1
|
Bu A, Afghah F, Castro N, Bawa M, Kohli S, Shah K, Rios B, Butty V, Raman R. Actuating Extracellular Matrices Decouple the Mechanical and Biochemical Effects of Muscle Contraction on Motor Neurons. Adv Healthc Mater 2025; 14:e2403712. [PMID: 39523700 PMCID: PMC11874633 DOI: 10.1002/adhm.202403712] [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: 09/28/2024] [Revised: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Emerging in vivo evidence suggests that repeated muscle contraction, or exercise, impacts peripheral nerves. However, the difficulty of isolating the muscle-specific impact on motor neurons in vivo, as well as the inability to decouple the biochemical and mechanical impacts of muscle contraction in this setting, motivates investigating this phenomenon in vitro. This study demonstrates that tuning the mechanical properties of fibrin enables longitudinal culture of highly contractile skeletal muscle monolayers, enabling functional characterization of and long-term secretome harvesting from exercised tissues. Motor neurons stimulated with exercised muscle-secreted factors significantly upregulate neurite outgrowth and migration, with an effect size dependent on muscle contraction intensity. Actuating magnetic microparticles embedded within fibrin hydrogels enable dynamically stretching motor neurons and non-invasively mimicking the mechanical effects of muscle contraction. Interestingly, axonogenesis is similarly upregulated in both mechanically and biochemically stimulated motor neurons, but RNA sequencing reveals different transcriptomic signatures between groups, with biochemical stimulation having a greater impact on cell signaling related to axonogenesis and synapse maturation. This study leverages actuating extracellular matrices to robustly validate a previously hypothesized role for muscle contraction in regulating motor neuron growth and maturation from the bottom-up through both mechanical and biochemical signaling.
Collapse
Affiliation(s)
- Angel Bu
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ferdows Afghah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Nicolas Castro
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Maheera Bawa
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sonika Kohli
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Karina Shah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Brandon Rios
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Vincent Butty
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ritu Raman
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
2
|
Delpech C, Schaeffer J, Vilallongue N, Delaunay A, Benadjal A, Blot B, Excoffier B, Plissonnier E, Gascon E, Albert F, Paccard A, Saintpierre A, Gasnier C, Zagar Y, Castellani V, Belin S, Chédotal A, Nawabi H. Axon guidance during mouse central nervous system regeneration is required for specific brain innervation. Dev Cell 2024; 59:3213-3228.e8. [PMID: 39353435 DOI: 10.1016/j.devcel.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 07/11/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
Reconstructing functional neuronal circuits is one major challenge of central nervous system repair. Through activation of pro-growth signaling pathways, some neurons achieve long-distance axon regrowth. Yet, functional reconnection has hardly been obtained, as these regenerating axons fail to resume their initial trajectory and reinnervate their proper target. Axon guidance is considered to be active only during development. Here, using the mouse visual system, we show that axon guidance is still active in the adult brain in regenerative conditions. We highlight that regenerating retinal ganglion cell axons avoid one of their primary targets, the suprachiasmatic nucleus (SCN), due to Slit/Robo repulsive signaling. Together with promoting regeneration, silencing Slit/Robo in vivo enables regenerating axons to enter the SCN and form active synapses. The newly formed circuit is associated with neuronal activation and functional recovery. Our results provide evidence that axon guidance mechanisms are required to reconnect regenerating axons to specific brain nuclei.
Collapse
Affiliation(s)
- Céline Delpech
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Julia Schaeffer
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Noemie Vilallongue
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Apolline Delaunay
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Amin Benadjal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Beatrice Blot
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Blandine Excoffier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Elise Plissonnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eduardo Gascon
- Aix Marseille University, CNRS, INT, Institute of Neurosci Timone, Marseille, France
| | - Floriane Albert
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Antoine Paccard
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Ana Saintpierre
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Celestin Gasnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Valérie Castellani
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | - Stephane Belin
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France; Institut de pathologie, groupe hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Homaira Nawabi
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
3
|
Rapti G. Regulation of axon pathfinding by astroglia across genetic model organisms. Front Cell Neurosci 2023; 17:1241957. [PMID: 37941606 PMCID: PMC10628440 DOI: 10.3389/fncel.2023.1241957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/07/2023] [Indexed: 11/10/2023] Open
Abstract
Glia and neurons are intimately associated throughout bilaterian nervous systems, and were early proposed to interact for patterning circuit assembly. The investigations of circuit formation progressed from early hypotheses of intermediate guideposts and a "glia blueprint", to recent genetic and cell manipulations, and visualizations in vivo. An array of molecular factors are implicated in axon pathfinding but their number appears small relatively to circuit complexity. Comprehending this circuit complexity requires to identify unknown factors and dissect molecular topographies. Glia contribute to both aspects and certain studies provide molecular and functional insights into these contributions. Here, I survey glial roles in guiding axon navigation in vivo, emphasizing analogies, differences and open questions across major genetic models. I highlight studies pioneering the topic, and dissect recent findings that further advance our current molecular understanding. Circuits of the vertebrate forebrain, visual system and neural tube in zebrafish, mouse and chick, the Drosophila ventral cord and the C. elegans brain-like neuropil emerge as major contexts to study glial cell functions in axon navigation. I present astroglial cell types in these models, and their molecular and cellular interactions that drive axon guidance. I underline shared principles across models, conceptual or technical complications, and open questions that await investigation. Glia of the radial-astrocyte lineage, emerge as regulators of axon pathfinding, often employing common molecular factors across models. Yet this survey also highlights different involvements of glia in embryonic navigation or pioneer axon pathfinding, and unknowns in the molecular underpinnings of glial cell functions. Future cellular and molecular investigations should complete the comprehensive view of glial roles in circuit assembly.
Collapse
Affiliation(s)
- Georgia Rapti
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, Rome, Italy
- Interdisciplinary Center of Neurosciences, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
4
|
Guidance landscapes unveiled by quantitative proteomics to control reinnervation in adult visual system. Nat Commun 2022; 13:6040. [PMID: 36229455 PMCID: PMC9561644 DOI: 10.1038/s41467-022-33799-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
In the injured adult central nervous system (CNS), activation of pro-growth molecular pathways in neurons leads to long-distance regeneration. However, most regenerative fibers display guidance defects, which prevent reinnervation and functional recovery. Therefore, the molecular characterization of the proper target regions of regenerative axons is essential to uncover the modalities of adult reinnervation. In this study, we use mass spectrometry (MS)-based quantitative proteomics to address the proteomes of major nuclei of the adult visual system. These analyses reveal that guidance-associated molecules are expressed in adult visual targets. Moreover, we show that bilateral optic nerve injury modulates the expression of specific proteins. In contrast, the expression of guidance molecules remains steady. Finally, we show that regenerative axons are able to respond to guidance cues ex vivo, suggesting that these molecules possibly interfere with brain target reinnervation in adult. Using a long-distance regeneration model, we further demonstrate that the silencing of specific guidance signaling leads to rerouting of regenerative axons in vivo. Altogether, our results suggest ways to modulate axon guidance of regenerative neurons to achieve circuit repair in adult.
Collapse
|
5
|
Lan YX, Yang P, Zeng Z, Yadav N, Zhang LJ, Wang LB, Xia HC. Gene and protein expression profiles of olfactory ensheathing cells from olfactory bulb versus olfactory mucosa. Neural Regen Res 2022; 17:440-449. [PMID: 34269221 PMCID: PMC8463967 DOI: 10.4103/1673-5374.317986] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Olfactory ensheathing cells (OECs) from the olfactory bulb (OB) and the olfactory mucosa (OM) have the capacity to repair nerve injury. However, the difference in the therapeutic effect between OB-derived OECs and OM-derived OECs remains unclear. In this study, we extracted OECs from OB and OM and compared the gene and protein expression profiles of the cells using transcriptomics and non-quantitative proteomics techniques. The results revealed that both OB-derived OECs and OM-derived OECs highly expressed genes and proteins that regulate cell growth, proliferation, apoptosis and vascular endothelial cell regeneration. The differentially expressed genes and proteins of OB-derived OECs play a key role in regulation of nerve regeneration and axon regeneration and extension, transmission of nerve impulses and response to axon injury. The differentially expressed genes and proteins of OM-derived OECs mainly participate in the positive regulation of inflammatory response, defense response, cytokine binding, cell migration and wound healing. These findings suggest that differentially expressed genes and proteins may explain why OB-derived OECs and OM-derived OECs exhibit different therapeutic roles. This study was approved by the Animal Ethics Committee of the General Hospital of Ningxia Medical University (approval No. 2017-073) on February 13, 2017.
Collapse
Affiliation(s)
- Yuan-Xiang Lan
- School of Clinical Medicine, Ningxia Medical University; Ningxia Human Stem Cell Institute; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Ping Yang
- Clinical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhong Zeng
- School of Clinical Medicine, Ningxia Medical University; Ningxia Human Stem Cell Institute; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Neeraj Yadav
- Department of Neurosurgery, General Hospital of Ningxia Medical University; School of International Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Li-Jian Zhang
- School of Clinical Medicine, Ningxia Medical University; Ningxia Human Stem Cell Institute; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Li-Bin Wang
- Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - He-Chun Xia
- Ningxia Human Stem Cell Institute; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| |
Collapse
|
6
|
Muzyka VV, Badea TC. Genetic interplay between transcription factor Pou4f1/Brn3a and neurotrophin receptor Ret in retinal ganglion cell type specification. Neural Dev 2021; 16:5. [PMID: 34548095 PMCID: PMC8454062 DOI: 10.1186/s13064-021-00155-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background While the transcriptional code governing retinal ganglion cell (RGC) type specification begins to be understood, its interplay with neurotrophic signaling is largely unexplored. In mice, the transcription factor Brn3a/Pou4f1 is expressed in most RGCs, and is required for the specification of RGCs with small dendritic arbors. The Glial Derived Neurotrophic Factor (GDNF) receptor Ret is expressed in a subset of RGCs, including some expressing Brn3a, but its role in RGC development is not defined. Methods Here we use combinatorial genetic experiments using conditional knock-in reporter alleles at the Brn3a and Ret loci, in combination with retina- or Ret specific Cre drivers, to generate complete or mosaic genetic ablations of either Brn3a or Ret in RGCs. We then use sparse labelling to investigate Brn3a and Ret gene dosage effects on RGC dendritic arbor morphology. In addition, we use immunostaining and/or gene expression profiling by RNASeq to identify transcriptional targets relevant for the potential Brn3a-Ret interaction in RGC development. Results We find that mosaic gene dosage manipulation of the transcription factor Brn3a/Pou4f1 in neurotrophic receptor Ret heterozygote RGCs results in altered cell fate decisions and/or morphological dendritic defects. Specific RGC types are lost if Brn3a is ablated during embryogenesis and only mildly affected by postnatal Brn3a ablation. Sparse but not complete Brn3a heterozygosity combined with complete Ret heterozygosity has striking effects on RGC type distribution. Brn3a only mildly modulates Ret transcription, while Ret knockouts exhibit slightly skewed Brn3a and Brn3b expression during development that is corrected by adult age. Brn3a loss of function modestly but significantly affects distribution of Ret co-receptors GFRα1-3, and neurotrophin receptors TrkA and TrkC in RGCs. Conclusions Based on these observations, we propose that Brn3a and Ret converge onto developmental pathways that control RGC type specification, potentially through a competitive mechanism requiring signaling from the surrounding tissue. Supplementary Information The online version contains supplementary material available at 10.1186/s13064-021-00155-z.
