1
|
Le VH, Orniacki C, Murcia-Belmonte V, Denti L, Schütz D, Stumm R, Ruhrberg C, Erskine L. CXCL12 promotes the crossing of retinal ganglion cell axons at the optic chiasm. Development 2024; 151:dev202446. [PMID: 38095299 PMCID: PMC10820821 DOI: 10.1242/dev.202446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/07/2023] [Indexed: 01/13/2024]
Abstract
Binocular vision requires the segregation of retinal ganglion cell (RGC) axons extending from the retina into the ipsilateral and contralateral optic tracts. RGC axon segregation occurs at the optic chiasm, which forms at the ventral diencephalon midline. Using expression analyses, retinal explants and genetically modified mice, we demonstrate that CXCL12 (SDF1) is required for axon segregation at the optic chiasm. CXCL12 is expressed by the meninges bordering the optic pathway, and CXCR4 by both ipsilaterally and contralaterally projecting RGCs. CXCL12 or ventral diencephalon meninges potently promoted axon outgrowth from both ipsilaterally and contralaterally projecting RGCs. Further, a higher proportion of axons projected ipsilaterally in mice lacking CXCL12 or its receptor CXCR4 compared with wild-type mice as a result of misrouting of presumptive contralaterally specified RGC axons. Although RGCs also expressed the alternative CXCL12 receptor ACKR3, the optic chiasm developed normally in mice lacking ACKR3. Our data support a model whereby meningeal-derived CXCL12 helps drive axon growth from CXCR4-expressing RGCs towards the diencephalon midline, enabling contralateral axon growth. These findings further our understanding of the molecular and cellular mechanisms controlling optic pathway development.
Collapse
Affiliation(s)
- Viet-Hang Le
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| | - Clarisse Orniacki
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| | - Verónica Murcia-Belmonte
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Dagmar Schütz
- Institute for Pharmacology/Toxicology, Jena University Hospital,Drackendorfer Str. 1, D-07747 Jena, Germany
| | - Ralf Stumm
- Institute for Pharmacology/Toxicology, Jena University Hospital,Drackendorfer Str. 1, D-07747 Jena, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Lynda Erskine
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| |
Collapse
|
2
|
Soucy JR, Todd L, Kriukov E, Phay M, Malechka VV, Rivera JD, Reh TA, Baranov P. Controlling donor and newborn neuron migration and maturation in the eye through microenvironment engineering. Proc Natl Acad Sci U S A 2023; 120:e2302089120. [PMID: 37931105 PMCID: PMC10655587 DOI: 10.1073/pnas.2302089120] [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: 02/07/2023] [Accepted: 09/30/2023] [Indexed: 11/08/2023] Open
Abstract
Ongoing cell therapy trials have demonstrated the need for precision control of donor cell behavior within the recipient tissue. We present a methodology to guide stem cell-derived and endogenously regenerated neurons by engineering the microenvironment. Being an "approachable part of the brain," the eye provides a unique opportunity to study neuron fate and function within the central nervous system. Here, we focused on retinal ganglion cells (RGCs)-the neurons in the retina are irreversibly lost in glaucoma and other optic neuropathies but can potentially be replaced through transplantation or reprogramming. One of the significant barriers to successful RGC integration into the existing mature retinal circuitry is cell migration toward their natural position in the retina. Our in silico analysis of the single-cell transcriptome of the developing human retina identified six receptor-ligand candidates, which were tested in functional in vitro assays for their ability to guide human stem cell-derived RGCs. We used our lead molecule, SDF1, to engineer an artificial gradient in the retina, which led to a 2.7-fold increase in donor RGC migration into the ganglion cell layer (GCL) and a 3.3-fold increase in the displacement of newborn RGCs out of the inner nuclear layer. Only donor RGCs that migrated into the GCL were found to express mature RGC markers, indicating the importance of proper structure integration. Together, these results describe an "in silico-in vitro-in vivo" framework for identifying, selecting, and applying soluble ligands to control donor cell function after transplantation.
Collapse
Affiliation(s)
- Jonathan R. Soucy
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Levi Todd
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Emil Kriukov
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Monichan Phay
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Volha V. Malechka
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - John Dayron Rivera
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Thomas A. Reh
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Petr Baranov
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| |
Collapse
|
3
|
Halasy V, Szőcs E, Soós Á, Kovács T, Pecsenye-Fejszák N, Hotta R, Goldstein AM, Nagy N. CXCR4 and CXCL12 signaling regulates the development of extrinsic innervation to the colorectum. Development 2023; 150:dev201289. [PMID: 37039233 PMCID: PMC10263150 DOI: 10.1242/dev.201289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/25/2023] [Indexed: 04/12/2023]
Abstract
The gastrointestinal tract is innervated by an intrinsic neuronal network, known as the enteric nervous system (ENS), and by extrinsic axons arising from peripheral ganglia. The nerve of Remak (NoR) is an avian-specific sacral neural crest-derived ganglionated structure that extends from the cloaca to the proximal midgut and, similar to the pelvic plexus, provides extrinsic innervation to the distal intestine. The molecular mechanisms controlling extrinsic nerve fiber growth into the gut is unknown. In vertebrates, CXCR4, a cell-surface receptor for the CXCL12 chemokine, regulates migration of neural crest cells and axon pathfinding. We have employed chimeric tissue recombinations and organ culture assays to study the role of CXCR4 and CXCL12 molecules in the development of colorectal innervation. CXCR4 is specifically expressed in nerve fibers arising from the NoR and pelvic plexus, while CXCL12 is localized to the hindgut mesenchyme and enteric ganglia. Overexpression of CXCL12 results in significantly enhanced axonal projections to the gut from the NoR, while CXCR4 inhibition disrupts nerve fiber extension, supporting a previously unreported role for CXCR4 and CXCL12 signaling in extrinsic innervation of the colorectum.
Collapse
Affiliation(s)
- Viktória Halasy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Ádám Soós
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Tamás Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Nóra Pecsenye-Fejszák
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Ryo Hotta
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| |
Collapse
|
4
|
Dang P, Barnes DT, Cheng RP, Xu A, Moon YJ, Kodukula SS, Raper JA. Netrins and Netrin Receptors are Essential for Normal Targeting of Sensory Axons in the Zebrafish Olfactory Bulb. Neuroscience 2023; 508:19-29. [PMID: 35940453 PMCID: PMC9839495 DOI: 10.1016/j.neuroscience.2022.08.004] [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: 04/28/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 01/17/2023]
Abstract
Olfactory sensory neurons that express related odorant receptors specifically target large identifiable neuropils called protoglomeruli when they first reach the olfactory bulb in the zebrafish. This crude odorant receptor-related mapping is further refined as odorant receptor-specific glomeruli segregate from protoglomeruli later in development. Netrins are a prominent class of axon guidance molecules whose contribution to olfactory circuit formation is poorly studied. Morpholino knock down experiments have suggested that Netrin/Dcc signaling is involved in normal protoglomerular targeting. Here we extend these findings with more detailed characterization and modeling of netrin expression, and by examining protoglomerular targeting in mutant lines fornetrin1a (ntn1a), netrin1b (ntn1b), and their receptorsunc5b,dcc, andneo1a. We confirm thatntn1a,ntn1b, anddccare required for normal protoglomerular guidance of a subset of olfactory sensory neurons that are labeled with the Tg(or111-7:IRES:Gal4) transgene. We also observe errors in the targeting of these axons inunc5bmutants, but not inneo1a mutants. Our findings are consistent with ntn1a andntn1bacting primarily as attractants for olfactory sensory neurons targeting the central zone protoglomerulus.
Collapse
Affiliation(s)
- Puneet Dang
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Daniel T Barnes
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan P Cheng
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Alison Xu
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Yoon Ji Moon
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Sai Sripad Kodukula
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Jonathan A Raper
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Xu K, Yu L, Wang Z, Lin P, Zhang N, Xing Y, Yang N. Use of gene therapy for optic nerve protection: Current concepts. Front Neurosci 2023; 17:1158030. [PMID: 37090805 PMCID: PMC10117674 DOI: 10.3389/fnins.2023.1158030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
Gene therapy has become an essential treatment for optic nerve injury (ONI) in recent years, and great strides have been made using animal models. ONI, which is characterized by the loss of retinal ganglion cells (RGCs) and axons, can induce abnormalities in the pupil light reflex, visual field defects, and even vision loss. The eye is a natural organ to target with gene therapy because of its high accessibility and certain immune privilege. As such, numerous gene therapy trials are underway for treating eye diseases such as glaucoma. The aim of this review was to cover research progress made in gene therapy for ONI. Specifically, we focus on the potential of gene therapy to prevent the progression of neurodegenerative diseases and protect both RGCs and axons. We cover the basic information of gene therapy, including the classification of gene therapy, especially focusing on genome editing therapy, and then we introduce common editing tools and vector tools such as Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) -Cas9 and adeno-associated virus (AAV). We also summarize the progress made on understanding the roles of brain derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), phosphatase-tensin homolog (PTEN), suppressor of cytokine signal transduction 3 (SOCS3), histone acetyltransferases (HATs), and other important molecules in optic nerve protection. However, gene therapy still has many challenges, such as misalignment and mutations, immunogenicity of AAV, time it takes and economic cost involved, which means that these issues need to be addressed before clinical trials can be considered.
Collapse
Affiliation(s)
- Kexin Xu
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lu Yu
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Department of Ophthalmology, Aier Eye Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhiyi Wang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Pei Lin
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yiqiao Xing
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Department of Ophthalmology, Aier Eye Hospital of Wuhan University, Wuhan, Hubei, China
- *Correspondence: Yiqiao Xing,
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Ning Yang,
| |
Collapse
|
6
|
Zebrafish Slit2 and Slit3 Act Together to Regulate Retinal Axon Crossing at the Midline. J Dev Biol 2022; 10:jdb10040041. [PMID: 36278546 PMCID: PMC9590056 DOI: 10.3390/jdb10040041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Slit-Robo signaling regulates midline crossing of commissural axons in different systems. In zebrafish, all retinofugal axons cross at the optic chiasm to innervate the contralateral tectum. Here, the mutant for the Robo2 receptor presents severe axon guidance defects, which were not completely reproduced in a Slit2 ligand null mutant. Since slit3 is also expressed around this area at the stage of axon crossing, we decided to analyze the possibility that it collaborates with Slit2 in this process. We found that the disruption of slit3 expression by sgRNA-Cas9 injection caused similar, albeit slightly milder, defects than those of the slit2 mutant, while the same treatment in the slit2−/−mz background caused much more severe defects, comparable to those observed in robo2 mutants. Tracking analysis of in vivo time-lapse experiments indicated differential but complementary functions of these secreted factors in the correction of axon turn errors around the optic chiasm. Interestingly, RT-qPCR analysis showed a mild increase in slit2 expression in slit3-deficient embryos, but not the opposite. Our observations support the previously proposed “repulsive channel” model for Slit-Robo action at the optic chiasm, with both Slits acting in different manners, most probably relating to their different spatial expression patterns.
