|
26
|
Spurlin JW, Nelson CM. Building branched tissue structures: from single cell guidance to coordinated construction. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2015.0527. [PMID: 28348257 DOI: 10.1098/rstb.2015.0527] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2016] [Indexed: 12/15/2022] Open
Abstract
Branched networks are ubiquitous throughout nature, particularly found in tissues that require large surface area within a restricted volume. Many tissues with a branched architecture, such as the vasculature, kidney, mammary gland, lung and nervous system, function to exchange fluids, gases and information throughout the body of an organism. The generation of branched tissues requires regulation of branch site specification, initiation and elongation. Branching events often require the coordination of many cells to build a tissue network for material exchange. Recent evidence has emerged suggesting that cell cooperativity scales with the number of cells actively contributing to branching events. Here, we compare mechanisms that regulate branching, focusing on how cell cohorts behave in a coordinated manner to build branched tissues.This article is part of the themed issue 'Systems morphodynamics: understanding the development of tissue hardware'.
Collapse
Affiliation(s)
- James W Spurlin
- Departments of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Departments of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA .,Molecular Biology, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ 08544, USA
| |
Collapse
|
27
|
Smith GM, Gallo G. The role of mitochondria in axon development and regeneration. Dev Neurobiol 2017; 78:221-237. [PMID: 29030922 DOI: 10.1002/dneu.22546] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/12/2017] [Accepted: 10/08/2017] [Indexed: 12/26/2022]
Abstract
Mitochondria are dynamic organelles that undergo transport, fission, and fusion. The three main functions of mitochondria are to generate ATP, buffer cytosolic calcium, and generate reactive oxygen species. A large body of evidence indicates that mitochondria are either primary targets for neurological disease states and nervous system injury, or are major contributors to the ensuing pathologies. However, the roles of mitochondria in the development and regeneration of axons have just begun to be elucidated. Advances in the understanding of the functional roles of mitochondria in neurons had been largely impeded by insufficient knowledge regarding the molecular mechanisms that regulate mitochondrial transport, stalling, fission/fusion, and a paucity of approaches to image and analyze mitochondria in living axons at the level of the single mitochondrion. However, technical advances in the imaging and analysis of mitochondria in living neurons and significant insights into the mechanisms that regulate mitochondrial dynamics have allowed the field to advance. Mitochondria have now been attributed important roles in the mechanism of axon extension, regeneration, and axon branching. The availability of new experimental tools is expected to rapidly increase our understanding of the functions of axonal mitochondria during both development and later regenerative attempts. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 221-237, 2018.
Collapse
Affiliation(s)
- George M Smith
- Department of Neuroscience, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140.,Shriners Hospitals Pediatric Research Center, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Gianluca Gallo
- Department of Anatomy and Cell Biology, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140.,Shriners Hospitals Pediatric Research Center, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140
| |
Collapse
|
28
|
Sainath R, Armijo-Weingart L, Ketscheck A, Xu Z, Li S, Gallo G. Chondroitin sulfate proteoglycans negatively regulate the positioning of mitochondria and endoplasmic reticulum to distal axons. Dev Neurobiol 2017; 77:1351-1370. [PMID: 28901718 DOI: 10.1002/dneu.22535] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/23/2017] [Accepted: 09/08/2017] [Indexed: 12/20/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are components of the extracellular matrix that inhibit the extension and regeneration of axons. However, the underlying mechanism of action remains poorly understood. Mitochondria and endoplasmic reticulum (ER) are functionally inter-linked organelles important to axon development and maintenance. We report that CSPGs impair the targeting of mitochondria and ER to the growth cones of chicken embryonic sensory axons. The effect of CSPGs on the targeting of mitochondria is blocked by inhibition of the LAR receptor for CSPGs. The regulation of the targeting of mitochondria and ER to the growth cone by CSPGs is due to attenuation of PI3K signaling, which is known to be downstream of LAR receptor activation. Dynactin is a required component of the dynein motor complex that drives the normally occurring retrograde evacuation of mitochondria from growth cones. CSPGs elevate the levels of p150Glu dynactin found in distal axons, and inhibition of the interaction of dynactin with dynein increased axon lengths on CSPGs. CSPGs decreased the membrane potential of mitochondria, and pharmacological inhibition of mitochondria respiration at the growth cone independent of manipulation of mitochondria positioning impaired axon extension. Combined inhibition of dynactin and potentiation of mitochondria respiration further increased axon lengths on CSPGs relative to inhibition of dynactin alone. These data reveal that the regulation of the localization of mitochondria and ER to growth cones is a previously unappreciated aspect of the effects of CSPGs on embryonic axons. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1351-1370, 2017.
Collapse
Affiliation(s)
- Rajiv Sainath
- Department of Anatomy and Cell Biology, Medical Education and Research Building, 3500 North Brad St, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Lorena Armijo-Weingart
- Department of Anatomy and Cell Biology, Medical Education and Research Building, 3500 North Brad St, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Andrea Ketscheck
- Department of Anatomy and Cell Biology, Medical Education and Research Building, 3500 North Brad St, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Zhuxuan Xu
- Department of Anatomy and Cell Biology, Medical Education and Research Building, 3500 North Brad St, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Shuxin Li
- Department of Anatomy and Cell Biology, Medical Education and Research Building, 3500 North Brad St, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Gianluca Gallo
- Department of Anatomy and Cell Biology, Medical Education and Research Building, 3500 North Brad St, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| |
Collapse
|
29
|
Li R, Wu Y, Zou S, Wang X, Li Y, Xu K, Gong F, Liu Y, Wang J, Liao Y, Li X, Xiao J. NGF Attenuates High Glucose-Induced ER Stress, Preventing Schwann Cell Apoptosis by Activating the PI3K/Akt/GSK3β and ERK1/2 Pathways. Neurochem Res 2017; 42:3005-3018. [PMID: 28762104 DOI: 10.1007/s11064-017-2333-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/22/2017] [Accepted: 06/14/2017] [Indexed: 01/24/2023]
Abstract
Diabetic peripheral neuropathy (DPN) is one of the most common and troublesome complications of diabetes mellitus. It has been demonstrated that nerve growth factor (NGF) exerts a pivotal role in the regulation of neuronal growth and the promotion of DPN recovery. However, the exact molecular mechanisms are not well understood. Recent studies have indicated that as a novel therapeutic target, endoplasmic reticulum (ER) stress participates in the onset and progression of DPN. In the present study, it has been demonstrated that NGF prevents the sciatic nerve from degeneration and demyelination in DPN rats. Thus, RSC 96 cells, which retain the characteristic features of Schwann cells (SCs), were cultured in medium containing 30 mM glucose (high glucose, HG) to mimic SCs in DPN mice. The 50-ng/ml dose of NGF was identified to be the optimal concentration for treating an excessive ER stress level under HG conditions for 24 h. We found that NGF treatment significantly inhibits HG-induced ER stress and subsequently suppresses ER-related apoptosis. Further, NGF administration also activates the upstream signaling pathway of ER stress, PI3K/Akt/GSK3β signaling and ERK1/2 signaling. Co-treatment with the PI3K inhibitor LY294002 or ERK1/2 inhibitor U0126 significantly reverses the protective role of NGF on HG-induced excessive ER stress and subsequent apoptosis. These observations suggest that the neuroprotective role of NGF in DPN is mediated by the inhibition of excessive ER stress via the activation of the PI3K/Akt/GSK3β and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Rui Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Shuang Zou
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiaofang Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yiyang Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ke Xu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Fanghua Gong
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yanlong Liu
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jian Wang
- Department of Peripheral Neurosurgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yi Liao
- Department of Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaokun Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China. .,The Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China.
