1
|
Sullivan KG, Bashaw GJ. Commissureless acts as a substrate adapter in a conserved Nedd4 E3 ubiquitin ligase pathway to promote axon growth across the midline. eLife 2025; 13:RP92757. [PMID: 40407164 PMCID: PMC12101832 DOI: 10.7554/elife.92757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2025] Open
Abstract
In both vertebrates and invertebrates, commissural neurons prevent premature responsiveness to the midline repellant Slit by downregulating surface levels of its receptor Roundabout1 (Robo1). In Drosophila, Commissureless (Comm) plays a critical role in this process; however, there is conflicting data on the underlying molecular mechanism. Here, we demonstrate that the conserved PY motifs in the cytoplasmic domain of Comm are required allow the ubiquitination and lysosomal degradation of Robo1. Disruption of these motifs prevents Comm from localizing to Lamp1 positive late endosomes and to promote axon growth across the midline in vivo. In addition, we conclusively demonstrate a role for Nedd4 in midline crossing. Genetic analysis shows that nedd4 mutations result in midline crossing defects in the Drosophila embryonic nerve cord, which can be rescued by introduction of exogenous Nedd4. Biochemical evidence shows that Nedd4 incorporates into a three-member complex with Comm and Robo1 in a PY motif-dependent manner. Finally, we present genetic evidence that Nedd4 acts with Comm in the embryonic nerve cord to downregulate Robo1 levels. Taken together, these findings demonstrate that Comm promotes midline crossing in the nerve cord by facilitating Robo1 ubiquitination by Nedd4, ultimately leading to its degradation.
Collapse
Affiliation(s)
- Kelly G Sullivan
- Department of Neuroscience, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
2
|
Hardin KR, Penas AB, Joubert S, Ye C, Myers KR, Zheng JQ. A Critical Role for the Fascin Family of Actin Bundling Proteins in Axon Development, Brain Wiring and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639554. [PMID: 40027761 PMCID: PMC11870622 DOI: 10.1101/2025.02.21.639554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Actin-based cell motility drives many neurodevelopmental events including guided axonal growth. Fascin is a major family of F-actin bundling proteins, but its role in axon development in vivo and brain wiring remains unclear. Here, we report that fascin is required for axon development, brain wiring and function. We show that fascin is enriched in the motile filopodia of axonal growth cones and its inhibition impairs axonal extension and branching of hippocampal neurons in culture. We next provide evidence that fascin is essential for axon development and brain wiring in vivo using Drosophila melanogaster as a model. Drosophila expresses a single ortholog of mammalian fascin called Singed (SN), which is highly expressed in the mushroom body (MB) of the central nervous system. We observe that loss of SN results in drastic MB disruption, highlighted by α- and β-lobe defects that are consistent with altered axonal guidance. SN-null flies also exhibit defective sensorimotor behaviors as assessed by the negative geotaxis assay. MB- specific expression of SN in SN-null flies rescues MB structure and sensorimotor deficits, indicating that SN functions autonomously in MB neurons. Together, our data from primary neuronal culture and in vivo models highlight a critical role for fascin in brain development and function. Highlights Fascin regulates axonal growth and branching of hippocampal neurons in culture.Singed, Drosophila fascin, is enriched specifically in mushroom body (MB) axons.Singed loss causes axon guidance defects and sensorimotor issues in flies.MB-specific Singed re-expression rescues MB structure and behavior in flies.
Collapse
|
3
|
Carranza A, Howard LJ, Brown HE, Ametepe AS, Evans TA. Slit-independent guidance of longitudinal axons by Drosophila Robo3. Dev Biol 2025; 521:14-27. [PMID: 39909366 PMCID: PMC11908893 DOI: 10.1016/j.ydbio.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
Drosophila Robo3 is a member of the evolutionarily conserved Roundabout (Robo) receptor family and one of three Drosophila Robo paralogs. During embryonic ventral nerve cord development, Robo3 does not participate in canonical Slit-dependent midline repulsion, but instead regulates the formation of longitudinal axon pathways at specific positions along the medial-lateral axis. Longitudinal axon guidance by Robo3 is hypothesized to be Slit dependent, but this has not been directly tested. Here we create a series of Robo3 variants in which the N-terminal Ig1 domain is deleted or modified, in order to characterize the functional importance of Ig1 and Slit binding for Robo3's axon guidance activity. We show that Robo3 requires its Ig1 domain for interaction with Slit and for proper axonal localization in embryonic neurons, but deleting Ig1 from Robo3 only partially disrupts longitudinal pathway formation. Robo3 variants with modified Ig1 domains that cannot bind Slit retain proper localization and fully rescue longitudinal axon guidance. Our results indicate that Robo3 guides longitudinal axons independently of Slit, and that sequences both within and outside of Ig1 contribute to this Slit-independent activity.
Collapse
Affiliation(s)
- Abigail Carranza
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, 72701, USA
| | - LaFreda J Howard
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Haley E Brown
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Ayawovi Selom Ametepe
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Timothy A Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, 72701, USA.
| |
Collapse
|
4
|
Wang KT, Chen YC, Tsai FY, Judy CP, Adler CE. Pluripotent Stem Cell Plasticity is Sculpted by a Slit-Independent Robo Pathway in a Regenerative Animal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.14.648795. [PMID: 40376085 PMCID: PMC12080947 DOI: 10.1101/2025.04.14.648795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Whole-body regeneration requires adult stem cells with high plasticity to differentiate into missing cell types. Planarians possess a unique configuration of organs embedded in a vast pool of pluripotent stem cells. How stem cells integrate positional information with discrete fates remains unknown. Here, we use the planarian pharynx to define the cell fates that depend on the pioneer transcription factor FoxA. We find that Roundabout receptor RoboA suppresses aberrant pharynx cell fates by altering foxA expression, independent of the canonical ligand Slit. An RNAi screen for extracellular proteins identifies Anosmin-1 as a potential partner of RoboA. Perturbing global patterning demonstrates that roboA / anosmin-1 functions locally in the brain. By contrast, altering pharynx fate with foxA knockdown induces head-specific neurons in the pharynx, indicating a latent plasticity of stem cells. Our data links critical extracellular cues with cell fate decisions of highly plastic stem cells, ensuring the fidelity of organ regeneration.
Collapse
|
5
|
Li Z, Lyu C, Xu C, Hu Y, Luginbuhl DJ, Caspi-Lebovic AB, Priest JM, Özkan E, Luo L. Repulsive interactions instruct synaptic partner matching in an olfactory circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.01.640985. [PMID: 40060423 PMCID: PMC11888401 DOI: 10.1101/2025.03.01.640985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Neurons exhibit extraordinary precision in selecting synaptic partners. Whereas cell-surface proteins (CSPs) mediating attractive interactions between developing axons and dendrites have been shown to instruct synaptic partner matching1,2, it is less clear the degree to which repulsive interactions play a role. Here, using a genetic screen guided by single cell transcriptomes3,4, we identified three CSP pairs-Toll2-Ptp10D, Fili-Kek1, and Hbs/Sns-Kirre-in mediating repulsive interactions between non-partner olfactory receptor neuron (ORN) axons and projection neuron (PN) dendrites in the developing Drosophila olfactory circuit. Each CSP pair exhibits inverse expression patterns in the select PN-ORN partners. Loss of each CSP in ORNs led to similar synaptic partner matching deficits as the loss of its partner CSP in PNs, and mistargeting phenotypes caused by overexpressing one CSP could be suppressed by loss of its partner CSP. Each CSP pair is also differentially expressed in other brain regions. Together, our data reveal that multiple repulsive CSP pairs work together to ensure precise synaptic partner matching during development by preventing neurons from forming connections with non-cognate partners.
Collapse
Affiliation(s)
- Zhuoran Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Cheng Lyu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Ying Hu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - David J. Luginbuhl
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Asaf B. Caspi-Lebovic
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Jessica M. Priest
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
6
|
Nelson KA, Lenhart KF, Anllo L, DiNardo S. The Drosophila hematopoietic niche assembles through collective cell migration controlled by neighbor tissues and Slit-Robo signaling. eLife 2025; 13:RP100455. [PMID: 39750120 PMCID: PMC11698496 DOI: 10.7554/elife.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Niches are often found in specific positions in tissues relative to the stem cells they support. Consistency of niche position suggests that placement is important for niche function. However, the complexity of most niches has precluded a thorough understanding of how their proper placement is established. To address this, we investigated the formation of a genetically tractable niche, the Drosophila Posterior Signaling Center (PSC), the assembly of which had not been previously explored. This niche controls hematopoietic progenitors of the lymph gland (LG). PSC cells were previously shown to be specified laterally in the embryo, but ultimately reside dorsally, at the LG posterior. Here, using live-imaging, we show that PSC cells migrate as a tight collective and associate with multiple tissues during their trajectory to the LG posterior. We find that Slit emanating from two extrinsic sources, visceral mesoderm and cardioblasts, is required for the PSC to remain a collective, and for its attachment to cardioblasts during migration. Without proper Slit-Robo signaling, PSC cells disperse, form aberrant contacts, and ultimately fail to reach their stereotypical position near progenitors. Our work characterizes a novel example of niche formation and identifies an extrinsic signaling relay that controls precise niche positioning.
Collapse
Affiliation(s)
- Kara A Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Kari F Lenhart
- Department of Biology, Drexel UniversityPhiladelphiaUnited States
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
7
|
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
|
8
|
Liu Y, Seguin C, Betzel RF, Han D, Akarca D, Di Biase MA, Zalesky A. A generative model of the connectome with dynamic axon growth. Netw Neurosci 2024; 8:1192-1211. [PMID: 39735503 PMCID: PMC11674315 DOI: 10.1162/netn_a_00397] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/03/2024] [Indexed: 12/31/2024] Open
Abstract
Connectome generative models, otherwise known as generative network models, provide insight into the wiring principles underpinning brain network organization. While these models can approximate numerous statistical properties of empirical networks, they typically fail to explicitly characterize an important contributor to brain organization-axonal growth. Emulating the chemoaffinity-guided axonal growth, we provide a novel generative model in which axons dynamically steer the direction of propagation based on distance-dependent chemoattractive forces acting on their growth cones. This simple dynamic growth mechanism, despite being solely geometry-dependent, is shown to generate axonal fiber bundles with brain-like geometry and features of complex network architecture consistent with the human brain, including lognormally distributed connectivity weights, scale-free nodal degrees, small-worldness, and modularity. We demonstrate that our model parameters can be fitted to individual connectomes, enabling connectome dimensionality reduction and comparison of parameters between groups. Our work offers an opportunity to bridge studies of axon guidance and connectome development, providing new avenues for understanding neural development from a computational perspective.