Collapse
Affiliation(s)
- Vladimir Vladimirovich Muzyka
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, MD, USA. .,Institute of Cytology and Genetics, Novosibirsk State University, Novosibirsk, Russia.
| | - Tudor Constantin Badea
- Retinal Circuit Development & Genetics Unit, Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, NIH, Bethesda, MD, USA. .,Research and Development Institute, School of Medicine, Transilvania University of Brasov, Brasov, Romania.
| |
Collapse
|
7
|
Onesto MM, Short CA, Rempel SK, Catlett TS, Gomez TM. Growth Factors as Axon Guidance Molecules: Lessons From in vitro Studies. Front Neurosci 2021; 15:678454. [PMID: 34093120 PMCID: PMC8175860 DOI: 10.3389/fnins.2021.678454] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Growth cones at the tips of extending axons navigate through developing organisms by probing extracellular cues, which guide them through intermediate steps and onto final synaptic target sites. Widespread focus on a few guidance cue families has historically overshadowed potentially crucial roles of less well-studied growth factors in axon guidance. In fact, recent evidence suggests that a variety of growth factors have the ability to guide axons, affecting the targeting and morphogenesis of growth cones in vitro. This review summarizes in vitro experiments identifying responses and signaling mechanisms underlying axon morphogenesis caused by underappreciated growth factors.
Collapse
Affiliation(s)
| | | | | | | | - Timothy M. Gomez
- Neuroscience Training Program and Cell and Molecular Biology Program, Department of Neuroscience, University of Wisconsin–Madison, Madison, WI, United States
| |
Collapse
|
8
|
Baeriswyl T, Dumoulin A, Schaettin M, Tsapara G, Niederkofler V, Helbling D, Avilés E, Frei JA, Wilson NH, Gesemann M, Kunz B, Stoeckli ET. Endoglycan plays a role in axon guidance by modulating cell adhesion. eLife 2021; 10:64767. [PMID: 33650489 PMCID: PMC7946425 DOI: 10.7554/elife.64767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/25/2021] [Indexed: 01/27/2023] Open
Abstract
Axon navigation depends on the interactions between guidance molecules along the trajectory and specific receptors on the growth cone. However, our in vitro and in vivo studies on the role of Endoglycan demonstrate that in addition to specific guidance cue – receptor interactions, axon guidance depends on fine-tuning of cell-cell adhesion. Endoglycan, a sialomucin, plays a role in axon guidance in the central nervous system of chicken embryos, but it is neither an axon guidance cue nor a receptor. Rather, Endoglycan acts as a negative regulator of molecular interactions based on evidence from in vitro experiments demonstrating reduced adhesion of growth cones. In the absence of Endoglycan, commissural axons fail to properly navigate the midline of the spinal cord. Taken together, our in vivo and in vitro results support the hypothesis that Endoglycan acts as a negative regulator of cell-cell adhesion in commissural axon guidance.
Collapse
Affiliation(s)
- Thomas Baeriswyl
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Alexandre Dumoulin
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Martina Schaettin
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Georgia Tsapara
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Vera Niederkofler
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Denise Helbling
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Evelyn Avilés
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Jeannine A Frei
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Nicole H Wilson
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Gesemann
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Beat Kunz
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Esther T Stoeckli
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Short CA, Onesto MM, Rempel SK, Catlett TS, Gomez TM. Familiar growth factors have diverse roles in neural network assembly. Curr Opin Neurobiol 2021; 66:233-239. [PMID: 33477094 PMCID: PMC8058242 DOI: 10.1016/j.conb.2020.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023]
Abstract
The assembly of neuronal circuits during development depends on guidance of axonal growth cones by molecular cues deposited in their environment. While a number of families of axon guidance molecules have been identified and reviewed, important and diverse activities of traditional growth factors are emerging. Besides clear and well recognized roles in the regulation of cell division, differentiation and survival, new research shows later phase roles for a number of growth factors in promoting neuronal migration, axon guidance and synapse formation throughout the nervous system.
Collapse
Affiliation(s)
- Caitlin A Short
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States
| | - Massimo M Onesto
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States; Stanford University School of Medicine, United States
| | - Sarah K Rempel
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States
| | - Timothy S Catlett
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States
| | - Timothy M Gomez
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States.
| |
Collapse
|
10
|
Limoni G, Niquille M. Semaphorins and Plexins in central nervous system patterning: the key to it all? Curr Opin Neurobiol 2021; 66:224-232. [PMID: 33513538 DOI: 10.1016/j.conb.2020.12.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Semaphorins and Plexins constitute one of the largest family of guidance molecules and receptors involved in setting critical biological steps for central nervous system development. The role of these molecules in axonal development has been extensively characterized but Semaphorins and Plexins are also involved in a variety of other developmental processes, spanning from cell polarization to migration, laminar segregation and neuronal maturation. In this review, we aim to gather discoveries carried in the field of neurodevelopment over the last decade, during which Semaphorin/Plexin complexes have emerged as key regulators of neurogenesis, neural cell migration and adult gliogenesis. As well, we report mechanisms that brought a better understanding of axonal midline crossing.
Collapse
Affiliation(s)
- Greta Limoni
- Department of Basic Neuroscience, University Medical Center, University of Geneva, Rue Michel-Servet 1, 1211 Genève 4, Switzerland.
| | - Mathieu Niquille
- Department of Basic Neuroscience, University Medical Center, University of Geneva, Rue Michel-Servet 1, 1211 Genève 4, Switzerland.
| |
Collapse
|
11
|
Huang Y, Zhang B, Haneke H, Haage V, Lubas M, Yuan Y, Xia P, Motta E, Nanvuma C, Dzaye O, Hu F, Kettenmann H. Glial cell line-derived neurotrophic factor increases matrix metallopeptidase 9 and 14 expression in microglia and promotes microglia-mediated glioma progression. J Neurosci Res 2021; 99:1048-1063. [PMID: 33404121 DOI: 10.1002/jnr.24768] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/09/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is released by glioma cells and promotes tumor growth. We have previously found that GDNF released from the tumor cells is a chemoattractant for microglial cells, the immune cells of the central nervous system. Here we show that GDNF increases matrix metalloproteinase (MMP) 9 and MMP14 expression in cultured microglial cells from mixed sexes of neonatal mice. The GDNF-induced microglial MMP9 and MMP14 upregulation is mediated by GDNF family receptor alpha 1 receptors and dependent on p38 mitogen-activated protein kinase signaling. In organotypic brain slices, GDNF promotes the growth of glioma and this effect depends on the presence of microglia. We also previously found that MMP9 and MMP14 upregulation can be mediated by Toll-like receptor (TLR) 2 signaling and here we demonstrate that GDNF increases the expression of TLR1 and TLR2. In conclusion, GDNF promotes the pro-tumorigenic phenotype of microglia.
Collapse
Affiliation(s)
- Yimin Huang
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin, Berlin, Germany
| | - Baole Zhang
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Department of Neurobiology and Cell Biology, Xuzhou Medical University, Xuzhou, China
| | - Hannah Haneke
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Verena Haage
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Malgorzata Lubas
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Yang Yuan
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Pengfei Xia
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Edyta Motta
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Cynthia Nanvuma
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Omar Dzaye
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Feng Hu
- Department of Neurosurgery, Tongji Hospital of Huazhong University of Science and Technology, Wuhan, China
| | - Helmut Kettenmann
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
12
|
Ducuing H, Gardette T, Pignata A, Kindbeiter K, Bozon M, Thoumine O, Delloye-Bourgeois C, Tauszig-Delamasure S, Castellani V. SlitC-PlexinA1 mediates iterative inhibition for orderly passage of spinal commissural axons through the floor plate. eLife 2020; 9:e63205. [PMID: 33345773 PMCID: PMC7775108 DOI: 10.7554/elife.63205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Spinal commissural axon navigation across the midline in the floor plate requires repulsive forces from local Slit repellents. The long-held view is that Slits push growth cones forward and prevent them from turning back once they became sensitized to these cues after midline crossing. We analyzed with fluorescent reporters Slits distribution and FP glia morphology. We observed clusters of Slit-N and Slit-C fragments decorating a complex architecture of glial basal process ramifications. We found that PC2 proprotein convertase activity contributes to this pattern of ligands. Next, we studied Slit-C acting via PlexinA1 receptor shared with another FP repellent, the Semaphorin3B, through generation of a mouse model baring PlexinA1Y1815F mutation abrogating SlitC but not Sema3B responsiveness, manipulations in the chicken embryo, and ex vivo live imaging. This revealed a guidance mechanism by which SlitC constantly limits growth cone exploration, imposing ordered and forward-directed progression through aligned corridors formed by FP basal ramifications.