Collapse
|
7
|
Knapp B, Roedig J, Roedig H, Krzysko J, Horn N, Güler BE, Kusuluri DK, Yildirim A, Boldt K, Ueffing M, Liebscher I, Wolfrum U. Affinity Proteomics Identifies Interaction Partners and Defines Novel Insights into the Function of the Adhesion GPCR VLGR1/ADGRV1. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103108. [PMID: 35630584 PMCID: PMC9146371 DOI: 10.3390/molecules27103108] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/20/2022]
Abstract
The very large G-protein-coupled receptor 1 (VLGR1/ADGRV1) is the largest member of the adhesion G-protein-coupled receptor (ADGR) family. Mutations in VLGR1/ADGRV1 cause human Usher syndrome (USH), a form of hereditary deaf-blindness, and have been additionally linked to epilepsy. In the absence of tangible knowledge of the molecular function and signaling of VLGR1, the pathomechanisms underlying the development of these diseases are still unknown. Our study aimed to identify novel, previously unknown protein networks associated with VLGR1 in order to describe new functional cellular modules of this receptor. Using affinity proteomics, we have identified numerous new potential binding partners and ligands of VLGR1. Tandem affinity purification hits were functionally grouped based on their Gene Ontology terms and associated with functional cellular modules indicative of functions of VLGR1 in transcriptional regulation, splicing, cell cycle regulation, ciliogenesis, cell adhesion, neuronal development, and retinal maintenance. In addition, we validated the identified protein interactions and pathways in vitro and in situ. Our data provided new insights into possible functions of VLGR1, related to the development of USH and epilepsy, and also suggest a possible role in the development of other neuronal diseases such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Barbara Knapp
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Jens Roedig
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Heiko Roedig
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Jacek Krzysko
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Nicola Horn
- Core Facility for Medical Bioanalytics, Institute for Ophthalmic Research, University of Tuebingen, 72076 Tuebingen, Germany; (N.H.); (K.B.); (M.U.)
| | - Baran E. Güler
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Deva Krupakar Kusuluri
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Adem Yildirim
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
| | - Karsten Boldt
- Core Facility for Medical Bioanalytics, Institute for Ophthalmic Research, University of Tuebingen, 72076 Tuebingen, Germany; (N.H.); (K.B.); (M.U.)
| | - Marius Ueffing
- Core Facility for Medical Bioanalytics, Institute for Ophthalmic Research, University of Tuebingen, 72076 Tuebingen, Germany; (N.H.); (K.B.); (M.U.)
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany;
| | - Uwe Wolfrum
- Institute of Molecular Physiology (ImP), Molecular Cell Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; (B.K.); (J.R.); (H.R.); (J.K.); (B.E.G.); (D.K.K.); (A.Y.)
- Correspondence:
| |
Collapse
|
8
|
Xie L, Cen LP, Li Y, Gilbert HY, Strelko O, Berlinicke C, Stavarache MA, Ma M, Wang Y, Cui Q, Kaplitt MG, Zack DJ, Benowitz LI, Yin Y. Monocyte-derived SDF1 supports optic nerve regeneration and alters retinal ganglion cells' response to Pten deletion. Proc Natl Acad Sci U S A 2022; 119:e2113751119. [PMID: 35394873 PMCID: PMC9169637 DOI: 10.1073/pnas.2113751119] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/23/2022] [Indexed: 12/22/2022] Open
Abstract
Although mammalian retinal ganglion cells (RGCs) normally cannot regenerate axons nor survive after optic nerve injury, this failure is partially reversed by inducing sterile inflammation in the eye. Infiltrative myeloid cells express the axogenic protein oncomodulin (Ocm) but additional, as-yet-unidentified, factors are also required. We show here that infiltrative macrophages express stromal cell–derived factor 1 (SDF1, CXCL12), which plays a central role in this regard. Among many growth factors tested in culture, only SDF1 enhances Ocm activity, an effect mediated through intracellular cyclic AMP (cAMP) elevation and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) activation. SDF1 deficiency in myeloid cells (CXCL12flx/flxLysM-Cre−/+ mice) or deletion of the SDF1 receptor CXCR4 in RGCs (intraocular AAV2-Cre in CXCR4flx/flx mice) or SDF1 antagonist AMD3100 greatly suppresses inflammation-induced regeneration and decreases RGC survival to baseline levels. Conversely, SDF1 induces optic nerve regeneration and RGC survival, and, when combined with Ocm/cAMP, SDF1 increases axon regeneration to levels similar to those induced by intraocular inflammation. In contrast to deletion of phosphatase and tensin homolog (Pten), which promotes regeneration selectively from αRGCs, SDF1 promotes regeneration from non-αRGCs and enables the latter cells to respond robustly to Pten deletion; however, SDF1 surprisingly diminishes the response of αRGCs to Pten deletion. When combined with inflammation and Pten deletion, SDF1 enables many RGCs to regenerate axons the entire length of the optic nerve. Thus, SDF1 complements the effects of Ocm in mediating inflammation-induced regeneration and enables different RGC subtypes to respond to Pten deletion.
Collapse
Affiliation(s)
- Lili Xie
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Ling-Ping Cen
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Joint Shantou International Eye Center, Shantou University and The Chinese University of Hong Kong, Shantou 515000, China
| | - Yiqing Li
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510085, China
| | - Hui-Ya Gilbert
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Oleksandr Strelko
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Cynthia Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Mihaela A. Stavarache
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065
| | - Madeline Ma
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Yongting Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Cui
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Joint Shantou International Eye Center, Shantou University and The Chinese University of Hong Kong, Shantou 515000, China
| | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Larry I. Benowitz
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115
| | - Yuqin Yin
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| |
Collapse
|
9
|
Cheng RP, Dang P, Taku AA, Moon YJ, Pham V, Sun X, Zhao E, Raper JA. Loss of Neuropilin2a/b or Sema3fa alters olfactory sensory axon dynamics and protoglomerular targeting. Neural Dev 2022; 17:1. [PMID: 34980234 PMCID: PMC8725463 DOI: 10.1186/s13064-021-00157-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/29/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Olfactory Sensory Neuron (OSN) axons project from the zebrafish olfactory epithelium to reproducible intermediate target locations in the olfactory bulb called protoglomeruli at early stages in development. Two classes of OSNs expressing either OMP or TRPC2 exclusively target distinct, complementary protoglomeruli. Using RNAseq, we identified axon guidance receptors nrp2a and nrp2b, and their ligand sema3fa, as potential guidance factors that are differentially expressed between these two classes of OSNs. METHODS To investigate their role in OSN axon guidance, we assessed the protoglomerular targeting fidelity of OSNs labeled by OMP:RFP and TRPC2:Venus transgenes in nrp2a, nrp2b, or sema3fa mutants. We used double mutant and genetic interaction experiments to interrogate the relationship between the three genes. We used live time-lapse imaging to compare the dynamic behaviors of OSN growth cones during protoglomerular targeting in heterozygous and mutant larvae. RESULTS The fidelity of protoglomerular targeting of TRPC2-class OSNs is degraded in nrp2a, nrp2b, or sema3fa mutants, as axons misproject into OMP-specific protoglomeruli and other ectopic locations in the bulb. These misprojections are further enhanced in nrp2a;nrp2b double mutants suggesting that nrp2s work at least partially in parallel in the same guidance process. Results from genetic interaction experiments are consistent with sema3fa acting in the same biological pathway as both nrp2a and nrp2b. Live time-lapse imaging was used to examine the dynamic behavior of TRPC2-class growth cones in nrp2a mutants compared to heterozygous siblings. Some TRPC2-class growth cones ectopically enter the dorsal-medial region of the bulb in both groups, but in fully mutant embryos, they are less likely to correct the error through retraction. The same result was observed when TRPC2-class growth cone behavior was compared between sema3fa heterozygous and sema3fa mutant larvae. CONCLUSIONS Our results suggest that nrp2a and nrp2b expressed in TRPC2-class OSNs help prevent their mixing with axon projections in OMP-specific protoglomeruli, and further, that sema3fa helps to exclude TRPC2-class axons by repulsion from the dorsal-medial bulb.
Collapse
Affiliation(s)
- Ryan P Cheng
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Puneet Dang
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Alemji A Taku
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Yoon Ji Moon
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Vi Pham
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Xiaohe Sun
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Ethan Zhao
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Jonathan A Raper
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
10
|
Nickoloff-Bybel EA, Festa L, Meucci O, Gaskill PJ. Co-receptor signaling in the pathogenesis of neuroHIV. Retrovirology 2021; 18:24. [PMID: 34429135 PMCID: PMC8385912 DOI: 10.1186/s12977-021-00569-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
The HIV co-receptors, CCR5 and CXCR4, are necessary for HIV entry into target cells, interacting with the HIV envelope protein, gp120, to initiate several signaling cascades thought to be important to the entry process. Co-receptor signaling may also promote the development of neuroHIV by contributing to both persistent neuroinflammation and indirect neurotoxicity. But despite the critical importance of CXCR4 and CCR5 signaling to HIV pathogenesis, there is only one therapeutic (the CCR5 inhibitor Maraviroc) that targets these receptors. Moreover, our understanding of co-receptor signaling in the specific context of neuroHIV is relatively poor. Research into co-receptor signaling has largely stalled in the past decade, possibly owing to the complexity of the signaling cascades and functions mediated by these receptors. Examining the many signaling pathways triggered by co-receptor activation has been challenging due to the lack of specific molecular tools targeting many of the proteins involved in these pathways and the wide array of model systems used across these experiments. Studies examining the impact of co-receptor signaling on HIV neuropathogenesis often show activation of multiple overlapping pathways by similar stimuli, leading to contradictory data on the effects of co-receptor activation. To address this, we will broadly review HIV infection and neuropathogenesis, examine different co-receptor mediated signaling pathways and functions, then discuss the HIV mediated signaling and the differences between activation induced by HIV and cognate ligands. We will assess the specific effects of co-receptor activation on neuropathogenesis, focusing on neuroinflammation. We will also explore how the use of substances of abuse, which are highly prevalent in people living with HIV, can exacerbate the neuropathogenic effects of co-receptor signaling. Finally, we will discuss the current state of therapeutics targeting co-receptors, highlighting challenges the field has faced and areas in which research into co-receptor signaling would yield the most therapeutic benefit in the context of HIV infection. This discussion will provide a comprehensive overview of what is known and what remains to be explored in regard to co-receptor signaling and HIV infection, and will emphasize the potential value of HIV co-receptors as a target for future therapeutic development. ![]()
Collapse
Affiliation(s)
- E A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - L Festa
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 S. 40th Street, Philadelphia, PA, 19104, USA
| | - O Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.,Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
11
|
Slit2 is necessary for optic axon organization in the zebrafish ventral midline. Cells Dev 2021; 166:203677. [PMID: 33994352 DOI: 10.1016/j.cdev.2021.203677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Slit-Robo signaling has been implicated in regulating several steps of retinal ganglion cell axon guidance, with a central role assigned to Slit2. We report here the phenotypical characterization of a CRISPR-Cas9-generated zebrafish null mutant for this gene, along with a detailed analysis of its expression pattern by WM-FISH. All evident defects in the optic axons in slit2-/- mutants were detected outside the retina, coincident with the major sites of expression at the ventral forebrain, around the developing optic nerve and anterior to the optic chiasm/proximal tract. Anterograde axon tracing experiments in zygotic and maternal-zygotic mutants, as well as morphants, showed the occurrence of axon sorting defects, which appeared mild at the optic nerve level, but more severe in the optic chiasm and the proximal tract. A remarkable sorting defect was the usual splitting of one of the optic nerves in two branches that surrounded the contralateral nerve at the chiasm. Although all axons eventually crossed the midline, the retinotopic order appeared lost at the proximal optic tract, to eventually correct distally. Time-lapse analysis demonstrated the sporadic occurrence of axon misrouting at the chiasm level, which could be responsible for the sorting errors. Our results support previous evidence of a channeling role for Slit molecules in retinal ganglion cell axons at the optic nerve, in addition to a function in the segregation of axons coming from each nerve and from different retinal regions at the medio-ventral area of the forebrain.