Collapse
Affiliation(s)
- Yuanzhe Liu
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Caio Seguin
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Richard F. Betzel
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Daniel Han
- School of Mathematics and Statistics, University of New South Wales, Sydney, NSW, Australia
| | - Danyal Akarca
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
- Department of Electrical and Electronic Engineering, Imperial College London, London, UK
| | - Maria A. Di Biase
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
- Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Zalesky
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Geraldo LH, Xu Y, Mouthon G, Furtado J, Leser FS, Blazer LL, Adams JJ, Zhang S, Zheng L, Song E, Robinson ME, Thomas JL, Sidhu SS, Eichmann A. Monoclonal antibodies that block Roundabout 1 and 2 signaling target pathological ocular neovascularization through myeloid cells. Sci Transl Med 2024; 16:eadn8388. [PMID: 39565875 PMCID: PMC11822886 DOI: 10.1126/scitranslmed.adn8388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 07/29/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
Roundabout (ROBO) 1 and 2 are transmembrane receptors that bind secreted SLIT ligands through their extracellular domains (ECDs) and signal through their cytoplasmic domains to modulate the cytoskeleton and regulate cell migration, adhesion, and proliferation. SLIT-ROBO signaling regulates pathological ocular neovascularization, which is a major cause of vision loss worldwide, but pharmacological tools to prevent SLIT-ROBO signaling are lacking. Here, we developed human monoclonal antibodies (mAbs) against the ROBO1 and ROBO2 ECDs. One antibody that inhibited in vitro SLIT2 signaling through ROBO1 and ROBO2 (anti-ROBO1/2) also reduced ocular neovascularization in oxygen-induced retinopathy (OIR) and laser-induced corneal neovascularization (CNV) mouse models in vivo. Single-cell RNA sequencing of OIR retinas revealed that antibody treatment affected several cell types relevant to physiological and pathological angiogenesis, including endothelial cells, pericytes, and a heterogeneous population of myeloid cells. mAb treatment improved blood-retina barrier integrity and prevented pathological pericyte activation in OIR. SLIT-ROBO signaling inhibition prevented pathological activation of myeloid cells and increased neuroprotective myeloid populations normally seen in the developing retina. Microglia/infiltrating macrophage-specific ablation of Robo1 and Robo2 or knockout of the downstream effector phosphatidylinositol 3-kinase (Pik3cg) encoding PI3Kγ in both OIR and CNV models phenocopied anti-ROBO1/2 treatment, further demonstrating the key role of myeloid cells as drivers of ocular neovascular diseases. ROBO1/2 blocking antibodies may thus provide a promising strategy to combat inflammation in blinding eye diseases.
Collapse
Affiliation(s)
- Luiz Henrique Geraldo
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yunling Xu
- Université de Paris, INSERM, PARCC, F-75015 Paris, France
| | - Gaspard Mouthon
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jessica Furtado
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | - Levi L. Blazer
- School of Pharmacy, University of Waterloo, Kitchener, Ontario N2G 1C5, Canada
| | - Jarrett J. Adams
- School of Pharmacy, University of Waterloo, Kitchener, Ontario N2G 1C5, Canada
| | - Sophia Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Lana Zheng
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Eric Song
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Ophthalmology and Visual Sciences, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Mark E. Robinson
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, F-75013 Paris, France
| | - Sachdev S. Sidhu
- School of Pharmacy, University of Waterloo, Kitchener, Ontario N2G 1C5, Canada
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Université de Paris, INSERM, PARCC, F-75015 Paris, France
| |
Collapse
|
10
|
Sullivan KG, Bashaw GJ. Commissureless acts as a substrate adapter in a conserved Nedd4 E3 ubiquitin ligase pathway to promote axon growth across the midline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.13.562283. [PMID: 37905056 PMCID: PMC10614773 DOI: 10.1101/2023.10.13.562283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
In both vertebrates and invertebrates, commissural neurons prevent premature responsiveness to the midline repellant Slit by downregulating surface levels of its receptor Roundabout1 (Robo1). In Drosophila, Commissureless (Comm) plays a critical role in this process; however, there is conflicting data on the underlying molecular mechanism. Here, we demonstrate that the conserved PY motifs in the cytoplasmic domain of Comm are required allow the ubiquitination and lysosomal degradation of Robo1. Disruption of these motifs prevents Comm from localizing to Lamp1 positive late endosomes and to promote axon growth across the midline in vivo. In addition, we conclusively demonstrate a role for Nedd4 in midline crossing. Genetic analysis shows that nedd4 mutations result in midline crossing defects in the Drosophila embryonic nerve cord, which can be rescued by introduction of exogenous Nedd4. Biochemical evidence shows that Nedd4 incorporates into a three-member complex with Comm and Robo1 in a PY motif-dependent manner. Finally, we present genetic evidence that Nedd4 acts with Comm in the embryonic nerve cord to downregulate Robo1 levels. Taken together, these findings demonstrate that Comm promotes midline crossing in the nerve cord by facilitating Robo1 ubiquitination by Nedd4, ultimately leading to its degradation.
Collapse
Affiliation(s)
- Kelly G. Sullivan
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, 415 Curie Blvd., Philadelphia, PA, 19104, USA
| | - Greg J. Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, 415 Curie Blvd., Philadelphia, PA, 19104, USA
| |
Collapse
|
11
|
Deng X, Sandoval IC, Zhu S. Slit regulates compartment-specific targeting of dendrites and axons in the Drosophila brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620851. [PMID: 39554193 PMCID: PMC11565903 DOI: 10.1101/2024.10.29.620851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Proper functioning of the nervous system requires precise neuronal connections at subcellular domains, which can be achieved by projection of axons or dendrites to subcellular domains of target neurons. Here we studied subcellular-specific targeting of dendrites and axons in the Drosophila mushroom body (MB), where mushroom body output neurons (MBONs) and local dopaminergic neurons (DAN) project their dendrites and axons, respectively, to specific compartments of MB axons. Through genetic ablation, we demonstrate that compartment-specific targeting of MBON dendrites and DAN axons involves mutual repulsion of MBON dendrites and/or DAN axons between neighboring compartments. We further show that Slit expressed in subset of DANs mediates such repulsion by acting through different Robo receptors in different neurons. Loss of Slit-mediated repulsion leads to projection of MBON dendrites and DAN axons into neighboring compartments, resulting formation of ectopic synaptic contacts between MBONs and DANs and changes in olfactory-associative learning. Together, our findings suggest that Slit-mediated repulsion controls compartment-specific targeting of MBON dendrites and DAN axons, which ensures precise connections between MBON dendrites and DAN axons and proper learning and memory formation.
Collapse
|
12
|
Nelson KA, Lenhart KF, Anllo L, DiNardo S. The Drosophila hematopoietic niche assembles through collective cell migration controlled by neighbor tissues and Slit-Robo signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600069. [PMID: 38979182 PMCID: PMC11230208 DOI: 10.1101/2024.06.21.600069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Niches are often found in specific positions in tissues relative to the stem cells they support. Consistency of niche position suggests that placement is important for niche function. However, the complexity of most niches has precluded a thorough understanding of how their proper placement is established. To address this, we investigated the formation of a genetically tractable niche, the Drosophila Posterior Signaling Center (PSC), the assembly of which had not been previously explored. This niche controls hematopoietic progenitors of the lymph gland (LG). PSC cells were previously shown to be specified laterally in the embryo, but ultimately reside dorsally, at the LG posterior. Here, using live-imaging, we show that PSC cells migrate as a tight collective and associate with multiple tissues during their trajectory to the LG posterior. We find that Slit emanating from two extrinsic sources, visceral mesoderm and cardioblasts, is required for the PSC to remain a collective, and for its attachment to cardioblasts during migration. Without proper Slit-Robo signaling, PSC cells disperse, form aberrant contacts, and ultimately fail to reach their stereotypical position near progenitors. Our work characterizes a novel example of niche formation and identifies an extrinsic signaling relay that controls precise niche positioning.
Collapse
Affiliation(s)
- Kara A Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
| | - Kari F Lenhart
- Department of Biology, Drexel University, 3245 Chestnut St. Philadelphia, PA 19104, United States
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
- Present address: Department of Biology, East Carolina University, 458 Science & Tech Bldg. Greenville, NC 27858, United States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
| |
Collapse
|
13
|
Bui KC, Kamiyama D. Adjacent Neuronal Fascicle Guides Motoneuron 24 Dendritic Branching and Axonal Routing Decisions through Dscam1 Signaling. eNeuro 2024; 11:ENEURO.0130-24.2024. [PMID: 39349058 PMCID: PMC11495862 DOI: 10.1523/eneuro.0130-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/08/2024] [Accepted: 08/27/2024] [Indexed: 10/02/2024] Open
Abstract
The formation and precise positioning of axons and dendrites are crucial for the development of neural circuits. Although juxtacrine signaling via cell-cell contact is known to influence these processes, the specific structures and mechanisms regulating neuronal process positioning within the central nervous system (CNS) remain to be fully identified. Our study investigates motoneuron 24 (MN24) in the Drosophila embryonic CNS, which is characterized by a complex yet stereotyped axon projection pattern, known as "axonal routing." In this motoneuron, the primary dendritic branches project laterally toward the midline, specifically emerging at the sites where axons turn. We observed that Scp2-positive neurons contribute to the lateral fascicle structure in the ventral nerve cord (VNC) near MN24 dendrites. Notably, the knockout of the Down syndrome cell adhesion molecule (Dscam1) results in the loss of dendrites and disruption of proper axonal routing in MN24, while not affecting the formation of the fascicle structure. Through cell-type specific knockdown and rescue experiments of Dscam1, we have determined that the interaction between MN24 and Scp2-positive fascicle, mediated by Dscam1, promotes the development of both dendrites and axonal routing. Our findings demonstrate that the holistic configuration of neuronal structures, such as axons and dendrites, within single motoneurons can be governed by local contact with the adjacent neuron fascicle, a novel reference structure for neural circuitry wiring.
Collapse
Affiliation(s)
- Kathy Clara Bui
- Department of Cellular Biology, University of Georgia, Athens, Georgia 30605
| | - Daichi Kamiyama
- Department of Cellular Biology, University of Georgia, Athens, Georgia 30605
| |
Collapse
|
14
|
Johnson AN. Myotube Guidance: Shaping up the Musculoskeletal System. J Dev Biol 2024; 12:25. [PMID: 39311120 PMCID: PMC11417883 DOI: 10.3390/jdb12030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/20/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Myofibers are highly specialized contractile cells of skeletal muscles, and dysregulation of myofiber morphogenesis is emerging as a contributing cause of myopathies and structural birth defects. Myotubes are the myofiber precursors and undergo a dramatic morphological transition into long bipolar myofibers that are attached to tendons on two ends. Similar to axon growth cones, myotube leading edges navigate toward target cells and form cell-cell connections. The process of myotube guidance connects myotubes with the correct tendons, orients myofiber morphology with the overall body plan, and generates a functional musculoskeletal system. Navigational signaling, addition of mass and volume, and identification of target cells are common events in myotube guidance and axon guidance, but surprisingly, the mechanisms regulating these events are not completely overlapping in myotubes and axons. This review summarizes the strategies that have evolved to direct myotube leading edges to predetermined tendon cells and highlights key differences between myotube guidance and axon guidance. The association of myotube guidance pathways with developmental disorders is also discussed.
Collapse
Affiliation(s)
- Aaron N Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
15
|
Kidd T, Evans T. Analysis of Axon Guidance in the Drosophila Embryo. Cold Spring Harb Protoc 2024; 2024:pdb.top108109. [PMID: 37419653 DOI: 10.1101/pdb.top108109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
The establishment of neural connectivity is a major part of neural development. The central nervous system (CNS) midline is the most characterized axon guidance choice point, and work in Drosophila has played a pivotal role in understanding the molecular mechanisms responsible. Axons respond to attractive cues such as Netrin via the Frazzled receptor, and repulsive cues such as Slit via Robo receptors. Both signals are expressed at the CNS midline, affect pioneer axons, and have dramatic effects on the axon scaffold as a whole. Here, we focus on previous research analyzing classic mutants in the Slit/Robo pathway, which can readily be detected with a dissecting microscope. We also discuss analyzing these mutants in a teaching lab situation. The combination of sophisticated genetics and reliable axonal markers in Drosophila allows phenotypic analysis to be performed at the single-cell level. The elaborate architecture of neurons is very sensitive to disruption by genetic mutations, allowing the effects of novel mutations to be easily detected and assessed.