Collapse
Affiliation(s)
- Hugo Ducuing
- Institut NeuroMyoGène - CNRS UMR 5310 - INSERM U1217 de Lyon- UCBL Lyon 1, Faculté de Médecine et de PharmacieLyonFrance
| | - Thibault Gardette
- Institut NeuroMyoGène - CNRS UMR 5310 - INSERM U1217 de Lyon- UCBL Lyon 1, Faculté de Médecine et de PharmacieLyonFrance
| | - Aurora Pignata
- Institut NeuroMyoGène - CNRS UMR 5310 - INSERM U1217 de Lyon- UCBL Lyon 1, Faculté de Médecine et de PharmacieLyonFrance
| | - Karine Kindbeiter
- Institut NeuroMyoGène - CNRS UMR 5310 - INSERM U1217 de Lyon- UCBL Lyon 1, Faculté de Médecine et de PharmacieLyonFrance
| | - Muriel Bozon
- Institut NeuroMyoGène - CNRS UMR 5310 - INSERM U1217 de Lyon- UCBL Lyon 1, Faculté de Médecine et de PharmacieLyonFrance
| | - Olivier Thoumine
- Interdisciplinary Institute for Neuroscience, UMR CNRS 5297 - University of BordeauxBordeauxFrance
| | - Céline Delloye-Bourgeois
- Institut NeuroMyoGène - CNRS UMR 5310 - INSERM U1217 de Lyon- UCBL Lyon 1, Faculté de Médecine et de PharmacieLyonFrance
| | - Servane Tauszig-Delamasure
- Institut NeuroMyoGène - CNRS UMR 5310 - INSERM U1217 de Lyon- UCBL Lyon 1, Faculté de Médecine et de PharmacieLyonFrance
| | - Valerie Castellani
- Institut NeuroMyoGène - CNRS UMR 5310 - INSERM U1217 de Lyon- UCBL Lyon 1, Faculté de Médecine et de PharmacieLyonFrance
| |
Collapse
|
13
|
RET-independent signaling by GDNF ligands and GFRα receptors. Cell Tissue Res 2020; 382:71-82. [PMID: 32737575 PMCID: PMC7529620 DOI: 10.1007/s00441-020-03261-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022]
Abstract
The discovery in the late 1990s of the partnership between the RET receptor tyrosine kinase and the GFRα family of GPI-anchored co-receptors as mediators of the effects of GDNF family ligands galvanized the field of neurotrophic factors, firmly establishing a new molecular framework besides the ubiquitous neurotrophins. Soon after, however, it was realized that many neurons and brain areas expressed GFRα receptors without expressing RET. These observations led to the formulation of two new concepts in GDNF family signaling, namely, the non-cell-autonomous functions of GFRα molecules, so-called trans signaling, as well as cell-autonomous functions mediated by signaling receptors distinct from RET, which became known as RET-independent signaling. To date, the best studied RET-independent signaling pathway for GDNF family ligands involves the neural cell adhesion molecule NCAM and its association with GFRα co-receptors. Among the many functions attributed to this signaling system are neuronal migration, neurite outgrowth, dendrite branching, spine formation, and synaptogenesis. This review summarizes our current understanding of this and other mechanisms of RET-independent signaling by GDNF family ligands and GFRα receptors, as well as their physiological importance.
Collapse
|
14
|
Abstract
The spinal cord receives, relays and processes sensory information from the periphery and integrates this information with descending inputs from supraspinal centres to elicit precise and appropriate behavioural responses and orchestrate body movements. Understanding how the spinal cord circuits that achieve this integration are wired during development is the focus of much research interest. Several families of proteins have well-established roles in guiding developing spinal cord axons, and recent findings have identified new axon guidance molecules. Nevertheless, an integrated view of spinal cord network development is lacking, and many current models have neglected the cellular and functional diversity of spinal cord circuits. Recent advances challenge the existing spinal cord axon guidance dogmas and have provided a more complex, but more faithful, picture of the ontogenesis of vertebrate spinal cord circuits.
Collapse
|
15
|
Roig-Puiggros S, Vigouroux RJ, Beckman D, Bocai NI, Chiou B, Davimes J, Gomez G, Grassi S, Hoque A, Karikari TK, Kiffer F, Lopez M, Lunghi G, Mazengenya P, Meier S, Olguín-Albuerne M, Oliveira MM, Paraíso-Luna J, Pradhan J, Radiske A, Ramos-Hryb AB, Ribeiro MC, Schellino R, Selles MC, Singh S, Theotokis P, Chédotal A. Construction and reconstruction of brain circuits: normal and pathological axon guidance. J Neurochem 2019; 153:10-32. [PMID: 31630412 DOI: 10.1111/jnc.14900] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
Perception of our environment entirely depends on the close interaction between the central and peripheral nervous system. In order to communicate each other, both systems must develop in parallel and in coordination. During development, axonal projections from the CNS as well as the PNS must extend over large distances to reach their appropriate target cells. To do so, they read and follow a series of axon guidance molecules. Interestingly, while these molecules play critical roles in guiding developing axons, they have also been shown to be critical in other major neurodevelopmental processes, such as the migration of cortical progenitors. Currently, a major hurdle for brain repair after injury or neurodegeneration is the absence of axonal regeneration in the mammalian CNS. By contrasts, PNS axons can regenerate. Many hypotheses have been put forward to explain this paradox but recent studies suggest that hacking neurodevelopmental mechanisms may be the key to promote CNS regeneration. Here we provide a seminar report written by trainees attending the second Flagship school held in Alpbach, Austria in September 2018 organized by the International Society for Neurochemistry (ISN) together with the Journal of Neurochemistry (JCN). This advanced school has brought together leaders in the fields of neurodevelopment and regeneration in order to discuss major keystones and future challenges in these respective fields.
Collapse
Affiliation(s)
| | - Robin J Vigouroux
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Danielle Beckman
- California National Primate Research Center, UC Davis, Davis, California, USA
| | - Nadia I Bocai
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Brian Chiou
- Department of Pediatrics, University of California - San Francisco, San Francisco, California, USA
| | - Joshua Davimes
- Faculty of Health Sciences School of Anatomical Sciences, University of the Witwatersrand, Parktown Johannesburg, South Africa
| | - Gimena Gomez
- Laboratorio de Parkinson Experimental, Instituto de Investigaciones Farmacológicas (ININFA-CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Ashfaqul Hoque
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,School of Life Sciences, University of Warwick, Coventry, UK.,Midlands Integrative Biosciences Training Partnership, University of Warwick, Coventry, UK
| | - Frederico Kiffer
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Mary Lopez
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicin, University of Milano, Segrate, Italy
| | - Pedzisai Mazengenya
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sonja Meier
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mauricio M Oliveira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juan Paraíso-Luna
- Ramón y Cajal Institute of Health Research (IRYCIS), Department of Biochemistry and Molecular Biology and University Research Institute in Neurochemistry (IUIN), Complutense University, Madrid, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jonu Pradhan
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Andressa Radiske
- Memory Research Laboratory, Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Ana Belén Ramos-Hryb
- Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina.,Grupo de Neurociencia de Sistemas, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Mayara C Ribeiro
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York, USA
| | - Roberta Schellino
- Neuroscience Department "Rita Levi-Montalcini" and Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Torino, Italy
| | - Maria Clara Selles
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Shripriya Singh
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Paschalis Theotokis
- Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Macedonia, Greece
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
16
|
Pignata A, Ducuing H, Boubakar L, Gardette T, Kindbeiter K, Bozon M, Tauszig-Delamasure S, Falk J, Thoumine O, Castellani V. A Spatiotemporal Sequence of Sensitization to Slits and Semaphorins Orchestrates Commissural Axon Navigation. Cell Rep 2019; 29:347-362.e5. [PMID: 31597096 DOI: 10.1016/j.celrep.2019.08.098] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/05/2019] [Accepted: 08/28/2019] [Indexed: 01/24/2023] Open
Abstract
Accurate perception of guidance cues is crucial for cell and axon migration. During initial navigation in the spinal cord, commissural axons are kept insensitive to midline repellents. Upon midline crossing in the floor plate, they switch on responsiveness to Slit and Semaphorin repulsive signals and are thus propelled away and prevented from crossing back. Whether and how the different midline repellents control specific aspects of this navigation remain to be elucidated. We set up a paradigm for live-imaging and super-resolution analysis of PlexinA1, Neuropilin-2, and Robo1/2 receptor dynamics during commissural growth cone navigation in chick and mouse embryos. We uncovered a remarkable program of sensitization to midline cues achieved by unique spatiotemporal sequences of receptor allocation at the growth-cone surface that orchestrates receptor-specific growth-cone behavior changes. This reveals post-translational mechanisms whereby coincident guidance signals are temporally resolved to allow the generation of specific guidance responses.
Collapse
Affiliation(s)
- Aurora Pignata
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France
| | - Hugo Ducuing
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France
| | - Leila Boubakar
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France
| | - Thibault Gardette
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France
| | - Karine Kindbeiter
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France
| | - Muriel Bozon
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France
| | - Servane Tauszig-Delamasure
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France
| | - Julien Falk
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France
| | - Olivier Thoumine
- Interdisciplinary Institute for Neuroscience, UMR CNRS 5297, University of Bordeaux 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Valérie Castellani
- University of Lyon, University of Lyon 1 Claude Bernard Lyon1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, 8 Avenue Rockefeller, 69008 Lyon, France.
| |
Collapse
|
17
|
Duquette PM, Lamarche-Vane N. The calcium-activated protease calpain regulates netrin-1 receptor deleted in colorectal cancer-induced axon outgrowth in cortical neurons. J Neurochem 2019; 152:315-332. [PMID: 31344270 DOI: 10.1111/jnc.14837] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 07/02/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022]
Abstract
During development, neurons extend axons toward their appropriate synaptic targets to establish functional neuronal connections. The growth cone, a highly motile structure at the tip of the axon, is capable of recognizing extracellular guidance cues and translating them into directed axon outgrowth through modulation of the actin cytoskeleton. Netrin-1 mediates its attractive function through the receptor deleted in colorectal cancer (DCC) to promote axon outgrowth and guidance. The calcium-activated protease calpain is involved in the cleavage of cytoskeletal proteins, which plays an important role during adhesion turnover and cell migration. However, its function during neuronal development is less understood. Here we demonstrate that netrin-1 activated calpain in embryonic rat cortical neurons in an extracellular-regulated kinase 1/2-dependent manner. In addition, we found that netrin-1 stimulation led to an increase in calpain-1 localization in the axon, whereas its endogenous inhibitor calpastatin was decreased in the growth cones of cortical neurons by indirect immunofluorescence. Interestingly, calpain-1 was able to cleave DCC in vitro. Furthermore, netrin-1 induced the cleavage of the cytoskeletal proteins spectrin and focal adhesion kinase concomitantly with the intracellular domain of DCC in a calpain-dependent manner in embryonic rat cortical neurons. Cortical neurons over-expressing calpastatin or calpain-depleted neurons displayed increased basal axon length and were unresponsive to netrin-1 stimulation. Altogether, we propose a novel model whereby netrin-1/DCC-mediated axon outgrowth is modulated by calpain-mediated proteolysis of DCC and cytoskeletal targets in embryonic cortical neurons. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Philippe M Duquette
- Cancer Research Program, Research Institute of the McGill University Health Center (RI-MUHC), Montréal, Québec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Nathalie Lamarche-Vane
- Cancer Research Program, Research Institute of the McGill University Health Center (RI-MUHC), Montréal, Québec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| |
Collapse
|
18
|
Ye X, Qiu Y, Gao Y, Wan D, Zhu H. A Subtle Network Mediating Axon Guidance: Intrinsic Dynamic Structure of Growth Cone, Attractive and Repulsive Molecular Cues, and the Intermediate Role of Signaling Pathways. Neural Plast 2019; 2019:1719829. [PMID: 31097955 PMCID: PMC6487106 DOI: 10.1155/2019/1719829] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 01/01/2023] Open
Abstract
A fundamental feature of both early nervous system development and axon regeneration is the guidance of axonal projections to their targets in order to assemble neural circuits that control behavior. In the navigation process where the nerves grow toward their targets, the growth cones, which locate at the tips of axons, sense the environment surrounding them, including varies of attractive or repulsive molecular cues, then make directional decisions to adjust their navigation journey. The turning ability of a growth cone largely depends on its highly dynamic skeleton, where actin filaments and microtubules play a very important role in its motility. In this review, we summarize some possible mechanisms underlying growth cone motility, relevant molecular cues, and signaling pathways in axon guidance of previous studies and discuss some questions regarding directions for further studies.