Collapse
|
12
|
Yin Y, De Lima S, Gilbert HY, Hanovice NJ, Peterson SL, Sand RM, Sergeeva EG, Wong KA, Xie L, Benowitz LI. Optic nerve regeneration: A long view. Restor Neurol Neurosci 2020; 37:525-544. [PMID: 31609715 DOI: 10.3233/rnn-190960] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The optic nerve conveys information about the outside world from the retina to multiple subcortical relay centers. Until recently, the optic nerve was widely believed to be incapable of re-growing if injured, with dire consequences for victims of traumatic, ischemic, or neurodegenerative diseases of this pathway. Over the past 10-20 years, research from our lab and others has made considerable progress in defining factors that normally suppress axon regeneration and the ability of retinal ganglion cells, the projection neurons of the retina, to survive after nerve injury. Here we describe research from our lab on the role of inflammation-derived growth factors, suppression of inter-cellular signals among diverse retinal cell types, and combinatorial therapies, along with related studies from other labs, that enable animals with optic nerve injury to regenerate damaged retinal axons back to the brain. These studies raise the possibility that vision might one day be restored to people with optic nerve damage.
Collapse
Affiliation(s)
- Yuqin Yin
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Silmara De Lima
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Hui-Ya Gilbert
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - Nicholas J Hanovice
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Sheri L Peterson
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Rheanna M Sand
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Elena G Sergeeva
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kimberly A Wong
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Lili Xie
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Larry I Benowitz
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Zamproni LN, Teixeira D, Alliegro AA, Maugéri IL, des Rieux A, Porcionatto MA. Decreased viability and neurite length in neural cells treated with chitosan-dextran sulfate nanocomplexes. Neurotoxicology 2020; 76:33-43. [DOI: 10.1016/j.neuro.2019.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 01/25/2023]
|
14
|
Liu ZZ, Guo J, Lu Y, Liu W, Fu X, Yao T, Zhou Y, Xu HA. Sema3E is required for migration of cranial neural crest cells in zebrafish: Implications for the pathogenesis of CHARGE syndrome. Int J Exp Pathol 2019; 100:234-243. [PMID: 31464029 DOI: 10.1111/iep.12331] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 05/12/2019] [Accepted: 05/24/2019] [Indexed: 12/24/2022] Open
Abstract
CHARGE syndrome is a congenital disorder with multiple malformations in the craniofacial structures, and cardiovascular and genital systems, which are mainly affected by neural crest defects caused by loss-of-function mutations within chromodomain helicase DNA-binding protein 7 (CHD7). However, many patients with CHARGE syndrome test negative for CHD7. Semaphorin 3E (sema3E) is a gene reported to be mutated in patients with CHARGE syndrome. However, its role in the pathogenesis of CHARGE syndrome has not been verified experimentally. Here, we report that the knockdown of sema3E results in severe craniofacial malformations, including small eyes, defective cartilage and an abnormal number of otoliths in zebrafish embryos, which resemble the major features of CHARGE syndrome. Further analysis reveals that the migratory cranial neural crest cells are scattered in the region of the hindbrain, and the postmigratory neural crest cells are reduced in the pharyngeal arches upon sema3E knockdown. Notably, immunostaining and time-lapse imaging analyses of a neural crest cell-labelled transgenic fish line, sox10:EGFP, show that the migration of cranial neural crest cells is severely impaired, and many of these cells are misrouted upon sema3E knockdown. Furthermore, the sox10-expressing cranial neural crest cells are scattered in chd7 homozygous mutants, which phenocopied the phenotype in sema3E morphants. Overexpression of sema3E rescues the phenotype of scattered cranial neural crest cells in chd7 homozygotes, indicating that chd7 may control the expression of sema3E to regulate cranial neural crest cell migration. Collectively, our data demonstrate that sema3E is involved in the pathogenesis of CHARGE syndrome by modulating cranial neural crest cell migration.
Collapse
Affiliation(s)
- Zhi-Zhi Liu
- Lab of Neural Development and Diseases, Institute of Life Science, Nanchang University, Nanchang, China.,School of Life Sciences, Nanchang University, Nanchang, China.,Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Jingjing Guo
- The First Clinical Medical College of Nanchang University, Nanchang University, Nanchang, China
| | - Yanli Lu
- Children's Hospital of Jiang Xi, Nanchang, China
| | - Wenfeng Liu
- Lab of Neural Development and Diseases, Institute of Life Science, Nanchang University, Nanchang, China
| | - Xiaofeng Fu
- Lab of Neural Development and Diseases, Institute of Life Science, Nanchang University, Nanchang, China
| | - Tianbing Yao
- Lab of Neural Development and Diseases, Institute of Life Science, Nanchang University, Nanchang, China
| | - Yanjun Zhou
- Lab of Neural Development and Diseases, Institute of Life Science, Nanchang University, Nanchang, China
| | - Hong A Xu
- Lab of Neural Development and Diseases, Institute of Life Science, Nanchang University, Nanchang, China.,School of Life Sciences, Nanchang University, Nanchang, China.,Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| |
Collapse
|
15
|
Alassaf M, Daykin EC, Mathiaparanam J, Wolman MA. Pregnancy-associated plasma protein-aa supports hair cell survival by regulating mitochondrial function. eLife 2019; 8:47061. [PMID: 31205004 PMCID: PMC6594750 DOI: 10.7554/elife.47061] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022] Open
Abstract
To support cell survival, mitochondria must balance energy production with oxidative stress. Inner ear hair cells are particularly vulnerable to oxidative stress; thus require tight mitochondrial regulation. We identified a novel molecular regulator of the hair cells’ mitochondria and survival: Pregnancy-associated plasma protein-aa (Pappaa). Hair cells in zebrafish pappaa mutants exhibit mitochondrial defects, including elevated mitochondrial calcium, transmembrane potential, and reactive oxygen species (ROS) production and reduced antioxidant expression. In pappaa mutants, hair cell death is enhanced by stimulation of mitochondrial calcium or ROS production and suppressed by a mitochondrial ROS scavenger. As a secreted metalloprotease, Pappaa stimulates extracellular insulin-like growth factor 1 (IGF1) bioavailability. We found that the pappaa mutants’ enhanced hair cell loss can be suppressed by stimulation of IGF1 availability and that Pappaa-IGF1 signaling acts post-developmentally to support hair cell survival. These results reveal Pappaa as an extracellular regulator of hair cell survival and essential mitochondrial function.
Collapse
Affiliation(s)
- Mroj Alassaf
- Department of Integrative Biology, University of Wisconsin, Madison, United States.,Neuroscience Training Program, University of Wisconsin, Madison, United States
| | - Emily C Daykin
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| | - Jaffna Mathiaparanam
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| | - Marc A Wolman
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| |
Collapse
|
16
|
Guidance of motor axons: where do we stand? CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Pregnancy-Associated Plasma Protein-aa Regulates Photoreceptor Synaptic Development to Mediate Visually Guided Behavior. J Neurosci 2018; 38:5220-5236. [PMID: 29739870 DOI: 10.1523/jneurosci.0061-18.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/04/2018] [Accepted: 04/30/2018] [Indexed: 02/08/2023] Open
Abstract
To guide behavior, sensory systems detect the onset and offset of stimuli and process these distinct inputs via parallel pathways. In the retina, this strategy is implemented by splitting neural signals for light onset and offset via synapses connecting photoreceptors to ON and OFF bipolar cells, respectively. It remains poorly understood which molecular cues establish the architecture of this synaptic configuration to split light-onset and light-offset signals. A mutant with reduced synapses between photoreceptors and one bipolar cell type, but not the other, could reveal a critical cue. From this approach, we report a novel synaptic role for pregnancy-associated plasma protein aa (pappaa) in promoting the structure and function of cone synapses that transmit light-offset information. Electrophysiological and behavioral analyses indicated pappaa mutant zebrafish have dysfunctional cone-to-OFF bipolar cell synapses and impaired responses to light offset, but intact cone-to-ON bipolar cell synapses and light-onset responses. Ultrastructural analyses of pappaa mutant cones showed a lack of presynaptic domains at synapses with OFF bipolar cells. pappaa is expressed postsynaptically to the cones during retinal synaptogenesis and encodes a secreted metalloprotease known to stimulate insulin-like growth factor 1 (IGF1) signaling. Induction of dominant-negative IGF1 receptor expression during synaptogenesis reduced light-offset responses. Conversely, stimulating IGF1 signaling at this time improved pappaa mutants' light-offset responses and cone presynaptic structures. Together, our results indicate Pappaa-regulated IGF1 signaling as a novel pathway that establishes how cone synapses convey light-offset signals to guide behavior.SIGNIFICANCE STATEMENT Distinct sensory inputs, like stimulus onset and offset, are often split at distinct synapses into parallel circuits for processing. In the retina, photoreceptors and ON and OFF bipolar cells form discrete synapses to split neural signals coding light onset and offset, respectively. The molecular cues that establish this synaptic configuration to specifically convey light onset or offset remain unclear. Our work reveals a novel cue: pregnancy-associated plasma protein aa (pappaa), which regulates photoreceptor synaptic structure and function to specifically transmit light-offset information. Pappaa is a metalloprotease that stimulates local insulin-like growth factor 1 (IGF1) signaling. IGF1 promotes various aspects of synaptic development and function and is broadly expressed, thus requiring local regulators, like Pappaa, to govern its specificity.