Collapse
Affiliation(s)
- Thomas Kidd
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Timothy Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, USA
| |
Collapse
|
16
|
Liu J, Wang Y, Liu X, Han J, Tian Y. Spatiotemporal changes in Netrin/Dscam1 signaling dictate axonal projection direction in Drosophila small ventral lateral clock neurons. eLife 2024; 13:RP96041. [PMID: 39052321 PMCID: PMC11272162 DOI: 10.7554/elife.96041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
Axon projection is a spatial- and temporal-specific process in which the growth cone receives environmental signals guiding axons to their final destination. However, the mechanisms underlying changes in axonal projection direction without well-defined landmarks remain elusive. Here, we present evidence showcasing the dynamic nature of axonal projections in Drosophila's small ventral lateral clock neurons (s-LNvs). Our findings reveal that these axons undergo an initial vertical projection in the early larval stage, followed by a subsequent transition to a horizontal projection in the early-to-mid third instar larvae. The vertical projection of s-LNv axons correlates with mushroom body calyx expansion, while the s-LNv-expressed Down syndrome cell adhesion molecule (Dscam1) interacts with Netrins to regulate the horizontal projection. During a specific temporal window, locally newborn dorsal clock neurons secrete Netrins, facilitating the transition of axonal projection direction in s-LNvs. Our study establishes a compelling in vivo model to probe the mechanisms of axonal projection direction switching in the absence of clear landmarks. These findings underscore the significance of dynamic local microenvironments in the complementary regulation of axonal projection direction transitions.
Collapse
Affiliation(s)
- Jingjing Liu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
| | - Yuedong Wang
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
| | - Xian Liu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
- Co-innovation Center of Neuroregeneration, Nantong UniversityNantongChina
| | - Yao Tian
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast UniversityNanjingChina
| |
Collapse
|
17
|
Bui KC, Kamiyama D. Adjacent Neuronal Fascicle Guides Motoneuron 24 Dendritic Branching and Axonal Routing Decisions through Dscam1 Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588591. [PMID: 38645010 PMCID: PMC11030417 DOI: 10.1101/2024.04.08.588591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The formation and precise positioning of axons and dendrites are crucial for the development of neural circuits. Although juxtracrine signaling via cell-cell contact is known to influence these processes, the specific structures and mechanisms regulating neuronal process positioning within the central nervous system (CNS) remain to be fully identified. Our study investigates motoneuron 24 (MN24) in the Drosophila embryonic CNS, which is characterized by a complex yet stereotyped axon projection pattern, known as 'axonal routing.' In this motoneuron, the primary dendritic branches project laterally toward the midline, specifically emerging at the sites where axons turn. We observed that Scp2-positive neurons contribute to the lateral fascicle structure in the ventral nerve cord (VNC) near MN24 dendrites. Notably, the knockout of the Down syndrome cell adhesion molecule (dscam1) results in the loss of dendrites and disruption of proper axonal routing in MN24, while not affecting the formation of the fascicle structure. Through cell-type specific knockdown and rescue experiments of dscam1, we have determined that the interaction between MN24 and Scp2-positive fascicle, mediated by Dscam1, promotes the development of both dendrites and axonal routing. Our findings demonstrate that the holistic configuration of neuronal structures, such as axons and dendrites, within single motoneurons can be governed by local contact with the adjacent neuron fascicle, a novel reference structure for neural circuitry wiring.
Collapse
Affiliation(s)
- Kathy Clara Bui
- Department of Cellular Biology, University of Georgia, Athens, GA 30605, USA
| | - Daichi Kamiyama
- Department of Cellular Biology, University of Georgia, Athens, GA 30605, USA
| |
Collapse
|
18
|
Hoving JJA, Harford-Wright E, Wingfield-Digby P, Cattin AL, Campana M, Power A, Morgan T, Torchiaro E, Quereda V, Lloyd AC. N-cadherin directs the collective Schwann cell migration required for nerve regeneration through Slit2/3-mediated contact inhibition of locomotion. eLife 2024; 13:e88872. [PMID: 38591541 PMCID: PMC11052573 DOI: 10.7554/elife.88872] [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: 04/25/2023] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
Collective cell migration is fundamental for the development of organisms and in the adult for tissue regeneration and in pathological conditions such as cancer. Migration as a coherent group requires the maintenance of cell-cell interactions, while contact inhibition of locomotion (CIL), a local repulsive force, can propel the group forward. Here we show that the cell-cell interaction molecule, N-cadherin, regulates both adhesion and repulsion processes during Schwann cell (SC) collective migration, which is required for peripheral nerve regeneration. However, distinct from its role in cell-cell adhesion, the repulsion process is independent of N-cadherin trans-homodimerisation and the associated adherens junction complex. Rather, the extracellular domain of N-cadherin is required to present the repulsive Slit2/Slit3 signal at the cell surface. Inhibiting Slit2/Slit3 signalling inhibits CIL and subsequently collective SC migration, resulting in adherent, nonmigratory cell clusters. Moreover, analysis of ex vivo explants from mice following sciatic nerve injury showed that inhibition of Slit2 decreased SC collective migration and increased clustering of SCs within the nerve bridge. These findings provide insight into how opposing signals can mediate collective cell migration and how CIL pathways are promising targets for inhibiting pathological cell migration.
Collapse
Affiliation(s)
- Julian JA Hoving
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Elizabeth Harford-Wright
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Patrick Wingfield-Digby
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Anne-Laure Cattin
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Mariana Campana
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Alex Power
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Toby Morgan
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Erica Torchiaro
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Victor Quereda
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College LondonLondonUnited Kingdom
| |
Collapse
|
19
|
Liu X, Li B, Wang S, Zhang E, Schultz M, Touma M, Monteiro Da Rocha A, Evans SM, Eichmann A, Herron T, Chen R, Xiong D, Jaworski A, Weiss S, Si MS. Stromal Cell-SLIT3/Cardiomyocyte-ROBO1 Axis Regulates Pressure Overload-Induced Cardiac Hypertrophy. Circ Res 2024; 134:913-930. [PMID: 38414132 PMCID: PMC10977056 DOI: 10.1161/circresaha.122.321292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Recently shown to regulate cardiac development, the secreted axon guidance molecule SLIT3 maintains its expression in the postnatal heart. Despite its known expression in the cardiovascular system after birth, SLIT3's relevance to cardiovascular function in the postnatal state remains unknown. As such, the objectives of this study were to determine the postnatal myocardial sources of SLIT3 and to evaluate its functional role in regulating the cardiac response to pressure overload stress. METHODS We performed in vitro studies on cardiomyocytes and myocardial tissue samples from patients and performed in vivo investigation with SLIT3 and ROBO1 (roundabout homolog 1) mutant mice undergoing transverse aortic constriction to establish the role of SLIT3-ROBO1 in adverse cardiac remodeling. RESULTS We first found that SLIT3 transcription was increased in myocardial tissue obtained from patients with congenital heart defects that caused ventricular pressure overload. Immunostaining of hearts from WT (wild-type) and reporter mice revealed that SLIT3 is secreted by cardiac stromal cells, namely fibroblasts and vascular mural cells, within the heart. Conditioned media from cardiac fibroblasts and vascular mural cells both stimulated cardiomyocyte hypertrophy in vitro, an effect that was partially inhibited by an anti-SLIT3 antibody. Also, the N-terminal, but not the C-terminal, fragment of SLIT3 and the forced overexpression of SLIT3 stimulated cardiomyocyte hypertrophy and the transcription of hypertrophy-related genes. We next determined that ROBO1 was the most highly expressed roundabout receptor in cardiomyocytes and that ROBO1 mediated SLIT3's hypertrophic effects in vitro. In vivo, Tcf21+ fibroblast and Tbx18+ vascular mural cell-specific knockout of SLIT3 in mice resulted in decreased left ventricular hypertrophy and cardiac fibrosis after transverse aortic constriction. Furthermore, α-MHC+ cardiomyocyte-specific deletion of ROBO1 also preserved left ventricular function and abrogated hypertrophy, but not fibrosis, after transverse aortic constriction. CONCLUSIONS Collectively, these results indicate a novel role for the SLIT3-ROBO1-signaling axis in regulating postnatal cardiomyocyte hypertrophy induced by pressure overload.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, China (X.L., R.C.)
| | - Baolei Li
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, China (B.L.)
| | - Shuyun Wang
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
| | - Erge Zhang
- Division of Cardiac Surgery, Department of Surgery (E.Z., M.S., M.-S.S.), David Geffen School of Medicine University of California, Los Angeles
| | - Megan Schultz
- Division of Cardiac Surgery, Department of Surgery (E.Z., M.S., M.-S.S.), David Geffen School of Medicine University of California, Los Angeles
| | - Marlin Touma
- Department of Pediatrics (M.T.), David Geffen School of Medicine University of California, Los Angeles
| | - Andre Monteiro Da Rocha
- Division of Cardiovascular Medicine, Department of Internal Medicine (A.M.D.R., T.H.), Michigan Medicine, Ann Arbor
| | - Sylvia M. Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences (S.M.E.), University of California, San Diego, La Jolla
- Department of Medicine, School of Medicine (S.M.E.), University of California, San Diego, La Jolla
| | - Anne Eichmann
- Department of Internal Medicine, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.E.)
- INSERM, Paris Cardiovascular Research Center (PARCC), Université de Paris, France (A.E.)
| | - Todd Herron
- Division of Cardiovascular Medicine, Department of Internal Medicine (A.M.D.R., T.H.), Michigan Medicine, Ann Arbor
| | - Ruizhen Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, China (X.L., R.C.)
| | - Dingding Xiong
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
| | - Alexander Jaworski
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI (A.J.)
| | - Stephen Weiss
- Life Sciences Institute, University of Michigan, Ann Arbor (S.W.)
| | - Ming-Sing Si
- Department of Cardiac Surgery (X.L., B.L., S.W., D.X., M.-S.S.), Michigan Medicine, Ann Arbor
- Division of Cardiac Surgery, Department of Surgery (E.Z., M.S., M.-S.S.), David Geffen School of Medicine University of California, Los Angeles
| |
Collapse
|
20
|
Liu Y, Seguin C, Betzel RF, Akarca D, Di Biase MA, Zalesky A. A generative model of the connectome with dynamic axon growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581824. [PMID: 38464116 PMCID: PMC10925171 DOI: 10.1101/2024.02.23.581824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Connectome generative models, otherwise known as generative network models, provide insight into the wiring principles underpinning brain network organization. While these models can approximate numerous statistical properties of empirical networks, they typically fail to explicitly characterize an important contributor to brain organization - axonal growth. Emulating the chemoaffinity guided axonal growth, we provide a novel generative model in which axons dynamically steer the direction of propagation based on distance-dependent chemoattractive forces acting on their growth cones. This simple dynamic growth mechanism, despite being solely geometry-dependent, is shown to generate axonal fiber bundles with brain-like geometry and features of complex network architecture consistent with the human brain, including lognormally distributed connectivity weights, scale-free nodal degrees, small-worldness, and modularity. We demonstrate that our model parameters can be fitted to individual connectomes, enabling connectome dimensionality reduction and comparison of parameters between groups. Our work offers an opportunity to bridge studies of axon guidance and connectome development, providing new avenues for understanding neural development from a computational perspective.