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China. .,The Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China.
| |
Collapse
|
30
|
Zhang X, Cheng Q, Yin H, Yang G. Regulation of autophagy and EMT by the interplay between p53 and RAS during cancer progression (Review). Int J Oncol 2017; 51:18-24. [PMID: 28560457 DOI: 10.3892/ijo.2017.4025] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/05/2017] [Indexed: 11/06/2022] Open
Abstract
Cellular autophagy and epithelial-mesenchymal transition (EMT) are key events mostly resulted from the interplay of tumor suppressors and oncogenes during cancer progression. The master tumor suppressor p53 may control tumor cell autophagy and EMT through the transcriptional induction of multiple target genes, while the activated oncogene RAS may also play a critical role in regulating mitogenic signaling to tumor cell autophagy and EMT. Although the fundamental functions of p53 and RAS are well understood, the interactive effects of p53 and RAS on autophagy and EMT are still unclear. In this review, we highlight the recent advances in the regulation of autophagy and EMT by p53 and RAS, aiming to explore novel therapeutic targets and biomarkers in cancer treatment and prevention.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Qian Cheng
- Department of Orthopedics, the Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Huijing Yin
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Gong Yang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| |
Collapse
|
31
|
Chen CH, He CW, Liao CP, Pan CL. A Wnt-planar polarity pathway instructs neurite branching by restricting F-actin assembly through endosomal signaling. PLoS Genet 2017; 13:e1006720. [PMID: 28384160 PMCID: PMC5398721 DOI: 10.1371/journal.pgen.1006720] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/20/2017] [Accepted: 03/29/2017] [Indexed: 02/01/2023] Open
Abstract
Spatial arrangement of neurite branching is instructed by both attractive and repulsive cues. Here we show that in C. elegans, the Wnt family of secreted glycoproteins specify neurite branching sites in the PLM mechanosensory neurons. Wnts function through MIG-1/Frizzled and the planar cell polarity protein (PCP) VANG-1/Strabismus/Vangl2 to restrict the formation of F-actin patches, which mark branching sites in nascent neurites. We find that VANG-1 promotes Wnt signaling by facilitating Frizzled endocytosis and genetically acts in a common pathway with arr-1/β-arrestin, whose mutation results in defective PLM branching and F-actin patterns similar to those in the Wnt, mig-1 or vang-1 mutants. On the other hand, the UNC-6/Netrin pathway intersects orthogonally with Wnt-PCP signaling to guide PLM branch growth along the dorsal-ventral axis. Our study provides insights for how attractive and repulsive signals coordinate to sculpt neurite branching patterns, which are critical for circuit connectivity.
Collapse
Affiliation(s)
- Chun-Hao Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Wei He
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Po Liao
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Liang Pan
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
32
|
Armijo-Weingart L, Gallo G. It takes a village to raise a branch: Cellular mechanisms of the initiation of axon collateral branches. Mol Cell Neurosci 2017; 84:36-47. [PMID: 28359843 DOI: 10.1016/j.mcn.2017.03.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/03/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022] Open
Abstract
The formation of axon collateral branches from the pre-existing shafts of axons is an important aspect of neurodevelopment and the response of the nervous system to injury. This article provides an overview of the role of the cytoskeleton and signaling mechanisms in the formation of axon collateral branches. Both the actin filament and microtubule components of the cytoskeleton are required for the formation of axon branches. Recent work has begun to shed light on how these two elements of the cytoskeleton are integrated by proteins that functionally or physically link the cytoskeleton. While a number of signaling pathways have been determined as having a role in the formation of axon branches, the complexity of the downstream mechanisms and links to specific signaling pathways remain to be fully determined. The regulation of intra-axonal protein synthesis and organelle function are also emerging as components of signal-induced axon branching. Although much has been learned in the last couple of decades about the mechanistic basis of axon branching we can look forward to continue elucidating this complex biological phenomenon with the aim of understanding how multiple signaling pathways, cytoskeletal regulators and organelles are coordinated locally along the axon to give rise to a branch.
Collapse
Affiliation(s)
- Lorena Armijo-Weingart
- Shriners Pediatric Research Center, Temple University, Department of Anatomy and Cell Biology, 3500 North Broad St, Philadelphia, PA 19140, United States
| | - Gianluca Gallo
- Shriners Pediatric Research Center, Temple University, Department of Anatomy and Cell Biology, 3500 North Broad St, Philadelphia, PA 19140, United States.
| |
Collapse
|
33
|
Lombardi L, Persiconi I, Gallo A, Hoogenraad CC, De Stefano ME. NGF-dependent axon growth and regeneration are altered in sympathetic neurons of dystrophic mdx mice. Mol Cell Neurosci 2017; 80:1-17. [PMID: 28161362 DOI: 10.1016/j.mcn.2017.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 12/09/2016] [Accepted: 01/29/2017] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal disease, determined by lack of dystrophin (Dp427), a muscular cytoskeletal protein also expressed by selected neuronal populations. Consequently, besides muscular wasting, both human patients and DMD animal models suffer several neural disorders. In previous studies on the superior cervical ganglion (SCG) of wild type and dystrophic mdx mice (Lombardi et al. 2008), we hypothesized that Dp427 could play some role in NGF-dependent axonal growth, both during development and adulthood. To address this issue, we first analyzed axon regeneration potentials of SCG neurons of both genotypes after axotomy in vivo. While noradrenergic innervation of mdx mouse submandibular gland, main source of nerve growth factor (NGF), recovered similarly to wild type, iris innervation (muscular target) never did. We, therefore, evaluated whether dystrophic SCG neurons were poorly responsive to NGF, especially at low concentration. Following in vitro axotomy in the presence of either 10 or 50ng/ml NGF, the number of regenerated axons in mdx mouse neuron cultures was indeed reduced, compared to wild type, at the lower concentration. Neurite growth parameters (i.e. number, length), growth cone dynamics and NGF/TrkA receptor signaling in differentiating neurons (not injured) were also significantly reduced when cultured with 10ng/ml NGF, but also with higher NGF concentrations. In conclusion, we propose a role for Dp427 in NGF-dependent cytoskeletal dynamics associated to growth cone advancement, possibly through indirect stabilization of TrkA receptors. Considering NGF activity in nervous system development/remodeling, this aspect could concur in some of the described DMD-associated neural dysfunctions.