Collapse
|
18
|
Liu ZZ, Wang ZL, Choi TI, Huang WT, Wang HT, Han YY, Zhu LY, Kim HT, Choi JH, Lee JS, Kim HG, Zhao J, Chen Y, Lu Z, Tian XL, Pan BX, Li BM, Kim CH, Xu HA. Chd7 Is Critical for Early T-Cell Development and Thymus Organogenesis in Zebrafish. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1043-1058. [PMID: 29353058 DOI: 10.1016/j.ajpath.2017.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
Abstract
Coloboma, heart defect, atresia choanae, retarded growth and development, genital hypoplasia, ear anomalies/deafness (CHARGE) syndrome is a congenital disorder affecting multiple organs and mainly caused by mutations in CHD7, a gene encoding a chromatin-remodeling protein. Immunodeficiency and reduced T cells have been noted in CHARGE syndrome. However, the mechanisms underlying T lymphopenia are largely unexplored. Herein, we observed dramatic decrease of T cells in both chd7knockdown and knockout zebrafish embryos. Unexpectedly, hematopoietic stem and progenitor cells and, particularly, lymphoid progenitor cells were increased peripherally in nonthymic areas in chd7-deficient embryos, unlikely to contribute to the T-cell decrease. Further analysis demonstrated that both the organogenesis and homing function of the thymus were seriously impaired. Chd7 might regulate thymus organogenesis through modulating the development of both neural crest cell-derived mesenchyme and pharyngeal endoderm-derived thymic epithelial cells. The expression of foxn1, a central regulator of thymic epithelium, was remarkably down-regulated in the pharyngeal region in chd7-deficient embryos. Moreover, the T-cell reduction in chd7-deficient embryos was partially rescued by overexpressing foxn1, suggesting that restoring thymic epithelium may be a potential therapeutic strategy for treating immunodeficiency in CHARGE syndrome. Collectively, the results indicated that chd7 was critical for thymic development and T-lymphopenia in CHARGE syndrome may be mainly attributed to the defects of thymic organogenesis. The current finding may benefit the diagnosis and therapy of T lymphopenia and immunodeficiency in CHARGE syndrome.
Collapse
Affiliation(s)
- Zhi-Zhi Liu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Zi-Long Wang
- Institute of Life Science, Nanchang University, Nanchang, China; Queen Mary School, Nanchang University, Nanchang, China
| | - Tae-Ik Choi
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Wen-Ting Huang
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Han-Tsing Wang
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Ying-Ying Han
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Lou-Yin Zhu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Hyun-Taek Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jung-Hwa Choi
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jin-Soo Lee
- National Cancer Center, Goyang, Republic of Korea
| | - Hyung-Goo Kim
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia; Children's Hospital of Jiang Xi, Nanchang, China; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia
| | - Jian Zhao
- Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Yue Chen
- Children's Hospital of Jiang Xi, Nanchang, China
| | - Zhuo Lu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Xiao-Li Tian
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Bing-Xing Pan
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Bao-Ming Li
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea.
| | - Hong A Xu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China.
| |
Collapse
|
19
|
Shao X, Lakhina V, Dang P, Cheng RP, Marcaccio CL, Raper JA. Olfactory sensory axons target specific protoglomeruli in the olfactory bulb of zebrafish. Neural Dev 2017; 12:18. [PMID: 29020985 PMCID: PMC5637265 DOI: 10.1186/s13064-017-0095-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/24/2017] [Indexed: 01/08/2023] Open
Abstract
Background The axons of Olfactory Sensory Neurons (OSNs) project to reproducible target locations within the Olfactory Bulb (OB), converting odorant experience into a spatial map of neural activity. We characterized the initial targeting of OSN axons in the zebrafish, a model system suitable for studying axonal targeting early in development. In this system the initial targets of OSN axons are a small number of distinct, individually identifiable neuropilar regions called protoglomeruli. Previously, Olfactory Marker Protein-expressing and TRPC2-expressing classes of OSNs were shown to project to specific, non-overlapping sets of protoglomeruli, indicating that particular subsets of OSNs project to specific protoglomerular targets. We set out to map the relationship between the classical Odorant Receptor (OR) an OSN chooses to express and the protoglomerulus its axon targets. Methods A panel of BACs were recombineered so that the axons of OSNs choosing to express modified ORs were fluorescently labeled. Axon projections were followed into the olfactory bulb to determine the protoglomeruli in which they terminated. Results RNA-seq demonstrates that OSNs express a surprisingly wide variety of ORs and Trace Amine Associated Receptors (TAARs) very early when sensory axons are arriving in the bulb. Only a single OR is expressed in any given OSN even at these early developmental times. We used a BAC expression technique to map the trajectories of OSNs expressing specific odorant receptors. ORs can be divided into three clades based upon their sequence similarities. OSNs expressing ORs from two of these clades project to the CZ protoglomerulus, while OSNs expressing ORs from the third clade project to the DZ protoglomerulus. In contrast, OSNs expressing a particular TAAR project to multiple protoglomeruli. Neither OR choice nor axonal targeting are related to the position an OSN occupies within the olfactory pit. Conclusions Our results demonstrate that it is not the choice of a particular OR, but of one from a category of ORs, that is related to initial OSN target location within the olfactory bulb. These choices are not related to OSN position within the olfactory epithelium.
Collapse
Affiliation(s)
- Xin Shao
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Vanisha Lakhina
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.,Department of Molecular Biology, Princeton University, Princeton, NJ, 08540, USA
| | - Puneet Dang
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ryan P Cheng
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Christina L Marcaccio
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jonathan A Raper
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA. .,, 105 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
20
|
Roberson S, Halpern ME. Convergence of signaling pathways underlying habenular formation and axonal outgrowth in zebrafish. Development 2017; 144:2652-2662. [PMID: 28619821 DOI: 10.1242/dev.147751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 06/07/2017] [Indexed: 12/20/2022]
Abstract
The habenular nuclei are a conserved integrating center in the vertebrate epithalamus, where they modulate diverse behaviors. Despite their importance, our understanding of habenular development is incomplete. Time-lapse imaging and fate mapping demonstrate that the dorsal habenulae (dHb) of zebrafish are derived from dbx1b-expressing (dbx1b+ ) progenitors, which transition into cxcr4b-expressing neuronal precursors. The precursors give rise to differentiated neurons, the axons of which innervate the midbrain interpeduncular nucleus (IPN). Formation of the dbx1b+ progenitor population relies on the activity of the Shh, Wnt and Fgf signaling pathways. Wnt and Fgf function additively to generate dHb progenitors. Surprisingly, Wnt signaling also negatively regulates fgf8a, confining expression to a discrete dorsal diencephalic domain. Moreover, the Wnt and Fgf pathways have opposing roles in transcriptional regulation of components of the Cxcr4-chemokine signaling pathway. The chemokine pathway, in turn, directs the posterior outgrowth of dHb efferents toward the IPN and, when disrupted, results in ectopic, anteriorly directed axonal projections. The results define a signaling network underlying the generation of dHb neurons and connectivity with their midbrain target.
Collapse
Affiliation(s)
- Sara Roberson
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA.,Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
| | - Marnie E Halpern
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA .,Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
| |
Collapse
|
21
|
Bjorke B, Shoja-Taheri F, Kim M, Robinson GE, Fontelonga T, Kim KT, Song MR, Mastick GS. Contralateral migration of oculomotor neurons is regulated by Slit/Robo signaling. Neural Dev 2016; 11:18. [PMID: 27770832 PMCID: PMC5075191 DOI: 10.1186/s13064-016-0073-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 10/11/2016] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Oculomotor neurons develop initially like typical motor neurons, projecting axons out of the ventral midbrain to their ipsilateral targets, the extraocular muscles. However, in all vertebrates, after the oculomotor nerve (nIII) has reached the extraocular muscle primordia, the cell bodies that innervate the superior rectus migrate to join the contralateral nucleus. This motor neuron migration represents a unique strategy to form a contralateral motor projection. Whether migration is guided by diffusible cues remains unknown. METHODS We examined the role of Slit chemorepellent signals in contralateral oculomotor migration by analyzing mutant mouse embryos. RESULTS We found that the ventral midbrain expresses high levels of both Slit1 and 2, and that oculomotor neurons express the repellent Slit receptors Robo1 and Robo2. Therefore, Slit signals are in a position to influence the migration of oculomotor neurons. In Slit 1/2 or Robo1/2 double mutant embryos, motor neuron cell bodies migrated into the ventral midbrain on E10.5, three days prior to normal migration. These early migrating neurons had leading projections into and across the floor plate. In contrast to the double mutants, embryos which were mutant for single Slit or Robo genes did not have premature migration or outgrowth on E10.5, demonstrating a cooperative requirement of Slit1 and 2, as well as Robo1 and 2. To test how Slit/Robo midline repulsion is modulated, we found that the normal migration did not require the receptors Robo3 and CXCR4, or the chemoattractant, Netrin 1. The signal to initiate contralateral migration is likely autonomous to the midbrain because oculomotor neurons migrate in embryos that lack either nerve outgrowth or extraocular muscles, or in cultured midbrains that lacked peripheral tissue. CONCLUSION Overall, our results demonstrate that a migratory subset of motor neurons respond to floor plate-derived Slit repulsion to properly control the timing of contralateral migration.
Collapse
Affiliation(s)
- Brielle Bjorke
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | | | - Minkyung Kim
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - G Eric Robinson
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | | | - Kyung-Tai Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Mi-Ryoung Song
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Grant S Mastick
- Department of Biology, University of Nevada, Reno, NV, 89557, USA.
| |
Collapse
|
22
|
Abstract
Slits are secreted proteins that bind to Roundabout (Robo) receptors. Slit-Robo signaling is best known for mediating axon repulsion in the developing nervous system. However, in recent years the functional repertoire of Slits and Robo has expanded tremendously and Slit-Robo signaling has been linked to roles in neurogenesis, angiogenesis and cancer progression among other processes. Likewise, our mechanistic understanding of Slit-Robo signaling has progressed enormously. Here, we summarize new insights into Slit-Robo evolutionary and system-dependent diversity, receptor-ligand interactions, signaling crosstalk and receptor activation.
Collapse
Affiliation(s)
- Heike Blockus
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, Paris 75012, France Ecole des Neurosciences de Paris, Paris F-75005, France
| | - Alain Chédotal
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, Paris 75012, France
| |
Collapse
|
23
|
Taku AA, Marcaccio CL, Ye W, Krause GJ, Raper JA. Attractant and repellent cues cooperate in guiding a subset of olfactory sensory axons to a well-defined protoglomerular target. Development 2016; 143:123-32. [PMID: 26732841 DOI: 10.1242/dev.127985] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Olfactory sensory axons target well-defined intermediate targets in the zebrafish olfactory bulb called protoglomeruli well before they form odorant receptor-specific glomeruli. A subset of olfactory sensory neurons are labeled by expression of the or111-7:IRES:GAL4 transgene whose axons terminate in the central zone (CZ) protoglomerulus. Previous work has shown that some of these axons misproject to the more dorsal and anterior dorsal zone (DZ) protoglomerulus in the absence of Netrin 1/Dcc signaling. In search of additional cues that guide these axons to the CZ, we found that Semaphorin 3D (Sema3D) is expressed in the anterior bulb and acts as a repellent that pushes them towards the CZ. Further analysis indicates that Sema3D signaling is mediated through Nrp1a, while Nrp2b also promotes CZ targeting but in a Sema3D-independent manner. nrp1a, nrp2b and dcc transcripts are detected in or111-7 transgene-expressing neurons early in development and both Nrp1a and Dcc act cell-autonomously in sensory neurons to promote accurate targeting to the CZ. dcc and nrp1a double mutants have significantly more DZ misprojections than either single mutant, suggesting that the two signaling systems act independently and in parallel to direct a specific subset of sensory axons to their initial protoglomerular target.
