1
|
c-Abl Tyrosine Kinase Is Required for BDNF-Induced Dendritic Branching and Growth. Int J Mol Sci 2023; 24:ijms24031944. [PMID: 36768268 PMCID: PMC9916151 DOI: 10.3390/ijms24031944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) induces activation of the TrkB receptor and several downstream pathways (MAPK, PI3K, PLC-γ), leading to neuronal survival, growth, and plasticity. It has been well established that TrkB signaling regulation is required for neurite formation and dendritic arborization, but the specific mechanism is not fully understood. The non-receptor tyrosine kinase c-Abl is a possible candidate regulator of this process, as it has been implicated in tyrosine kinase receptors' signaling and trafficking, as well as regulation of neuronal morphogenesis. To assess the role of c-Abl in BDNF-induced dendritic arborization, wild-type and c-Abl-KO neurons were stimulated with BDNF, and diverse strategies were employed to probe the function of c-Abl, including the use of pharmacological inhibitors, an allosteric c-Abl activator, and shRNA to downregulates c-Abl expression. Surprisingly, BDNF promoted c-Abl activation and interaction with TrkB receptors. Furthermore, pharmacological c-Abl inhibition and genetic ablation abolished BDNF-induced dendritic arborization and increased the availability of TrkB in the cell membrane. Interestingly, inhibition or genetic ablation of c-Abl had no effect on the classic TrkB downstream pathways. Together, our results suggest that BDNF/TrkB-dependent c-Abl activation is a novel and essential mechanism in TrkB signaling.
Collapse
|
2
|
A CRISPR-Cas9-engineered mouse model for GPI-anchor deficiency mirrors human phenotypes and exhibits hippocampal synaptic dysfunctions. Proc Natl Acad Sci U S A 2021; 118:2014481118. [PMID: 33402532 PMCID: PMC7812744 DOI: 10.1073/pnas.2014481118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inherited GPI-anchor biosynthesis deficiencies (IGDs) explain many cases of syndromic intellectual disability. Although diagnostic methods are improving, the pathophysiology underlying the disease remains unclear. Furthermore, we lack rodent models suitable for characterizing cognitive and social disabilities. To address this issue, we generated a viable mouse model for an IGD that mirrors the condition in human patients with a behavioral phenotype and susceptibility to epilepsy. Using this model, we obtained neurological insights such as deficits in synaptic transmission that will facilitate understanding of the pathophysiology of IGDs. Pathogenic germline mutations in PIGV lead to glycosylphosphatidylinositol biosynthesis deficiency (GPIBD). Individuals with pathogenic biallelic mutations in genes of the glycosylphosphatidylinositol (GPI)-anchor pathway exhibit cognitive impairments, motor delay, and often epilepsy. Thus far, the pathophysiology underlying the disease remains unclear, and suitable rodent models that mirror all symptoms observed in human patients have not been available. Therefore, we used CRISPR-Cas9 to introduce the most prevalent hypomorphic missense mutation in European patients, Pigv:c.1022C > A (p.A341E), at a site that is conserved in mice. Mirroring the human pathology, mutant Pigv341E mice exhibited deficits in motor coordination, cognitive impairments, and alterations in sociability and sleep patterns, as well as increased seizure susceptibility. Furthermore, immunohistochemistry revealed reduced synaptophysin immunoreactivity in Pigv341E mice, and electrophysiology recordings showed decreased hippocampal synaptic transmission that could underlie impaired memory formation. In single-cell RNA sequencing, Pigv341E-hippocampal cells exhibited changes in gene expression, most prominently in a subtype of microglia and subicular neurons. A significant reduction in Abl1 transcript levels in several cell clusters suggested a link to the signaling pathway of GPI-anchored ephrins. We also observed elevated levels of Hdc transcripts, which might affect histamine metabolism with consequences for circadian rhythm. This mouse model will not only open the doors to further investigation into the pathophysiology of GPIBD, but will also deepen our understanding of the role of GPI-anchor–related pathways in brain development.
Collapse
|
3
|
Protein Tyrosine Phosphatase Receptor Type J (PTPRJ) Regulates Retinal Axonal Projections by Inhibiting Eph and Abl Kinases in Mice. J Neurosci 2018; 38:8345-8363. [PMID: 30082414 DOI: 10.1523/jneurosci.0128-18.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/05/2018] [Accepted: 07/30/2018] [Indexed: 12/28/2022] Open
Abstract
Eph receptors play pivotal roles in the axon guidance of retinal ganglion cells (RGCs) at the optic chiasm and the establishment of the topographic retinocollicular map. We previously demonstrated that protein tyrosine phosphatase receptor type O (PTPRO) is specifically involved in the control of retinotectal projections in chicks through the dephosphorylation of EphA and EphB receptors. We subsequently revealed that all the mouse R3 subfamily members (PTPRB, PTPRH, PTPRJ, and PTPRO) of the receptor protein tyrosine phosphatase (RPTP) family inhibited Eph receptors as their substrates in cultured mammalian cells. We herein investigated the functional roles of R3 RPTPs in the projection of mouse retinal axon of both sexes. Ptpro and Ptprj were expressed in mouse RGCs; however, Ptprj expression levels were markedly higher than those of Ptpro Consistent with their expression levels, Eph receptor activity was significantly enhanced in Ptprj-knock-out (Ptprj-KO) retinas. In Ptprj-KO and Ptprj/Ptpro-double-KO (DKO) mice, the number of retinal axons that projected ipsilaterally or to the contralateral eye was significantly increased. Furthermore, retinal axons in Ptprj-KO and DKO mice formed anteriorly shifted ectopic terminal zones in the superior colliculus (SC). We found that c-Abl (Abelson tyrosine kinase) was downstream of ephrin-Eph signaling for the repulsion of retinal axons at the optic chiasm and in the SC. c-Abl was identified as a novel substrate for PTPRJ and PTPRO, and the phosphorylation of c-Abl was upregulated in Ptprj-KO and DKO retinas. Thus, PTPRJ regulates retinocollicular projections in mice by controlling the activity of Eph and c-Abl kinases.SIGNIFICANCE STATEMENT Correct retinocollicular projection is a prerequisite for proper vision. Eph receptors have been implicated in retinal axon guidance at the optic chiasm and the establishment of the topographic retinocollicular map. We herein demonstrated that protein tyrosine phosphatase receptor type J (PTPRJ) regulated retinal axonal projections by controlling Eph activities. The retinas of Ptprj-knock-out (KO) and Ptpro/Ptprj double-KO mice exhibited significantly enhanced Eph activities over those in wild-type mice, and their axons showed defects in pathfinding at the chiasm and retinocollicular topographic map formation. We also revealed that c-Abl (Abelson tyrosine kinase) downstream of Eph receptors was regulated by PTPRJ. These results indicate that the regulation of the ephrin-Eph-c-Abl axis by PTPRJ plays pivotal roles in the proper central projection of retinal axons during development.