Collapse
Affiliation(s)
- Loredana Lombardi
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185 Roma, Italy
| | - Irene Persiconi
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185 Roma, Italy
| | - Alessandra Gallo
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185 Roma, Italy
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, 3584CH Utrecht, The Netherlands
| | - Maria Egle De Stefano
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza Università di Roma, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185 Roma, Italy; Center for Research in Neurobiology "Daniel Bovet", Sapienza Università di Roma, 00185 Roma, Italy.
| |
Collapse
|
34
|
Autophagy and epithelial-mesenchymal transition: an intricate interplay in cancer. Cell Death Dis 2016; 7:e2520. [PMID: 27929542 PMCID: PMC5260980 DOI: 10.1038/cddis.2016.415] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 12/19/2022]
Abstract
Autophagy and epithelial to mesenchymal transition (EMT) are major biological processes in cancer. Autophagy is a catabolic pathway that aids cancer cells to overcome intracellular or environmental stress, including nutrient deprivation, hypoxia and drugs effect. EMT is a complex transdifferentiation through which cancer cells acquire mesenchymal features, including motility and metastatic potential. Recent observations indicate that these two processes are linked in a complex relationship. On the one side, cells that underwent EMT require autophagy activation to survive during the metastatic spreading. On the other side, autophagy, acting as oncosuppressive signal, tends to inhibit the early phases of metastasization, contrasting the activation of the EMT mainly by selectively destabilizing crucial mediators of this process. Currently, still limited information is available regarding the molecular hubs at the interplay between autophagy and EMT. However, a growing number of evidence points to the functional interaction between cytoskeleton and mitochondria as one of the crucial regulatory center at the crossroad between these two biological processes. Cytoskeleton and mitochondria are linked in a tight functional relationship. Controlling mitochondria dynamics, the cytoskeleton cooperates to dictate mitochondria availability for the cell. Vice versa, the number and structure of mitochondria, which are primarily affected by autophagy-related processes, define the energy supply that cancer cells use to reorganize the cytoskeleton and to sustain cell movement during EMT. In this review, we aim to revise the evidence on the functional crosstalk between autophagy and EMT in cancer and to summarize the data supporting a parallel regulation of these two processes through shared signaling pathways. Furthermore, we intend to highlight the relevance of cytoskeleton and mitochondria in mediating the interaction between autophagy and EMT in cancer.
Collapse
|
35
|
Neuritin Mediates Activity-Dependent Axonal Branch Formation in Part via FGF Signaling. J Neurosci 2016; 36:4534-48. [PMID: 27098696 DOI: 10.1523/jneurosci.1715-15.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 03/09/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Aberrant branch formation of granule cell axons (mossy fiber sprouting) is observed in the dentate gyrus of many patients with temporal lobe epilepsy and in animal models of epilepsy. However, the mechanisms underlying mossy fiber sprouting remain elusive. Based on the hypothesis that seizure-mediated gene expression induces abnormal mossy fiber growth, we screened activity-regulated genes in the hippocampus and found that neuritin, an extracellular protein anchored to the cell surface, was rapidly upregulated after electroconvulsive seizures. Overexpression of neuritin in the cultured rat granule cells promoted their axonal branching. Also, kainic acid-dependent axonal branching was abolished in the cultured granule cells fromneuritinknock-out mice, suggesting that neuritin may be involved in activity-dependent axonal branching. Moreover,neuritinknock-out mice showed less-severe seizures in chemical kindling probably by reduced mossy fiber sprouting and/or increased seizure resistance. We found that inhibition of the fibroblast growth factor (FGF) receptor attenuated the neuritin-dependent axonal branching. FGF administration also increased branching in granule neurons, whereasneuritinknock-out mice did not show FGF-dependent axonal branching. In addition, FGF and neuritin treatment enhanced the recruitment of FGF receptors to the cell surface. These findings suggest that neuritin and FGF cooperate in inducing mossy fiber sprouting through FGF signaling. Together, these results suggest that FGF and neuritin-mediated axonal branch induction are involved in the aggravation of epilepsy. SIGNIFICANCE STATEMENT This study reveals the molecular mechanism underlying mossy fiber sprouting. Mossy fiber sprouting is the aberrant axonal branching of granule neurons in the hippocampus, which is observed in patients with epilepsy. Excess amounts of neuritin, a protein upregulated by neural activity, promoted axonal branching in granule neurons. A deficiency of neuritin suppressed mossy fiber sprouting and resulted in mitigation of seizure severity. Neuritin and fibroblast growth factor (FGF) cooperated in stimulating FGF signaling and enhancing axonal branching. Neuritin is necessary for FGF-mediated recruitment of FGF receptors to the cell surface. The recruitment of FGF receptors would promote axonal branching. The discovery of this new mechanism should contribute to the development of novel antiepileptic drugs to inhibit axonal branching via neuritin-FGF signaling.
Collapse
|
36
|
The Adaptor Protein CD2AP Is a Coordinator of Neurotrophin Signaling-Mediated Axon Arbor Plasticity. J Neurosci 2016; 36:4259-75. [PMID: 27076424 DOI: 10.1523/jneurosci.2423-15.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 02/14/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Growth of intact axons of noninjured neurons, often termed collateral sprouting, contributes to both adaptive and pathological plasticity in the adult nervous system, but the intracellular factors controlling this growth are largely unknown. An automated functional assay of genes regulated in sensory neurons from the rat in vivo spared dermatome model of collateral sprouting identified the adaptor protein CD2-associated protein (CD2AP; human CMS) as a positive regulator of axon growth. In non-neuronal cells, CD2AP, like other adaptor proteins, functions to selectively control the spatial/temporal assembly of multiprotein complexes that transmit intracellular signals. Although CD2AP polymorphisms are associated with increased risk of late-onset Alzheimer's disease, its role in axon growth is unknown. Assessments of neurite arbor structure in vitro revealed CD2AP overexpression, and siRNA-mediated knockdown, modulated (1) neurite length, (2) neurite complexity, and (3) growth cone filopodia number, in accordance with CD2AP expression levels. We show, for the first time, that CD2AP forms a novel multiprotein complex with the NGF receptor TrkA and the PI3K regulatory subunit p85, with the degree of TrkA:p85 association positively regulated by CD2AP levels. CD2AP also regulates NGF signaling through AKT, but not ERK, and regulates long-range signaling though TrkA(+)/RAB5(+) signaling endosomes. CD2AP mRNA and protein levels were increased in neurons during collateral sprouting but decreased following injury, suggesting that, although typically considered together, these two adult axonal growth processes are fundamentally different. These data position CD2AP as a major intracellular signaling molecule coordinating NGF signaling to regulate collateral sprouting and structural plasticity of intact adult axons. SIGNIFICANCE STATEMENT Growth of noninjured axons in the adult nervous system contributes to adaptive and maladaptive plasticity, and dysfunction of this process may contribute to neurologic pathologies. Functional screening of genes regulated during growth of noninjured axons revealed CD2AP as a positive regulator of axon outgrowth. A novel association of CD2AP with TrkA and p85 suggests a distinct intracellular signaling pathway regulating growth of noninjured axons. This may also represent a novel mechanism of generating specificity in multifunctional NGF signaling. Divergent regulation of CD2AP in different axon growth conditions suggests that separate mechanisms exist for different modes of axon growth. CD2AP is the first signaling molecule associated with adult sensory axonal collateral sprouting, and this association may offer new insights for NGF/TrkA-related Alzheimer's disease mechanisms.