Collapse
Affiliation(s)
- Yuanzhe Liu
- Department of Biomedical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Caio Seguin
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Richard F. Betzel
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Danyal Akarca
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, UK
| | - Maria A. Di Biase
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
- Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Zalesky
- Department of Biomedical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
21
|
Wang M, Fan J, Shao Z. Cellular and Molecular Mechanisms Underlying Synaptic Subcellular Specificity. Brain Sci 2024; 14:155. [PMID: 38391729 PMCID: PMC10886843 DOI: 10.3390/brainsci14020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 02/24/2024] Open
Abstract
Chemical synapses are essential for neuronal information storage and relay. The synaptic signal received or sent from spatially distinct subcellular compartments often generates different outcomes due to the distance or physical property difference. Therefore, the final output of postsynaptic neurons is determined not only by the type and intensity of synaptic inputs but also by the synaptic subcellular location. How synaptic subcellular specificity is determined has long been the focus of study in the neurodevelopment field. Genetic studies from invertebrates such as Caenorhabditis elegans (C. elegans) have uncovered important molecular and cellular mechanisms required for subcellular specificity. Interestingly, similar molecular mechanisms were found in the mammalian cerebellum, hippocampus, and cerebral cortex. This review summarizes the comprehensive advances in the cellular and molecular mechanisms underlying synaptic subcellular specificity, focusing on studies from C. elegans and rodents.
Collapse
Affiliation(s)
- Mengqing Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, 131 Dong An Rd, Research Building B4017, Shanghai 200032, China
| | - Jiale Fan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, 131 Dong An Rd, Research Building B4017, Shanghai 200032, China
| | - Zhiyong Shao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, 131 Dong An Rd, Research Building B4017, Shanghai 200032, China
| |
Collapse
|
22
|
Due Tankmar M, Reichel M, Arribas‐Hernández L, Brodersen P. A YTHDF-PABP interaction is required for m 6 A-mediated organogenesis in plants. EMBO Rep 2023; 24:e57741. [PMID: 38009565 PMCID: PMC10702811 DOI: 10.15252/embr.202357741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/29/2023] Open
Abstract
N6-methyladenosine (m6 A) in mRNA is key to eukaryotic gene regulation. Many m6 A functions involve RNA-binding proteins that recognize m6 A via a YT521-B Homology (YTH) domain. YTH domain proteins contain long intrinsically disordered regions (IDRs) that may mediate phase separation and interaction with protein partners, but whose precise biochemical functions remain largely unknown. The Arabidopsis thaliana YTH domain proteins ECT2, ECT3, and ECT4 accelerate organogenesis through stimulation of cell division in organ primordia. Here, we use ECT2 to reveal molecular underpinnings of this function. We show that stimulation of leaf formation requires the long N-terminal IDR, and we identify two short IDR elements required for ECT2-mediated organogenesis. Of these two, a 19-amino acid region containing a tyrosine-rich motif conserved in both plant and metazoan YTHDF proteins is necessary for binding to the major cytoplasmic poly(A)-binding proteins PAB2, PAB4, and PAB8. Remarkably, overexpression of PAB4 in leaf primordia partially rescues the delayed leaf formation in ect2 ect3 ect4 mutants, suggesting that the ECT2-PAB2/4/8 interaction on target mRNAs of organogenesis-related genes may overcome limiting PAB concentrations in primordial cells.
Collapse
Affiliation(s)
| | - Marlene Reichel
- Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | | | - Peter Brodersen
- Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
23
|
Gong L, Si MS. SLIT3-mediated fibroblast signaling: a promising target for antifibrotic therapies. Am J Physiol Heart Circ Physiol 2023; 325:H1400-H1411. [PMID: 37830982 PMCID: PMC11932536 DOI: 10.1152/ajpheart.00216.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023]
Abstract
The SLIT family (SLIT1-3) of highly conserved glycoproteins was originally identified as ligands for the Roundabout (ROBO) family of single-pass transmembrane receptors, serving to provide repulsive axon guidance cues in the nervous system. Intriguingly, studies involving SLIT3 mutant mice suggest that SLIT3 might have crucial biological functions outside the neural context. Although these mutant mice display no noticeable neurological abnormalities, they present pronounced connective tissue defects, including congenital central diaphragmatic hernia, membranous ventricular septal defect, and osteopenia. We recently hypothesized that the phenotype observed in SLIT3-deficient mice may be tied to abnormalities in fibrillar collagen-rich connective tissue. Further research by our group indicates that both SLIT3 and its primary receptor, ROBO1, are expressed in fibrillar collagen-producing cells across various nonneural tissues. Global and constitutive SLIT3 deficiency not only reduces the synthesis and content of fibrillar collagen in various organs but also alleviates pressure overload-induced fibrosis in both the left and right ventricles. This review delves into the known phenotypes of SLIT3 mutants and the debated role of SLIT3 in vasculature and bone. Present evidence hints at SLIT3 acting as an autocrine regulator of fibrillar collagen synthesis, suggesting it as a potential antifibrotic treatment. However, the precise pathway and mechanisms through which SLIT3 regulates fibrillar collagen synthesis remain uncertain, presenting an intriguing avenue for future research.
Collapse
Affiliation(s)
- Lianghui Gong
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People's Republic of China
| | - Ming-Sing Si
- Division of Cardiac Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| |
Collapse
|
24
|
Kerstein PC, Agreda YS, Curran BM, Ma L, Wright KM. Gbx2 controls amacrine cell dendrite stratification through Robo1/2 receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.03.551861. [PMID: 37577554 PMCID: PMC10418232 DOI: 10.1101/2023.08.03.551861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Within the neuronal classes of the retina, amacrine cells (ACs) exhibit the greatest neuronal diversity in morphology and function. We show that the selective expression of the transcription factor Gbx2 is required for cell fate specification and dendritic stratification of an individual AC subtype in the mouse retina. We identify Robo1 and Robo2 as downstream effectors that when deleted, phenocopy the dendritic misprojections seen in Gbx2 mutants. Slit1 and Slit2, the ligands of Robo receptors, are localized to the OFF layers of the inner plexiform layer where we observe the dendritic misprojections in both Gbx2 and Robo1/2 mutants. We show that Robo receptors also are required for the proper dendritic stratification of additional AC subtypes, such as Vglut3+ ACs. These results show both that Gbx2 functions as a terminal selector in a single AC subtype and identify Slit-Robo signaling as a developmental mechanism for ON-OFF pathway segregation in the retina.
Collapse
|
25
|
Hanson-Drury S, Patni AP, Lee DL, Alghadeer A, Zhao YT, Ehnes DD, Vo VN, Kim SY, Jithendra D, Phal A, Edman NI, Schlichthaerle T, Baker D, Young JE, Mathieu J, Ruohola-Baker H. Single Cell RNA Sequencing Reveals Human Tooth Type Identity and Guides In Vitro hiPSC Derived Odontoblast Differentiation (iOB). FRONTIERS IN DENTAL MEDICINE 2023; 4:1209503. [PMID: 38259324 PMCID: PMC10802932 DOI: 10.3389/fdmed.2023.1209503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/29/2023] [Indexed: 01/24/2024] Open
Abstract
Over 90% of the U.S. adult population suffers from tooth structure loss due to caries. Most of the mineralized tooth structure is composed of dentin, a material produced and mineralized by ectomesenchyme derived cells known as odontoblasts. Clinicians, scientists, and the general public share the desire to regenerate this missing tooth structure. To bioengineer missing dentin, increased understanding of human tooth development is required. Here we interrogate at the single cell level the signaling interactions that guide human odontoblast and ameloblast development and which determine incisor or molar tooth germ type identity. During human odontoblast development, computational analysis predicts that early FGF and BMP activation followed by later HH signaling is crucial. Application of this sci-RNA-seq analysis generates a differentiation protocol to produce mature hiPSC derived odontoblasts in vitro (iOB). Further, we elucidate the critical role of FGF signaling in odontoblast maturation and its biomineralization capacity using the de novo designed FGFR1/2c isoform specific minibinder scaffolded as a C6 oligomer that acts as a pathway agonist. We find that FGFR1c is upregulated in functional odontoblasts and specifically plays a crucial role in driving odontoblast maturity. Using computational tools, we show on a molecular level how human molar development is delayed compared to incisors. We reveal that enamel knot development is guided by FGF and WNT in incisors and BMP and ROBO in the molars, and that incisor and molar ameloblast development is guided by FGF, EGF and BMP signaling, with tooth type specific intensity of signaling interactions. Dental ectomesenchyme derived cells are the primary source of signaling ligands responsible for both enamel knot and ameloblast development.
Collapse
Affiliation(s)
- Sesha Hanson-Drury
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Anjali P. Patni
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, India
| | - Deborah L. Lee
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Ammar Alghadeer
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Yan Ting Zhao
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Devon Duron Ehnes
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Vivian N. Vo
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Biology, University of Washington, Seattle, WA, United States
| | - Sydney Y. Kim
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Druthi Jithendra
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Biotechnology, SRM Institute of Science and Technology, Chennai, India
| | - Ashish Phal
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Natasha I. Edman
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Protein Design, University of Washington, Seattle, WA, United States
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, United States
- Medical Scientist Training Program, University of Washington, Seattle, WA, United States
| | - Thomas Schlichthaerle
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Protein Design, University of Washington, Seattle, WA, United States
| | - David Baker
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Protein Design, University of Washington, Seattle, WA, United States
| | - Jessica E. Young
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Julie Mathieu
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Hannele Ruohola-Baker
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Biology, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
26
|
Feng L, Shu HP, Sun LL, Tu YC, Liao QQ, Yao LJ. Role of the SLIT-ROBO signaling pathway in renal pathophysiology and various renal diseases. Front Physiol 2023; 14:1226341. [PMID: 37497439 PMCID: PMC10366692 DOI: 10.3389/fphys.2023.1226341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 06/30/2023] [Indexed: 07/28/2023] Open
Abstract
SLIT ligand and its receptor ROBO were initially recognized for their role in axon guidance in central nervous system development. In recent years, as research has advanced, the role of the SLIT-ROBO signaling pathway has gradually expanded from axonal repulsion to cell migration, tumor development, angiogenesis, and bone metabolism. As a secreted protein, SLIT regulates various pathophysiological processes in the kidney, such as proinflammatory responses and fibrosis progression. Many studies have shown that SLIT-ROBO is extensively involved in various aspects of kidney development and maintenance of structure and function. The SLIT-ROBO signaling pathway also plays an important role in different types of kidney disease. This article reviews the advances in the study of the SLIT-ROBO pathway in various renal pathophysiological and kidney disorders and proposes new directions for further research in this field.
Collapse
|
27
|
Tang L, Liu C, Rosenberger P. Platelet formation and activation are influenced by neuronal guidance proteins. Front Immunol 2023; 14:1206906. [PMID: 37398659 PMCID: PMC10310924 DOI: 10.3389/fimmu.2023.1206906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Platelets are anucleate blood cells derived from megakaryocytes. They link the fundamental functions of hemostasis, inflammation and host defense. They undergo intracellular calcium flux, negatively charged phospholipid translocation, granule release and shape change to adhere to collagen, fibrin and each other, forming aggregates, which are key to several of their functions. In all these dynamic processes, the cytoskeleton plays a crucial role. Neuronal guidance proteins (NGPs) form attractive and repulsive signals to drive neuronal axon navigation and thus refine neuronal circuits. By binding to their target receptors, NGPs rearrange the cytoskeleton to mediate neuron motility. In recent decades, evidence has indicated that NGPs perform important immunomodulatory functions and influence platelet function. In this review, we highlight the roles of NGPs in platelet formation and activation.