Collapse
|
4
|
Liu T, Zeng X, Sun F, Hou H, Guan Y, Guo D, Ai H, Wang W, Zhang G. EphB4 Regulates Self-Renewal, Proliferation and Neuronal Differentiation of Human Embryonic Neural Stem Cells in Vitro. Cell Physiol Biochem 2017; 41:819-834. [DOI: 10.1159/000459693] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/19/2016] [Indexed: 11/19/2022] Open
Abstract
Background/Aims: EphB4 belongs to the largest family of Eph receptor tyrosine kinases. It contributes to a variety of pathological progresses of cancer malignancy. However, little is known about its role in neural stem cells (NSCs). This study examined whether EphB4 is required for proliferation and differentiation of human embryonic neural stem cells (hNSCs) in vitro. Methods: We up- and down-regulated EphB4 expression in hNSCs using lentiviral over-expression and shRNA knockdown constructs and then investigated the influence of EphB4 on the properties of hNSCs. Results: Our results show that shRNA-mediated EphB4 reduction profoundly impaired hNSCs self-renewal and proliferation. Furthermore, detection of differentiation revealed that knockdown of EphB4 inhibited hNSCs differentiation towards a neuronal lineage and promoted hNSCs differentiation to glial cells. In contrast, EphB4 overexpression promoted hNSCs self-renewal and proliferation, further induced hNSCs differentiation towards a neuronal lineage and inhibited hNSCs differentiation to glial cells. Moreover, we found that EphB4 regulates cell proliferation mediated by the Abl-CyclinD1 pathway. Conclusion: These studies provide strong evidence that fine tuning of EphB4 expression is crucial for the proliferation and neuronal differentiation of hNSCs, suggesting that EphB4 might be an interesting target for overcoming some of the therapeutic limitations of neuronal loss in brain diseases.
Collapse
|
5
|
Vuong TA, Leem YE, Kim BG, Cho H, Lee SJ, Bae GU, Kang JS. A Sonic hedgehog coreceptor, BOC regulates neuronal differentiation and neurite outgrowth via interaction with ABL and JNK activation. Cell Signal 2016; 30:30-40. [PMID: 27871935 DOI: 10.1016/j.cellsig.2016.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022]
Abstract
Neurite outgrowth is a critical step for neurogenesis and remodeling synaptic circuitry during neuronal development and regeneration. An immunoglobulin superfamily member, BOC functions as Sonic hedgehog (Shh) coreceptor in canonical and noncanonical Shh signaling in neuronal development and axon outgrowth/guidance. However signaling mechanisms responsible for BOC action during these processes remain unknown. In our previous studies, a multiprotein complex containing BOC and a closely related protein CDO promotes myogenic differentiation through activation of multiple signaling pathways, including non-receptor tyrosine kinase ABL. Given that ABL and Jun. N-terminal kinase (JNK) are implicated in actin cytoskeletal dynamics required for neurogenesis, we investigated the relationship between BOC, ABL and JNK during neuronal differentiation. Here, we demonstrate that BOC and ABL are induced in P19 embryonal carcinoma (EC) cells and cortical neural progenitor cells (NPCs) during neuronal differentiation. BOC-depleted EC cells or Boc-/- NPCs exhibit impaired neuronal differentiation with shorter neurite formation. BOC interacts with ABL through its putative SH2 binding domain and seems to be phosphorylated in an ABL activity-dependent manner. Unlike wildtype BOC, ABL-binding defective BOC mutants exhibit impaired JNK activation and neuronal differentiation. Finally, Shh treatment enhances JNK activation which is diminished by BOC depletion. These data suggest that BOC interacts with ABL and activates JNK thereby promoting neuronal differentiation and neurite outgrowth.
Collapse
Affiliation(s)
- Tuan Anh Vuong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Bok-Geon Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea
| | - Sang-Jin Lee
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Gyu-Un Bae
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon 16419, Republic of Korea.
| |
Collapse
|
6
|
Harding MJ, McGraw HF, Nechiporuk A. The roles and regulation of multicellular rosette structures during morphogenesis. Development 2014; 141:2549-58. [PMID: 24961796 DOI: 10.1242/dev.101444] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multicellular rosettes have recently been appreciated as important cellular intermediates that are observed during the formation of diverse organ systems. These rosettes are polarized, transient epithelial structures that sometimes recapitulate the form of the adult organ. Rosette formation has been studied in various developmental contexts, such as in the zebrafish lateral line primordium, the vertebrate pancreas, the Drosophila epithelium and retina, as well as in the adult neural stem cell niche. These studies have revealed that the cytoskeletal rearrangements responsible for rosette formation appear to be conserved. By contrast, the extracellular cues that trigger these rearrangements in vivo are less well understood and are more diverse. Here, we review recent studies of the genetic regulation and cellular transitions involved in rosette formation. We discuss and compare specific models for rosette formation and highlight outstanding questions in the field.
Collapse
Affiliation(s)
- Molly J Harding
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR 97239, USA Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239, USA
| | - Hillary F McGraw
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alex Nechiporuk
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
7
|
M. Vargas L, Leal N, Estrada LD, González A, Serrano F, Araya K, Gysling K, Inestrosa NC, Pasquale EB, Alvarez AR. EphA4 activation of c-Abl mediates synaptic loss and LTP blockade caused by amyloid-β oligomers. PLoS One 2014; 9:e92309. [PMID: 24658113 PMCID: PMC3962387 DOI: 10.1371/journal.pone.0092309] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/21/2014] [Indexed: 01/04/2023] Open
Abstract
The early stages of Alzheimer's disease are characterised by impaired synaptic plasticity and synapse loss. Here, we show that amyloid-β oligomers (AβOs) activate the c-Abl kinase in dendritic spines of cultured hippocampal neurons and that c-Abl kinase activity is required for AβOs-induced synaptic loss. We also show that the EphA4 receptor tyrosine kinase is upstream of c-Abl activation by AβOs. EphA4 tyrosine phosphorylation (activation) is increased in cultured neurons and synaptoneurosomes exposed to AβOs, and in Alzheimer-transgenic mice brain. We do not detect c-Abl activation in EphA4-knockout neurons exposed to AβOs. More interestingly, we demonstrate EphA4/c-Abl activation is a key-signalling event that mediates the synaptic damage induced by AβOs. According to this results, the EphA4 antagonistic peptide KYL and c-Abl inhibitor STI prevented i) dendritic spine reduction, ii) the blocking of LTP induction and iii) neuronal apoptosis caused by AβOs. Moreover, EphA4-/- neurons or sh-EphA4-transfected neurons showed reduced synaptotoxicity by AβOs. Our results are consistent with EphA4 being a novel receptor that mediates synaptic damage induced by AβOs. EphA4/c-Abl signalling could be a relevant pathway involved in the early cognitive decline observed in Alzheimer's disease patients.