Collapse
Affiliation(s)
- Xiyue Ye
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yan Qiu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yuqing Gao
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Dong Wan
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Huifeng Zhu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| |
Collapse
|
19
|
Kinoshita-Kawada M, Hasegawa H, Hongu T, Yanagi S, Kanaho Y, Masai I, Mishima T, Chen X, Tsuboi Y, Rao Y, Yuasa-Kawada J, Wu JY. A crucial role for Arf6 in the response of commissural axons to Slit. Development 2019; 146:dev172106. [PMID: 30674481 PMCID: PMC6382006 DOI: 10.1242/dev.172106] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 01/14/2019] [Indexed: 12/23/2022]
Abstract
A switch in the response of commissural axons to the repellent Slit is crucial for ensuring that they cross the ventral midline only once. However, the underlying mechanisms remain to be elucidated. We have found that both endocytosis and recycling of Robo1 receptor are crucial for modulating Slit sensitivity in vertebrate commissural axons. Robo1 endocytosis and its recycling back to the cell surface maintained the stability of axonal Robo1 during Slit stimulation. We identified Arf6 guanosine triphosphatase and its activators, cytohesins, as previously unknown components in Slit-Robo1 signalling in vertebrate commissural neurons. Slit-Robo1 signalling activated Arf6. The Arf6-deficient mice exhibited marked defects in commissural axon midline crossing. Our data showed that a Robo1 endocytosis-triggered and Arf6-mediated positive-feedback strengthens the Slit response in commissural axons upon their midline crossing. Furthermore, the cytohesin-Arf6 pathways modulated this self-enhancement of the Slit response before and after midline crossing, resulting in a switch that reinforced robust regulation of axon midline crossing. Our study provides insights into endocytic trafficking-mediated mechanisms for spatiotemporally controlled axonal responses and uncovers new players in the midline switch in Slit responsiveness of commissural axons.
Collapse
Affiliation(s)
- Mariko Kinoshita-Kawada
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
- Department of Neurology, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Hiroshi Hasegawa
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Tsunaki Hongu
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Yasunori Kanaho
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
| | - Takayasu Mishima
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Xiaoping Chen
- Department of Neurology, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yoshio Tsuboi
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Yi Rao
- State Key Laboratory of Biomembrane and Membrane Biology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University School of Life Sciences, Beijing 100871, China
| | - Junichi Yuasa-Kawada
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
- Department of Neurology, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka 812-8582, Japan
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Jane Y Wu
- Department of Neurology, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
20
|
Ducuing H, Gardette T, Pignata A, Tauszig-Delamasure S, Castellani V. Commissural axon navigation in the spinal cord: A repertoire of repulsive forces is in command. Semin Cell Dev Biol 2019; 85:3-12. [DOI: 10.1016/j.semcdb.2017.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 01/31/2023]
|
21
|
Ayanlaja AA, Zhang B, Ji G, Gao Y, Wang J, Kanwore K, Gao D. The reversible effects of glial cell line-derived neurotrophic factor (GDNF) in the human brain. Semin Cancer Biol 2018; 53:212-222. [PMID: 30059726 DOI: 10.1016/j.semcancer.2018.07.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 12/20/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent survival factor, and a member of the transforming growth factor β (TGF-β) superfamily acting on different neuronal activities. GDNF was originally identified as a neurotrophic factor crucially involved in the survival of dopaminergic neurons of the nigrostriatal pathway and is currently an established therapeutic target in Parkinson's disease. However, GDNF was later reported to be highly expressed in gliomas, especially in glioblastomas, and was demonstrated as a potent proliferation factor involved in the development and migration of gliomas. Here, we review our current understanding and progress made so far by researchers in our laboratories with references to relevant articles to support our discoveries. We present past and recent discoveries on the mechanisms involved in the protection of neurons by GDNF and examine its emerging roles in gliomas, as well as reasons for the abnormal expression in Glioblastoma Multiforme (GBM). Collectively, our work establishes a paradigm by which the ability of GDNF to protect dopaminergic neurons from degradation and its corresponding effects on glioma cells points to an underlying biological vulnerability in the effects of GDNF in the normal brain which can be subverted for use by cancer cells. Hence, presenting novel opportunities for intervention in glioma therapies.
Collapse
Affiliation(s)
- Abiola Abdulrahman Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Baole Zhang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - GuangQuan Ji
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yue Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jie Wang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - DianShuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
22
|
Schaeffer J, Tannahill D, Cioni JM, Rowlands D, Keynes R. Identification of the extracellular matrix protein Fibulin-2 as a regulator of spinal nerve organization. Dev Biol 2018; 442:101-114. [PMID: 29944871 DOI: 10.1016/j.ydbio.2018.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 05/24/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022]
Abstract
During amniote peripheral nervous system development, segmentation ensures the correct patterning of the spinal nerves relative to the vertebral column. Along the antero-posterior (rostro-caudal) axis, each somite-derived posterior half-sclerotome expresses repellent molecules to restrict axon growth and neural crest migration to the permissive anterior half-segment. To identify novel regulators of spinal nerve patterning, we investigated the differential gene expression of anterior and posterior half-sclerotomes in the chick embryo by RNA-sequencing. Several genes encoding extracellular matrix proteins were found to be enriched in either anterior (e.g. Tenascin-C, Laminin alpha 4) or posterior (e.g. Fibulin-2, Fibromodulin, Collagen VI alpha 2) half-sclerotomes. Among them, the extracellular matrix protein Fibulin-2 was found specifically restricted to the posterior half-sclerotome. By using in ovo ectopic expression in chick somites, we found that Fibulin-2 modulates spinal axon growth trajectories in vivo. While no intrinsic axon repellent activity of Fibulin-2 was found, we showed that it enhances the growth cone repulsive activity of Semaphorin 3A in vitro. Some molecules regulating axon growth during development are found to be upregulated in the adult central nervous system (CNS) following traumatic injury. Here, we found increased Fibulin-2 protein levels in reactive astrocytes at the lesion site of a mouse model of CNS injury. Together, these results suggest that the developing vertebral column and the adult CNS share molecular features that control axon growth and plasticity, which may open up the possibility for the identification of novel therapeutic targets for brain and spinal cord injury.
Collapse
Affiliation(s)
- Julia Schaeffer
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK.
| | | | - Jean-Michel Cioni
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | | | - Roger Keynes
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| |
Collapse
|
23
|
Ros O, Barrecheguren PJ, Cotrufo T, Schaettin M, Roselló-Busquets C, Vílchez-Acosta A, Hernaiz-Llorens M, Martínez-Marmol R, Ulloa F, Stoeckli ET, Araújo SJ, Soriano E. A conserved role for Syntaxin-1 in pre- and post-commissural midline axonal guidance in fly, chick, and mouse. PLoS Genet 2018; 14:e1007432. [PMID: 29912942 PMCID: PMC6029812 DOI: 10.1371/journal.pgen.1007432] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 07/03/2018] [Accepted: 05/18/2018] [Indexed: 02/03/2023] Open
Abstract
Axonal growth and guidance rely on correct growth cone responses to guidance cues. Unlike the signaling cascades that link axonal growth to cytoskeletal dynamics, little is known about the crosstalk mechanisms between guidance and membrane dynamics and turnover. Recent studies indicate that whereas axonal attraction requires exocytosis, chemorepulsion relies on endocytosis. Indeed, our own studies have shown that Netrin-1/Deleted in Colorectal Cancer (DCC) signaling triggers exocytosis through the SNARE Syntaxin-1 (STX1). However, limited in vivo evidence is available about the role of SNARE proteins in axonal guidance. To address this issue, here we systematically deleted SNARE genes in three species. We show that loss-of-function of STX1 results in pre- and post-commissural axonal guidance defects in the midline of fly, chick, and mouse embryos. Inactivation of VAMP2, Ti-VAMP, and SNAP25 led to additional abnormalities in axonal guidance. We also confirmed that STX1 loss-of-function results in reduced sensitivity of commissural axons to Slit-2 and Netrin-1. Finally, genetic interaction studies in Drosophila show that STX1 interacts with both the Netrin-1/DCC and Robo/Slit pathways. Our data provide evidence of an evolutionarily conserved role of STX1 and SNARE proteins in midline axonal guidance in vivo, by regulating both pre- and post-commissural guidance mechanisms. Syntaxin-1 is a core factor in tethering synaptic vesicles and mediating their fusion to the cell membrane at the synapse. Thus, Syntaxin-1 mediates neurotransmission in the adult nervous system. Here we show that this protein is also involved in axonal guidance in the CNS of vertebrates and invertebrates during the development of the nervous system: our systematic analysis of the phenotypes in the nervous system midline of fly, chick, and mouse embryos mutant for Syntaxin-1 unveils an evolutionarily conserved role for this protein in midline axonal guidance. Further, we also dissect the contribution of other proteins regulating neuronal exocytosis in axonal development. We propose that the coupling of the guidance molecule machinery to proteins that regulate exocytosis is a general mechanism linking chemotropism to axonal growth.