Collapse
Affiliation(s)
- Alemji A Taku
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Christina L Marcaccio
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Wenda Ye
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Gregory J Krause
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Jonathan A Raper
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Bussmann J, Raz E. Chemokine-guided cell migration and motility in zebrafish development. EMBO J 2015; 34:1309-18. [PMID: 25762592 DOI: 10.15252/embj.201490105] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/04/2015] [Indexed: 12/29/2022] Open
Abstract
Chemokines are vertebrate-specific, structurally related proteins that function primarily in controlling cell movements by activating specific 7-transmembrane receptors. Chemokines play critical roles in a large number of biological processes and are also involved in a range of pathological conditions. For these reasons, chemokines are at the focus of studies in developmental biology and of clinically oriented research aimed at controlling cancer, inflammation, and immunological diseases. The small size of the zebrafish embryos, their rapid external development, and optical properties as well as the large number of eggs and the fast expansion in genetic tools available make this model an extremely useful one for studying the function of chemokines and chemokine receptors in an in vivo setting. Here, we review the findings relevant to the role that chemokines play in the context of directed single-cell migration, primarily in neutrophils and germ cells, and compare it to the collective cell migration of the zebrafish lateral line. We present the current knowledge concerning the formation of the chemokine gradient, its interpretation within the cell, and the molecular mechanisms underlying the cellular response to chemokine signals during directed migration.
Collapse
Affiliation(s)
- Jeroen Bussmann
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands Gorlaeus Laboratories, Department of Molecular Cell Biology, Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Erez Raz
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| |
Collapse
|
25
|
Abstract
The visual system is beautifully crafted to transmit information of the external world to visual processing and cognitive centers in the brain. For visual information to be relayed to the brain, a series of axon pathfinding events must take place to ensure that the axons of retinal ganglion cells, the only neuronal cell type in the retina that sends axons out of the retina, find their way out of the eye to connect with targets in the brain. In the past few decades, the power of molecular and genetic tools, including the generation of genetically manipulated mouse lines, have multiplied our knowledge about the molecular mechanisms involved in the sculpting of the visual system. Here, we review major advances in our understanding of the mechanisms controlling the differentiation of RGCs, guidance of their axons from the retina to the primary visual centers, and the refinement processes essential for the establishment of topographic maps and eye-specific axon segregation. Human disorders, such as albinism and achiasmia, that impair RGC axon growth and guidance and, thus, the establishment of a fully functioning visual system will also be discussed.
Collapse
Affiliation(s)
- Lynda Erskine
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Scotland, UK
| | - Eloisa Herrera
- Instituto de Neurosciencias de Alicante, CSIC-UMH, San Juan de Alicante, Spain
| |
Collapse
|
26
|
Abstract
Roundabout receptors (Robo) and their Slit ligands were discovered in the 1990s and found to be key players in axon guidance. Slit was initially described s an extracellular matrix protein that was expressed by midline glia in Drosophila. A few years later, it was shown that, in vertebrates and invertebrates, Slits acted as chemorepellents for axons crossing the midline. Robo proteins were originally discovered in Drosophila in a mutant screen for genes involved in the regulation of midline crossing. This ligand-receptor pair has since been implicated in a variety of other neuronal and non-neuronal processes ranging from cell migration to angiogenesis, tumourigenesis and even organogenesis of tissues such as kidneys, lungs and breasts.
Collapse
|
27
|
Mao S, Huang S. The signaling pathway of stromal cell-derived factor-1 and its role in kidney diseases. J Recept Signal Transduct Res 2013; 34:85-91. [PMID: 24303939 DOI: 10.3109/10799893.2013.865746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The chemokine stromal cell-derived factor-1 (SDF-1) regulates the trafficking of progenitor cell (PGC) during embryonic development, cell chemotaxis, and postnatal homing into injury sites. SDF-1 also regulates cell growth, survival, adhesion and angiogenesis. However, in different tissues/cells, the role of SDF-1 is different, such as that it is increased in most of the tumors and associated with cancer metastasis, whereas it is essential for the development of vasculature. For kidney diseases, its role remains controversial. Signaling pathways might be very important in the pathogenesis of kidney diseases. We performed this review to provide a relatively complete signaling pathway flowchart for SDF-1 to the investigators who were interested in the role of SDF-1 in the pathogenesis of kidney diseases. Here, we reviewed the signal transduction pathway of SDF-1 and its role in the pathogenesis of kidney diseases.
Collapse
Affiliation(s)
- Song Mao
- Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University , Nanjing , China
| | | |
Collapse
|
28
|
Glawe JD, Mijalis EM, Davis WC, Barlow SC, Gungor N, McVie R, Kevil CG. SDF-1-CXCR4 differentially regulates autoimmune diabetogenic T cell adhesion through ROBO1-SLIT2 interactions in mice. Diabetologia 2013; 56:2222-30. [PMID: 23811810 DOI: 10.1007/s00125-013-2978-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/07/2013] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS We had previously reported that stromal cell-derived factor 1 (SDF-1) mediates chemorepulsion of diabetogenic T cell adhesion to islet microvascular endothelium through unknown mechanisms in NOD mice. Here we report that SDF-1-mediated chemorepulsion occurs through slit homologue (SLIT)2-roundabout, axon guidance receptor, homologue 1 (Drosophila) (ROBO1) interactions. METHODS C-X-C receptor (CXCR)4 and ROBO1 protein expression was measured in mouse and human T cells. Parallel plate flow chamber adhesion and detachment studies were performed to examine the molecular importance of ROBO1 and SLIT2 for SDF-1-mediated T cell chemorepulsion. Diabetogenic splenocyte transfer was performed in NOD/LtSz Rag1(-/-) mice to examine the effect of the SDF-1 mimetic CTCE-0214 on adoptive transfer of diabetes. RESULTS CXCR4 and ROBO1 protein expression was elevated in diabetic NOD/ShiLtJ T cells over time and coincided with the onset of hyperglycaemia. CXCR4 and ROBO1 expression was also increased in human type 1 diabetic T cells, with ROBO1 expression maximal at less than 1 year post diagnosis. Cell detachment studies revealed that immunoneutralisation of ROBO1 prevented SDF-1-mediated chemorepulsion of NOD T cell firm adhesion to TNFα-stimulated islet endothelial cells. SDF-1 increased NOD T cell adhesion to recombinant adhesion molecules, a phenomenon that was reversed by recombinant SLIT2. Finally, we found that an SDF-1 peptide mimetic prevented NOD T cell adhesion in vitro and significantly delayed adoptive transfer of autoimmune diabetes in vivo. CONCLUSIONS/INTERPRETATION These data reveal a novel molecular pathway, which regulates diabetogenic T cell recruitment and may be useful in modulating autoimmune diabetes.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Adhesion/physiology
- Cells, Cultured
- Chemokine CXCL12/genetics
- Chemokine CXCL12/metabolism
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Female
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Protein Binding
- Receptors, CXCR/genetics
- Receptors, CXCR/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Roundabout Proteins
Collapse
Affiliation(s)
- John D Glawe
- Department of Pathology, LSU Health-Shreveport, 1501 Kings Highway, Shreveport, LA 71130, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
cAMP-induced expression of neuropilin1 promotes retinal axon crossing in the zebrafish optic chiasm. J Neurosci 2013; 33:11076-88. [PMID: 23825413 DOI: 10.1523/jneurosci.0197-13.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Growing axons navigate a complex environment as they respond to attractive and repellent guidance cues. Axons can modulate their responses to cues through a G-protein-coupled, cAMP-dependent signaling pathway. To examine the role of G-protein signaling in axon guidance in vivo, we used the GAL4/UAS system to drive expression of dominant-negative heterotrimeric G-proteins (DNG) in retinal ganglion cells (RGCs) of embryonic zebrafish. Retinal axons normally cross at the ventral midline and project to the contralateral tectum. Expression of DNGα(S) in RGCs causes retinal axons to misproject to the ipsilateral tectum. These errors resemble misprojections in adcy1, adcy8, nrp1a, sema3D, or sema3E morphant embryos, as well as in sema3D mutant embryos. nrp1a is expressed in RGCs as their axons extend toward and across the midline. sema3D and sema3E are expressed adjacent to the chiasm, suggesting that they facilitate retinal midline crossing. We demonstrate synergistic induction of ipsilateral misprojections between adcy8 knockdown and transgenic DNGα(S) expression, adcy8 and nrp1a morphants, or nrp1a morphants and transgenic DNGα(S) expression. Using qPCR analysis, we show that either transgenic DNGα(S)-expressing embryos or adcy8 morphant embryos have decreased levels of nrp1a and nrp1b mRNA. Ipsilateral misprojections in adcy8 morphants are corrected by the expression of an nrp1a rescue construct expressed in RGCs. These findings are consistent with the idea that elevated cAMP levels promote Neuropilin1a expression in RGCs, increasing the sensitivity of retinal axons to Sema3D, Sema3E, or other neuropilin ligands at the midline, and consequently facilitate retinal axon crossing in the chiasm.
Collapse
|
30
|
Yang XT, Pan DC, Chen ET, Bi YY, Feng DF. Glial cells activation potentially contributes to the upregulation of stromal cell-derived factor-1α after optic nerve crush in rats. Neurochem Res 2013; 38:1996-2008. [PMID: 23832528 DOI: 10.1007/s11064-013-1106-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 06/17/2013] [Accepted: 06/29/2013] [Indexed: 02/04/2023]
Abstract
Stromal cell-derived factor-1α (SDF-1α) plays an important role after injury. However, little is known regarding its temporal and spatial expression patterns or how it interacts with glial cells after optic nerve crush injury. We characterized the temporal and spatial expression pattern of SDF-1α in the retina and optic nerve following optic nerve crush and demonstrated that SDF-1α is localized to the glial cells that are distributed in the retina and optic nerve. CXCR4, the receptor for SDF-1α, is expressed along the ganglion cell layer (GCL). The relative expression levels of Sdf-1α mRNA and SDF-1α protein in the retina and optic nerve 1, 2, 3, 5, 7, 10 and 14 days after injury were determined using real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay, respectively, and the Cxcr4 mRNA expression was determined using real-time PCR. Immunofluorescence and immunohistochemical approaches were used to detect the localization of SDF-1α and CXCR4 after injury. The upregulation of Sdf-1α and Cxcr4 mRNA was detected as early as day one after injury in the retina and day two in the optic nerve, the expression peaks 5-7 days after injury. The expression of Sdf-1α and Cxcr4 mRNA was maintained for at least 14 days after the optic nerve crush injury. Furthermore, SDF-1α-positive zones were distributed locally in the reactive glial cells, which suggested potential autocrine stimulation. CXCR4 was mainly expressed in the GCL, which was also adjacent to the the glial cells. These findings suggest that following optic nerve crush, the levels of endogenous SDF-1α and CXCR4 increase in the retina and optic nerve, where activated glial cells may act as a source of increased SDF-1α protein.