Collapse
Affiliation(s)
- Lina M. Vargas
- Departamento de Biología Celular y Molecular, Laboratorio de Señalización Celular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Nancy Leal
- Departamento de Biología Celular y Molecular, Laboratorio de Señalización Celular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Lisbell D. Estrada
- Departamento de Biología Celular y Molecular, Laboratorio de Señalización Celular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Adrian González
- Departamento de Biología Celular y Molecular, Laboratorio de Señalización Celular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Felipe Serrano
- Departamento de Biología Celular y Molecular, Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Katherine Araya
- Departamento de Biología Celular y Molecular, Millenium Nucleus in Stress and Addiction, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Katia Gysling
- Departamento de Biología Celular y Molecular, Millenium Nucleus in Stress and Addiction, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C. Inestrosa
- Departamento de Biología Celular y Molecular, Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Elena B. Pasquale
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Alejandra R. Alvarez
- Departamento de Biología Celular y Molecular, Laboratorio de Señalización Celular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
8
|
Engel U, Zhan Y, Long JB, Boyle SN, Ballif BA, Dorey K, Gygi SP, Koleske AJ, Vanvactor D. Abelson phosphorylation of CLASP2 modulates its association with microtubules and actin. Cytoskeleton (Hoboken) 2014; 71:195-209. [PMID: 24520051 PMCID: PMC4054870 DOI: 10.1002/cm.21164] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 12/21/2013] [Accepted: 12/30/2013] [Indexed: 11/20/2022]
Abstract
The Abelson (Abl) non-receptor tyrosine kinase regulates the cytoskeleton during multiple stages of neural development, from neurulation, to the articulation of axons and dendrites, to synapse formation and maintenance. We previously showed that Abl is genetically linked to the microtubule (MT) plus end tracking protein (+TIP) CLASP in Drosophila. Here we show in vertebrate cells that Abl binds to CLASP and phosphorylates it in response to serum or PDGF stimulation. In vitro, Abl phosphorylates CLASP with a Km of 1.89 µM, indicating that CLASP is a bona fide substrate. Abl-phosphorylated tyrosine residues that we detect in CLASP by mass spectrometry lie within previously mapped F-actin and MT plus end interaction domains. Using purified proteins, we find that Abl phosphorylation modulates direct binding between purified CLASP2 with both MTs and actin. Consistent with these observations, Abl-induced phosphorylation of CLASP2 modulates its localization as well as the distribution of F-actin structures in spinal cord growth cones. Our data suggest that the functional relationship between Abl and CLASP2 is conserved and provides a means to control the CLASP2 association with the cytoskeleton. © 2014 The Authors. Cytoskeleton Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ulrike Engel
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts; Nikon Imaging Center, the University of Heidelberg, Bioquant, 69120, Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Grossman EN, Giurumescu CA, Chisholm AD. Mechanisms of ephrin receptor protein kinase-independent signaling in amphid axon guidance in Caenorhabditis elegans. Genetics 2013; 195:899-913. [PMID: 23979582 PMCID: PMC3813872 DOI: 10.1534/genetics.113.154393] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/16/2013] [Indexed: 12/30/2022] Open
Abstract
Eph receptors and their ephrin ligands are key conserved regulators of axon guidance and can function in a variety of signaling modes. Here we analyze the genetic and cellular requirements for Eph signaling in a Caenorhabditis elegans axon guidance choice point, the ventral guidance of axons in the amphid commissure. The C. elegans Eph receptor EFN-1 has both kinase-dependent and kinase-independent roles in amphid ventral guidance. Of the four C. elegans ephrins, we find that only EFN-1 has a major role in amphid axon ventral guidance, and signals in both a receptor kinase-dependent and kinase-independent manner. Analysis of EFN-1 and EFN-1 expression and tissue-specific requirements is consistent with a model in which VAB-1 acts in amphid neurons, interacting with EFN-1 expressed on surrounding cells. Unexpectedly, left-hand neurons are more strongly affected than right-hand neurons by loss of Eph signaling, indicating a previously undetected left-right asymmetry in the requirement for Eph signaling. By screening candidate genes involved in Eph signaling, we find that the Eph kinase-independent pathway involves the ABL-1 nonreceptor tyrosine kinase and possibly the phosphatidylinositol 3-kinase pathway. Overexpression of ABL-1 is sufficient to rescue EFN-1 ventral guidance defects cell autonomously. Our results reveal new aspects of Eph signaling in a single axon guidance decision in vivo.
Collapse
Affiliation(s)
- Emily N. Grossman
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093
| | - Claudiu A. Giurumescu
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093
| | - Andrew D. Chisholm
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
10
|
Toyoda Y, Shinohara R, Thumkeo D, Kamijo H, Nishimaru H, Hioki H, Kaneko T, Ishizaki T, Furuyashiki T, Narumiya S. EphA4-dependent axon retraction and midline localization of Ephrin-B3 are disrupted in the spinal cord of mice lacking mDia1 and mDia3 in combination. Genes Cells 2013; 18:873-85. [PMID: 23890216 DOI: 10.1111/gtc.12081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 06/04/2013] [Indexed: 01/13/2023]
Abstract
mDia is an actin nucleator and polymerization factor regulated by the small GTPase Rho and consists of three isoforms. Here, we found that mice lacking mDia1 and mDia3, two isoforms expressed in the brain, in combination (mDia-DKO mice) show impaired left-right limb coordination during locomotion and aberrant midline crossing of axons of corticospinal neurons and spinal cord interneurons. Given that mice lacking Ephrin-B3-EphA4 signaling show a similar impairment in locomotion, we examined whether mDia is involved in Ephrin-B3-EphA4 signaling for axon repulsion. In primary cultured neurons, mDia deficiency impairs growth cone collapse and axon retraction induced by chemo-repellants including EphA ligands. In mDia-DKO mice, the Ephrin-B3-expressing midline structure in the spinal cord is disrupted, and axons aberrantly cross the spinal cord midline preferentially through the region devoid of Ephrin-B3. Therefore, mDia plays multiple roles in the proper formation of the neural network in vivo.
Collapse
Affiliation(s)
- Yosuke Toyoda
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Yoshida-konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Srivastava N, Robichaux MA, Chenaux G, Henkemeyer M, Cowan CW. EphB2 receptor forward signaling controls cortical growth cone collapse via Nck and Pak. Mol Cell Neurosci 2012; 52:106-16. [PMID: 23147113 DOI: 10.1016/j.mcn.2012.11.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 10/03/2012] [Accepted: 11/02/2012] [Indexed: 02/08/2023] Open
Abstract
EphB receptors and their ephrinB ligands transduce bidirectional signals that mediate contact-dependent axon guidance primarily by promoting growth cone repulsion. However, how EphB receptor-mediated forward signaling induces axonal repulsion remains poorly understood. Here, we identify Nck and Pak proteins as essential forward signaling components of EphB2-dependent growth cone collapse in cortical neurons. We show that kinase-active EphB2 binds to Pak and promotes growth cone repulsion via Pak kinase activity, Pak-Nck binding, RhoA signaling and endocytosis. However, Pak's function in this context appears to be independent of Rac/Cdc42-GTP, consistent with the absence of Rac-GTP production after ephrinB treatment of cortical neurons. Taken together, our findings suggest that ephrinB-activated EphB2 receptors recruit a novel Nck/Pak signaling complex to mediate repulsive cortical growth cone guidance, which may be relevant for EphB forward signaling-dependent axon guidance in vivo.
Collapse
Affiliation(s)
- Nishi Srivastava
- Department of Psychiatry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, United States
| | | | | | | | | |
Collapse
|
12
|
Ball SG, Shuttleworth A, Kielty CM. Inhibition of platelet-derived growth factor receptor signaling regulates Oct4 and Nanog expression, cell shape, and mesenchymal stem cell potency. Stem Cells 2012; 30:548-60. [PMID: 22213560 PMCID: PMC3537888 DOI: 10.1002/stem.1015] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Defining the signaling mechanisms that regulate the fate of adult stem cells is an essential step toward their use in regenerative medicine. Platelet-derived growth factor receptor (PDGFR) signaling plays a crucial role in specifying mesenchymal stem cell (MSC) commitment to mesenchymal lineages. Based on the hypothesis that selective inhibition of signaling pathways involved in differentiation may increase stem cell potency, we examined the role of PDGFR signaling in controlling the fate of human MSCs. Using a small molecular PDGFR inhibitor that induced MSCs toward a more rounded shape, expression of Oct4 and Nanog were markedly upregulated. In these PDGFR inhibitor-treated MSCs, Oct4 and Nanog expression and cell shape were regulated by janus kinase (JAK), MAPK kinase (MEK), and epidermal growth factor receptor (EGFR) signaling. Under defined differentiation conditions, these PDGFR-inhibited MSCs expressed definitive endodermal, ectodermal, and mesodermal markers. We also confirmed that depletion of individual PDGF receptors upregulated expression of Oct4A and Nanog. This study identifies PDGFR signaling as a key regulator of Oct4 and Nanog expression and of MSC potency. Thus, inhibiting these specific receptor tyrosine kinases, which play essential roles in tissue formation, offers a novel approach to unlock the therapeutic capacity of MSCs. STEM CELLS 2012;30:548–560
Collapse
Affiliation(s)
- Stephen G Ball
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
13
|
Marsick BM, Roche FK, Letourneau PC. Repulsive axon guidance cues ephrin-A2 and slit3 stop protrusion of the growth cone leading margin concurrently with inhibition of ADF/cofilin and ERM proteins. Cytoskeleton (Hoboken) 2012; 69:496-505. [PMID: 22328420 DOI: 10.1002/cm.21016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 02/01/2012] [Indexed: 11/08/2022]
Abstract
Axonal growth cones turn away from repulsive guidance cues. This may start with reduced protrusive motility in the region the growth cone leading margin that is closer to the source of repulsive cue. Using explants of E7 chick temporal retina, we examine the effects of two repulsive guidance cues, ephrin-A2 and slit3, on retinal ganglion cell growth cone protrusive activity, total F-actin, free F-actin barbed ends, and the activities (phosphorylation states) of actin regulatory proteins, ADF/cofilin and ezrin, radixin, moesin (ERM) proteins. Ephrin-A2 rapidly stops protrusive activity simultaneously with reducing F-actin, free barbed ends and the activities of ADF/cofilin and ERM proteins. Slit3 also stops protrusion and reduces the activities of ADF/cofilin and ERM proteins. We interpret these results as indicating that repulsive guidance cues inhibit actin polymerization and actin-membrane linkage to stop protrusive activity. Retrograde F-actin flow withdraws actin to the C-domain, where F-actin bundles interact with myosin II to generate contractile forces that can collapse and retract the growth cone. Our results suggest that common mechanisms are used by repulsive guidance cue to disable growth cone motility and remodel growing axon terminals.