Collapse
|
37
|
Saha T, Rathmann I, Viplav A, Panzade S, Begemann I, Rasch C, Klingauf J, Matis M, Galic M. Automated analysis of filopodial length and spatially resolved protein concentration via adaptive shape tracking. Mol Biol Cell 2016; 27:3616-3626. [PMID: 27535428 PMCID: PMC5221593 DOI: 10.1091/mbc.e16-06-0406] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/11/2016] [Indexed: 12/05/2022] Open
Abstract
A novel approach based on the convex-hull algorithm is used for parallel analysis of growth dynamics and relative spatiotemporal protein concentration along flexible filopodial protrusions. Testing of filopodia formation in silico, in vitro, and in vivo validates the robustness and sensitivity of the proposed approach. Filopodia are dynamic, actin-rich structures that transiently form on a variety of cell types. To understand the underlying control mechanisms requires precise monitoring of localization and concentration of individual regulatory and structural proteins as filopodia elongate and subsequently retract. Although several methods exist that analyze changes in filopodial shape, a software solution to reliably correlate growth dynamics with spatially resolved protein concentration along the filopodium independent of bending, lateral shift, or tilting is missing. Here we introduce a novel approach based on the convex-hull algorithm for parallel analysis of growth dynamics and relative spatiotemporal protein concentration along flexible filopodial protrusions. Detailed in silico tests using various geometries confirm that our technique accurately tracks growth dynamics and relative protein concentration along the filopodial length for a broad range of signal distributions. To validate our technique in living cells, we measure filopodial dynamics and quantify spatiotemporal localization of filopodia-associated proteins during the filopodial extension–retraction cycle in a variety of cell types in vitro and in vivo. Together these results show that the technique is suitable for simultaneous analysis of growth dynamics and spatiotemporal protein enrichment along filopodia. To allow readily application by other laboratories, we share source code and instructions for software handling.
Collapse
Affiliation(s)
- Tanumoy Saha
- DFG Cluster of Excellence Cells in Motion (EXC 1003), University of Münster, 48149 Münster, Germany.,Institute of Medical Physics and Biophysics, University of Münster, 48149 Münster, Germany
| | - Isabel Rathmann
- DFG Cluster of Excellence Cells in Motion (EXC 1003), University of Münster, 48149 Münster, Germany.,Institute of Medical Physics and Biophysics, University of Münster, 48149 Münster, Germany
| | - Abhiyan Viplav
- DFG Cluster of Excellence Cells in Motion (EXC 1003), University of Münster, 48149 Münster, Germany.,Institute of Medical Physics and Biophysics, University of Münster, 48149 Münster, Germany
| | - Sadhana Panzade
- DFG Cluster of Excellence Cells in Motion (EXC 1003), University of Münster, 48149 Münster, Germany.,Institute of Cell Biology, University of Münster, 48149 Münster, Germany
| | - Isabell Begemann
- DFG Cluster of Excellence Cells in Motion (EXC 1003), University of Münster, 48149 Münster, Germany.,Institute of Medical Physics and Biophysics, University of Münster, 48149 Münster, Germany
| | - Christiane Rasch
- Institute of Medical Physics and Biophysics, University of Münster, 48149 Münster, Germany
| | - Jürgen Klingauf
- DFG Cluster of Excellence Cells in Motion (EXC 1003), University of Münster, 48149 Münster, Germany.,Institute of Medical Physics and Biophysics, University of Münster, 48149 Münster, Germany
| | - Maja Matis
- DFG Cluster of Excellence Cells in Motion (EXC 1003), University of Münster, 48149 Münster, Germany.,Institute of Cell Biology, University of Münster, 48149 Münster, Germany
| | - Milos Galic
- DFG Cluster of Excellence Cells in Motion (EXC 1003), University of Münster, 48149 Münster, Germany .,Institute of Medical Physics and Biophysics, University of Münster, 48149 Münster, Germany
| |
Collapse
|
38
|
Sainath R, Ketschek A, Grandi L, Gallo G. CSPGs inhibit axon branching by impairing mitochondria-dependent regulation of actin dynamics and axonal translation. Dev Neurobiol 2016; 77:454-473. [PMID: 27429169 DOI: 10.1002/dneu.22420] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/28/2016] [Accepted: 07/14/2016] [Indexed: 12/27/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) inhibit the formation of axon collateral branches. The regulation of the axonal cytoskeleton and mitochondria are important components of the mechanism of branching. Actin-dependent axonal plasticity, reflected in the dynamics of axonal actin patches and filopodia, is greatest along segments of the axon populated by mitochondria. It is reported that CSPGs partially depolarize the membrane potential of axonal mitochondria, which impairs the dynamics of the axonal actin cytoskeleton and decreases the formation and duration of axonal filopodia, the first steps in the mechanism of branching. The effects of CSPGs on actin cytoskeletal dynamics are specific to axon segments populated by mitochondria. In contrast, CSPGs do not affect the microtubule content of axons, or the localization of microtubules into axonal filopodia, a required step in the mechanism of branch formation. It is also reported that CSPGs decrease the mitochondria-dependent axonal translation of cortactin, an actin associated protein involved in branching. Finally, the inhibitory effects of CSPGs on axon branching, actin cytoskeletal dynamics and the axonal translation of cortactin are reversed by culturing neurons with acetyl-l-carnitine, which promotes mitochondrial respiration. Collectively these data indicate that CSPGs impair mitochondrial function in axons, an effect which contributes to the inhibition of axon branching. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.