Collapse
|
28
|
Carranza A, Howard LJ, Brown HE, Ametepe AS, Evans TA. Slit-independent guidance of longitudinal axons by Drosophila Robo3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.539901. [PMID: 37214810 PMCID: PMC10197545 DOI: 10.1101/2023.05.08.539901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Drosophila Robo3 is a member of the evolutionarily conserved Roundabout (Robo) receptor family and one of three Drosophila Robo paralogs. During embryonic ventral nerve cord development, Robo3 does not participate in canonical Slit-dependent midline repulsion, but instead regulates the formation of longitudinal axon pathways at specific positions along the medial-lateral axis. Longitudinal axon guidance by Robo3 is hypothesized to be Slit dependent, but this has not been directly tested. Here we create a series of Robo3 variants in which the N-terminal Ig1 domain is deleted or modified, in order to characterize the functional importance of Ig1 and Slit binding for Robo3's axon guidance activity. We show that Robo3 requires its Ig1 domain for interaction with Slit and for proper axonal localization in embryonic neurons, but deleting Ig1 from Robo3 only partially disrupts longitudinal pathway formation. Robo3 variants with modified Ig1 domains that cannot bind Slit retain proper localization and fully rescue longitudinal axon guidance. Our results indicate that Robo3 guides longitudinal axons independently of Slit, and that sequences both within and outside of Ig1 contribute to this Slit-independent activity.
Collapse
Affiliation(s)
- Abigail Carranza
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701
- Current Address: Texas A&M University School of Medicine, Bryan, TX 77807
| | - LaFreda J. Howard
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701
- Current Address: NAVA PBC, Washington, DC 20005
| | - Haley E. Brown
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701
- Current Address: Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Timothy A. Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701
| |
Collapse
|
29
|
Basha S, Jin-Smith B, Sun C, Pi L. The SLIT/ROBO Pathway in Liver Fibrosis and Cancer. Biomolecules 2023; 13:785. [PMID: 37238655 PMCID: PMC10216401 DOI: 10.3390/biom13050785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Liver fibrosis is a common outcome of most chronic liver insults/injuries that can develop into an irreversible process of cirrhosis and, eventually, liver cancer. In recent years, there has been significant progress in basic and clinical research on liver cancer, leading to the identification of various signaling pathways involved in tumorigenesis and disease progression. Slit glycoprotein (SLIT)1, SLIT2, and SLIT3 are secreted members of a protein family that accelerate positional interactions between cells and their environment during development. These proteins signal through Roundabout receptor (ROBO) receptors (ROBO1, ROBO2, ROBO3, and ROBO4) to achieve their cellular effects. The SLIT and ROBO signaling pathway acts as a neural targeting factor regulating axon guidance, neuronal migration, and axonal remnants in the nervous system. Recent findings suggest that various tumor cells differ in SLIT/ROBO signaling levels and show varying degrees of expression patterns during tumor angiogenesis, cell invasion, metastasis, and infiltration. Emerging roles of the SLIT and ROBO axon-guidance molecules have been discovered in liver fibrosis and cancer development. Herein, we examined the expression patterns of SLIT and ROBO proteins in normal adult livers and two types of liver cancers: hepatocellular carcinoma and cholangiocarcinoma. This review also summarizes the potential therapeutics of this pathway for anti-fibrosis and anti-cancer drug development.
Collapse
Affiliation(s)
| | | | | | - Liya Pi
- Department of Pathology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
| |
Collapse
|
30
|
Kwak HJ, Medina-Jiménez BI, Park SC, Kim JH, Jeong GH, Jeon MJ, Kim S, Kim JW, Weisblat DA, Cho SJ. Slit-Robo expression in the leech nervous system: insights into eyespot evolution. Cell Biosci 2023; 13:70. [PMID: 37013648 PMCID: PMC10071614 DOI: 10.1186/s13578-023-01019-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/26/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Slit and Robo are evolutionarily conserved ligand and receptor proteins, respectively, but the number of slit and robo gene paralogs varies across recent bilaterian genomes. Previous studies indicate that this ligand-receptor complex is involved in axon guidance. Given the lack of data regarding Slit/Robo in the Lophotrochozoa compared to Ecdysozoa and Deuterostomia, the present study aims to identify and characterize the expression of Slit/Robo orthologs in leech development. RESULTS We identified one slit (Hau-slit), and two robo genes (Hau-robo1 and Hau-robo2), and characterized their expression spatiotemporally during the development of the glossiphoniid leech Helobdella austinensis. Throughout segmentation and organogenesis, Hau-slit and Hau-robo1 are broadly expressed in complex and roughly complementary patterns in the ventral and dorsal midline, nerve ganglia, foregut, visceral mesoderm and/or endoderm of the crop, rectum and reproductive organs. Before yolk exhaustion, Hau-robo1 is also expressed where the pigmented eye spots will later develop, and Hau-slit is expressed in the area between these future eye spots. In contrast, Hau-robo2 expression is extremely limited, appearing first in the developing pigmented eye spots, and later in the three additional pairs of cryptic eye spots in head region that never develop pigment. Comparing the expression of robo orthologs between H. austinensis and another glossiphoniid leech, Alboglossiphonia lata allows to that robo1 and robo2 operate combinatorially to differentially specify pigmented and cryptic eyespots within the glossiphoniid leeches. CONCLUSIONS Our results support a conserved role in neurogenesis, midline formation and eye spot development for Slit/Robo in the Lophotrochozoa, and provide relevant data for evo-devo studies related to nervous system evolution.
Collapse
Affiliation(s)
- Hee-Jin Kwak
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Department of Ecology, Evolution and Behavior, Faculty of Science, Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, 9190401, Jerusalem, Israel
| | - Brenda I Medina-Jiménez
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Department of Earth Sciences, Paleobiology, Geocentrum, Uppsala University, Villavägen 16, 75236, Uppsala, Sweden
| | - Soon Cheol Park
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Hyeuk Kim
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Wildlife Disease Response Team, National Institute of Wildlife Disease Control and Prevention, Incheon, 22689, Republic of Korea
| | - Geon-Hwi Jeong
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Mi-Jeong Jeon
- National Institute of Biological Resources, Environmental Research Complex, Incheon, 22689, Republic of Korea
| | - Sangil Kim
- Museum of Comparative Zoology and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Jung-Woong Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - David A Weisblat
- Department of Molecular and Cell Biology, University of California, 385 Weill Hall, Berkeley, CA, 94720-3200, USA.
| | - Sung-Jin Cho
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
31
|
Cline MM, Juarez B, Hunker A, Regiarto EG, Hariadi B, Soden ME, Zweifel LS. Netrin-1 regulates the balance of synaptic glutamate signaling in the adult ventral tegmental area. eLife 2023; 12:e83760. [PMID: 36927614 PMCID: PMC10023152 DOI: 10.7554/elife.83760] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
The axonal guidance cue netrin-1 serves a critical role in neural circuit development by promoting growth cone motility, axonal branching, and synaptogenesis. Within the adult mouse brain, expression of the gene encoding (Ntn1) is highly enriched in the ventral midbrain where it is expressed in both GABAergic and dopaminergic neurons, but its function in these cell types in the adult system remains largely unknown. To address this, we performed viral-mediated, cell-type specific CRISPR-Cas9 mutagenesis of Ntn1 in the ventral tegmental area (VTA) of adult mice. Ntn1 loss-of-function in either cell type resulted in a significant reduction in excitatory postsynaptic connectivity. In dopamine neurons, the reduced excitatory tone had a minimal phenotypic behavioral outcome; however, reduced glutamatergic tone on VTA GABA neurons induced behaviors associated with a hyperdopaminergic phenotype. Simultaneous loss of Ntn1 function in both cell types largely rescued the phenotype observed in the GABA-only mutagenesis. These findings demonstrate an important role for Ntn1 in maintaining excitatory connectivity in the adult midbrain and that a balance in this connectivity within two of the major cell types of the VTA is critical for the proper functioning of the mesolimbic system.
Collapse
Affiliation(s)
- Marcella M Cline
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Molecular and Cellular Biology Program, University of WashingtonSeattleUnited States
| | - Barbara Juarez
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Avery Hunker
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Ernesto G Regiarto
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Bryan Hariadi
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Marta E Soden
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| |
Collapse
|
32
|
Schwend T. Wiring the ocular surface: A focus on the comparative anatomy and molecular regulation of sensory innervation of the cornea. Differentiation 2023:S0301-4681(23)00010-5. [PMID: 36997455 DOI: 10.1016/j.diff.2023.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
The cornea is richly innervated with sensory nerves that function to detect and clear harmful debris from the surface of the eye, promote growth and survival of the corneal epithelium and hasten wound healing following ocular disease or trauma. Given their importance to eye health, the neuroanatomy of the cornea has for many years been a source of intense investigation. Resultantly, complete nerve architecture maps exist for adult human and many animal models and these maps reveal few major differences across species. Interestingly, recent work has revealed considerable variation across species in how sensory nerves are acquired during developmental innervation of the cornea. Highlighting such species-distinct key differences, but also similarities, this review provides a full, comparative anatomy analysis of sensory innervation of the cornea for all species studied to date. Further, this article comprehensively describes the molecules that have been shown to guide and direct nerves toward, into and through developing corneal tissue as the final architectural pattern of the cornea's neuroanatomy is established. Such knowledge is useful for researchers and clinicians seeking to better understand the anatomical and molecular basis of corneal nerve pathologies and to hasten neuro-regeneration following infection, trauma or surgery that damage the ocular surface and its corneal nerves.
Collapse
|
33
|
Sullivan KG, Bashaw GJ. Intracellular Trafficking Mechanisms that Regulate Repulsive Axon Guidance. Neuroscience 2023; 508:123-136. [PMID: 35863679 PMCID: PMC9839465 DOI: 10.1016/j.neuroscience.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 01/17/2023]
Abstract
Friedrich Bonhoeffer made seminal contributions to the study of axon guidance in the developing nervous system. His discoveries of key cellular and molecular mechanisms that dictate wiring specificity laid the foundation for countless investigators who have followed in his footsteps. Perhaps his most significant contribution was the cloning and characterization of members of the conserved ephrin family of repulsive axon guidance cues. In this review, we highlight the major contributions that Bonhoeffer and his colleagues made to the field of axon guidance, and discuss ongoing investigations into the diverse array of mechanisms that ensure that axon repulsion is precisely regulated to allow for accurate pathfinding. Specifically, we focus our discussion on the post-translational regulation of two major families of repulsive axon guidance factors: ephrin ligands and their Eph receptors, and slit ligands and their Roundabout (Robo) receptors. We will give special emphasis to the ways in which regulated endocytic trafficking events allow navigating axons to adjust their responses to repellant signals and how these trafficking events are intimately related to receptor signaling. By highlighting parallels and differences between the regulation of these two important repulsive axon guidance pathways, we hope to identify key outstanding questions for future investigation.
Collapse
Affiliation(s)
- Kelly G Sullivan
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, United States
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, United States.
| |
Collapse
|
34
|
Leahy SN, Song C, Vita DJ, Broadie K. FMRP activity and control of Csw/SHP2 translation regulate MAPK-dependent synaptic transmission. PLoS Biol 2023; 21:e3001969. [PMID: 36701299 PMCID: PMC9879533 DOI: 10.1371/journal.pbio.3001969] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/16/2022] [Indexed: 01/27/2023] Open
Abstract
Noonan syndrome (NS) and NS with multiple lentigines (NSML) cognitive dysfunction are linked to SH2 domain-containing protein tyrosine phosphatase-2 (SHP2) gain-of-function (GoF) and loss-of-function (LoF), respectively. In Drosophila disease models, we find both SHP2 mutations from human patients and corkscrew (csw) homolog LoF/GoF elevate glutamatergic transmission. Cell-targeted RNAi and neurotransmitter release analyses reveal a presynaptic requirement. Consistently, all mutants exhibit reduced synaptic depression during high-frequency stimulation. Both LoF and GoF mutants also show impaired synaptic plasticity, including reduced facilitation, augmentation, and post-tetanic potentiation. NS/NSML diseases are characterized by elevated MAPK/ERK signaling, and drugs suppressing this signaling restore normal neurotransmission in mutants. Fragile X syndrome (FXS) is likewise characterized by elevated MAPK/ERK signaling. Fragile X Mental Retardation Protein (FMRP) binds csw mRNA and neuronal Csw protein is elevated in Drosophila fragile X mental retardation 1 (dfmr1) nulls. Moreover, phosphorylated ERK (pERK) is increased in dfmr1 and csw null presynaptic boutons. We find presynaptic pERK activation in response to stimulation is reduced in dfmr1 and csw nulls. Trans-heterozygous csw/+; dfmr1/+ recapitulate elevated presynaptic pERK activation and function, showing FMRP and Csw/SHP2 act within the same signaling pathway. Thus, a FMRP and SHP2 MAPK/ERK regulative mechanism controls basal and activity-dependent neurotransmission strength.