Collapse
Affiliation(s)
- Bonnie M Marsick
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
14
|
Kim D, Song J, Kim S, Chun CH, Jin EJ. MicroRNA-34a regulates migration of chondroblast and IL-1β-induced degeneration of chondrocytes by targeting EphA5. Biochem Biophys Res Commun 2011; 415:551-7. [DOI: 10.1016/j.bbrc.2011.10.087] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 10/18/2011] [Indexed: 12/31/2022]
|
15
|
Hester SD, Belmonte JM, Gens JS, Clendenon SG, Glazier JA. A multi-cell, multi-scale model of vertebrate segmentation and somite formation. PLoS Comput Biol 2011; 7:e1002155. [PMID: 21998560 PMCID: PMC3188485 DOI: 10.1371/journal.pcbi.1002155] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 06/27/2011] [Indexed: 12/23/2022] Open
Abstract
Somitogenesis, the formation of the body's primary segmental structure common to all vertebrate development, requires coordination between biological mechanisms at several scales. Explaining how these mechanisms interact across scales and how events are coordinated in space and time is necessary for a complete understanding of somitogenesis and its evolutionary flexibility. So far, mechanisms of somitogenesis have been studied independently. To test the consistency, integrability and combined explanatory power of current prevailing hypotheses, we built an integrated clock-and-wavefront model including submodels of the intracellular segmentation clock, intercellular segmentation-clock coupling via Delta/Notch signaling, an FGF8 determination front, delayed differentiation, clock-wavefront readout, and differential-cell-cell-adhesion-driven cell sorting. We identify inconsistencies between existing submodels and gaps in the current understanding of somitogenesis mechanisms, and propose novel submodels and extensions of existing submodels where necessary. For reasonable initial conditions, 2D simulations of our model robustly generate spatially and temporally regular somites, realistic dynamic morphologies and spontaneous emergence of anterior-traveling stripes of Lfng. We show that these traveling stripes are pseudo-waves rather than true propagating waves. Our model is flexible enough to generate interspecies-like variation in somite size in response to changes in the PSM growth rate and segmentation-clock period, and in the number and width of Lfng stripes in response to changes in the PSM growth rate, segmentation-clock period and PSM length.
Collapse
Affiliation(s)
- Susan D Hester
- Biocomplexity Institute and Department of Physics, Indiana University Bloomington, Bloomington, Indiana, United States of America.
| | | | | | | | | |
Collapse
|
16
|
Kadir S, Astin JW, Tahtamouni L, Martin P, Nobes CD. Microtubule remodelling is required for the front-rear polarity switch during contact inhibition of locomotion. J Cell Sci 2011; 124:2642-53. [PMID: 21750190 DOI: 10.1242/jcs.087965] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
When migrating mesenchymal cells collide, they exhibit a 'contact inhibition of locomotion' response that results in reversal of their front-rear polarity by extension of a new leading edge, which enables their migration away from the opposing contacted cell. The critical cytoskeletal rearrangements underpinning these mutual repulsion events are currently unknown. We found that during fibroblast cell-cell collisions, microtubules at the region of contact increase their frequency of catastrophe, their rates of shrinkage and growth, and concomitantly, a new microtubule array is established at a new leading edge. We show that Rho and ROCK activity is necessary for this repulsion response, and we observed increased microtubule stabilisation as a consequence of ROCK inhibition. Importantly, partial destabilisation of microtubules, by co-treatment with a low dose of nocodazole, restored microtubule dynamics to that of untreated cells and rescued contact inhibition of locomotion in ROCK-inhibited cells. Although there was an increase in microtubule growth or shrinkage rates in Y27632 cell-cell collisions, these failed to reach the same level of dynamicity compared with untreated collisions. Our data suggest that microtubule dynamics at contact sites must increase beyond a threshold for a cell to switch its front-rear polarity, and that microtubule stabilisation can lead to a failure of contact inhibition of locomotion.
Collapse
Affiliation(s)
- Shereen Kadir
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | | | |
Collapse
|
17
|
Akay C, Lindl KA, Wang Y, White MG, Isaacman-Beck J, Kolson DL, Jordan-Sciutto KL. Site-specific hyperphosphorylation of pRb in HIV-induced neurotoxicity. Mol Cell Neurosci 2011; 47:154-65. [PMID: 21504794 DOI: 10.1016/j.mcn.2011.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 03/25/2011] [Accepted: 04/04/2011] [Indexed: 12/18/2022] Open
Abstract
HIV-Associated Neurocognitive Disorder (HAND) remains a serious complication of HIV infection, despite combined Anti-Retroviral Therapy (cART). Neuronal dysfunction and death are attributed to soluble factors released from activated and/or HIV-infected macrophages. Most of these factors affect the cell cycle machinery, determining cellular outcomes even in the absence of cell division. One of the earliest events in cell cycle activation is hyperphosphorylation of the retinoblastoma protein, pRb (ppRb). We and others have previously shown increased ppRb expression in the CNS of patients with HIV encephalitis (HIVE) and in neurons in an in vitro model of HIV-induced neurodegeneration. However, trophic factors also lead to an increase in neuronal ppRb with an absence of cell death, suggesting that, depending on the stimulus, hyperphosphorylation of pRb can have different outcomes on neuronal fate. pRb has multiple serines and threonines targeted for phosphorylation by distinct kinases, and we hypothesized that different stimuli may target separate sites for phosphorylation. Thus, to determine whether pRb is differentially phosphorylated in response to different stimuli and whether any of these sites is preferentially phosphorylated in association with HIV-induced neurotoxicity, we treated primary rat mixed cortical cultures with trophic factors, BDNF or RANTES, or with the neurotoxic factor, N-methyl-d-aspartate (NMDA), or with supernatants containing factors secreted by HIV-infected monocyte-derived macrophages (HIV-MDM), our in vitro model of HIV-induced neurodegeneration. We found that, while BDNF and RANTES phosphorylated serine807/811 and serine608 in vitro, treatment with HIV-MDM did not, even though these trophic factors are components of HIV-MDM. Rather, HIV-MDM targets a specific phosphorylation site, serine795, of pRb for phosphorylation in vitro and this ppRb isoform is also increased in HIV-infected brains in vivo. Further, overexpression of a nonphosphorylatable pRb (ppRb S795A) attenuated HIV-MDM-induced neurotoxicity. These findings indicate that HIV-infection in the brain is associated with site-specific hyperphosphorylation of pRb at serine795, which is not induced by other tested stimuli, and that this phosphorylation contributes to neuronal death in this disease, demonstrating that specific pRb sites are differentially targeted and may have diverse impacts on the viability of post-mitotic neurons.