Collapse
Affiliation(s)
- Xi-Tao Yang
- Department of Neurosurgery, Shanghai Third People's Hospital, Shanghai Jiaotong University School of Medicine, 280 Mo-He Road, Shanghai, 201900, China,
| | | | | | | | | |
Collapse
|
31
|
CXCL12/SDF-1 facilitates optic nerve regeneration. Neurobiol Dis 2013; 55:76-86. [PMID: 23578489 DOI: 10.1016/j.nbd.2013.04.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 03/08/2013] [Accepted: 04/01/2013] [Indexed: 01/30/2023] Open
Abstract
Mature retinal ganglion cells (RGCs) do not normally regenerate injured axons, but undergo apoptosis soon after axotomy. Besides the insufficient intrinsic capability of mature neurons to regrow axons inhibitory molecules located in myelin of the central nervous system as well as the glial scar forming at the site of injury strongly limit axon regeneration. Nevertheless, RGCs can be transformed into a regenerative state upon inflammatory stimulation (IS), enabling these neurons to grow axons into the injured optic nerve. The outcome of IS stimulated regeneration is, however, still limited by the inhibitory extracellular environment. Here, we report that the chemokine CXCL12/SDF-1 moderately stimulates neurite growth of mature RGCs on laminin in culture and, in contrast to CNTF, exerts potent disinhibitory effects towards myelin. Consistently, co-treatment of RGCs with CXCL12 facilitated CNTF stimulated neurite growth of RGCs on myelin. Mature RGCs express CXCR4, the cognate CXCL12 receptor. Furthermore, the neurite growth promoting and disinhibitory effects of CXCL12 were abrogated by a specific CXCR4 antagonist and by inhibition of the PI3K/AKT/mTOR-, but not the JAK/STAT3-pathway. In vivo, intravitreal application of CXCL12 sustained mTOR activity in RGCs upon optic nerve injury and moderately stimulated axon regeneration in the optic nerve without affecting the survival of RGCs. Importantly, intravitreal application of CXCL12 also significantly increased IS triggered axon regeneration in vivo. These data suggest that the disinhibitory effect of CXCL12 towards myelin may be a useful feature to facilitate optic nerve regeneration, particularly in combination with other axon growth stimulatory treatments.
Collapse
|
32
|
Chen Z, Lee H, Henle SJ, Cheever TR, Ekker SC, Henley JR. Primary neuron culture for nerve growth and axon guidance studies in zebrafish (Danio rerio). PLoS One 2013; 8:e57539. [PMID: 23469201 PMCID: PMC3587632 DOI: 10.1371/journal.pone.0057539] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/25/2013] [Indexed: 12/21/2022] Open
Abstract
Zebrafish (Danio rerio) is a widely used model organism in genetics and developmental biology research. Genetic screens have proven useful for studying embryonic development of the nervous system in vivo, but in vitro studies utilizing zebrafish have been limited. Here, we introduce a robust zebrafish primary neuron culture system for functional nerve growth and guidance assays. Distinct classes of central nervous system neurons from the spinal cord, hindbrain, forebrain, and retina from wild type zebrafish, and fluorescent motor neurons from transgenic reporter zebrafish lines, were dissociated and plated onto various biological and synthetic substrates to optimize conditions for axon outgrowth. Time-lapse microscopy revealed dynamically moving growth cones at the tips of extending axons. The mean rate of axon extension in vitro was 21.4±1.2 µm hr−1 s.e.m. for spinal cord neurons, which corresponds to the typical ∼0.5 mm day−1 growth rate of nerves in vivo. Fluorescence labeling and confocal microscopy demonstrated that bundled microtubules project along axons to the growth cone central domain, with filamentous actin enriched in the growth cone peripheral domain. Importantly, the growth cone surface membrane expresses receptors for chemotropic factors, as detected by immunofluorescence microscopy. Live-cell functional assays of axon extension and directional guidance demonstrated mammalian brain-derived neurotrophic factor (BDNF)-dependent stimulation of outgrowth and growth cone chemoattraction, whereas mammalian myelin-associated glycoprotein inhibited outgrowth. High-resolution live-cell Ca2+-imaging revealed local elevation of cytoplasmic Ca2+ concentration in the growth cone induced by BDNF application. Moreover, BDNF-induced axon outgrowth, but not basal outgrowth, was blocked by treatments to suppress cytoplasmic Ca2+ signals. Thus, this primary neuron culture model system may be useful for studies of neuronal development, chemotropic axon guidance, and mechanisms underlying inhibition of neural regeneration in vitro, and complement observations made in vivo.
Collapse
Affiliation(s)
- Zheyan Chen
- Mayo Graduate School, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Han Lee
- Mayo Graduate School, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Steven J. Henle
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Thomas R. Cheever
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Stephen C. Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - John R. Henley
- Mayo Graduate School, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
33
|
Schweitzer J, Löhr H, Bonkowsky JL, Hübscher K, Driever W. Sim1a and Arnt2 contribute to hypothalamo-spinal axon guidance by regulating Robo2 activity via a Robo3-dependent mechanism. Development 2013; 140:93-106. [PMID: 23222439 DOI: 10.1242/dev.087825] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Precise spatiotemporal control of axon guidance factor expression is a prerequisite for formation of functional neuronal connections. Although Netrin/Dcc- and Robo/Slit-mediated attractive and repulsive guidance of commissural axons have been extensively studied, little is known about mechanisms controlling mediolateral positioning of longitudinal axons in vertebrates. Here, we use a genetic approach in zebrafish embryos to study pathfinding mechanisms of dopaminergic and neuroendocrine longitudinal axons projecting from the hypothalamus into hindbrain and spinal cord. The transcription factors Sim1a and Arnt2 contribute to differentiation of a defined population of dopaminergic and neuroendocrine neurons. We show that both factors also control aspects of axon guidance: Sim1a or Arnt2 depletion results in displacement of hypothalamo-spinal longitudinal axons towards the midline. This phenotype is suppressed in robo3 guidance receptor mutant embryos. In the absence of Sim1a and Arnt2, expression of the robo3 splice isoform robo3a.1 is increased in the hypothalamus, indicating negative control of robo3a.1 transcription by these factors. We further provide evidence that increased Robo3a.1 levels interfere with Robo2-mediated repulsive axon guidance. Finally, we show that the N-terminal domain unique to Robo3a.1 mediates the block of Robo2 repulsive activity. Therefore, Sim1a and Arnt2 contribute to control of lateral positioning of longitudinal hypothalamic-spinal axons by negative regulation of robo3a.1 expression, which in turn attenuates the repulsive activity of Robo2.
Collapse
Affiliation(s)
- Jörn Schweitzer
- Developmental Biology, Institute Biology 1, Faculty of Biology, University of Freiburg, Hauptstrasse 1, D-79104 Freiburg, Germany.
| | | | | | | | | |
Collapse
|
34
|
Zhu Y, Murakami F. Chemokine CXCL12 and its receptors in the developing central nervous system: emerging themes and future perspectives. Dev Neurobiol 2012; 72:1349-62. [PMID: 22689506 DOI: 10.1002/dneu.22041] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 05/25/2012] [Accepted: 06/01/2012] [Indexed: 01/24/2023]
Abstract
Homeostatic chemokine CXCL12 (also known as SDF-1) and its receptor CXCR4 are indispensable for the normal development of the nervous system. This chemokine system plays a plethora of functions in numerous neural developmental processes, from which the underlying molecular and cellular mechanisms are beginning to be unravelled. Recent identification of CXCR7 as a second receptor for CXCL12 provides opportunities to gain deeper insights into how CXCL12 operates in the nervous system. Here, we review the diverse roles of CXCL12 in the developing central nervous system, summarize the recent progress in uncovering CXCR7 functions, and discuss the emerging common themes from these works and future perspectives.
Collapse
Affiliation(s)
- Yan Zhu
- Graduate School of Frontier Biosciences, Osaka University, Yamadaoka 1-3, Suita, Osaka 565-0871, Japan.
| | | |
Collapse
|
35
|
Geutskens SB, Andrews WD, van Stalborch AMD, Brussen K, Holtrop-de Haan SE, Parnavelas JG, Hordijk PL, van Hennik PB. Control of human hematopoietic stem/progenitor cell migration by the extracellular matrix protein Slit3. J Transl Med 2012; 92:1129-39. [PMID: 22614124 DOI: 10.1038/labinvest.2012.81] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Patients whose hematopoietic system is compromised by chemo- and/or radiotherapy require transplantation of hematopoietic stem and progenitor cells (HSPCs) to restore hematopoiesis. Successful homing of transplanted HSPCs to the bone marrow (BM) largely depends on their migratory potential, which is critically regulated by the chemokine CXCL12. In this study, we have investigated the expression and function of Slit proteins and their corresponding Roundabout (Robo) receptors in human HSPC migration. Slit proteins are extracellular matrix proteins that can modulate the (chemoattractant-induced) migration of mature leukocytes. We show that mRNAs for all Slits (Slit1-3) are expressed in primary BM stroma and BM-derived endothelial and stromal cell lines, but not in CD34⁺ HSPCs. Human CD34⁺ HSPCs expressed mRNAs for all Robos (Robo1-4), but only the Robo1 protein was detected on their cell surface. Functionally, Slit3 treatment increased the in vivo homing efficiency of CD34⁺ HSPCs to the BM in NOD/SCID mice, whereas Slit3-exposed HSPC migration in vitro was inhibited. These effects do not appear to result from modulated CXCL12 responsiveness as CXCR4 expression, CXCL12-induced actin polymerization or the basal and CXCL12-induced adhesion to fibronectin or BM-derived endothelial cells of CD34⁺ HSPC were not altered by Slit3 exposure. However, we show that Slit3 rapidly reduced the levels of active RhoA in HL60 cells and primary CD34⁺ HSPC, directly affecting a pathway involved in actin cytoskeleton remodeling and HSPC migration. Together, our results support a role for Slit3 in human HSPC migration in vitro and homing in vivo and might contribute to the design of future approaches aimed at improving transplantation efficiency of human CD34⁺ HSPCs.