Collapse
Affiliation(s)
- Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Chunzhu Song
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Dominic J. Vita
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
35
|
Han P, She Y, Yang Z, Zhuang M, Wang Q, Luo X, Yin C, Zhu J, Jaffrey SR, Ji SJ. Cbln1 regulates axon growth and guidance in multiple neural regions. PLoS Biol 2022; 20:e3001853. [PMID: 36395107 PMCID: PMC9671368 DOI: 10.1371/journal.pbio.3001853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
The accurate construction of neural circuits requires the precise control of axon growth and guidance, which is regulated by multiple growth and guidance cues during early nervous system development. It is generally thought that the growth and guidance cues that control the major steps of axon development have been defined. Here, we describe cerebellin-1 (Cbln1) as a novel cue that controls diverse aspects of axon growth and guidance throughout the central nervous system (CNS) by experiments using mouse and chick embryos. Cbln1 has previously been shown to function in late neural development to influence synapse organization. Here, we find that Cbln1 has an essential role in early neural development. Cbln1 is expressed on the axons and growth cones of developing commissural neurons and functions in an autocrine manner to promote axon growth. Cbln1 is also expressed in intermediate target tissues and functions as an attractive guidance cue. We find that these functions of Cbln1 are mediated by neurexin-2 (Nrxn2), which functions as the Cbln1 receptor for axon growth and guidance. In addition to the developing spinal cord, we further show that Cbln1 functions in diverse parts of the CNS with major roles in cerebellar parallel fiber growth and retinal ganglion cell axon guidance. Despite the prevailing role of Cbln1 as a synaptic organizer, our study discovers a new and unexpected function for Cbln1 as a general axon growth and guidance cue throughout the nervous system.
Collapse
Affiliation(s)
- Peng Han
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yuanchu She
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhuoxuan Yang
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Mengru Zhuang
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Qingjun Wang
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiaopeng Luo
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Chaoqun Yin
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Junda Zhu
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Samie R. Jaffrey
- Department of Pharmacology, Weill Cornell Medicine, Cornell University, New York, New York, United States of America
- * E-mail: (SRJ); (SJJ)
| | - Sheng-Jian Ji
- School of Life Sciences, Department of Neuroscience and Department of Biology, Brain Research Center, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
- * E-mail: (SRJ); (SJJ)
| |
Collapse
|
36
|
Şen S, Erber R. Neuronal Guidance Molecules in Bone Remodeling and Orthodontic Tooth Movement. Int J Mol Sci 2022; 23:ijms231710077. [PMID: 36077474 PMCID: PMC9456342 DOI: 10.3390/ijms231710077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
During orthodontic tooth movement, mechanically induced remodeling occurs in the alveolar bone due to the action of orthodontic forces. The number of factors identified to be involved in mechanically induced bone remodeling is growing steadily. With the uncovering of the functions of neuronal guidance molecules (NGMs) for skeletal development as well as for bone homeostasis, NGMs are now also among the potentially significant factors for the regulation of bone remodeling during orthodontic tooth movement. This narrative review attempts to summarize the functions of NGMs in bone homeostasis and provides insight into the currently sparse literature on the functions of these molecules during orthodontic tooth movement. Presently, four families of NGMs are known: Netrins, Slits, Semaphorins, ephrins and Eph receptors. A search of electronic databases revealed roles in bone homeostasis for representatives from all four NGM families. Functions during orthodontic tooth movement, however, were only identified for Semaphorins, ephrins and Eph receptors. For these, crucial prerequisites for participation in the regulation of orthodontically induced bone remodeling, such as expression in cells of the periodontal ligament and in the alveolar bone, as well as mechanical inducibility, were shown, which suggests that the importance of NGMs in orthodontic tooth movement may be underappreciated to date and further research might be warranted.
Collapse
Affiliation(s)
- Sinan Şen
- Department of Orthodontics, University Medical Center Schleswig-Holstein, Campus Kiel, Christian Albrechts University, 24105 Kiel, Germany
- Correspondence: ; Tel.: +49-431-5002-6301
| | - Ralf Erber
- Department of Orthodontics and Dentofacial Orthopedics, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| |
Collapse
|
37
|
Huang Y, Ma M, Mao X, Pehlivan D, Kanca O, Un-Candan F, Shu L, Akay G, Mitani T, Lu S, Candan S, Wang H, Xiao B, Lupski JR, Bellen HJ. Novel dominant and recessive variants in human ROBO1 cause distinct neurodevelopmental defects through different mechanisms. Hum Mol Genet 2022; 31:2751-2765. [PMID: 35348658 PMCID: PMC9402236 DOI: 10.1093/hmg/ddac070] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/17/2022] [Accepted: 03/20/2022] [Indexed: 07/27/2023] Open
Abstract
The Roundabout (Robo) receptors, located on growth cones of neurons, induce axon repulsion in response to the extracellular ligand Slit. The Robo family of proteins controls midline crossing of commissural neurons during development in flies. Mono- and bi-allelic variants in human ROBO1 (HGNC: 10249) have been associated with incomplete penetrance and variable expressivity for a breath of phenotypes, including neurodevelopmental defects such as strabismus, pituitary defects, intellectual impairment, as well as defects in heart and kidney. Here, we report two novel ROBO1 variants associated with very distinct phenotypes. A homozygous missense p.S1522L variant in three affected siblings with nystagmus; and a monoallelic de novo p.D422G variant in a proband who presented with early-onset epileptic encephalopathy. We modeled these variants in Drosophila and first generated a null allele by inserting a CRIMIC T2A-GAL4 in an intron. Flies that lack robo1 exhibit reduced viability but have very severe midline crossing defects in the central nervous system. The fly wild-type cDNA driven by T2A-Gal4 partially rescues both defects. Overexpression of the human reference ROBO1 with T2A-GAL4 is toxic and reduces viability, whereas the recessive p.S1522L variant is less toxic, suggesting that it is a partial loss-of-function allele. In contrast, the dominant variant in fly robo1 (p.D413G) affects protein localization, impairs axonal guidance activity and induces mild phototransduction defects, suggesting that it is a neomorphic allele. In summary, our studies expand the phenotypic spectrum associated with ROBO1 variant alleles.
Collapse
Affiliation(s)
- Yan Huang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mengqi Ma
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiao Mao
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital, Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Feride Un-Candan
- Department of Neuroloy, Balikesir Ataturk Public Hospital, Balikesir 10100, Turkey
| | - Li Shu
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Gulsen Akay
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sukru Candan
- Department of Medical Genetics, Balikesir Ataturk Public Hospital, Balikesir 10100, Turkey
| | - Hua Wang
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Bo Xiao
- Neurology Department, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital, Houston, TX 77030, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
38
|
Le TM, Nguyen HDT, Lee E, Lee D, Choi YS, Cho J, Park NJY, Han HS, Chong GO. Transcriptomic Immune Profiles Can Represent the Tumor Immune Microenvironment Related to the Tumor Budding Histology in Uterine Cervical Cancer. Genes (Basel) 2022; 13:1405. [PMID: 36011316 PMCID: PMC9407871 DOI: 10.3390/genes13081405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 11/30/2022] Open
Abstract
Tumor budding (TB) histology has become a critical biomarker for several solid cancers. Despite the accumulating evidence for the association of TB histology with poor prognosis, the biological characteristics of TB are little known about in the context related to the tumor immune microenvironment (TIME) in uterine cervical cancer (CC). Therefore, this study aimed to identify the transcriptomic immune profiles related to TB status and further provide robust medical evidence for clinical application. In our study, total RNA was extracted and sequenced from 21 CC tissue specimens. As such, 1494 differentially expressed genes (DEGs) between the high- and low-TB groups were identified by DESeq2. After intersecting the list of DEGs and public immune genes, we selected 106 immune-related DEGs. Then, hub genes were obtained using Least Absolute Shrinkage and Selection Operator regression. Finally, the correlation between the hub genes and immune cell types was analyzed and four candidate genes were identified (one upregulated (FCGR3B) and three downregulated (ROBO2, OPRL1, and NR4A2) genes). These gene expression levels were highly accurate in predicting TB status (area under the curve >80%). Interestingly, FCGR3B is a hub gene of several innate immune pathways; its expression significantly differed in the overall survival analysis (p = 0.0016). In conclusion, FCGR3B, ROBO2, OPRL1, and NR4A2 expression can strongly interfere with TB growth and replace TB to stratify CC patients.
Collapse
Affiliation(s)
- Tan Minh Le
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hong Duc Thi Nguyen
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Eunmi Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Donghyeon Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Ye Seul Choi
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Junghwan Cho
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
| | - Nora Jee-Young Park
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
- Department of Pathology, Kyungpook National University, Chilgok Hospital, Daegu 41404, Korea
| | - Hyung Soo Han
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Gun Oh Chong
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
- Department of Obstetrics and Gynecology, Kyungpook National University, Chilgok Hospital, Daegu 41404, Korea
| |
Collapse
|
39
|
Mahabaleshwar H, Asharani PV, Loo TY, Koh SY, Pitman MR, Kwok S, Ma J, Hu B, Lin F, Li Lok X, Pitson SM, Saunders TE, Carney TJ. Slit‐Robo signalling establishes a Sphingosine‐1‐phosphate gradient to polarise fin mesenchyme. EMBO Rep 2022; 23:e54464. [DOI: 10.15252/embr.202154464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/12/2022] [Accepted: 05/18/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Harsha Mahabaleshwar
- Lee Kong Chian School of Medicine Experimental Medicine Building Nanyang Technological University Singapore City Singapore
| | - PV Asharani
- Institute of Molecular and Cell Biology (IMCB) A*STAR (Agency for Science, Technology and Research) Singapore City Singapore
| | - Tricia Yi Loo
- Mechanobiology Institute National University of Singapore Singapore City Singapore
| | - Shze Yung Koh
- Lee Kong Chian School of Medicine Experimental Medicine Building Nanyang Technological University Singapore City Singapore
| | - Melissa R Pitman
- Centre for Cancer Biology University of South Australia, and SA Pathology North Tce Adelaide SA Australia
- School of Biological Sciences University of Adelaide Adelaide South Australia Australia
| | - Samuel Kwok
- Lee Kong Chian School of Medicine Experimental Medicine Building Nanyang Technological University Singapore City Singapore
| | - Jiajia Ma
- Lee Kong Chian School of Medicine Experimental Medicine Building Nanyang Technological University Singapore City Singapore
| | - Bo Hu
- Department of Anatomy & Cell Biology Carver College of Medicine The University of Iowa Iowa City IA USA
| | - Fang Lin
- Department of Anatomy & Cell Biology Carver College of Medicine The University of Iowa Iowa City IA USA
| | - Xue Li Lok
- Institute of Molecular and Cell Biology (IMCB) A*STAR (Agency for Science, Technology and Research) Singapore City Singapore
| | - Stuart M Pitson
- Centre for Cancer Biology University of South Australia, and SA Pathology North Tce Adelaide SA Australia
| | - Timothy E Saunders
- Institute of Molecular and Cell Biology (IMCB) A*STAR (Agency for Science, Technology and Research) Singapore City Singapore
- Mechanobiology Institute National University of Singapore Singapore City Singapore
- Warwick Medical School University of Warwick Coventry UK
| | - Tom J Carney
- Lee Kong Chian School of Medicine Experimental Medicine Building Nanyang Technological University Singapore City Singapore
- Institute of Molecular and Cell Biology (IMCB) A*STAR (Agency for Science, Technology and Research) Singapore City Singapore
| |
Collapse
|
40
|
González-Ramírez MC, Rojo-Cortés F, Candia N, Garay-Montecinos J, Guzmán-Palma P, Campusano JM, Oliva C. Autocrine/Paracrine Slit–Robo Signaling Controls Optic Lobe Development in Drosophila melanogaster. Front Cell Dev Biol 2022; 10:874362. [PMID: 35982851 PMCID: PMC9380857 DOI: 10.3389/fcell.2022.874362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022] Open
Abstract
Cell segregation mechanisms play essential roles during the development of the central nervous system (CNS) to support its organization into distinct compartments. The Slit protein is a secreted signal, classically considered a paracrine repellent for axonal growth through Robo receptors. However, its function in the compartmentalization of CNS is less explored. In this work, we show that Slit and Robo3 are expressed in the same neuronal population of the Drosophila optic lobe, where they are required for the correct compartmentalization of optic lobe neuropils by the action of an autocrine/paracrine mechanism. We characterize the endocytic route followed by the Slit/Robo3 complex and detected genetic interactions with genes involved in endocytosis and actin dynamics. Thus, we report that the Slit-Robo3 pathway regulates the morphogenesis of the optic lobe through an atypical autocrine/paracrine mechanism in addition to its role in axon guidance, and in association with proteins of the endocytic pathway and small GTPases.