Collapse
Affiliation(s)
- C Akay
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Dent EW, Gupton SL, Gertler FB. The growth cone cytoskeleton in axon outgrowth and guidance. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a001800. [PMID: 21106647 DOI: 10.1101/cshperspect.a001800] [Citation(s) in RCA: 402] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Axon outgrowth and guidance to the proper target requires the coordination of filamentous (F)-actin and microtubules (MTs), the dynamic cytoskeletal polymers that promote shape change and locomotion. Over the past two decades, our knowledge of the many guidance cues, receptors, and downstream signaling cascades involved in neuronal outgrowth and guidance has increased dramatically. Less is known, however, about how those cascades of information converge and direct appropriate remodeling and interaction of cytoskeletal polymers, the ultimate effectors of movement and guidance. During development, much of the communication that occurs between environmental guidance cues and the cytoskeleton takes place at the growing tip of the axon, the neuronal growth cone. Several articles on this topic focus on the "input" to the growth cone, the myriad of receptor types, and their corresponding cognate ligands. Others investigate the signaling cascades initiated by receptors and propagated by second messenger pathways (i.e., kinases, phosphatases, GTPases). Ultimately, this plethora of information converges on proteins that associate directly with the actin and microtubule cytoskeletons. The role of these cytoskeletal-associated proteins, as well as the cytoskeleton itself in axon outgrowth and guidance, is the subject of this article.
Collapse
Affiliation(s)
- Erik W Dent
- Department of Anatomy, University of Wisconsin-Madison, 53706, USA
| | | | | |
Collapse
|
19
|
Petros TJ, Bryson JB, Mason C. Ephrin-B2 elicits differential growth cone collapse and axon retraction in retinal ganglion cells from distinct retinal regions. Dev Neurobiol 2010; 70:781-94. [PMID: 20629048 DOI: 10.1002/dneu.20821] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The circuit for binocular vision and stereopsis is established at the optic chiasm, where retinal ganglion cell (RGC) axons diverge into the ipsilateral and contralateral optic tracts. In the mouse retina, ventrotemporal (VT) RGCs express the guidance receptor EphB1, which interacts with the repulsive guidance cue ephrin-B2 on radial glia at the optic chiasm to direct VT RGC axons ipsilaterally. RGCs in the ventral retina also express EphB2, which interacts with ephrin-B2, whereas dorsal RGCs express low levels of EphB receptors. To investigate how growth cones of RGCs from different retinal regions respond upon initial contact with ephrin-B2, we utilized time-lapse imaging to characterize the effects of ephrin-B2 on growth cone collapse and axon retraction in real time. We demonstrate that bath application of ephrin-B2 induces rapid and sustained growth cone collapse and axon retraction in VT RGC axons, whereas contralaterally-projecting dorsotemporal RGCs display moderate growth cone collapse and little axon retraction. Dose response curves reveal that contralaterally-projecting ventronasal axons are less sensitive to ephrin-B2 treatment compared to VT axons. Additionally, we uncovered a specific role for Rho kinase signaling in the retraction of VT RGC axons but not in growth cone collapse. The detailed characterization of growth cone behavior in this study comprises an assay for the study of Eph signaling in RGCs, and provides insight into the phenomena of growth cone collapse and axon retraction in general.
Collapse
Affiliation(s)
- Timothy J Petros
- Department of Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | |
Collapse
|
20
|
Regulation of axonal development by the nuclear protein hindsight (pebbled) in the Drosophila visual system. Dev Biol 2010; 344:911-21. [PMID: 20541542 DOI: 10.1016/j.ydbio.2010.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 05/14/2010] [Accepted: 06/03/2010] [Indexed: 11/24/2022]
Abstract
The molecules and networks involved in the process of acquisition and maintenance of the form of a mature neuron are not completely known. Using a misexpression screen we identified the gene hindsight as a gene involved in the process of acquisition of the neuronal morphogenesis in the Drosophila adult nervous system. hindsight encodes a transcription factor known for its role in early developmental processes such as embryonic germ band retraction and dorsal closure, as well as in the establishment of cell morphology, planar cell polarity, and epithelial integrity during retinal development. We describe here a novel function for HNT by showing that both loss and gain of function of HNT affects the pathfinding of the photoreceptors axons. By manipulating the timing and level of HNT expression, together with the number of cells manipulated we show here that the function of HNT in axonal guidance is independent of the HNT functions previously reported in retinal cells. Based on genetic interaction experiments we show that part of HNT function in axonal development is exerted through the regulation of genes involved in the dynamics of the actin cytoskeleton.
Collapse
|
21
|
Genander M, Halford MM, Xu NJ, Eriksson M, Yu Z, Qiu Z, Martling A, Greicius G, Thakar S, Catchpole T, Chumley MJ, Zdunek S, Wang C, Holm T, Goff SP, Pettersson S, Pestell RG, Henkemeyer M, Frisén J. Dissociation of EphB2 signaling pathways mediating progenitor cell proliferation and tumor suppression. Cell 2009; 139:679-92. [PMID: 19914164 DOI: 10.1016/j.cell.2009.08.048] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 05/20/2009] [Accepted: 08/27/2009] [Indexed: 01/25/2023]
Abstract
Signaling proteins driving the proliferation of stem and progenitor cells are often encoded by proto-oncogenes. EphB receptors represent a rare exception; they promote cell proliferation in the intestinal epithelium and function as tumor suppressors by controlling cell migration and inhibiting invasive growth. We show that cell migration and proliferation are controlled independently by the receptor EphB2. EphB2 regulated cell positioning is kinase-independent and mediated via phosphatidylinositol 3-kinase, whereas EphB2 tyrosine kinase activity regulates cell proliferation through an Abl-cyclin D1 pathway. Cyclin D1 regulation becomes uncoupled from EphB signaling during the progression from adenoma to colon carcinoma in humans, allowing continued proliferation with invasive growth. The dissociation of EphB2 signaling pathways enables the selective inhibition of the mitogenic effect without affecting the tumor suppressor function and identifies a pharmacological strategy to suppress adenoma growth.
Collapse
Affiliation(s)
- Maria Genander
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Bradley WD, Koleske AJ. Regulation of cell migration and morphogenesis by Abl-family kinases: emerging mechanisms and physiological contexts. J Cell Sci 2009; 122:3441-54. [PMID: 19759284 DOI: 10.1242/jcs.039859] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Abl-family non-receptor tyrosine kinases are essential regulators of the cytoskeleton. They transduce diverse extracellular cues into cytoskeletal rearrangements that have dramatic effects on cell motility and morphogenesis. Recent biochemical and genetic studies have revealed several mechanisms that Abl-family kinases use to mediate these effects. Abl-family kinases stimulate actin polymerization through the activation of cortactin, hematopoietic lineage cell-specific protein (HS1), WASp- and WAVE-family proteins, and Rac1. They also attenuate cell contractility by inhibiting RhoA and altering adhesion dynamics. These pathways impinge on several physiological processes, including development and maintenance of the nervous and immune systems, and epithelial morphogenesis. Elucidating how Abl-family kinases are regulated, and where and when they coordinate cytoskeletal changes, is essential for garnering a better understanding of these complex processes.