Collapse
Affiliation(s)
- Rajiv Sainath
- Temple University School of Medicine, Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, 3500 N Broad St, Philadelphia, Pennsylvania
| | - Andrea Ketschek
- Temple University School of Medicine, Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, 3500 N Broad St, Philadelphia, Pennsylvania
| | - Leah Grandi
- Temple University School of Medicine, Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, 3500 N Broad St, Philadelphia, Pennsylvania
| | - Gianluca Gallo
- Temple University School of Medicine, Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, 3500 N Broad St, Philadelphia, Pennsylvania
| |
Collapse
|
39
|
Winkle CC, Taylor KL, Dent EW, Gallo G, Greif KF, Gupton SL. Beyond the cytoskeleton: The emerging role of organelles and membrane remodeling in the regulation of axon collateral branches. Dev Neurobiol 2016; 76:1293-1307. [PMID: 27112549 DOI: 10.1002/dneu.22398] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/11/2016] [Accepted: 04/21/2016] [Indexed: 12/19/2022]
Abstract
The generation of axon collateral branches is a fundamental aspect of the development of the nervous system and the response of axons to injury. Although much has been discovered about the signaling pathways and cytoskeletal dynamics underlying branching, additional aspects of the cell biology of axon branching have received less attention. This review summarizes recent advances in our understanding of key factors involved in axon branching. This article focuses on how cytoskeletal mechanisms, intracellular organelles, such as mitochondria and the endoplasmic reticulum, and membrane remodeling (exocytosis and endocytosis) contribute to branch initiation and formation. Together this growing literature provides valuable insight as well as a platform for continued investigation into how multiple aspects of axonal cell biology are spatially and temporally orchestrated to give rise to axon branches. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1293-1307, 2016.
Collapse
Affiliation(s)
- Cortney C Winkle
- Neurobiology Curriculum, University of North Carolina, Chapel Hill, North Carolina, 27599
| | - Kendra L Taylor
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Erik W Dent
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Gianluca Gallo
- Lewis Katz School of Medicine, Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, Pennsylvania, 19140
| | - Karen F Greif
- Department of Biology, Bryn Mawr College, Bryn Mawr, Pennsylvania, 19010
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, 27599
| |
Collapse
|
40
|
Hotulainen P, Saarikangas J. The initiation of post-synaptic protrusions. Commun Integr Biol 2016; 9:e1125053. [PMID: 27489575 PMCID: PMC4951170 DOI: 10.1080/19420889.2015.1125053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 11/20/2015] [Accepted: 11/20/2015] [Indexed: 02/02/2023] Open
Abstract
The post-synaptic spines of neuronal dendrites are highly elaborate membrane protrusions. Their anatomy, stability and density are intimately linked to cognitive performance. The morphological transitions of spines are powered by coordinated polymerization of actin filaments against the plasma membrane, but how the membrane-associated polymerization is spatially and temporally regulated has remained ill defined. Here, we discuss our recent findings showing that dendritic spines can be initiated by direct membrane bending by the I-BAR protein MIM/Mtss1. This lipid phosphatidylinositol (PI(4,5)P2) signaling-activated membrane bending coordinated spatial actin assembly and promoted spine formation. From recent advances, we formulate a general model to discuss how spatially concentrated protein-lipid microdomains formed by multivalent interactions between lipids and actin/membrane regulatory proteins might launch cell protrusions.
Collapse
Affiliation(s)
- Pirta Hotulainen
- Neuroscience Center, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | | |
Collapse
|
41
|
O'Neill E, Kwok B, Day JS, Connor TJ, Harkin A. Amitriptyline protects against TNF-α-induced atrophy and reduction in synaptic markers via a Trk-dependent mechanism. Pharmacol Res Perspect 2016; 4:e00195. [PMID: 27069625 PMCID: PMC4804321 DOI: 10.1002/prp2.195] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/09/2015] [Indexed: 12/20/2022] Open
Abstract
Neuritic degeneration and synaptic loss are features of both neuroinflammation and neurodegenerative disease. The tricyclic antidepressant amitriptyline has neurotrophic and anti-inflammatory properties and acts as a novel agonist of the neurotrophin Trk receptors. Primary cortical neurons were treated with amitriptyline, nortriptyline and NGF and tested for neuronal complexity by Sholl analysis, protein expression by Western immunoblotting, and synapse number by colocalization of pre and postsynaptic makers. Amitriptyline (500 nmol/L) and its active metabolite nortriptyline (50 nmol/L) are found to induce neurite outgrowth in rat primary cortical neurons. Amitriptyline-induced neurite outgrowth is blocked by inhibition of Trk signaling using Trk antagonist K252a (200 nmol/L) but not by the neurotrophin inhibitor Y1036 (40 μmol/L), indicating that amitriptyline binds directly to the Trk receptor to initiate neurite outgrowth. MEK inhibitor PD98059 (10 μmol/L) also blocks amitriptyline-induced neurite outgrowth, implicating activation of the MAPK signaling pathway downstream of Trk receptor activation. Furthermore, pretreatment of primary cortical neurons with amitriptyline and nortriptyline prevents the effects of the proinflammatory cytokine TNF-α (10 ng/mL) on neurite outgrowth and colocalization of synaptic proteins. These findings suggest that amitriptyline and nortriptyline can exert neurotrophic effects in primary cortical neurons via activation of a Trk/MAPK signaling pathway. These compounds therefore have significant potential to be used in the treatment of neurodegenerative conditions where atrophy and loss of synaptic connections contribute to progression of disease.