Collapse
|
41
|
Rekler D, Kalcheim C. Completion of neural crest cell production and emigration is regulated by retinoic-acid-dependent inhibition of BMP signaling. eLife 2022; 11:72723. [PMID: 35394423 PMCID: PMC8993216 DOI: 10.7554/elife.72723] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 03/02/2022] [Indexed: 12/21/2022] Open
Abstract
Production and emigration of neural crest cells is a transient process followed by the emergence of the definitive roof plate. The mechanisms regulating the end of neural crest ontogeny are poorly understood. Whereas early crest development is stimulated by mesoderm-derived retinoic acid, we report that the end of the neural crest period is regulated by retinoic acid synthesized in the dorsal neural tube. Inhibition of retinoic acid signaling in the neural tube prevents the normal upregulation of BMP inhibitors in the nascent roof plate and prolongs the period of BMP responsiveness which otherwise ceases close to roof plate establishment. Consequently, neural crest production and emigration are extended well into the roof plate stage. In turn, extending the activity of neural crest-specific genes inhibits the onset of retinoic acid synthesis in roof plate suggesting a mutual repressive interaction between neural crest and roof plate traits. Although several roof plate-specific genes are normally expressed in the absence of retinoic acid signaling, roof plate and crest markers are co-expressed in single cells and this domain also contains dorsal interneurons. Hence, the cellular and molecular architecture of the roof plate is compromised. Collectively, our results demonstrate that neural tube-derived retinoic acid, via inhibition of BMP signaling, is an essential factor responsible for the end of neural crest generation and the proper segregation of dorsal neural lineages. The division between the central nervous system – formed by the brain and spinal cord – and the peripheral nervous system – which consists of the neurons that sense and relay information to and from the body – takes place early during embryonic development. Initially, the nervous system consists of a tube of cells called the neural tube. From the top region of this tube, some cells change their shape, exit the tube and migrate to different places in the developing body. These cells are called the ‘neural crest’, and they form many different structures, including the peripheral nervous system. Neural crest cells keep leaving the neural tube for a period of time, but after that, the neural tube stops producing them. At this point, the region of the neural tube that had been producing neural crest cells becomes the ‘roof plate’ of the central nervous system, a structure that is essential for the development of specific groups of neurons in the brain and spinal cord. In bird embryos, a protein called bone morphogenetic protein (BMP) is essential for neural crest production because it triggers the migration of these cells away from the neural tube. Before the roof plate is formed, the activity of BMP is blocked by proteins known as BMP inhibitors, which stop more cells from leaving the neural tube. Around the time when neural crest formation stops, another molecule called retinoic acid begins to be synthesized in the top region of the neural tube. Rekler and Kalcheim asked whether retinoic acid is involved in the transition from neural crest to roof plate. To test this hypothesis, Rekler and Kalcheim blocked the activity of retinoic acid in the neural tube of quail embryos at the time when they should stop producing neural crest cells. This resulted in embryos in which the neural tube keeps producing neural crest cells after the roof plate has formed. In these embryos, individual cells in the resulting ‘roof plate’ produced both proteins that are normally only found in neural crest cells, and proteins typically exclusive to the roof plate. This suggests that, in the absence of retinoic acid activity, the segregation of neural crest identity from roof plate identity is compromised. Rekler and Kalcheim also found that, in the embryos where retinoic acid activity had been blocked, the cells in the area where the roof plate should be produced virtually no BMP inhibitors, and exhibited extended BMP activity. This allowed neural crest cells to continue forming and migrating away from the neural tube well after the period when they would stop in a normal embryo. These results indicate that retinoic acid stops the production of neural crest cells by repressing BMP activity in the roof plate of the neural tube. Rekler and Kalcheim’s experiments shed light on the mechanisms that allow the central and peripheral nervous systems to become segregated. This could increase our understanding of the origin of several neurodevelopmental disorders, potentially providing insights into their treatment or prevention. Additionally, the process of neural crest production and exit from the neural tube is highly similar to the process of metastasis in many invasive cancers. Thus, by understanding how the production of neural crest cells is terminated, it may be possible to learn how to prevent malignant cancer cells from spreading through the body.
Collapse
Affiliation(s)
- Dina Rekler
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
42
|
Walker SE, Senatore A, Carlone RL, Spencer GE. Context-Dependent Role of miR-124 in Retinoic Acid-Induced Growth Cone Attraction of Regenerating Motorneurons. Cell Mol Neurobiol 2022; 42:847-869. [PMID: 33094464 PMCID: PMC11441188 DOI: 10.1007/s10571-020-00982-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/14/2020] [Indexed: 10/23/2022]
Abstract
During development and regeneration, growth cones at the tips of extending axons navigate through a complex environment to establish accurate connections with appropriate targets. Growth cones can respond rapidly to classical and non-classical guidance cues in their environment, often requiring local protein synthesis. In vertebrate growth cones, local protein synthesis in response to classical cues can require regulation by microRNAs (miRNAs), a class of small, conserved, non-coding RNAs that post-transcriptionally regulate gene expression. However, less is known of how miRNAs mediate growth cone responses to non-classical cues (such as retinoic acid (RA)), specifically in invertebrates. Here, we utilized adult regenerating invertebrate motorneurons to study miRNA regulation of growth cone attraction to RA, shown to require local protein synthesis. In situ hybridization revealed the presence of miR-124 in growth cones of regenerating ciliary motorneurons of the mollusc Lymnaea stagnalis. Changes in the spatiotemporal distribution of miR-124 occurred following application of RA, and dysregulation of miR-124 (with mimic injection), disrupted RA-induced growth cone turning in a time-dependent manner. This behavioural regulation by miR-124 was altered when the neurite was transected, and the growth cone completely separated from the soma. miR-124 did not, however, appear to be involved in growth cone attraction to serotonin, a response independent of local protein synthesis. Finally, we provide evidence that a downstream effector of RhoGTPases, ROCK, is a potential target of miR-124 during RA-induced growth cone responses. These data advance our current understanding of how microRNAs might mediate cue- and context-dependent behaviours during axon guidance.
Collapse
Affiliation(s)
- Sarah E Walker
- Department of Biological Sciences, Brock University, St Catharines, ON, L2S 3A1, Canada
| | - Adriano Senatore
- University of Toronto Mississauga, Mississauga, ON, L2L 1C6, Canada
| | - Robert L Carlone
- Department of Biological Sciences, Brock University, St Catharines, ON, L2S 3A1, Canada
| | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, St Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
43
|
Abstract
PURPOSE Our understanding of thyroid-associated ophthalmopathy (TAO, A.K.A Graves' orbitopathy, thyroid eye disease) has advanced substantially, since one of us (TJS) wrote the 2010 update on TAO, appearing in this journal. METHODS PubMed was searched for relevant articles. RESULTS Recent insights have resulted from important studies conducted by many different laboratory groups around the World. A clearer understanding of autoimmune diseases in general and TAO specifically emerged from the use of improved research methodologies. Several key concepts have matured over the past decade. Among them, those arising from the refinement of mouse models of TAO, early stage investigation into restoring immune tolerance in Graves' disease, and a hard-won acknowledgement that the insulin-like growth factor-I receptor (IGF-IR) might play a critical role in the development of TAO, stand out as important. The therapeutic inhibition of IGF-IR has blossomed into an effective and safe medical treatment. Teprotumumab, a β-arrestin biased agonist monoclonal antibody inhibitor of IGF-IR has been studied in two multicenter, double-masked, placebo-controlled clinical trials demonstrated both effectiveness and a promising safety profile in moderate-to-severe, active TAO. Those studies led to the approval by the US FDA of teprotumumab, currently marketed as Tepezza for TAO. We have also learned far more about the putative role that CD34+ fibrocytes and their derivatives, CD34+ orbital fibroblasts, play in TAO. CONCLUSION The past decade has been filled with substantial scientific advances that should provide the necessary springboard for continually accelerating discovery over the next 10 years and beyond.
Collapse
Affiliation(s)
- E J Neag
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Brehm Tower, 1000 Wall Street, Ann Arbor, MI, 48105, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Michigan State University College of Osteopathic Medicine, East Lansing, MI, USA
| | - T J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Brehm Tower, 1000 Wall Street, Ann Arbor, MI, 48105, USA.
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
44
|
Fernando R, Caldera O, Smith TJ. Therapeutic IGF-I receptor inhibition alters fibrocyte immune phenotype in thyroid-associated ophthalmopathy. Proc Natl Acad Sci U S A 2021; 118:e2114244118. [PMID: 34949642 PMCID: PMC8719891 DOI: 10.1073/pnas.2114244118] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2021] [Indexed: 01/20/2023] Open
Abstract
Thyroid-associated ophthalmopathy (TAO) represents a disfiguring and potentially blinding autoimmune component of Graves' disease. It appears to be driven, at least in part, by autoantibodies targeting the thyrotropin receptor (TSHR)/insulin-like growth factor I receptor (IGF-IR) complex. Actions mediated through either TSHR or IGF-IR are dependent on IGF-IR activity. CD34+ fibrocytes, monocyte lineage cells, reside uniquely in the TAO orbit, where they masquerade as CD34+ orbital fibroblasts. Fibrocytes present antigens to T cells through their display of the major histocompatibility complex class II (MHC II) while providing costimulation through B7 proteins (CD80, CD86, and programmed death-ligand 1 [PD-L1]). Here, we demonstrate that teprotumumab, an anti-IGF-IR inhibitor, attenuates constitutive expression and induction by the thyroid-stimulating hormone of MHC II and these B7 members in CD34+ fibrocytes. These actions are mediated through reduction of respective gene transcriptional activity. Other IGF-IR inhibitors (1H7 and linsitinib) and knocking down IGF-IR gene expression had similar effects. Interrogation of circulating fibrocytes collected from patients with TAO, prior to and following teprotumumab treatment in vivo during a phase 2 clinical trial, demonstrated reductions in cell-surface MHC II and B7 proteins similar to those found following IGF-IR inhibitor treatment in vitro. Teprotumumab therapy reduces levels of interferon-γ and IL-17A expression in circulating CD4+ T cells, effects that may be indirect and mediated through actions of the drug on fibrocytes. Teprotumumab was approved by the US Food and Drug Administration for TAO. Our current findings identify potential mechanisms through which teprotumumab might be eliciting its clinical response systemically in patients with TAO, potentially by restoring immune tolerance.