Collapse
Affiliation(s)
- Sacha B Geutskens
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Gensel J, Kigerl K, Mandrekar-Colucci S, Gaudet A, Popovich P. Achieving CNS axon regeneration by manipulating convergent neuro-immune signaling. Cell Tissue Res 2012; 349:201-13. [PMID: 22592625 PMCID: PMC10881271 DOI: 10.1007/s00441-012-1425-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 04/02/2012] [Indexed: 12/20/2022]
Abstract
After central nervous system (CNS) trauma, axons have a low capacity for regeneration. Regeneration failure is associated with a muted regenerative response of the neuron itself, combined with a growth-inhibitory and cytotoxic post-injury environment. After spinal cord injury (SCI), resident and infiltrating immune cells (especially microglia/macrophages) contribute significantly to the growth-refractory milieu near the lesion. By targeting both the regenerative potential of the axon and the cytotoxic phenotype of microglia/macrophages, we may be able to improve CNS repair after SCI. In this review, we discuss molecules shown to impact CNS repair by affecting both immune cells and neurons. Specifically, we provide examples of pattern recognition receptors, integrins, cytokines/chemokines, nuclear receptors and galectins that could improve CNS repair. In many cases, signaling by these molecules is complex and may have contradictory effects on recovery depending on the cell types involved or the model studied. Despite this caveat, deciphering convergent signaling pathways on immune cells (which affect axon growth indirectly) and neurons (direct effects on axon growth) could improve repair and recovery after SCI. Future studies must continue to consider how regenerative therapies targeting neurons impact other cells in the pathological CNS. By identifying molecules that simultaneously improve axon regenerative capacity and drive the protective, growth-promoting phenotype of immune cells, we may discover SCI therapies that act synergistically to improve CNS repair and functional recovery.
Collapse
Affiliation(s)
- J.C. Gensel
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - K.A. Kigerl
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - S. Mandrekar-Colucci
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - A.D. Gaudet
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - P.G. Popovich
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| |
Collapse
|
37
|
Stacher Hörndli C, Chien CB. Sonic hedgehog is indirectly required for intraretinal axon pathfinding by regulating chemokine expression in the optic stalk. Development 2012; 139:2604-13. [PMID: 22696293 DOI: 10.1242/dev.077594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Successful axon pathfinding requires both correct patterning of tissues, which will later harbor axonal tracts, and precise localization of axon guidance cues along these tracts at the time of axon outgrowth. Retinal ganglion cell (RGC) axons grow towards the optic disc in the central retina, where they turn to exit the eye through the optic nerve. Normal patterning of the optic disc and stalk and the expression of guidance cues at this choice point are necessary for the exit of RGC axons out of the eye. Sonic hedgehog (Shh) has been implicated in both patterning of ocular tissue and direct guidance of RGC axons. Here, we examine the precise spatial and temporal requirement for Hedgehog (Hh) signaling for intraretinal axon pathfinding and show that Shh acts to pattern the optic stalk in zebrafish but does not guide RGC axons inside the eye directly. We further reveal an interaction between the Hh and chemokine pathways for axon guidance and show that cxcl12a functions downstream of Shh and depends on Shh for its expression at the optic disc. Together, our results support a model in which Shh acts in RGC axon pathfinding indirectly by regulating axon guidance cues at the optic disc through patterning of the optic stalk.
Collapse
Affiliation(s)
- Cornelia Stacher Hörndli
- Program in Neuroscience, Department of Neurobiology and Anatomy, University of Utah Medical Center, 20 North 1900 East, Salt Lake City, UT 84132, USA.
| | | |
Collapse
|
38
|
Bonner J, Letko M, Nikolaus OB, Krug L, Cooper A, Chadwick B, Conklin P, Lim A, Chien CB, Dorsky RI. Midline crossing is not required for subsequent pathfinding decisions in commissural neurons. Neural Dev 2012; 7:18. [PMID: 22672767 PMCID: PMC3507651 DOI: 10.1186/1749-8104-7-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 05/03/2012] [Indexed: 11/16/2022] Open
Abstract
Background Growth cone navigation across the vertebrate midline is critical in the establishment of nervous system connectivity. While midline crossing is achieved through coordinated signaling of attractive and repulsive cues, this has never been demonstrated at the single cell level. Further, though growth cone responsiveness to guidance cues changes after crossing the midline, it is unclear whether midline crossing itself is required for subsequent guidance decisions in vivo. In the zebrafish, spinal commissures are initially formed by a pioneer neuron called CoPA (Commissural Primary Ascending). Unlike in other vertebrate models, CoPA navigates the midline alone, allowing for single-cell analysis of axon guidance mechanisms. Results We provide evidence that CoPA expresses the known axon guidance receptors dcc, robo3 and robo2. Using loss of function mutants and gene knockdown, we show that the functions of these genes are evolutionarily conserved in teleosts and that they are used consecutively by CoPA neurons. We also reveal novel roles for robo2 and robo3 in maintaining commissure structure. When midline crossing is prevented in robo3 mutants and dcc gene knockdown, ipsilaterally projecting neurons respond to postcrossing guidance cues. Furthermore, DCC inhibits Robo2 function before midline crossing to allow a midline approach and crossing. Conclusions Our results demonstrate that midline crossing is not required for subsequent guidance decisions by pioneer axons and that this is due, in part, to DCC inhibition of Robo2 function prior to midline crossing.
Collapse
Affiliation(s)
- Jennifer Bonner
- Biology Department, Skidmore College, Saratoga Springs, NY 12866, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Netrin/DCC signaling guides olfactory sensory axons to their correct location in the olfactory bulb. J Neurosci 2012; 32:4440-56. [PMID: 22457493 DOI: 10.1523/jneurosci.4442-11.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Olfactory sensory neurons expressing particular olfactory receptors project to specific reproducible locations within the bulb. The axonal guidance cues that organize this precise projection pattern are only beginning to be identified. To aid in their identification and characterization, we generated a transgenic zebrafish line, OR111-7:IRES:Gal4, in which a small subset of olfactory sensory neurons is labeled. Most sensory neurons expressing the OR111-7 transgene project to a specific location within the bulb, the central zone protoglomerulus, while a smaller number project to the lateral glomerulus 1 protoglomerulus. Inhibiting Netrin/DCC (deleted in colorectal cancer) signaling perturbs the ability of OR111-7-expressing axons to enter the olfactory bulb and alters their patterns of termination within the bulb. The Netrin receptor DCC is expressed in olfactory sensory neurons around the time that they elaborate their axons, netrin1a is expressed near the medial-most margin of the olfactory bulb, and netrin1b is expressed within the ventral region of the bulb. Loss of Netrin/DCC signaling components causes some OR111-7-expressing sensory axons to wander posteriorly after exiting the olfactory pit, away from netrin-expressing areas in the bulb. OR111-7-expressing axons that enter the bulb target the central zone less precisely than normal, spreading away from netrin-expressing regions. These pathfinding errors can be corrected by the reexpression of DCC within OR111-7 transgene-expressing neurons in DCC morphant embryos. These findings implicate Netrins as the only known attractants for olfactory sensory neurons, first drawing OR111-7-expressing axons into the bulb and then into the ventromedially positioned central zone protoglomerulus.
Collapse
|
40
|
Mithal DS, Banisadr G, Miller RJ. CXCL12 signaling in the development of the nervous system. J Neuroimmune Pharmacol 2012; 7:820-34. [PMID: 22270883 DOI: 10.1007/s11481-011-9336-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 12/14/2011] [Indexed: 11/30/2022]
Abstract
Chemokines are small, secreted proteins that have been shown to be important regulators of leukocyte trafficking and inflammation. All the known effects of chemokines are transduced by action at a family of G protein coupled receptors. Two of these receptors, CCR5 and CXCR4, are also known to be the major cellular receptors for HIV-1. Consideration of the evolution of the chemokine family has demonstrated that the chemokine Stromal cell Derived Factor-1 or SDF1 (CXCL12) and its receptor CXCR4 are the most ancient members of the family and existed in animals prior to the development of a sophisticated immune system. Thus, it appears that the original function of chemokine signaling was in the regulation of stem cell trafficking and development. CXCR4 signaling is important in the development of many tissues including the nervous system. Here we discuss the manner in which CXCR4 signaling can regulate the development of different structures in the central and peripheral nervous systems and the different strategies employed to achieve these effects.
Collapse
Affiliation(s)
- Divakar S Mithal
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | |
Collapse
|
41
|
Mortazavi MM, Verma K, Deep A, Esfahani FB, Pritchard PR, Tubbs RS, Theodore N. Chemical priming for spinal cord injury: a review of the literature part II-potential therapeutics. Childs Nerv Syst 2011; 27:1307-16. [PMID: 21174102 DOI: 10.1007/s00381-010-1365-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 12/07/2010] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Spinal cord injury is a complex cascade of reactions secondary to the initial mechanical trauma that puts into action the innate properties of the injured cells, the circulatory, inflammatory, and chemical status around them, into a non-permissive and destructive environment for neuronal function and regeneration. Priming means putting a cell, in a state of "arousal" towards better function. Priming can be mechanical as trauma is known to enhance activity in cells. MATERIALS AND METHODS A comprehensive review of the literature was performed to better understand the possible chemical primers used for spinal cord injuries. CONCLUSIONS Taken together, many studies have shown various promising results using the substances outlined herein for treating SCI.
Collapse
Affiliation(s)
- Martin M Mortazavi
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AR, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Mortazavi MM, Verma K, Deep A, Esfahani FB, Pritchard PR, Tubbs RS, Theodore N. Chemical priming for spinal cord injury: a review of the literature. Part I-factors involved. Childs Nerv Syst 2011; 27:1297-306. [PMID: 21170536 DOI: 10.1007/s00381-010-1364-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 12/07/2010] [Indexed: 12/29/2022]
Abstract
INTRODUCTION There are significant differences between the propensity of neural regeneration between the central and peripheral nervous systems. MATERIALS AND METHODS Following a review of the literature, we describe the role of growth factors, guiding factors, and neurite outgrowth inhibitors in the physiology and development of the nervous system as well as the pathophysiology of the spinal cord. We also detail their therapeutic role as well as those of other chemical substances that have recently been found to modify regrowth following cord injury. CONCLUSIONS Multiple factors appear to have promising futures for the possibility of improving spinal cord injury following injury.
Collapse
Affiliation(s)
- Martin M Mortazavi
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AR, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Twery EN, Raper JA. SDF1-induced antagonism of axonal repulsion requires multiple G-protein coupled signaling components that work in parallel. PLoS One 2011; 6:e18896. [PMID: 21556147 PMCID: PMC3083402 DOI: 10.1371/journal.pone.0018896] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 03/24/2011] [Indexed: 11/18/2022] Open
Abstract
SDF1 reduces the responsiveness of axonal growth cones to repellent guidance cues in a pertussis-toxin-sensitive, cAMP-dependent manner. Here, we show that SDF1's antirepellent effect can be blocked in embryonic chick dorsal root ganglia (DRGs) by expression of peptides or proteins inhibiting either Gαi, Gαq, or Gβγ. SDF1 antirepellent activity is also blocked by pharmacological inhibition of PLC, a common effector protein for Gαq. We also show that SDF1 antirepellent activity can be mimicked by overexpression of constitutively active Gαi, Gαq, or Gαs. These results suggest a model in which multiple G protein components cooperate to produce the cAMP levels required for SDF1 antirepellent activity.