Collapse
Affiliation(s)
- William D Bradley
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA.
| | | |
Collapse
|
23
|
Reverse signaling by glycosylphosphatidylinositol-linked Manduca ephrin requires a SRC family kinase to restrict neuronal migration in vivo. J Neurosci 2009; 29:3404-18. [PMID: 19295147 DOI: 10.1523/jneurosci.5464-08.2009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Reverse signaling via glycosylphosphatidylinositol (GPI)-linked Ephrins may help control cell proliferation and outgrowth within the nervous system, but the mechanisms underlying this process remain poorly understood. In the embryonic enteric nervous system (ENS) of the moth Manduca sexta, migratory neurons forming the enteric plexus (EP cells) express a single Ephrin ligand (GPI-linked MsEphrin), whereas adjacent midline cells that are inhibitory to migration express the cognate receptor (MsEph). Knocking down MsEph receptor expression in cultured embryos with antisense morpholino oligonucleotides allowed the EP cells to cross the midline inappropriately, consistent with the model that reverse signaling via MsEphrin mediates a repulsive response in the ENS. Src family kinases have been implicated in reverse signaling by type-A Ephrins in other contexts, and MsEphrin colocalizes with activated forms of endogenous Src in the leading processes of the EP cells. Pharmacological inhibition of Src within the developing ENS induced aberrant midline crossovers, similar to the effect of blocking MsEphrin reverse signaling. Hyperstimulating MsEphrin reverse signaling with MsEph-Fc fusion proteins induced the rapid activation of endogenous Src specifically within the EP cells, as assayed by Western blots of single embryonic gut explants and by whole-mount immunostaining of cultured embryos. In longer cultures, treatment with MsEph-Fc caused a global inhibition of EP cell migration and outgrowth, an effect that was prevented by inhibiting Src activation. These results support the model that MsEphrin reverse signaling induces the Src-dependent retraction of EP cell processes away from the enteric midline, thereby helping to confine the neurons to their appropriate pathways.
Collapse
|
24
|
Huang X, Wu D, Jin H, Stupack D, Wang JYJ. Induction of cell retraction by the combined actions of Abl-CrkII and Rho-ROCK1 signaling. ACTA ACUST UNITED AC 2008; 183:711-23. [PMID: 19001122 PMCID: PMC2582888 DOI: 10.1083/jcb.200801192] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Dynamic modulation of cell adhesion is integral to a wide range of biological processes. The small guanosine triphosphatase (GTPase) Rap1 is an important regulator of cell–cell and cell–matrix adhesions. We show here that induced expression of activated Abl tyrosine kinase reduces Rap1-GTP levels through phosphorylation of Tyr221 of CrkII, which disrupts interaction of CrkII with C3G, a guanine nucleotide exchange factor for Rap1. Abl-dependent down-regulation of Rap1-GTP causes cell rounding and detachment only when the Rho–ROCK1 pathway is also activated, for example, by lysophosphatidic acid (LPA). During ephrin-A1–induced retraction of PC3 prostate cancer cells, we show that endogenous Abl is activated and disrupts the CrkII–C3G complex to reduce Rap1-GTP. Interestingly, ephrin-A1–induced PC3 cell retraction also requires LPA, which stimulates Rho to a much higher level than that is activated by ephrin-A1. Our results establish Rap1 as another downstream target of the Abl–CrkII signaling module and show that Abl–CrkII collaborates with Rho–ROCK1 to stimulate cell retraction.
Collapse
Affiliation(s)
- XiaoDong Huang
- Division of Biological Sciences, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
25
|
Reverse signaling via a glycosyl-phosphatidylinositol-linked ephrin prevents midline crossing by migratory neurons during embryonic development in Manduca. J Neurosci 2008; 28:3846-60. [PMID: 18400884 DOI: 10.1523/jneurosci.5691-07.2008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have investigated whether reverse signaling via a glycosyl-phosphatidylinositol (GPI)-linked ephrin controls the behavior of migratory neurons in vivo. During the formation of the enteric nervous system (ENS) in the moth Manduca, approximately 300 neurons [enteric plexus (EP) cells] migrate onto the midgut via bilaterally paired muscle bands but avoid adjacent midline regions. As they migrate, the EP cells express a single ephrin ligand (MsEphrin; a GPI-linked ligand), whereas the midline cells express the corresponding Eph receptor (MsEph). Blocking endogenous MsEphrin-MsEph receptor interactions in cultured embryos resulted in aberrant midline crossing by the neurons and their processes. In contrast, activating endogenous MsEphrin on the EP cells with dimeric MsEph-Fc constructs inhibited their migration and outgrowth, supporting a role for MsEphrin-dependent reverse signaling in this system. In short-term cultures, blocking endogenous MsEph receptors allowed filopodia from the growth cones of the neurons to invade the midline, whereas activating neuronal MsEphrin led to filopodial retraction. MsEphrin-dependent signaling may therefore guide the migratory enteric neurons by restricting the orientation of their leading processes. Knocking down MsEphrin expression in the EP cells with morpholino antisense oligonucleotides also induced aberrant midline crossing, consistent with the effects of blocking endogenous MsEphrin-MsEph interactions. Unexpectedly, this treatment enhanced the overall extent of migration, indicating that MsEphrin-dependent signaling may also modulate the general motility of the EP cells. These results demonstrate that MsEphrin-MsEph receptor interactions normally prevent midline crossing by migratory neurons within the developing ENS, an effect that is most likely mediated by reverse signaling through this GPI-linked ephrin ligand.
Collapse
|
26
|
Ichikawa M, Yoshida J, Saito K, Sagawa H, Tokita Y, Watanabe M. Differential effects of two ROCK inhibitors, Fasudil and Y-27632, on optic nerve regeneration in adult cats. Brain Res 2008; 1201:23-33. [PMID: 18313036 DOI: 10.1016/j.brainres.2008.01.063] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 01/23/2008] [Accepted: 01/24/2008] [Indexed: 11/19/2022]
Abstract
A ROCK inhibitor Fasudil is widely administered to relieve vasospasm in patients after subarachnoid hemorrhage in Japan. We investigated the difference of Fasudil and Y-27632, a common ROCK inhibitor, on neurite regeneration in culture and axonal regeneration after injuring the optic nerve (OpN) in cats. The optimal dose of Y-27632, determined by counting the number and length of neurites in retinal explants, was found to be 100 microM: the only effect of Fasudil was to promote extension of glial processes. We next examined the effects of Fasudil (10 microM-100 microM) and Y-27632 (10 microM-300 microM) on axonal regeneration in the crushed OpN model in vivo. Immediately after crushing the left OpN, Fasudil or Y-27632 was injected into the vitreous and the crushed site. Injection of 10 microM and 100 microM Y-27632 induced extension of the optic axons beyond the crush site, with the latter dosage giving stronger regeneration. Very few axons passed beyond the crush site in the optic nerve with phosphate-buffered saline injection, and no axons elongated in the OpN with Fasudil injection.