Collapse
Affiliation(s)
- Roshini Fernando
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Oshadi Caldera
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Terry J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105
| |
Collapse
|
45
|
Landínez-Macías M, Urwyler O. The Fine Art of Writing a Message: RNA Metabolism in the Shaping and Remodeling of the Nervous System. Front Mol Neurosci 2021; 14:755686. [PMID: 34916907 PMCID: PMC8670310 DOI: 10.3389/fnmol.2021.755686] [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: 08/09/2021] [Accepted: 10/18/2021] [Indexed: 01/25/2023] Open
Abstract
Neuronal morphogenesis, integration into circuits, and remodeling of synaptic connections occur in temporally and spatially defined steps. Accordingly, the expression of proteins and specific protein isoforms that contribute to these processes must be controlled quantitatively in time and space. A wide variety of post-transcriptional regulatory mechanisms, which act on pre-mRNA and mRNA molecules contribute to this control. They are thereby critically involved in physiological and pathophysiological nervous system development, function, and maintenance. Here, we review recent findings on how mRNA metabolism contributes to neuronal development, from neural stem cell maintenance to synapse specification, with a particular focus on axon growth, guidance, branching, and synapse formation. We emphasize the role of RNA-binding proteins, and highlight their emerging roles in the poorly understood molecular processes of RNA editing, alternative polyadenylation, and temporal control of splicing, while also discussing alternative splicing, RNA localization, and local translation. We illustrate with the example of the evolutionary conserved Musashi protein family how individual RNA-binding proteins are, on the one hand, acting in different processes of RNA metabolism, and, on the other hand, impacting multiple steps in neuronal development and circuit formation. Finally, we provide links to diseases that have been associated with the malfunction of RNA-binding proteins and disrupted post-transcriptional regulation.
Collapse
Affiliation(s)
- María Landínez-Macías
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Molecular Life Sciences Program, Life Science Zurich Graduate School, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Olivier Urwyler
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Molecular Life Sciences Program, Life Science Zurich Graduate School, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland.,Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
Ahmed G, Shinmyo Y. Multiple Functions of Draxin/Netrin-1 Signaling in the Development of Neural Circuits in the Spinal Cord and the Brain. Front Neuroanat 2021; 15:766911. [PMID: 34899198 PMCID: PMC8655782 DOI: 10.3389/fnana.2021.766911] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/22/2021] [Indexed: 11/30/2022] Open
Abstract
Axon guidance proteins play key roles in the formation of neural circuits during development. We previously identified an axon guidance cue, named draxin, that has no homology with other axon guidance proteins. Draxin is essential for the development of various neural circuits including the spinal cord commissure, corpus callosum, and thalamocortical projections. Draxin has been shown to not only control axon guidance through netrin-1 receptors, deleted in colorectal cancer (Dcc), and neogenin (Neo1) but also modulate netrin-1-mediated axon guidance and fasciculation. In this review, we summarize the multifaceted functions of draxin and netrin-1 signaling in neural circuit formation in the central nervous system. Furthermore, because recent studies suggest that the distributions and functions of axon guidance cues are highly regulated by glycoproteins such as Dystroglycan and Heparan sulfate proteoglycans, we discuss a possible function of glycoproteins in draxin/netrin-1-mediated axon guidance.
Collapse
Affiliation(s)
- Giasuddin Ahmed
- Department of Neuroscience and Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
47
|
Jeong S. Molecular Mechanisms Underlying Motor Axon Guidance in Drosophila. Mol Cells 2021; 44:549-556. [PMID: 34385406 PMCID: PMC8424136 DOI: 10.14348/molcells.2021.0129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 11/30/2022] Open
Abstract
Decoding the molecular mechanisms underlying axon guidance is key to precise understanding of how complex neural circuits form during neural development. Although substantial progress has been made over the last three decades in identifying numerous axon guidance molecules and their functional roles, little is known about how these guidance molecules collaborate to steer growth cones to their correct targets. Recent studies in Drosophila point to the importance of the combinatorial action of guidance molecules, and further show that selective fasciculation and defasciculation at specific choice points serve as a fundamental strategy for motor axon guidance. Here, I discuss how attractive and repulsive guidance cues cooperate to ensure the recognition of specific choice points that are inextricably linked to selective fasciculation and defasciculation, and correct pathfinding decision-making.
Collapse
Affiliation(s)
- Sangyun Jeong
- Division of Life Sciences (Molecular Biology Major), Department of Bioactive Material Sciences, and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
48
|
Geraldo LH, Xu Y, Jacob L, Pibouin-Fragner L, Rao R, Maissa N, Verreault M, Lemaire N, Knosp C, Lesaffre C, Daubon T, Dejaegher J, Solie L, Rudewicz J, Viel T, Tavitian B, De Vleeschouwer S, Sanson M, Bikfalvi A, Idbaih A, Lu QR, Lima FR, Thomas JL, Eichmann A, Mathivet T. SLIT2/ROBO signaling in tumor-associated microglia and macrophages drives glioblastoma immunosuppression and vascular dysmorphia. J Clin Invest 2021; 131:141083. [PMID: 34181595 PMCID: PMC8363292 DOI: 10.1172/jci141083] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/22/2021] [Indexed: 12/27/2022] Open
Abstract
SLIT2 is a secreted polypeptide that guides migration of cells expressing Roundabout 1 and 2 (ROBO1 and ROBO2) receptors. Herein, we investigated SLIT2/ROBO signaling effects in gliomas. In patients with glioblastoma (GBM), SLIT2 expression increased with malignant progression and correlated with poor survival and immunosuppression. Knockdown of SLIT2 in mouse glioma cells and patient-derived GBM xenografts reduced tumor growth and rendered tumors sensitive to immunotherapy. Tumor cell SLIT2 knockdown inhibited macrophage invasion and promoted a cytotoxic gene expression profile, which improved tumor vessel function and enhanced efficacy of chemotherapy and immunotherapy. Mechanistically, SLIT2 promoted microglia/macrophage chemotaxis and tumor-supportive polarization via ROBO1- and ROBO2-mediated PI3K-γ activation. Macrophage Robo1 and Robo2 deletion and systemic SLIT2 trap delivery mimicked SLIT2 knockdown effects on tumor growth and the tumor microenvironment (TME), revealing SLIT2 signaling through macrophage ROBOs as a potentially novel regulator of the GBM microenvironment and immunotherapeutic target for brain tumors.
Collapse
Affiliation(s)
- Luiz H. Geraldo
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Brazil
| | - Yunling Xu
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
| | - Laurent Jacob
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
| | | | - Rohit Rao
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nawal Maissa
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
| | - Maïté Verreault
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - Nolwenn Lemaire
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - Camille Knosp
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
| | - Corinne Lesaffre
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
| | | | - Joost Dejaegher
- Department of Neurosciences and
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Lien Solie
- Department of Neurosciences and
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | | | - Thomas Viel
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
| | - Bertrand Tavitian
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
| | | | - Marc Sanson
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
- Onconeurotek Tumor Bank, Institut du Cerveau et de la Moelle épinière-ICM, Paris, France
| | | | - Ahmed Idbaih
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - Q. Richard Lu
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Flavia R.S. Lima
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Brazil
| | - Jean-Leon Thomas
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
- Department of Neurology
| | - Anne Eichmann
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
- Cardiovascular Research Center, Department of Internal Medicine, and
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thomas Mathivet
- Université de Paris, Paris Cardiovascular Research Center, INSERM, Paris, France
| |
Collapse
|
49
|
Malheiro A, Wieringa P, Moroni L. Peripheral neurovascular link: an overview of interactions and in vitro models. Trends Endocrinol Metab 2021; 32:623-638. [PMID: 34127366 DOI: 10.1016/j.tem.2021.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/23/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022]
Abstract
Nerves and blood vessels (BVs) establish extensive arborized networks to innervate tissues and deliver oxygen/metabolic support. Developmental cues direct the formation of these intricate and often overlapping patterns, which reflect close interactions within the peripheral neurovascular system. Besides the mutual dependence to survive and function, nerves and BVs share several receptors and ligands, as well as principles of differentiation, growth and pathfinding. Neurovascular (NV) interactions are maintained in adult life and are essential for certain regenerative mechanisms, such as wound healing. In pathological situations (e.g., type 2 diabetes mellitus), the NV system can be severely perturbed and become dysfunctional. Unwanted neural growth and vascularization are also associated with the progression of some pathologies, such as cancer and endometriosis. In this review, we describe the fundamental NV interactions in development, highlighting the similarities between both networks and wiring mechanisms. We also describe the NV contribution to regenerative processes and potential pathological dysfunctions. Finally, we provide an overview of current in vitro models used to replicate and investigate the NV ecosystem, addressing present limitations and future perspectives.
Collapse
Affiliation(s)
- Afonso Malheiro
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands
| | - Paul Wieringa
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands.
| |
Collapse
|
50
|
Guzmán-Palma P, Contreras EG, Mora N, Smith M, González-Ramírez MC, Campusano JM, Sierralta J, Hassan BA, Oliva C. Slit/Robo Signaling Regulates Multiple Stages of the Development of the Drosophila Motion Detection System. Front Cell Dev Biol 2021; 9:612645. [PMID: 33968921 PMCID: PMC8097104 DOI: 10.3389/fcell.2021.612645] [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: 09/30/2020] [Accepted: 03/24/2021] [Indexed: 11/29/2022] Open
Abstract
Neurogenesis is achieved through a sequence of steps that include specification and differentiation of progenitors into mature neurons. Frequently, precursors migrate to distinct positions before terminal differentiation. The Slit-Robo pathway, formed by the secreted ligand Slit and its membrane bound receptor Robo, was first discovered as a regulator of axonal growth. However, today, it is accepted that this pathway can regulate different cellular processes even outside the nervous system. Since most of the studies performed in the nervous system have been focused on axonal and dendritic growth, it is less clear how versatile is this signaling pathway in the developing nervous system. Here we describe the participation of the Slit-Robo pathway in the development of motion sensitive neurons of the Drosophila visual system. We show that Slit and Robo receptors are expressed in different stages during the neurogenesis of motion sensitive neurons. Furthermore, we find that Slit and Robo regulate multiple aspects of their development including neuronal precursor migration, cell segregation between neural stem cells and daughter cells and formation of their connectivity pattern. Specifically, loss of function of slit or robo receptors in differentiated motion sensitive neurons impairs dendritic targeting, while knocking down robo receptors in migratory progenitors or neural stem cells leads to structural defects in the adult optic lobe neuropil, caused by migration and cell segregation defects during larval development. Thus, our work reveals the co-option of the Slit-Robo signaling pathway in distinct developmental stages of a neural lineage.
Collapse
Affiliation(s)
- Pablo Guzmán-Palma
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban G Contreras
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Natalia Mora
- Institut du Cerveau-Paris Brain Institute (ICM), Inserm, CNRS, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Macarena Smith
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M Constanza González-Ramírez
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge M Campusano
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jimena Sierralta
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Bassem A Hassan
- Institut du Cerveau-Paris Brain Institute (ICM), Inserm, CNRS, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Carlos Oliva
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|