Collapse
Affiliation(s)
- Eimear O'Neill
- Neuropsychopharmacology Research Group School of Pharmacy & Pharmaceutical Sciences and Trinity College Institute of Neuroscience Dublin Ireland; Neuroimmunology Research Group Department of Physiology School of Medicine and Trinity College Institute of Neuroscience Dublin Ireland
| | - Billy Kwok
- Neuroimmunology Research Group Department of Physiology School of Medicine and Trinity College Institute of Neuroscience Dublin Ireland
| | - Jennifer S Day
- Neuroimmunology Research Group Department of Physiology School of Medicine and Trinity College Institute of Neuroscience Dublin Ireland
| | - Thomas J Connor
- Neuroimmunology Research Group Department of Physiology School of Medicine and Trinity College Institute of Neuroscience Dublin Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group School of Pharmacy & Pharmaceutical Sciences and Trinity College Institute of Neuroscience Dublin Ireland
| |
Collapse
|
42
|
Ketschek A, Spillane M, Dun XP, Hardy H, Chilton J, Gallo G. Drebrin coordinates the actin and microtubule cytoskeleton during the initiation of axon collateral branches. Dev Neurobiol 2016; 76:1092-110. [PMID: 26731339 DOI: 10.1002/dneu.22377] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/07/2015] [Accepted: 01/01/2016] [Indexed: 11/10/2022]
Abstract
Drebrin is a cytoskeleton-associated protein which can interact with both actin filaments and the tips of microtubules. Its roles have been studied mostly in dendrites, and the functions of drebrin in axons are less well understood. In this study, we analyzed the role of drebrin, through shRNA-mediated depletion and overexpression, in the collateral branching of chicken embryonic sensory axons. We report that drebrin promotes the formation of axonal filopodia and collateral branches in vivo and in vitro. Live imaging of cytoskeletal dynamics revealed that drebrin promotes the formation of filopodia from precursor structures termed axonal actin patches. Endogenous drebrin localizes to actin patches and depletion studies indicate that drebrin contributes to the development of patches. In filopodia, endogenous drebrin localizes to the proximal portion of the filopodium. Drebrin was found to promote the stability of axonal filopodia and the entry of microtubule plus tips into axonal filopodia. The effects of drebrin on the stabilization of filopodia are independent of its effects on promoting microtubule targeting to filopodia. Inhibition of myosin II induces a redistribution of endogenous drebrin distally into filopodia, and further increases branching in drebrin overexpressing neurons. Finally, a 30 min treatment with the branch-inducing signal nerve growth factor increases the levels of axonal drebrin. This study determines the specific roles of drebrin in the regulation of the axonal cytoskeleton, and provides evidence that drebrin contributes to the coordination of the actin and microtubule cytoskeleton during the initial stages of axon branching. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1092-1110, 2016.
Collapse
Affiliation(s)
- Andrea Ketschek
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, 3500 N. Broad St, Philadelphia, Pennsylvania, 19140
| | - Mirela Spillane
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, 3500 N. Broad St, Philadelphia, Pennsylvania, 19140
| | - Xin-Peng Dun
- Peninsula Schools of Medicine and Dentistry, University of Plymouth, Plymouth Science Park, Research Way, Plymouth, PL6 8BU, United Kingdom
| | - Holly Hardy
- RILD Building, University of Exeter Medical School, Wellcome Wolfson Medical Research Centre, Barrack Road, Exeter, EX2 5DW, United Kingdom
| | - John Chilton
- RILD Building, University of Exeter Medical School, Wellcome Wolfson Medical Research Centre, Barrack Road, Exeter, EX2 5DW, United Kingdom
| | - Gianluca Gallo
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, 3500 N. Broad St, Philadelphia, Pennsylvania, 19140
| |
Collapse
|
43
|
Zhang SX, Duan LH, Qian H, Yu X. Actin Aggregations Mark the Sites of Neurite Initiation. Neurosci Bull 2016; 32:1-15. [PMID: 26779918 DOI: 10.1007/s12264-016-0012-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/25/2015] [Indexed: 12/25/2022] Open
Abstract
A salient feature of neurons is their intrinsic ability to grow and extend neurites, even in the absence of external cues. Compared to the later stages of neuronal development, such as neuronal polarization and dendrite morphogenesis, the early steps of neuritogenesis remain relatively unexplored. Here we showed that redistribution of cortical actin into large aggregates preceded neuritogenesis and determined the site of neurite initiation. Enhancing actin polymerization by jasplakinolide treatment effectively blocked actin redistribution and neurite initiation, while treatment with the actin depolymerizing agents latrunculin A or cytochalasin D accelerated neurite formation. Together, these results demonstrate a critical role of actin dynamics and reorganization in neurite initiation. Further experiments showed that microtubule dynamics and protein synthesis are not required for neurite initiation, but are required for later neurite stabilization. The redistribution of actin during early neuronal development was also observed in the cerebral cortex and hippocampus in vivo.
Collapse
Affiliation(s)
- Shu-Xin Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Li-Hui Duan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Qian
- Department of Applied Mathematics, University of Washington, Seattle, Washington, 98195, USA
| | - Xiang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
44
|
Eckharter C, Junker N, Winter L, Fischer I, Fogli B, Kistner S, Pfaller K, Zheng B, Wiche G, Klimaschewski L, Schweigreiter R. Schwann Cell Expressed Nogo-B Modulates Axonal Branching of Adult Sensory Neurons Through the Nogo-B Receptor NgBR. Front Cell Neurosci 2015; 9:454. [PMID: 26635533 PMCID: PMC4655273 DOI: 10.3389/fncel.2015.00454] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/05/2015] [Indexed: 11/13/2022] Open
Abstract
In contrast to the central nervous system (CNS) nerve fibers do regenerate in the peripheral nervous system (PNS) although in a clinically unsatisfying manner. A major problem is excessive sprouting of regenerating axons which results in aberrant reinnervation of target tissue and impaired functional recovery. In the CNS, the reticulon protein Nogo-A has been identified as a prominent oligodendrocyte expressed inhibitor of long-distance growth of regenerating axons. We show here that the related isoform Nogo-B is abundantly expressed in Schwann cells in the PNS. Other than Nogo-A in oligodendrocytes, Nogo-B does not localize to the myelin sheath but is detected in the ER and the plasma membrane of Schwann cells. Adult sensory neurons that are cultured on nogo-a/b deficient Schwann cells form significantly fewer axonal branches vs. those on wildtype Schwann cells, while their maximal axonal extension is unaffected. We demonstrate that this effect of Nogo-B on neuronal morphology is restricted to undifferentiated Schwann cells and is mediated by direct physical contact between these two cell types. Moreover, we show that blocking the Nogo-B specific receptor NgBR, which we find expressed on sensory neurons and to interact with Schwann cell expressed Nogo-B, produces the same branching phenotype as observed after deletion of Nogo-B. These data provide evidence for a novel function of the nogo gene that is implemented by the Nogo-B isoform. The remarkably specific effects of Nogo-B/NgBR on axonal branching, while leaving axonal extension unaffected, are of potential clinical relevance in the context of excessive axonal sprouting after peripheral nerve injury.