Collapse
Affiliation(s)
- Masahiro Ichikawa
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Tsuruma-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Glazier JA, Zhang Y, Swat M, Zaitlen B, Schnell S. Coordinated action of N-CAM, N-cadherin, EphA4, and ephrinB2 translates genetic prepatterns into structure during somitogenesis in chick. Curr Top Dev Biol 2008; 81:205-47. [PMID: 18023729 PMCID: PMC2556964 DOI: 10.1016/s0070-2153(07)81007-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
During gastrulation in vertebrates, mesenchymal cells at the anterior end of the presomitic mesoderm (PSM) periodically compact, transiently epithelialize and detach from the posterior PSM to form somites. In the prevailing clock-and-wavefront model of somitogenesis, periodic gene expression, particularly of Notch and Wnt, interacts with an FGF8-based thresholding mechanism to determine cell fates. However, this model does not explain how cell determination and subsequent differentiation translates into somite morphology. In this paper, we use computer simulations of chick somitogenesis to show that experimentally-observed temporal and spatial patterns of adhesive N-CAM and N-cadherin and repulsive EphA4-ephrinB2 pairs suffice to reproduce the complex dynamic morphological changes of somitogenesis in wild-type and N-cadherin (-/-) chick, including intersomitic separation, boundary-shape evolution and sorting of misdifferentiated cells across compartment boundaries. Since different models of determination yield the same, experimentally-observed, distribution of adhesion and repulsion molecules, the patterning is independent of the details of this mechanism.
Collapse
Affiliation(s)
- James A Glazier
- Biocomplexity Institute and Department of Physics, 727 East Third Street, Indiana University, Bloomington, Indiana 47405, USA
| | | | | | | | | |
Collapse
|
28
|
Shaut CA, Saneyoshi C, Morgan EA, Knosp WM, Sexton DR, Stadler HS. HOXA13 directly regulates EphA6 and EphA7 expression in the genital tubercle vascular endothelia. Dev Dyn 2007; 236:951-60. [PMID: 17304517 DOI: 10.1002/dvdy.21077] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hypospadias, a common defect affecting the growth and closure of the external genitalia, is often accompanied by gross enlargements of the genital tubercle (GT) vasculature. Because Hoxa13 homozygous mutant mice also exhibit hypospadias and GT vessel expansion, we examined whether genes playing a role in angiogenesis exhibit reduced expression in the GT. From this analysis, reductions in EphA6 and EphA7 were detected. Characterization of EphA6 and EphA7 expression in the GT confirmed colocalization with HOXA13 in the GT vascular endothelia. Analysis of the EphA6 and EphA7 promoter regions revealed a series of highly conserved cis-regulatory elements bound by HOXA13 with high affinity. GT chromatin immunoprecipitation confirmed that HOXA13 binds these gene-regulatory elements in vivo. In vitro, HOXA13 activates gene expression through the EphA6 and EphA7 gene-regulatory elements. Together these findings indicate that HOXA13 directly regulates EphA6 and EphA7 in the developing GT and identifies the GT vascular endothelia as a novel site for HOXA13-dependent expression of EphA6 and EphA7.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites
- Cells, Cultured
- Endothelium, Vascular/embryology
- Endothelium, Vascular/metabolism
- Gene Expression Regulation, Developmental
- Genitalia/blood supply
- Genitalia/embryology
- Genitalia/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homeodomain Proteins/physiology
- Mice
- Mice, Mutant Strains
- Molecular Sequence Data
- Promoter Regions, Genetic
- Receptor, EphA6/genetics
- Receptor, EphA6/metabolism
- Receptor, EphA7/genetics
- Receptor, EphA7/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Sequence Homology, Nucleic Acid
- Transfection
Collapse
Affiliation(s)
- Carley A Shaut
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon
| | | | | | | | | | | |
Collapse
|
29
|
Mann F, Chauvet S, Rougon G. Semaphorins in development and adult brain: Implication for neurological diseases. Prog Neurobiol 2007; 82:57-79. [PMID: 17537564 DOI: 10.1016/j.pneurobio.2007.02.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 01/18/2007] [Accepted: 02/26/2007] [Indexed: 01/17/2023]
Abstract
As a group, Semaphorins are expressed in most tissues and this distribution varies considerably with age. Semaphorins are dynamically expressed during embryonic development and their expression is often associated with growing axons. This expression decreases with maturity and several observations support the idea that in adult brain the expression of secreted Semaphorins is sensitive to electrical activity and experience. The functional role of Semaphorins in guiding axonal projections is well established and more recent evidence points to additional roles in the development, function and reorganization of synaptic complexes. Semaphorins exert the majority of their effects by binding to cognate receptor proteins through their extracellular domains. A common theme is that Semaphorin-triggered signalling induces the rearrangement of the actin and microtubule cytoskeleton. Mutations in Semaphorin genes are linked to several human diseases associated with neurological changes, but their actual influence in the pathogenesis of these diseases remains to be demonstrated. In addition, Semaphorins and their receptors are likely to mediate cross-talk between neurons and other cell types, including in pathological situations where their influence can be damaging or favourable depending on the context. We discuss how the manipulation of Semaphorin function might be crucial for future clinical studies.
Collapse
Affiliation(s)
- Fanny Mann
- CNRS UMR 6216, Université de la Méditerranée, Developmental Biology Institute of Marseille Luminy, Case 907, Parc Scientifique de Luminy, 13288 Marseille Cedex 09, France
| | | | | |
Collapse
|
30
|
Groeger G, Nobes C. Co-operative Cdc42 and Rho signalling mediates ephrinB-triggered endothelial cell retraction. Biochem J 2007; 404:23-9. [PMID: 17300218 PMCID: PMC1868826 DOI: 10.1042/bj20070146] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cell repulsion responses to Eph receptor activation are linked to rapid actin cytoskeletal reorganizations, which in turn are partially mediated by Rho-ROCK (Rho kinase) signalling, driving actomyosin contractility. In the present study, we show that Rho alone is not sufficient for this repulsion response. Rather, Cdc42 (cell division cycle 42) and its effector MRCK (myotonic dystrophy kinase-related Cdc42-binding kinase) are also critical for ephrinB-induced cell retraction. Stimulation of endothelial cells with ephrinB2 triggers rapid, but transient, cell retraction. We show that, although membrane retraction is fully blocked by blebbistatin (a myosin-II ATPase inhibitor), it is only partially blocked by inhibiting Rho-ROCK signalling, suggesting that there is ROCK-independent signalling to actomyosin contractility downstream of EphBs. We find that a combination of either Cdc42 or MRCK inhibition with ROCK inhibition completely abolishes the repulsion response. Additionally, endocytosis of ephrin-Eph complexes is not required for initial cell retraction, but is essential for subsequent Rac-mediated re-spreading of cells. Our data reveal a complex interplay of Rho, Rac and Cdc42 in the process of EphB-mediated cell retraction-recovery responses.
Collapse
Affiliation(s)
- Gillian Groeger
- *Department of Physiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, U.K
| | - Catherine D. Nobes
- *Department of Physiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, U.K
- †Department of Biochemistry, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, U.K
- To whom correspondence should be addressed (email )
| |
Collapse
|
31
|
Dorsten JN, Kolodziej PA, VanBerkum MFA. Frazzled regulation of myosin II activity in the Drosophila embryonic CNS. Dev Biol 2007; 308:120-32. [PMID: 17568577 DOI: 10.1016/j.ydbio.2007.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Revised: 05/10/2007] [Accepted: 05/14/2007] [Indexed: 12/30/2022]
Abstract
Frazzled (Fra) is a chemoattractive guidance receptor regulating the cytoskeletal dynamics underlying growth cone steering at the Drosophila embryonic midline. Here, by genetically evaluating the role of Rho GTPases in Fra signaling in vivo, we uncover a Rho-dependent pathway apparently regulating conventional myosin II activity. Midline crossing errors induced by expressing activated Cdc42(v12) or Rac(v12) are suppressed by a heterozygous loss of fra(4) signaling but, in a Fra(wt) gain-of-function condition, no interaction is detected. In contrast, the frequency of crossovers is enhanced approximately 5-fold when Fra(wt) is co-expressed with activated Rho(v14) and this interaction specifically requires the cytoplasmic P3 motif of Fra. Expression of Rho(v14) and activated MLCK (ctMLCK) synergistically increase ectopic crossovers and both require phosphorylation of the regulatory light chain (Sqh) of myosin II. Abelson tyrosine kinase may also help regulate myosin II activity. Heterozygous abl(4) abolishes the midline crossing errors induced by ctMLCK alone or in combination with Fra(wt); suppression of Rho(v14) crossovers is not observed. Interestingly, an interaction between Fra and an activated Abl (Bcr-Abl) also specifically requires the P3 motif. Therefore, the P3 motif of Frazzled appears to initiate Rho and Abl dependent signals to directly or indirectly regulate myosin II activity in growth cones.