Collapse
Affiliation(s)
- E. Naomi Twery
- Neuroscience Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jonathan A. Raper
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
44
|
Regulation of neuronal ferritin heavy chain, a new player in opiate-induced chemokine dysfunction. J Neuroimmune Pharmacol 2011; 6:466-76. [PMID: 21465240 DOI: 10.1007/s11481-011-9278-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 03/21/2011] [Indexed: 12/19/2022]
Abstract
The heavy chain subunit of ferritin (FHC), a ubiquitous protein best known for its iron-sequestering activity as part of the ferritin complex, has recently been described as a novel inhibitor of signaling through the chemokine receptor CXCR4. Levels of FHC as well as its effects on CXCR4 activation increase in cortical neurons exposed to mu-opioid receptor agonists such as morphine, an effect likely specific to neurons. Major actions of CXCR4 signaling in the mature brain include a promotion of neurogenesis, activation of pro-survival signals, and modulation of excitotoxic pathways; thus, FHC up-regulation may contribute to the neuronal dysfunction often associated with opiate drug abuse. This review summarizes our knowledge of neuronal CXCR4 function, its regulation by opiates and the role of FHC in this process, and known mechanisms controlling FHC production. We speculate on the mechanism involved in FHC regulation by opiates and offer FHC as a new target in opioid-induced neuropathology.
Collapse
|
45
|
McIntyre JC, Titlow WB, McClintock TS. Axon growth and guidance genes identify nascent, immature, and mature olfactory sensory neurons. J Neurosci Res 2011; 88:3243-56. [PMID: 20882566 DOI: 10.1002/jnr.22497] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Neurogenesis of projection neurons requires that axons be initiated, extended, and connected. Differences in the expression of axon growth and guidance genes must drive these events, but comprehensively characterizing these differences in a single neuronal type has not been accomplished. Guided by a catalog of gene expression in olfactory sensory neurons (OSNs), in situ hybridization and immunohistochemistry revealed that Cxcr4 and Dbn1, two axon initiation genes, marked the developmental transition from basal progenitor cells to immature OSNs in the olfactory epithelium. The CXCR4 immunoreactivity of these nascent OSNs overlapped partially with markers of proliferation of basal progenitor cells and partially with immunoreactivity for GAP43, the canonical marker of immature OSNs. Intracellular guidance cue signaling transcripts Ablim1, Crmp1, Dypsl2, Dpysl3, Dpysl5, Gap43, Marcskl1, and Stmn1-4 were specific to, or much more abundant in, the immature OSN layer. Receptors that mediate axonal inhibition or repulsion tended to be expressed in both immature and mature OSNs (Plxna1, Plxna4, Nrp2, Efna5) or specifically in mature OSNs (Plxna3, Unc5b, Efna3, Epha5, Epha7), although some were specific to immature OSNs (Plxnb1, Plxnb2, Plxdc2, Nrp1). Cell adhesion molecules were expressed either by both immature and mature OSNs (Dscam, Ncam1, Ncam2, Nrxn1) or solely by immature OSNs (Chl1, Nfasc1, Dscaml1). Given the loss of intracellular signaling protein expression, the continued expression of guidance cue receptors in mature OSNs is consistent with a change in the role of these receptors, perhaps to sending signals back to the cell body and nucleus.
Collapse
Affiliation(s)
- Jeremy C McIntyre
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536-0298, USA
| | | | | |
Collapse
|
46
|
Lerner O, Davenport D, Patel P, Psatha M, Lieberam I, Guthrie S. Stromal cell-derived factor-1 and hepatocyte growth factor guide axon projections to the extraocular muscles. Dev Neurobiol 2010; 70:549-64. [PMID: 20506246 DOI: 10.1002/dneu.20796] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Vertebrate eye movements depend on the co-ordinated function of six extraocular muscles that are innervated by the oculomotor, trochlear, and abducens nerves. Here, we show that the diffusible factors, stromal cell-derived factor-1 (SDF-1) and hepatocyte growth factor (HGF), guide the development of these axon projections. SDF-1 is expressed in the mesenchyme around the oculomotor nerve exit point, and oculomotor axons fail to exit the neuroepithelium in mice mutant for the SDF-1 receptor CXCR4. Both SDF-1 and HGF are expressed in or around the ventral and dorsal oblique muscles, which are distal targets for the oculomotor and trochlear nerves, respectively, as well as in the muscles which are later targets for oculomotor axon branches. We find that in vitro SDF-1 and HGF promote the growth of oculomotor and trochlear axons, whereas SDF-1 also chemoattracts oculomotor axons. Oculomotor neurons show increased branching in the presence of SDF-1 and HGF singly or together. HGF promotes the growth of trochlear axons more than that of oculomotor axons. Taken together, these data point to a role for both SDF-1 and HGF in extraocular nerve projections and indicate that SDF-1 functions specifically in the development of the oculomotor nerve, including oculomotor axon branch formation to secondary muscle targets. HGF shows some specificity in preferentially enhancing development of the trochlear nerve.
Collapse
Affiliation(s)
- Oleg Lerner
- MRC Centre for Developmental Neurobiology, 4th Floor New Hunt's House, King's College, Guy's Campus, London SE1 1UL, United Kingdom
| | | | | | | | | | | |
Collapse
|
47
|
Hall A, Lalli G. Rho and Ras GTPases in axon growth, guidance, and branching. Cold Spring Harb Perspect Biol 2010; 2:a001818. [PMID: 20182621 DOI: 10.1101/cshperspect.a001818] [Citation(s) in RCA: 318] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The establishment of precise neuronal cell morphology provides the foundation for all aspects of neurobiology. During development, axons emerge from cell bodies after an initial polarization stage, elongate, and navigate towards target regions guided by a range of environmental cues. The Rho and Ras families of small GTPases have emerged as critical players at all stages of axonogenesis. Their ability to coordinately direct multiple signal transduction pathways with precise spatial control drives many of the activities that underlie this morphogenetic program: the dynamic assembly, disassembly, and reorganization of the actin and microtubule cytoskeletons, the interaction of the growing axon with other cells and extracellular matrix, the delivery of lipids and proteins to the axon through the exocytic machinery, and the internalization of membrane and proteins at the leading edge of the growth cone through endocytosis. This article highlights the contribution of Rho and Ras GTPases to axonogenesis.
Collapse
Affiliation(s)
- Alan Hall
- Memorial Sloan-Kettering Cancer Center, Cell Biology Program, New York, New York 10065, USA
| | | |
Collapse
|
48
|
Abstract
Motor neurons are functionally related, but represent a diverse collection of cells that show strict preferences for specific axon pathways during embryonic development. In this article, we describe the ligands and receptors that guide motor axons as they extend toward their peripheral muscle targets. Motor neurons share similar guidance molecules with many other neuronal types, thus one challenge in the field of axon guidance has been to understand how the vast complexity of brain connections can be established with a relatively small number of factors. In the context of motor guidance, we highlight some of the temporal and spatial mechanisms used to optimize the fidelity of pathfinding and increase the functional diversity of the signaling proteins.
Collapse
Affiliation(s)
- Dario Bonanomi
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | |
Collapse
|
49
|
Huang M, Li Y, Zhang H, Nan F. Breast cancer stromal fibroblasts promote the generation of CD44+CD24- cells through SDF-1/CXCR4 interaction. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:80. [PMID: 20569497 PMCID: PMC2911413 DOI: 10.1186/1756-9966-29-80] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2010] [Accepted: 06/22/2010] [Indexed: 12/21/2022]
Abstract
Background Breast cancer stem cells (BCSCs) have been recently identified in breast carcinoma as CD44+CD24- cells, which exclusively retain tumorigenic activity and display stem cell-like properties. Using a mammosphere culture technique, MCF7 mammosphere cells are found to enrich breast cancer stem-like cells expressing CD44+CD24-. The stromal cells are mainly constituted by fibroblasts within a breast carcinoma, yet little is known of the contributions of the stromal cells to BCSCs. Methods Carcinoma-associated fibroblasts (CAFs) and normal fibroblasts (NFs) were isolated and identified by immunohistochemistry. MCF7 mammosphere cells were co-cultured with different stromal fibroblasts by a transwell cocultured system. Flow cytometry was used to measure CD44 and CD24 expression status on MCF7. ELISA (enzyme-linked immunosorbent assay) was performed to investigate the production of stromal cell-derived factor 1 (SDF-1) in mammosphere cultures subject to various treatments. Mammosphere cells were injected with CAFs and NFs to examine the efficiency of tumorigenity in NOD/SCID mice. Results CAFs derived from breast cancer patients were found to be positive for α-smooth muscle actin (α-SMA), exhibiting the traits of myofibroblasts. In addition, CAFs played a central role in promoting the proliferation of CD44+CD24- cells through their ability to secrete SDF-1, which may be mediated to SDF-1/CXCR4 signaling. Moreover, the tumorigenicity of mammosphere cells with CAFs significantly increased as compared to that of mammosphere cells alone or with NFs. Conclusion We for the first time investigated the effects of stromal fibroblasts on CD44+CD24- cells and our findings indicated that breast CAFs contribute to CD44+CD24- cell proliferation through the secretion of SDF-1, and which may be important target for therapeutic approaches.
Collapse
Affiliation(s)
- Mingzhu Huang
- Department of Oncology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | | | | | | |
Collapse
|
50
|
The calmodulin-stimulated adenylate cyclase ADCY8 sets the sensitivity of zebrafish retinal axons to midline repellents and is required for normal midline crossing. J Neurosci 2010; 30:7423-33. [PMID: 20505109 DOI: 10.1523/jneurosci.0699-10.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The chemokine SDF1 activates a cAMP-mediated signaling pathway that antagonizes retinal responses to the midline repellent slit. We show that knocking down the calmodulin-activated adenylate cyclase ADCY8 makes retinal axons insensitive to SDF1. Experiments in vivo using male and female zebrafish (Danio rerio) confirm a mutual antagonism between slit signaling and ADCY8-mediated signaling. Unexpectedly, knockdown of ADCY8 or another calmodulin-activated cyclase, ADCY1, induces ipsilateral misprojections of retinal axons that would normally cross the ventral midline. We demonstrate a cell-autonomous requirement for ADCY8 in retinal neurons for normal midline crossing. These findings are the first to show that ADCY8 is required for axonal pathfinding before axons reach their targets. They support a model in which ADCY8 is an essential component of a signaling pathway that opposes repellent signaling. Finally, they demonstrate that ADCY8 helps regulate retinal sensitivity to midline guidance cues.
Collapse
|