Collapse
Affiliation(s)
- Christoph Eckharter
- Division of Neurobiochemistry, Biocenter, Innsbruck Medical University Innsbruck, Austria
| | - Nina Junker
- Division of Neurobiochemistry, Biocenter, Innsbruck Medical University Innsbruck, Austria
| | - Lilli Winter
- Max F. Perutz Laboratories, Department of Biochemistry and Cell Biology, University of Vienna Vienna, Austria
| | - Irmgard Fischer
- Max F. Perutz Laboratories, Department of Biochemistry and Cell Biology, University of Vienna Vienna, Austria
| | - Barbara Fogli
- Department of Anatomy, Histology and Embryology, Division of Neuroanatomy, Innsbruck Medical University Innsbruck, Austria
| | - Steffen Kistner
- Division of Neurobiochemistry, Biocenter, Innsbruck Medical University Innsbruck, Austria
| | - Kristian Pfaller
- Department of Anatomy, Histology and Embryology, Division of Histology and Embryology, Innsbruck Medical University Innsbruck, Austria
| | - Binhai Zheng
- Department of Neurosciences and Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego La Jolla, CA, USA
| | - Gerhard Wiche
- Max F. Perutz Laboratories, Department of Biochemistry and Cell Biology, University of Vienna Vienna, Austria
| | - Lars Klimaschewski
- Department of Anatomy, Histology and Embryology, Division of Neuroanatomy, Innsbruck Medical University Innsbruck, Austria
| | - Rüdiger Schweigreiter
- Division of Neurobiochemistry, Biocenter, Innsbruck Medical University Innsbruck, Austria
| |
Collapse
|
45
|
Chetta J, Love JM, Bober BG, Shah SB. Bidirectional actin transport is influenced by microtubule and actin stability. Cell Mol Life Sci 2015; 72:4205-20. [PMID: 26043972 PMCID: PMC11113749 DOI: 10.1007/s00018-015-1933-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 05/14/2015] [Accepted: 05/19/2015] [Indexed: 12/16/2022]
Abstract
Local and long-distance transport of cytoskeletal proteins is vital to neuronal maintenance and growth. Though recent progress has provided insight into the movement of microtubules and neurofilaments, mechanisms underlying the movement of actin remain elusive, in large part due to rapid transitions between its filament states and its diverse cellular localization and function. In this work, we integrated live imaging of rat sensory neurons, image processing, multiple regression analysis, and mathematical modeling to perform the first quantitative, high-resolution investigation of GFP-actin identity and movement in individual axons. Our data revealed that filamentous actin densities arise along the length of the axon and move short but significant distances bidirectionally, with a net anterograde bias. We directly tested the role of actin and microtubules in this movement. We also confirmed a role for actin densities in extension of axonal filopodia, and demonstrated intermittent correlation of actin and mitochondrial movement. Our results support a novel mechanism underlying slow component axonal transport, in which the stability of both microtubule and actin cytoskeletal components influence the mobility of filamentous actin.
Collapse
Affiliation(s)
- Joshua Chetta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - James M Love
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Brian G Bober
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Sameer B Shah
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Departments of Orthopaedic Surgery and Bioengineering, University of California, San Diego, 9500 Gilman Drive, MC 0863, La Jolla, CA, 92093, USA.
| |
Collapse
|
46
|
Gallo G. Localized regulation of the axon shaft during the emergence of collateral branches. Neural Regen Res 2015; 10:1206-8. [PMID: 26487837 PMCID: PMC4590222 DOI: 10.4103/1673-5374.162694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Department of Anatomy and Cell Biology, Philadelphia, PA, USA
| |
Collapse
|
47
|
Leondaritis G, Eickholt BJ. Short Lives with Long-Lasting Effects: Filopodia Protrusions in Neuronal Branching Morphogenesis. PLoS Biol 2015; 13:e1002241. [PMID: 26334727 PMCID: PMC4559444 DOI: 10.1371/journal.pbio.1002241] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The branching behaviors of both dendrites and axons are part of a neuronal maturation process initiated by the generation of small and transient membrane protrusions. These are highly dynamic, actin-enriched structures, collectively called filopodia, which can mature in neurons to form stable branches. Consequently, the generation of filopodia protrusions is crucial during the formation of neuronal circuits and involves the precise control of an interplay between the plasma membrane and actin dynamics. In this issue of PLOS Biology, Hou and colleagues identify a Ca2+/CaM-dependent molecular machinery in dendrites that ensures proper targeting of branch formation by activation of the actin nucleator Cobl. A new study provides novel insight into how calcium signalling can control the branching of dendrites during nervous system development.
Collapse
Affiliation(s)
- George Leondaritis
- Department of Pharmacology, Medical School, University of Ioannina, Ioannina, Greece
| | - Britta Johanna Eickholt
- Institute of Biochemistry & Neuro Cure Cluster of Excellence, Charité – Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
48
|
Kelamangalath L, Tang X, Bezik K, Sterling N, Son YJ, Smith GM. Neurotrophin selectivity in organizing topographic regeneration of nociceptive afferents. Exp Neurol 2015; 271:262-78. [PMID: 26054884 DOI: 10.1016/j.expneurol.2015.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/22/2015] [Accepted: 06/03/2015] [Indexed: 01/28/2023]
Abstract
Neurotrophins represent some of the best candidates to enhance regeneration. In the current study, we investigated the effects of artemin, a member of the glial derived neurotrophic factor (GDNF) family, on sensory axon regeneration following a lumbar dorsal root injury and compared these effects with that observed after either NGF or GDNF expression in the rat spinal cord. Unlike previously published data, artemin failed to induce regeneration of large-diameter myelinated sensory afferents when expressed within either the spinal cord or DRG. However, artemin or NGF induced regeneration of calcitonin gene related peptide positive (CGRP(+)) axons only when expressed within the spinal cord. Accordingly, artemin or NGF enhanced recovery of only nociceptive behavior and showed a cFos distribution similar to the topography of regenerating axons. Artemin and GDNF signaling requires binding to different co-receptors (GFRα3 or GFRα1, respectively) prior to binding to the signaling receptor, cRet. Approximately 70% of DRG neurons express cRet, but only 35% express either co-receptor. To enhance artemin-induced regeneration, we co-expressed artemin with either GFRα3 or GDNF. Co-expression of artemin and GFRα3 only slightly enhanced regeneration of IB4(+) non-peptidergic nociceptive axons, but not myelinated axons. Interestingly, this co-expression also disrupted the ability of artemin to produce topographic targeting and lead to significant increases in cFos immunoreactivity within the deep dorsal laminae. This study failed to demonstrate artemin-induced regeneration of myelinated axons, even with co-expression of GFRα3, which only promoted mistargeted regeneration.