Collapse
Affiliation(s)
- Joy N Dorsten
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | | | | |
Collapse
|
32
|
Evans IR, Renne T, Gertler FB, Nobes CD. Ena/VASP proteins mediate repulsion from ephrin ligands. J Cell Sci 2006; 120:289-98. [PMID: 17179204 DOI: 10.1242/jcs.03333] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Ena/VASP proteins negatively regulate cell motility and contribute to repulsion from several guidance cues; however, there is currently no evidence for a role downstream of Eph receptors. Eph receptors mediate repulsion from ephrins at sites of intercellular contact during several developmental migrations. For example, the expression of ephrin-Bs in posterior halves of somites restricts neural crest cell migration to the anterior halves. Here we show that ephrin-B2 destabilises neural crest cell lamellipodia when presented in a substrate-bound or soluble form. Our timelapse studies show that repulsive events are associated with the rearward collapse and subsequent loss of lamellipodia as membrane ruffles. We hypothesise that Ena/VASP proteins contribute to repulsion from ephrins by destabilising cellular protrusions and show that Ena/VASP-deficient fibroblasts exhibit reduced repulsion from both ephrin-A and ephrin-B stripes compared to wild-type controls. Moreover, when EphB4 and ephrin-B2 were expressed in neighbouring Swiss 3T3 fibroblasts, VASP and Mena co-accumulated with activated Eph receptors at protrusions formed by EphB4-expressing cells. Sequestration of Ena/VASP proteins away from the periphery of these cells inhibited Eph receptor internalisation, a process that facilitates repulsion. Our results suggest that Ena/VASP proteins regulate ephrin-induced Eph receptor signalling events, possibly by destabilising lamellipodial protrusions.
Collapse
Affiliation(s)
- Iwan R Evans
- Department of Biochemistry, School of Medical Sciences, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | | | | | | |
Collapse
|
33
|
Lauterbach J, Klein R. Release of full-length EphB2 receptors from hippocampal neurons to cocultured glial cells. J Neurosci 2006; 26:11575-81. [PMID: 17093078 PMCID: PMC6674799 DOI: 10.1523/jneurosci.2697-06.2006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glial cells are known to actively participate in neuronal development by shaping neuronal connections through axon pruning and by controlling dendritic spine morphology. These functions may in part be mediated by engulfment of neuronal structures and trans-endocytosis of neuronal material into glial cells. These processes are not well understood, and the molecular components that mediate these events have primarily been elusive. Here, we implicate the Eph/ephrin signaling system in trans-endocytosis events at the neuron-to-glia interface. Using time-lapse microscopy, we show that hippocampal neurons exogenously expressing EphB2 receptors release or pinch-off EphB2-containing vesicles at sites of neuron-to-glia contact. Cocultured glial cells endogenously express the corresponding ephrinB ligands and are able to trans-endocytose full-length EphB2 from neighboring cells. Although Eph/ephrin signaling often occurs in a bidirectional manner, the observed vesicle release from neurons to glia was only observed in a unidirectional manner, i.e., when the neurons expressed EphB2, but not ephrinBs. These findings suggest that Eph/ephrin signaling is involved in the glial cell-mediated fine sculpting of neuronal structures.
Collapse
Affiliation(s)
- Jenny Lauterbach
- Max-Planck Institute of Neurobiology, D-82152 Martinsried, Germany
| | - Rüdiger Klein
- Max-Planck Institute of Neurobiology, D-82152 Martinsried, Germany
| |
Collapse
|
34
|
Noren NK, Foos G, Hauser CA, Pasquale EB. The EphB4 receptor suppresses breast cancer cell tumorigenicity through an Abl–Crk pathway. Nat Cell Biol 2006; 8:815-25. [PMID: 16862147 DOI: 10.1038/ncb1438] [Citation(s) in RCA: 232] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Accepted: 04/07/2006] [Indexed: 01/06/2023]
Abstract
Recent evidence supports a role for EphB receptor tyrosine kinases as tumour suppressors in colorectal and prostate cancer. However, it is unclear how these receptors inhibit cancer cell tumorigenicity - an activity that is highly unusual for a family of receptor tyrosine kinases. Here, we report that the EphB4 receptor can behave as a tumour suppressor in a mouse xenograft model of breast cancer when stimulated by its ligand, ephrin-B2. In breast cancer cells, EphB4 activates an antioncogenic pathway involving Abl family tyrosine kinases and the Crk adaptor protein. This Abl-Crk pathway inhibits breast cancer cell viability and proliferation in addition to motility and invasion, and also downregulates the pro-invasive matrix metalloprotease, MMP-2. Consistent with these effects, EphB4 and the Abl-Crk pathway are constitutively active in non-transformed mammary epithelial cells. These findings identify a novel Eph receptor signalling pathway with tumour-suppressor activity and predict that therapeutic intervention to activate EphB4 signalling will inhibit tumour progression.
Collapse
Affiliation(s)
- Nicole K Noren
- Burnham Institute for Medical Research, 10901 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
35
|
Schiller MR. Coupling receptor tyrosine kinases to Rho GTPases--GEFs what's the link. Cell Signal 2006; 18:1834-43. [PMID: 16725310 DOI: 10.1016/j.cellsig.2006.01.022] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Revised: 01/19/2006] [Accepted: 04/07/2006] [Indexed: 12/20/2022]
Abstract
Rho GTPases are molecular switches involved in the regulation of many cellular processes. This review summarizes work examining how stimulation of receptor tyrosine kinases (RTKs) leads to the activation of Rho guanine nucleotide exchange factors (GEFs) and their Rho GTPase substrates. The collective findings strongly suggest that RTK signaling to Rho proteins is a general signal transduction mechanism, like RTK mediated activation of phosphatidyl inositol 3-kinase, phospholipase Cgamma, and the mitogen activated protein kinase (MAPK) pathway. More than half of the 58 known human RTKs activate at least one Rho family member. Likewise, 16 Rho GEFs directly interact with and/or are phosphorylated by a RTK. The specificity of receptor tyrosine kinase/Rho GEF signaling seems to be somewhat promiscuous. There several cases where multiple RTKs activate the same Rho GEF and where a single RTK can activate multiple Rho GEFs. Expression analysis indicates that the average human tissue contains transcripts for 33 RTKs, 34 Rho GEFs, and 14 Rho GTPases with each tissue containing a unique complement of these proteins. Given the promiscuity of RTKs for Rho GEFs, Rho GEFs for Rho GTPases, and the large number of these proteins expressed in cells, a complex combinatorial network of proteins in these families may contribute to coding specific signals and cell responses from RTKs.
Collapse
Affiliation(s)
- Martin R Schiller
- Department of Neuroscience and Partnership for Excellence in Structural Biology, University of Connecticut Health Center, Farmington, CT 06030-4301, USA.
| |
Collapse
|