Collapse
Affiliation(s)
- Oriol Ros
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Pablo José Barrecheguren
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut de Recerca Biomedica de Barcelona (IRB Barcelona), Parc Cientific de Barcelona, Barcelona, Spain
| | - Tiziana Cotrufo
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Martina Schaettin
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Cristina Roselló-Busquets
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Alba Vílchez-Acosta
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Marc Hernaiz-Llorens
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Ramón Martínez-Marmol
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Fausto Ulloa
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Esther T. Stoeckli
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- * E-mail: (ETS); (SJA); (ES)
| | - Sofia J. Araújo
- Institut de Recerca Biomedica de Barcelona (IRB Barcelona), Parc Cientific de Barcelona, Barcelona, Spain
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Parc Cientific de Barcelona, Barcelona, Spain
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- * E-mail: (ETS); (SJA); (ES)
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology and Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
- Vall d´Hebron Institute of Research (VHIR), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- * E-mail: (ETS); (SJA); (ES)
| |
Collapse
|
24
|
Helmbacher F. Tissue-specific activities of the Fat1 cadherin cooperate to control neuromuscular morphogenesis. PLoS Biol 2018; 16:e2004734. [PMID: 29768404 PMCID: PMC5973635 DOI: 10.1371/journal.pbio.2004734] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 05/29/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
Muscle morphogenesis is tightly coupled with that of motor neurons (MNs). Both MNs and muscle progenitors simultaneously explore the surrounding tissues while exchanging reciprocal signals to tune their behaviors. We previously identified the Fat1 cadherin as a regulator of muscle morphogenesis and showed that it is required in the myogenic lineage to control the polarity of progenitor migration. To expand our knowledge on how Fat1 exerts its tissue-morphogenesis regulator activity, we dissected its functions by tissue-specific genetic ablation. An emblematic example of muscle under such morphogenetic control is the cutaneous maximus (CM) muscle, a flat subcutaneous muscle in which progenitor migration is physically separated from the process of myogenic differentiation but tightly associated with elongating axons of its partner MNs. Here, we show that constitutive Fat1 disruption interferes with expansion and differentiation of the CM muscle, with its motor innervation and with specification of its associated MN pool. Fat1 is expressed in muscle progenitors, in associated mesenchymal cells, and in MN subsets, including the CM-innervating pool. We identify mesenchyme-derived connective tissue (CT) as a cell type in which Fat1 activity is required for the non-cell-autonomous control of CM muscle progenitor spreading, myogenic differentiation, motor innervation, and for motor pool specification. In parallel, Fat1 is required in MNs to promote their axonal growth and specification, indirectly influencing muscle progenitor progression. These results illustrate how Fat1 coordinates the coupling of muscular and neuronal morphogenesis by playing distinct but complementary actions in several cell types.
Collapse
|
25
|
Abstract
During nervous system development, neurons extend axons to reach their targets and form functional circuits. The faulty assembly or disintegration of such circuits results in disorders of the nervous system. Thus, understanding the molecular mechanisms that guide axons and lead to neural circuit formation is of interest not only to developmental neuroscientists but also for a better comprehension of neural disorders. Recent studies have demonstrated how crosstalk between different families of guidance receptors can regulate axonal navigation at choice points, and how changes in growth cone behaviour at intermediate targets require changes in the surface expression of receptors. These changes can be achieved by a variety of mechanisms, including transcription, translation, protein-protein interactions, and the specific trafficking of proteins and mRNAs. Here, I review these axon guidance mechanisms, highlighting the most recent advances in the field that challenge the textbook model of axon guidance.
Collapse
Affiliation(s)
- Esther T Stoeckli
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
26
|
GFRA1: A Novel Molecular Target for the Prevention of Osteosarcoma Chemoresistance. Int J Mol Sci 2018; 19:ijms19041078. [PMID: 29617307 PMCID: PMC5979596 DOI: 10.3390/ijms19041078] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/26/2018] [Accepted: 03/31/2018] [Indexed: 12/11/2022] Open
Abstract
The glycosylphosphatidylinositol-linked GDNF (glial cell derived neurotrophic factor) receptor alpha (GFRA), a coreceptor that recognizes the GDNF family of ligands, has a crucial role in the development and maintenance of the nervous system. Of the four identified GFRA isoforms, GFRA1 specifically recognizes GDNF and is involved in the regulation of proliferation, differentiation, and migration of neuronal cells. GFRA1 has also been implicated in cancer cell progression and metastasis. Recent findings show that GFRA1 can contribute to the development of chemoresistance in osteosarcoma. GFRA1 expression was induced following treatment of osteosarcoma cells with the popular anticancer drug, cisplatin and induction of GFRA1 expression significantly suppressed apoptosis mediated by cisplatin in osteosarcoma cells. GFRA1 expression promotes autophagy by activating the SRC-AMPK signaling axis following cisplatin treatment, resulting in enhanced osteosarcoma cell survival. GFRA1-induced autophagy promoted tumor growth in mouse xenograft models, suggesting a novel function of GFRA1 in osteosarcoma chemoresistance.
Collapse
|
27
|
Bonafina A, Fontanet PA, Paratcha G, Ledda F. GDNF/GFRα1 Complex Abrogates Self-Renewing Activity of Cortical Neural Precursors Inducing Their Differentiation. Stem Cell Reports 2018; 10:1000-1015. [PMID: 29478900 PMCID: PMC5918270 DOI: 10.1016/j.stemcr.2018.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/30/2022] Open
Abstract
The balance between factors leading to proliferation and differentiation of cortical neural precursors (CNPs) determines the correct cortical development. In this work, we show that GDNF and its receptor GFRα1 are expressed in the neocortex during the period of cortical neurogenesis. We show that the GDNF/GFRα1 complex inhibits the self-renewal capacity of mouse CNP cells induced by fibroblast growth factor 2 (FGF2), promoting neuronal differentiation. While GDNF leads to decreased proliferation of cultured cortical precursor cells, ablation of GFRα1 in glutamatergic cortical precursors enhances its proliferation. We show that GDNF treatment of CNPs promoted morphological differentiation even in the presence of the self-renewal-promoting factor, FGF2. Analysis of GFRα1-deficient mice shows an increase in the number of cycling cells during cortical development and a reduction in dendrite development of cortical GFRα1-expressing neurons. Together, these results indicate that GDNF/GFRα1 signaling plays an essential role in regulating the proliferative condition and the differentiation of cortical progenitors. GFRα1 receptor is expressed in the neocortex during the period of neurogenesis GDNF/GFRα1 complex inhibits self-renewing of cortical neuronal precursors GDNF and GFRα1 promote neurogenic differentiation of cortical neural progenitors Requirement of GFRα1 for proper dendrite development of cortical neurons
Collapse
Affiliation(s)
- Antonela Bonafina
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET-UBA, School of Medicine, University of Buenos Aires (UBA), Buenos Aires CP 1121, Argentina
| | - Paula Aldana Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET-UBA, School of Medicine, University of Buenos Aires (UBA), Buenos Aires CP 1121, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET-UBA, School of Medicine, University of Buenos Aires (UBA), Buenos Aires CP 1121, Argentina
| | - Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET-UBA, School of Medicine, University of Buenos Aires (UBA), Buenos Aires CP 1121, Argentina.
| |
Collapse
|
28
|
Yoon J, Terman JR. Common effects of attractive and repulsive signaling: Further analysis of Mical-mediated F-actin disassembly and regulation by Abl. Commun Integr Biol 2018; 11:e1405197. [PMID: 29497471 PMCID: PMC5824934 DOI: 10.1080/19420889.2017.1405197] [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: 10/04/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 11/21/2022] Open
Abstract
To change their size, shape, and connectivity, cells require actin and tubulin proteins to assemble together into long polymers – and numerous extracellular stimuli have now been identified that alter the assembly and organization of these cytoskeletal structures. Yet, there remains a lack of defined signaling pathways from the cell surface to the cytoskeleton for many of these extracellular signals, and so we still know little of how they exert their precise structural effects. These extracellular cues may be soluble or substrate-bound and have historically been classified into two independently acting and antagonistic groups: growth-promoting/attractants (inducing turning toward the source of the factor/positive chemotropism) or growth-preventing/repellents (turning away from the source of the factor/negative chemotropism). Paradoxically, our recent results directly link the action of growth factors/chemoattractants and their signaling pathways to the promotion of the disassembly of the F-actin cytoskeleton (a defined readout of repellents/repulsive signaling). Herein, we add to this by simply driving a constitutively active form of Mical, which strongly disassembles F-actin/remodels cells in vivo independent of repulsive cues – and find that loss of Abl, which mediates growth factor signaling in these cells, decreases Mical's F-actin disassembly/cellular remodeling effects. Thus, our results are consistent with a hypothesis that cues defined as positive effectors of movement (growth factors/chemoattractants) can at least in some contexts enhance the F-actin disassembly and remodeling activity of repellents.
Collapse
Affiliation(s)
- Jimok Yoon
- Departments of Neuroscience and Pharmacology, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jonathan R Terman
- Departments of Neuroscience and Pharmacology, Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
29
|
Rosich K, Hanna BF, Ibrahim RK, Hellenbrand DJ, Hanna A. The Effects of Glial Cell Line-Derived Neurotrophic Factor after Spinal Cord Injury. J Neurotrauma 2017; 34:3311-3325. [DOI: 10.1089/neu.2017.5175] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Konstantin Rosich
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| | - Bishoy F. Hanna
- Department of Neurological Surgery, Ross University School of Medicine, Dominica, West Indies
| | - Rami K. Ibrahim
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| | - Daniel J. Hellenbrand
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin
| | - Amgad Hanna
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
30
|
Hernandez-Fleming M, Rohrbach EW, Bashaw GJ. Sema-1a Reverse Signaling Promotes Midline Crossing in Response to Secreted Semaphorins. Cell Rep 2017; 18:174-184. [PMID: 28052247 DOI: 10.1016/j.celrep.2016.12.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/21/2016] [Accepted: 12/08/2016] [Indexed: 11/26/2022] Open
Abstract
Commissural axons must cross the midline to form functional midline circuits. In the invertebrate nerve cord and vertebrate spinal cord, midline crossing is mediated in part by Netrin-dependent chemoattraction. Loss of crossing, however, is incomplete in mutants for Netrin or its receptor Frazzled/DCC, suggesting the existence of additional pathways. We identified the transmembrane Semaphorin, Sema-1a, as an important regulator of midline crossing in the Drosophila CNS. We show that in response to the secreted Semaphorins Sema-2a and Sema-2b, Sema-1a functions as a receptor to promote crossing independently of Netrin. In contrast to other examples of reverse signaling where Sema1a triggers repulsion through activation of Rho in response to Plexin binding, in commissural neurons Sema-1a acts independently of Plexins to inhibit Rho to promote attraction to the midline. These findings suggest that Sema-1a reverse signaling can elicit distinct axonal responses depending on differential engagement of distinct ligands and signaling effectors.
Collapse
Affiliation(s)
- Melissa Hernandez-Fleming
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Ethan W Rohrbach
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Greg J Bashaw
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
31
|
Sergaki MC, Ibáñez CF. GFRα1 Regulates Purkinje Cell Migration by Counteracting NCAM Function. Cell Rep 2017; 18:367-379. [PMID: 28076782 PMCID: PMC5263233 DOI: 10.1016/j.celrep.2016.12.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/17/2016] [Accepted: 12/09/2016] [Indexed: 02/06/2023] Open
Abstract
During embryonic development of the cerebellum, Purkinje cells (PCs) migrate away from the ventricular zone to form the PC plate. The mechanisms that regulate PC migration are incompletely understood. Here, we report that the neurotrophic receptor GFRα1 is transiently expressed in developing PCs and loss of GFRα1 delays PC migration. Neither GDNF nor RET, the canonical GFRα1 ligand and co-receptor, respectively, contribute to this process. Instead, we found that the neural cell adhesion molecule NCAM is co-expressed and directly interacts with GFRα1 in embryonic PCs. Genetic reduction of NCAM expression enhances wild-type PC migration and restores migration in Gfra1 mutants, indicating that NCAM restricts PC migration in the embryonic cerebellum. In vitro experiments indicated that GFRα1 can function both in cis and trans to counteract NCAM and promote PC migration. Collectively, our studies show that GFRα1 contributes to PC migration by limiting NCAM function.