Collapse
Affiliation(s)
- Lakshmi Kelamangalath
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xiaoqing Tang
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Kathleen Bezik
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Noelle Sterling
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Young-Jin Son
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - George M Smith
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
49
|
Ketschek A, Jones S, Spillane M, Korobova F, Svitkina T, Gallo G. Nerve growth factor promotes reorganization of the axonal microtubule array at sites of axon collateral branching. Dev Neurobiol 2015; 75:1441-61. [PMID: 25846486 DOI: 10.1002/dneu.22294] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 03/13/2015] [Accepted: 03/30/2015] [Indexed: 12/31/2022]
Abstract
The localized debundling of the axonal microtubule array and the entry of microtubules into axonal filopodia are two defining features of collateral branching. We report that nerve growth factor (NGF), a branch-inducing signal, increases the frequency of microtubule debundling along the axon shaft of chicken embryonic sensory neurons. Sites of debundling correlate strongly with the localized targeting of microtubules into filopodia. Platinum replica electron microscopy suggests physical interactions between debundled microtubules and axonal actin filaments. However, as evidenced by depolymerization of actin filaments and inhibition of myosin II, actomyosin force generation does not promote debundling. In contrast, loss of actin filaments or inhibition of myosin II activity promotes debundling, indicating that axonal actomyosin forces suppress debundling. MAP1B is a microtubule associated protein that represses axon branching. Following treatment with NGF, microtubules penetrating filopodia during the early stages of branching exhibited lower levels of associated MAP1B. NGF increased and decreased the levels of MAP1B phosphorylated at a GSK-3β site (pMAP1B) along the axon shaft and within axonal filopodia, respectively. The levels of MAP1B and pMAP1B were not altered at sites of debundling, relative to the rest of the axon. Unlike the previously determined effects of NGF on the axonal actin cytoskeleton, the effects of NGF on microtubule debundling were not affected by inhibition of protein synthesis. Collectively, these data indicate that NGF promotes localized axonal microtubule debundling, that actomyosin forces antagonize microtubule debundling, and that NGF regulates pMAP1B in axonal filopodia during the early stages of collateral branch formation.
Collapse
Affiliation(s)
- Andrea Ketschek
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, Pennsylvania, 19140
| | - Steven Jones
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Mirela Spillane
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, Pennsylvania, 19140
| | - Farida Korobova
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Tatyana Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Gianluca Gallo
- Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, Pennsylvania, 19140
| |
Collapse
|
50
|
Yu L, Reynaud F, Falk J, Spencer A, Ding YD, Baumlé V, Lu R, Castellani V, Yuan C, Rudkin BB. Highly efficient method for gene delivery into mouse dorsal root ganglia neurons. Front Mol Neurosci 2015; 8:2. [PMID: 25698920 PMCID: PMC4313362 DOI: 10.3389/fnmol.2015.00002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/06/2015] [Indexed: 01/21/2023] Open
Abstract
The development of gene transfection technologies has greatly advanced our understanding of life sciences. While use of viral vectors has clear efficacy, it requires specific expertise and biological containment conditions. Electroporation has become an effective and commonly used method for introducing DNA into neurons and in intact brain tissue. The present study describes the use of the Neon® electroporation system to transfect genes into dorsal root ganglia neurons isolated from embryonic mouse Day 13.5–16. This cell type has been particularly recalcitrant and refractory to physical or chemical methods for introduction of DNA. By optimizing the culture condition and parameters including voltage and duration for this specific electroporation system, high efficiency (60–80%) and low toxicity (>60% survival) were achieved with robust differentiation in response to Nerve growth factor (NGF). Moreover, 3–50 times fewer cells are needed (6 × 104) compared with other traditional electroporation methods. This approach underlines the efficacy of this type of electroporation, particularly when only limited amount of cells can be obtained, and is expected to greatly facilitate the study of gene function in dorsal root ganglia neuron cultures.
Collapse
Affiliation(s)
- Lingli Yu
- Differentiation and Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR 5239, Centre National de la Recherche Scientifique, Ecole normale Supérieure de Lyon, University of Lyon 1 Claude Bernard, University of Lyon Lyon, France ; Laboratory of Molecular and Cellular Neurophysiology, East China Normal University Shanghai, China ; Joint Laboratory of Neuropathogenesis, Key Laboratory of Brain Functional Genomics, Chinese Ministry of Education, East China Normal University, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon Shanghai, China
| | - Florie Reynaud
- Centre de Génétique et Physiologie Moléculaire et Cellulaire, UMR Centre National de la Recherche Scientifique 5534, University of Lyon 1 Claude Bernard, University of Lyon Villeurbanne, France
| | - Julien Falk
- Centre de Génétique et Physiologie Moléculaire et Cellulaire, UMR Centre National de la Recherche Scientifique 5534, University of Lyon 1 Claude Bernard, University of Lyon Villeurbanne, France
| | - Ambre Spencer
- Differentiation and Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR 5239, Centre National de la Recherche Scientifique, Ecole normale Supérieure de Lyon, University of Lyon 1 Claude Bernard, University of Lyon Lyon, France ; Laboratory of Molecular and Cellular Neurophysiology, East China Normal University Shanghai, China ; Joint Laboratory of Neuropathogenesis, Key Laboratory of Brain Functional Genomics, Chinese Ministry of Education, East China Normal University, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon Shanghai, China
| | - Yin-Di Ding
- Differentiation and Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR 5239, Centre National de la Recherche Scientifique, Ecole normale Supérieure de Lyon, University of Lyon 1 Claude Bernard, University of Lyon Lyon, France ; Laboratory of Molecular and Cellular Neurophysiology, East China Normal University Shanghai, China ; Joint Laboratory of Neuropathogenesis, Key Laboratory of Brain Functional Genomics, Chinese Ministry of Education, East China Normal University, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon Shanghai, China
| | - Véronique Baumlé
- Differentiation and Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR 5239, Centre National de la Recherche Scientifique, Ecole normale Supérieure de Lyon, University of Lyon 1 Claude Bernard, University of Lyon Lyon, France ; Joint Laboratory of Neuropathogenesis, Key Laboratory of Brain Functional Genomics, Chinese Ministry of Education, East China Normal University, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon Shanghai, China
| | - Ruisheng Lu
- Laboratory of Molecular and Cellular Neurophysiology, East China Normal University Shanghai, China ; Joint Laboratory of Neuropathogenesis, Key Laboratory of Brain Functional Genomics, Chinese Ministry of Education, East China Normal University, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon Shanghai, China
| | - Valérie Castellani
- Centre de Génétique et Physiologie Moléculaire et Cellulaire, UMR Centre National de la Recherche Scientifique 5534, University of Lyon 1 Claude Bernard, University of Lyon Villeurbanne, France
| | - Chonggang Yuan
- Differentiation and Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR 5239, Centre National de la Recherche Scientifique, Ecole normale Supérieure de Lyon, University of Lyon 1 Claude Bernard, University of Lyon Lyon, France ; Joint Laboratory of Neuropathogenesis, Key Laboratory of Brain Functional Genomics, Chinese Ministry of Education, East China Normal University, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon Shanghai, China
| | - Brian B Rudkin
- Laboratory of Molecular and Cellular Neurophysiology, East China Normal University Shanghai, China ; Joint Laboratory of Neuropathogenesis, Key Laboratory of Brain Functional Genomics, Chinese Ministry of Education, East China Normal University, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon Shanghai, China
| |
Collapse
|