Collapse
Affiliation(s)
| | - Carlos F Ibáñez
- Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden; Department of Physiology, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore.
| |
Collapse
|
32
|
Xiao N, Yu WY, Liu D. Glial cell‐derived neurotrophic factor promotes dental pulp stem cell migration. J Tissue Eng Regen Med 2017; 12:705-714. [DOI: 10.1002/term.2490] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 04/27/2017] [Accepted: 06/01/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Nan Xiao
- Department of Biomedical Sciences, Arthur A. Dugoni School of DentistryUniversity of the Pacific San Francisco CA USA
| | - Wei Ye Yu
- Department of Biomedical Sciences, Arthur A. Dugoni School of DentistryUniversity of the Pacific San Francisco CA USA
| | - Dawei Liu
- Department of Anatomy and Cell Biology, School of Dental MedicineUniversity of the Pennsylvania Philadelphia PA USA
| |
Collapse
|
33
|
Onishi K, Zou Y. Sonic Hedgehog switches on Wnt/planar cell polarity signaling in commissural axon growth cones by reducing levels of Shisa2. eLife 2017; 6:25269. [PMID: 28885142 PMCID: PMC5779225 DOI: 10.7554/elife.25269] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/17/2017] [Indexed: 01/06/2023] Open
Abstract
Commissural axons switch on responsiveness to Wnt attraction during midline crossing and turn anteriorly only after exiting the floor plate. We report here that Sonic Hedgehog (Shh)-Smoothened signaling downregulates Shisa2, which inhibits the glycosylation and cell surface presentation of Frizzled3 in rodent commissural axon growth cones. Constitutive Shisa2 expression causes randomized turning of post-crossing commissural axons along the anterior–posterior (A–P) axis. Loss of Shisa2 led to precocious anterior turning of commissural axons before or during midline crossing. Post-crossing commissural axon turning is completely randomized along the A–P axis when Wntless, which is essential for Wnt secretion, is conditionally knocked out in the floor plate. This regulatory link between Shh and planar cell polarity (PCP) signaling may also occur in other developmental processes.
Collapse
Affiliation(s)
- Keisuke Onishi
- Neurobiology Section, Biological Sciences Division, University of California, San Diego, San Diego, United States
| | - Yimin Zou
- Neurobiology Section, Biological Sciences Division, University of California, San Diego, San Diego, United States
| |
Collapse
|
34
|
Cortés D, Carballo-Molina OA, Castellanos-Montiel MJ, Velasco I. The Non-Survival Effects of Glial Cell Line-Derived Neurotrophic Factor on Neural Cells. Front Mol Neurosci 2017; 10:258. [PMID: 28878618 PMCID: PMC5572274 DOI: 10.3389/fnmol.2017.00258] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 07/31/2017] [Indexed: 01/23/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) was first characterized as a survival-promoting molecule for dopaminergic neurons (DANs). Afterwards, other cells were also discovered to respond to GDNF not only as a survival factor but also as a protein supporting other cellular functions, such as proliferation, differentiation, maturation, neurite outgrowth and other phenomena that have been less studied than survival and are now more extendedly described here in this review article. During development, GDNF favors the commitment of neural precursors towards dopaminergic, motor, enteric and adrenal neurons; in addition, it enhances the axonal growth of some of these neurons. GDNF also induces the acquisition of a dopaminergic phenotype by increasing the expression of Tyrosine Hydroxylase (TH), Nurr1 and other proteins that confer this identity and promote further dendritic and electrical maturation. In motor neurons (MNs), GDNF not only promotes proliferation and maturation but also participates in regenerating damaged axons and modulates the neuromuscular junction (NMJ) at both presynaptic and postsynaptic levels. Moreover, GDNF modulates the rate of neuroblastoma (NB) and glioblastoma cancer cell proliferation. Additionally, the presence or absence of GDNF has been correlated with conditions such as depression, pain, muscular soreness, etc. Although, the precise role of GDNF is unknown, it extends beyond a survival effect. The understanding of the complete range of properties of this trophic molecule will allow us to investigate its broad mechanisms of action to accelerate and/or improve therapies for the aforementioned pathological conditions.
Collapse
Affiliation(s)
- Daniel Cortés
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - Oscar A. Carballo-Molina
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - María José Castellanos-Montiel
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| |
Collapse
|
35
|
Sun S, Lei Y, Li Q, Wu Y, Zhang L, Mu PP, Ji GQ, Tang CX, Wang YQ, Gao J, Gao J, Li L, Zhuo L, Li YQ, Gao DS. Neuropilin-1 is a glial cell line-derived neurotrophic factor receptor in glioblastoma. Oncotarget 2017; 8:74019-74035. [PMID: 29088765 PMCID: PMC5650320 DOI: 10.18632/oncotarget.18630] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/12/2017] [Indexed: 01/13/2023] Open
Abstract
The aim of this study was to identify the receptor for glial cell line-derived neurotrophic factor (GDNF) in glioblastoma multiforme (GBM). After GST pull-down assays, membrane proteins purified from C6 rat glioma cells were subjected to liquid chromatography-tandem mass spectrometry (LC-MS/MS). The differentially expressed proteins were annotated using Gene Ontology, and neuropilin-1 (NRP1) was identified as the putative GDNF receptor in glioma. NRP1 was more highly expressed in human GBM brains and C6 rat glioma cells than in normal human brains or primary rat astrocytes. Immunofluorescence staining showed that NRP1 was recruited to the membrane by GDNF, and NRP1 co-immunoprecipitated with GDNF. Using the NRP1 and GDNF protein structures to assess molecular docking in the ZDOCK server and visualization with the PyMOL Molecular Graphics System revealed 8 H-bonds and stable positive and negative electrostatic interactions between NRP1 and GDNF. RNAi knockdown of NRP1 reduced proliferation of C6 glioma cells when stimulated with GDNF. NRP1 was an independent risk factor for both survival and recurrence in GBM patients. High NRP1 mRNA expression correlated with shorter OS and DFS (OS: χ2=4.6720, P=0.0307; DFS: χ2=11.013, P=0.0009). NRP1 is thus a GDNF receptor in glioma cells and a potential therapeutic target.
Collapse
Affiliation(s)
- Shen Sun
- Department of Anatomy and Histology, The Fourth Military Medical University, Xi'an, Shanxi, China.,Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Histology and Embryology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Lei
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Qi Li
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yue Wu
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Zhang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Pei-Pei Mu
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guang-Quan Ji
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chuan-Xi Tang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu-Qian Wang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jin Gao
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Li Li
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lang Zhuo
- Department of Epidemiology, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yun-Qing Li
- Department of Anatomy and Histology, The Fourth Military Medical University, Xi'an, Shanxi, China
| | - Dian-Shuai Gao
- Department of Anatomy and Histology, The Fourth Military Medical University, Xi'an, Shanxi, China.,Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
36
|
Charoy C, Dinvaut S, Chaix Y, Morlé L, Sanyas I, Bozon M, Kindbeiter K, Durand B, Skidmore JM, De Groef L, Seki M, Moons L, Ruhrberg C, Martin JF, Martin DM, Falk J, Castellani V. Genetic specification of left-right asymmetry in the diaphragm muscles and their motor innervation. eLife 2017. [PMID: 28639940 PMCID: PMC5481184 DOI: 10.7554/elife.18481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The diaphragm muscle is essential for breathing in mammals. Its asymmetric elevation during contraction correlates with morphological features suggestive of inherent left–right (L/R) asymmetry. Whether this asymmetry is due to L versus R differences in the muscle or in the phrenic nerve activity is unknown. Here, we have combined the analysis of genetically modified mouse models with transcriptomic analysis to show that both the diaphragm muscle and phrenic nerves have asymmetries, which can be established independently of each other during early embryogenesis in pathway instructed by Nodal, a morphogen that also conveys asymmetry in other organs. We further found that phrenic motoneurons receive an early L/R genetic imprint, with L versus R differences both in Slit/Robo signaling and MMP2 activity and in the contribution of both pathways to establish phrenic nerve asymmetry. Our study therefore demonstrates L–R imprinting of spinal motoneurons and describes how L/R modulation of axon guidance signaling helps to match neural circuit formation to organ asymmetry. DOI:http://dx.doi.org/10.7554/eLife.18481.001 The diaphragm is a dome-shaped muscle that forms the floor of the rib cage, separating the lungs from the abdomen. As we breathe in, the diaphragm contracts. This causes the chest cavity to expand, drawing air into the lungs. A pair of nerves called the phrenic nerves carry signals from the spinal cord to the diaphragm to tell it when to contract. These nerves project from the left and right sides of the spinal cord to the left and right sides of the diaphragm respectively. The left and right sides of the diaphragm are not entirely level, but it was not known why. To investigate, Charoy et al. studied how the diaphragm develops in mouse embryos. This revealed that the left and right phrenic nerves are not symmetrical. Neither are the muscles on each side of the diaphragm. Further investigation revealed that a genetic program that establishes other differences between the left and right sides of the embryo also gives rise to the differences between the left and right sides of the diaphragm. This program switches on different genes in the left and right phrenic nerves, which activate different molecular pathways in the left and right sides of the diaphragm muscle. The differences between the nerves and muscles on the left and right sides of the diaphragm could explain why some muscle disorders affect only one side of the diaphragm. Similarly, they could explain why congenital hernias caused by abdominal organs pushing through the diaphragm into the chest cavity mostly affect the left side of the diaphragm. Further studies are now needed to investigate these possibilities. The techniques used by Charoy et al. to map the molecular diversity of spinal cord neurons could also lead to new strategies for repairing damage to the spinal cord following injury or disease. DOI:http://dx.doi.org/10.7554/eLife.18481.002
Collapse
Affiliation(s)
- Camille Charoy
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Sarah Dinvaut
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Yohan Chaix
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Laurette Morlé
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Isabelle Sanyas
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Muriel Bozon
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Karine Kindbeiter
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Bénédicte Durand
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Jennifer M Skidmore
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, United States.,Department of Communicable Diseases, University of Michigan Medical Center, Ann Arbor, United States
| | - Lies De Groef
- Animal Physiology and Neurobiology Section, Department of Biology, Laboratory of Neural Circuit Development and Regeneration, Leuven, Belgium
| | - Motoaki Seki
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan
| | - Lieve Moons
- Animal Physiology and Neurobiology Section, Department of Biology, Laboratory of Neural Circuit Development and Regeneration, Leuven, Belgium
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | | | - Donna M Martin
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, United States.,Department of Communicable Diseases, University of Michigan Medical Center, Ann Arbor, United States.,Department of Human Genetics, University of Michigan Medical Center, Ann Arbor, United States
| | - Julien Falk
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Valerie Castellani
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| |
Collapse
|
37
|
Wang L, Yu C, Wang J, Leung P, Ma D, Zhao H, Taylor JSH, Chan SO. Nogo-B is the major form of Nogo at the floor plate and likely mediates crossing of commissural axons in the mouse spinal cord. J Comp Neurol 2017; 525:2915-2928. [DOI: 10.1002/cne.24246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/06/2017] [Accepted: 05/13/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Liqing Wang
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-Sen University; Guangzhou Guangdong China
| | - Chao Yu
- Center of Health Examination, The Third Affiliated Hospital of Sun Yat-Sen University; Guangzhou Guangdong China
| | - Jun Wang
- Department of Anatomy and Embryology; School of Basic Medical Sciences, Peking University; Beijing China
| | - Peggy Leung
- School of Biomedical Sciences; The Chinese University of Hong Kong, Shatin, N.T.; Hong Kong China
| | - Ding Ma
- School of Biomedical Sciences; The Chinese University of Hong Kong, Shatin, N.T.; Hong Kong China
| | - Hui Zhao
- School of Biomedical Sciences; The Chinese University of Hong Kong, Shatin, N.T.; Hong Kong China
| | - Jeremy S. H. Taylor
- Department of Physiology; Anatomy and Genetics, University of Oxford; Oxford United Kingdom
| | - Sun-On Chan
- School of Biomedical Sciences; The Chinese University of Hong Kong, Shatin, N.T.; Hong Kong China
| |
Collapse
|
38
|
Abstract
During neural circuit formation, axons need to navigate to their target cells in a complex, constantly changing environment. Although we most likely have identified most axon guidance cues and their receptors, we still cannot explain the molecular background of pathfinding for any subpopulation of axons. We lack mechanistic insight into the regulation of interactions between guidance receptors and their ligands. Recent developments in the field of axon guidance suggest that the regulation of surface expression of guidance receptors comprises transcriptional, translational, and post-translational mechanisms, such as trafficking of vesicles with specific cargos, protein-protein interactions, and specific proteolysis of guidance receptors. Not only axon guidance molecules but also the regulatory mechanisms that control their spatial and temporal expression are involved in synaptogenesis and synaptic plasticity. Therefore, it is not surprising that genes associated with axon guidance are frequently found in genetic and genomic studies of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Esther Stoeckli
- Department of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Calpain-Mediated Proteolysis of Talin and FAK Regulates Adhesion Dynamics Necessary for Axon Guidance. J Neurosci 2017; 37:1568-1580. [PMID: 28069919 DOI: 10.1523/jneurosci.2769-16.2016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/21/2016] [Accepted: 12/29/2016] [Indexed: 01/05/2023] Open
Abstract
Guidance of axons to their proper synaptic target sites requires spatially and temporally precise modulation of biochemical signals within growth cones. Ionic calcium (Ca2+) is an essential signal for axon guidance that mediates opposing effects on growth cone motility. The diverse effects of Ca2+ arise from the precise localization of Ca2+ signals into microdomains containing specific Ca2+ effectors. For example, differences in the mechanical and chemical composition of the underlying substrata elicit local Ca2+ signals within growth cone filopodia that regulate axon guidance through activation of the protease calpain. However, how calpain regulates growth cone motility remains unclear. Here, we identify the adhesion proteins talin and focal adhesion kinase (FAK) as proteolytic targets of calpain in Xenopus laevis spinal cord neurons both in vivo and in vitro Inhibition of calpain increases the localization of endogenous adhesion signaling to growth cone filopodia. Using live cell microscopy and specific calpain-resistant point-mutants of talin (L432G) and FAK (V744G), we find that calpain inhibits paxillin-based adhesion assembly through cleavage of talin and FAK, and adhesion disassembly through cleavage of FAK. Blocking calpain cleavage of talin and FAK inhibits repulsive turning from focal uncaging of Ca2+ within filopodia. In addition, blocking calpain cleavage of talin and FAK in vivo promotes Rohon-Beard peripheral axon extension into the skin. These data demonstrate that filopodial Ca2+ signals regulate axon outgrowth and guidance through calpain regulation of adhesion dynamics through specific cleavage of talin and FAK.SIGNIFICANCE STATEMENT The proper formation of neuronal networks requires accurate guidance of axons and dendrites during development by motile structures known as growth cones. Understanding the intracellular signaling mechanisms that govern growth cone motility will clarify how the nervous system develops and regenerates, and may identify areas of therapeutic intervention in disease or injury. One important signal that controls growth cones is that of local Ca2+ transients, which control the rate and direction of axon outgrowth. We demonstrate here that Ca2+-dependent inhibition axon outgrowth and guidance is mediated by calpain proteolysis of the adhesion proteins talin and focal adhesion kinase. Our findings provide mechanistic insight into Ca2+/calpain regulation of growth cone motility and axon guidance during neuronal development.
Collapse
|
40
|
Ibáñez CF, Andressoo JO. Biology of GDNF and its receptors — Relevance for disorders of the central nervous system. Neurobiol Dis 2017; 97:80-89. [DOI: 10.1016/j.nbd.2016.01.021] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/14/2016] [Accepted: 01/25/2016] [Indexed: 01/15/2023] Open
|
41
|
de Ramon Francàs G, Zuñiga NR, Stoeckli ET. The spinal cord shows the way - How axons navigate intermediate targets. Dev Biol 2016; 432:43-52. [PMID: 27965053 DOI: 10.1016/j.ydbio.2016.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/26/2016] [Accepted: 12/01/2016] [Indexed: 12/13/2022]
Abstract
Functional neural circuits depend on the establishment of specific connections between neurons and their target cells. To this end, many axons have to travel long distances to reach their target cells during development. Studies addressing the molecular mechanisms of axon guidance have to overcome the complexity of subpopulation-specific requirements with respect to pathways, guidance cues, and target recognition. Compared to the brain, the relatively simple structure of the spinal cord provides an advantage for experimental studies of axon guidance mechanisms. Therefore, the so far best understood model for axon guidance is the dI1 population of dorsal interneurons of the spinal cord. They extend their axons ventrally towards the floor plate. After midline crossing, they turn rostrally along the contralateral floor-plate border. Despite the fact that the trajectory of dI1 axons seems to be rather simple, the number of axon guidance molecules involved in the decisions taken by these axons is bewildering. Because guidance molecules and mechanisms are conserved throughout the developing nervous system, we can generalize what we have learned about the navigation of the floor plate as an intermediate target for commissural axons to the brain.
Collapse
Affiliation(s)
- Gemma de Ramon Francàs
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Nikole R Zuñiga
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Esther T Stoeckli
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
42
|
Irala D, Bonafina A, Fontanet PA, Alsina FC, Paratcha G, Ledda F. The GDNF-GFRα1 complex promotes the development of hippocampal dendritic arbors and spines via NCAM. Development 2016; 143:4224-4235. [PMID: 27707798 DOI: 10.1242/dev.140350] [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: 05/29/2016] [Accepted: 09/28/2016] [Indexed: 12/26/2022]
Abstract
The formation of synaptic connections during nervous system development requires the precise control of dendrite growth and synapse formation. Although glial cell line-derived neurotrophic factor (GDNF) and its receptor GFRα1 are expressed in the forebrain, the role of this system in the hippocampus remains unclear. Here, we investigated the consequences of GFRα1 deficiency for the development of hippocampal connections. Analysis of conditional Gfra1 knockout mice shows a reduction in dendritic length and complexity, as well as a decrease in postsynaptic density specializations and in the synaptic localization of postsynaptic proteins in hippocampal neurons. Gain- and loss-of-function assays demonstrate that the GDNF-GFRα1 complex promotes dendritic growth and postsynaptic differentiation in cultured hippocampal neurons. Finally, in vitro assays revealed that GDNF-GFRα1-induced dendrite growth and spine formation are mediated by NCAM signaling. Taken together, our results indicate that the GDNF-GFRα1 complex is essential for proper hippocampal circuit development.
Collapse
Affiliation(s)
- Dolores Irala
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Antonela Bonafina
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Paula Aldana Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Fernando Cruz Alsina
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| | - Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine, University of Buenos Aires (UBA), Buenos Aires 1121, Argentina
| |
Collapse
|
43
|
Furlan A, La Manno G, Lübke M, Häring M, Abdo H, Hochgerner H, Kupari J, Usoskin D, Airaksinen MS, Oliver G, Linnarsson S, Ernfors P. Visceral motor neuron diversity delineates a cellular basis for nipple- and pilo-erection muscle control. Nat Neurosci 2016; 19:1331-40. [PMID: 27571008 DOI: 10.1038/nn.4376] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/04/2016] [Indexed: 01/19/2023]
Abstract
Despite the variety of physiological and target-related functions, little is known regarding the cellular complexity in the sympathetic ganglion. We explored the heterogeneity of mouse stellate and thoracic ganglia and found an unexpected variety of cell types. We identified specialized populations of nipple- and pilo-erector muscle neurons. These neurons extended axonal projections and were born among other neurons during embryogenesis, but remained unspecialized until target organogenesis occurred postnatally. Target innervation and cell-type specification was coordinated by an intricate acquisition of unique combinations of growth factor receptors and the initiation of expression of concomitant ligands by the nascent erector muscles. Overall, our results provide compelling evidence for a highly sophisticated organization of the sympathetic nervous system into discrete outflow channels that project to well-defined target tissues and offer mechanistic insight into how diversity and connectivity are established during development.
Collapse
Affiliation(s)
- Alessandro Furlan
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Gioele La Manno
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Moritz Lübke
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Häring
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Hind Abdo
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Hannah Hochgerner
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jussi Kupari
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dmitry Usoskin
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Matti S Airaksinen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sten Linnarsson
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Pignata A, Ducuing H, Castellani V. Commissural axon navigation: Control of midline crossing in the vertebrate spinal cord by the semaphorin 3B signaling. Cell Adh Migr 2016; 10:604-617. [PMID: 27532244 PMCID: PMC5160037 DOI: 10.1080/19336918.2016.1212804] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The mechanisms governing the navigation of commissural axons during embryonic development have been extensively investigated in the past years, often using the drosophila ventral nerve cord and the spinal cord as model systems. Similarities but also specificities in the general strategies, the molecular signals as well as in the regulatory pathways controlling the response of commissural axons to the guidance cues have been found between species. Whether the semaphorin signaling contributes to midline crossing in the fly nervous system remains unknown, while in contrast, it does play a prominent contribution in vertebrates. In this review we discuss the functions of the semaphorins during commissural axon guidance in the developing spinal cord, focusing on the family member semaphorin 3B (Sema3B) in the context of midline crossing in the spinal cord.
Collapse
Affiliation(s)
- Aurora Pignata
- a University of Lyon, Université Claude Bernard Lyon 1, NeuroMyogene Institute (INMG), UMR CNRS 5310, INSERM U1217 Lyon , France
| | - Hugo Ducuing
- a University of Lyon, Université Claude Bernard Lyon 1, NeuroMyogene Institute (INMG), UMR CNRS 5310, INSERM U1217 Lyon , France
| | - Valérie Castellani
- a University of Lyon, Université Claude Bernard Lyon 1, NeuroMyogene Institute (INMG), UMR CNRS 5310, INSERM U1217 Lyon , France
| |
Collapse
|
45
|
Ortega-de San Luis C, Pascual A. Simultaneous Detection of Both GDNF and GFRα1 Expression Patterns in the Mouse Central Nervous System. Front Neuroanat 2016; 10:73. [PMID: 27445711 PMCID: PMC4919337 DOI: 10.3389/fnana.2016.00073] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/09/2016] [Indexed: 12/20/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is proposed as a therapeutic tool in Parkinson's disease, addiction-related disorders, and neurodegenerative conditions affecting motor neurons (MNs). Despite the high amount of work about GDNF therapeutic application, the neuronal circuits requiring GDNF trophic support in the brain and spinal cord (SC) are poorly characterized. Here, we defined GDNF and GDNF family receptor-α 1 (GFRα1) expression pattern in the brain and SC of newborn and adult mice. We performed systematic and simultaneous detection of EGFP and LacZ expressing alleles in reporter mice and asked whether modifications of this signaling pathway lead to a significant central nervous system (CNS) alteration. GFRα1 was predominantly expressed by neurons but also by an unexpected population of non-neuronal cells. GFRα1 expression pattern was wider in neonatal than in adult CNS and GDNF expression was restricted in comparison with GFRα1 at both developmental time points. The use of confocal microscopy to imaging X-gal deposits and EGFP allowed us to identify regions containing cells that expressed both proteins and to discriminate between auto and non-autotrophic signaling. We also suggested long-range GDNF-GFRα1 circuits taking advantage of the ability of the EGFP genetically encoded reporter to label long distance projecting axons. The complete elimination of either the ligand or the receptor during development did not produce major abnormalities, suggesting a preponderant role for GDNF signaling during adulthood. In the SC, our results pointed to local modulatory interneurons as the main target of GDNF produced by Clarke's column (CC) cells. Our work increases the understanding on how GDNF signals in the CNS and establish a crucial framework for posterior studies addressing either the biological role of GDNF or the optimization of trophic factor-based therapies.
Collapse
Affiliation(s)
- Clara Ortega-de San Luis
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| |
Collapse
|
46
|
Noraz N, Jaaoini I, Charoy C, Watrin C, Chounlamountri N, Benon A, Malleval C, Boudin H, Honnorat J, Castellani V, Pellier-Monnin V. Syk kinases are required for spinal commissural axon repulsion at the midline via the ephrin/Eph pathway. Development 2016; 143:2183-93. [PMID: 27122172 DOI: 10.1242/dev.128629] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 04/15/2016] [Indexed: 12/26/2022]
Abstract
In the hematopoietic system, Syk family tyrosine kinases are essential components of immunoreceptor ITAM-based signaling. While there is increasing data indicating the involvement of immunoreceptors in neural functions, the contribution of Syk kinases remains obscure. Previously, we identified phosphorylated forms of Syk kinases in specialized populations of migrating neurons or projecting axons. Moreover, we identified ephrin/Eph as guidance molecules utilizing the ITAM-bearing CD3zeta (Cd247) and associated Syk kinases for the growth cone collapse response induced in vitro Here, we show that in the developing spinal cord, Syk is phosphorylated in navigating commissural axons. By analyzing axon trajectories in open-book preparations of Syk(-/-); Zap70(-/-) mouse embryos, we show that Syk kinases are dispensable for attraction towards the midline but confer growth cone responsiveness to repulsive signals that expel commissural axons from the midline. Known to serve a repulsive function at the midline, ephrin B3/EphB2 are obvious candidates for driving the Syk-dependent repulsive response. Indeed, Syk kinases were found to be required for ephrin B3-induced growth cone collapse in cultured commissural neurons. In fragments of commissural neuron-enriched tissues, Syk is in a constitutively phosphorylated state and ephrin B3 decreased its level of phosphorylation. Direct pharmacological inhibition of Syk kinase activity was sufficient to induce growth cone collapse. In conclusion, Syk kinases act as a molecular switch of growth cone adhesive and repulsive responses.
Collapse
Affiliation(s)
- Nelly Noraz
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Iness Jaaoini
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Camille Charoy
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Chantal Watrin
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Naura Chounlamountri
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Aurélien Benon
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Céline Malleval
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Hélène Boudin
- INSERM U1064, Institut de Transplantation Urologie-Néphrologie, Nantes F-44035, France
| | - Jérôme Honnorat
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France Hospices Civils de Lyon, Lyon F-69000, France
| | - Valérie Castellani
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Véronique Pellier-Monnin
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| |
Collapse
|
47
|
NCAM-140 Translocation into Lipid Rafts Mediates the Neuroprotective Effects of GDNF. Mol Neurobiol 2016; 54:2739-2751. [PMID: 27003822 DOI: 10.1007/s12035-016-9749-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 01/26/2016] [Indexed: 10/22/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor for substantia nigra dopaminergic (DA) neuronal cells. Recent studies have demonstrated that neural cell adhesion molecule functions as a signal transduction receptor for GDNF. The purpose of this study is to reveal whether neural cell adhesion molecule (NCAM) mediates the protective effects of GDNF on DA neuronal cells and further explore the mechanisms involved. We utilized SH-SY5Y cell line to establish a model of 6-hydroxydopamine (6-OHDA)-injured DA neuronal cells. Lentiviral vectors were constructed to knockdown or overexpress NCAM-140, and a density gradient centrifugation method was employed to separate membrane lipid rafts. 3-(4,5-Dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), flow cytometric analysis, and western blotting were used to evaluate the protective effects of GDNF. The results showed that GDNF could protect 6-OHDA-injured SH-SY5Y cells via improving cell viability and decreasing the cell death rate and cleaved caspase-3 expression. NCAM-140 knockdown decreased cell viability and increased the cell death rate and cleaved caspase-3 expression, while its overexpression had the opposite effects. Notably, the amount of NCAM-140 located in lipid rafts increased after GDNF treatment. Pretreatment with 2-bromopalmitate, a specific inhibitor of protein palmitoylation, suppressed NCAM-140 translocation to lipid rafts and reduced the NCAM-mediated protective effects of GDNF on injured DA neuronal cells. Our results suggest that GDNF have the protective effects on injured DA cells by influencing NCAM-140 translocation into lipid rafts.
Collapse
|
48
|
Alther TA, Domanitskaya E, Stoeckli ET. Calsyntenin1-mediated trafficking of axon guidance receptors regulates the switch in axonal responsiveness at a choice point. Development 2016; 143:994-1004. [DOI: 10.1242/dev.127449] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 01/26/2016] [Indexed: 02/04/2023]
Abstract
Axon guidance at choice points depends on the precise regulation of guidance receptors on the growth cone surface. Upon arrival at the intermediate target or choice point, a switch from attraction to repulsion is required for the axon to move on. Dorsal commissural (dI1) axons crossing the ventral midline of the spinal cord in the floor plate represent a convenient model for the analysis of the molecular mechanism underlying the switch in axonal behavior.
We identified a role of Calsyntenin1 in the regulation of vesicular trafficking of guidance receptors in dI1 axons at choice points. In cooperation with RabGDI, Calsyntenin1 shuttles Rab11-positive vesicles containing Robo1 to the growth cone surface in a precisely regulated manner. In contrast, Calsyntenin1-mediated trafficking of Frizzled3, a guidance receptor in the Wnt pathway, is independent of RabGDI. Thus, tightly regulated insertion of guidance receptors, which is required for midline crossing and the subsequent turn into the longitudinal axis, is achieved by specific trafficking.
Collapse
Affiliation(s)
- Tobias A. Alther
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Elena Domanitskaya
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Esther T. Stoeckli
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| |
Collapse
|
49
|
Xiao N, Le QT. Neurotrophic Factors and Their Potential Applications in Tissue Regeneration. Arch Immunol Ther Exp (Warsz) 2015; 64:89-99. [PMID: 26611762 DOI: 10.1007/s00005-015-0376-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/02/2015] [Indexed: 12/24/2022]
Abstract
Neurotrophic factors are growth factors that can nourish neurons and promote neuron survival and regeneration. They have been studied as potential drug candidates for treating neurodegenerative diseases. Since their identification, there are more and more evidences to indicate that neurotrophic factors are also expressed in non-neuronal tissues and regulate the survival, anti-inflammation, proliferation and differentiation in these tissues. This mini review summarizes the characteristics of the neurotrophic factors and their potential clinical applications in the regeneration of neuronal and non-neuronal tissues.
Collapse
Affiliation(s)
- Nan Xiao
- Department of Biomedical Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA, USA.
| | - Quynh-Thu Le
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
50
|
Neuhaus-Follini A, Bashaw GJ. The Intracellular Domain of the Frazzled/DCC Receptor Is a Transcription Factor Required for Commissural Axon Guidance. Neuron 2015; 87:751-63. [PMID: 26291159 DOI: 10.1016/j.neuron.2015.08.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/24/2015] [Accepted: 08/03/2015] [Indexed: 10/23/2022]
Abstract
In commissural neurons of Drosophila, the conserved Frazzled (Fra)/Deleted in Colorectal Cancer (DCC) receptor promotes midline axon crossing by signaling locally in response to Netrin and by inducing transcription of commissureless (comm), an antagonist of Slit-Roundabout midline repulsion, through an unknown mechanism. Here, we show that Fra is cleaved to release its intracellular domain (ICD), which shuttles between the cytoplasm and the nucleus, where it functions as a transcriptional activator. Rescue and gain-of-function experiments demonstrate that the Fra ICD is sufficient to regulate comm expression and that both γ-secretase proteolysis of Fra and Fra's function as a transcriptional activator are required for its ability to regulate comm in vivo. Our data uncover an unexpected role for the Fra ICD as a transcription factor whose activity regulates the responsiveness of commissural axons at the midline and raise the possibility that nuclear signaling may be a common output of axon guidance receptors.
Collapse
Affiliation(s)
- Alexandra Neuhaus-Follini
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|