Gustavo Paratcha, Fernanda Ledda, Carlos F. Ibáñez  Cell 

Slides:



Advertisements
Similar presentations
Volume 133, Issue 1, Pages (July 2007)
Advertisements

Toshihide Yamashita, Kerry Lee Tucker, Yves-Alain Barde  Neuron 
Volume 35, Issue 4, Pages (August 2009)
Large Hepatitis Delta Antigen Modulates Transforming Growth Factor-β Signaling Cascades: Implication of Hepatitis Delta Virus–Induced Liver Fibrosis 
Volume 133, Issue 1, Pages (July 2007)
Volume 29, Issue 1, Pages (January 2001)
Takashi Tanaka, Michelle A. Soriano, Michael J. Grusby  Immunity 
Volume 36, Issue 5, Pages (December 2009)
Regulation of Mast Cell Survival by IgE
Volume 8, Issue 1, Pages (July 2001)
Volume 8, Issue 5, Pages (November 2001)
Volume 5, Issue 4, Pages (April 2004)
Haihong Ye, Rejji Kuruvilla, Larry S Zweifel, David D Ginty  Neuron 
Volume 23, Issue 6, Pages (December 2005)
Estela Jacinto, Guy Werlen, Michael Karin  Immunity 
Volume 87, Issue 7, Pages (December 1996)
Volume 45, Issue 3, Pages (February 2005)
Volume 8, Issue 16, Pages (July 1998)
Volume 22, Issue 5, Pages (May 2012)
ASK1 Is Essential for JNK/SAPK Activation by TRAF2
J. Zhu, N. Sun, L. Aoudjit, H. Li, H. Kawachi, S. Lemay, T. Takano 
Volume 15, Issue 22, Pages (November 2005)
Yongli Bai, Chun Yang, Kathrin Hu, Chris Elly, Yun-Cai Liu 
Volume 15, Issue 6, Pages (December 2001)
MUC1 Oncoprotein Stabilizes and Activates Estrogen Receptor α
Robert L.S Perry, Maura H Parker, Michael A Rudnicki  Molecular Cell 
MyD88: An Adapter That Recruits IRAK to the IL-1 Receptor Complex
Ashton Breitkreutz, Lorrie Boucher, Mike Tyers  Current Biology 
Neurotensin receptor–1 and –3 complex modulates the cellular signaling of neurotensin in the HT29 cell line  Stéphane Martin, Valérie Navarro, Jean Pierre.
Transcription Factor MIZ-1 Is Regulated via Microtubule Association
Jungmook Lyu, Vicky Yamamoto, Wange Lu  Developmental Cell 
Volume 6, Issue 6, Pages (December 2000)
Regulation of Mast Cell Survival by IgE
MUC1 Oncoprotein Stabilizes and Activates Estrogen Receptor α
Upregulation of Tenascin-C Expression by IL-13 in Human Dermal Fibroblasts via the Phosphoinositide 3-kinase/Akt and the Protein Kinase C Signaling Pathways 
Histamine Inhibits the Production of Interferon-induced Protein of 10 kDa in Human Squamous Cell Carcinoma and Melanoma  Naoko Kanda, Shinichi Watanabe 
Volume 25, Issue 5, Pages (November 2006)
Volume 18, Issue 11, Pages (March 2017)
MyoD Targets TAF3/TRF3 to Activate Myogenin Transcription
Pei-lin Cheng, Hui Lu, Maya Shelly, Hongfeng Gao, Mu-ming Poo  Neuron 
Paxillin Serves as an ERK-Regulated Scaffold for Coordinating FAK and Rac Activation in Epithelial Morphogenesis  Shuta Ishibe, Dominique Joly, Zhen-Xiang.
Lysine 63 Polyubiquitination of the Nerve Growth Factor Receptor TrkA Directs Internalization and Signaling  Thangiah Geetha, Jianxiong Jiang, Marie W.
GRM7 Regulates Embryonic Neurogenesis via CREB and YAP
Volume 34, Issue 6, Pages (June 2002)
Volume 103, Issue 6, Pages (December 2000)
Extracellular Regulated Kinase Phosphorylates Mitofusin 1 to Control Mitochondrial Morphology and Apoptosis  Aswin Pyakurel, Claudia Savoia, Daniel Hess,
EphB/Syndecan-2 Signaling in Dendritic Spine Morphogenesis
Volume 12, Issue 4, Pages (April 2007)
Rsk1 mediates a MEK–MAP kinase cell survival signal
Volume 96, Issue 6, Pages (March 1999)
ULK1 Phosphorylates and Regulates Mineralocorticoid Receptor
NGF Signaling from Clathrin-Coated Vesicles
Volume 25, Issue 5, Pages (March 2007)
Regulation of protein kinase C ζ by PI 3-kinase and PDK-1
Mst1 Is an Interacting Protein that Mediates PHLPPs' Induced Apoptosis
Volume 125, Issue 4, Pages (May 2006)
Raf and Akt Mediate Distinct Aspects of Sensory Axon Growth
Hua Gao, Yue Sun, Yalan Wu, Bing Luan, Yaya Wang, Bin Qu, Gang Pei 
Volume 119, Issue 5, Pages (November 2000)
Hui Jiang, Wei Guo, Xinhua Liang, Yi Rao  Cell 
Growth Factor-Dependent Trafficking of Cerebellar NMDA Receptors via Protein Kinase B/Akt Phosphorylation of NR2C  Bo-Shiun Chen, Katherine W. Roche 
Target-Derived GFRα1 as an Attractive Guidance Signal for Developing Sensory and Sympathetic Axons via Activation of Cdk5  Fernanda Ledda, Gustavo Paratcha,
In Vitro Analysis of Huntingtin-Mediated Transcriptional Repression Reveals Multiple Transcription Factor Targets  Weiguo Zhai, Hyunkyung Jeong, Libin.
Volume 16, Issue 16, Pages (August 2006)
Volume 23, Issue 2, Pages (August 2005)
Volume 18, Issue 2, Pages (February 1997)
Volume 129, Issue 5, Pages (June 2007)
Volume 21, Issue 2, Pages (January 2006)
Volume 10, Issue 2, Pages (February 1999)
Volume 45, Issue 3, Pages (February 2005)
Presentation transcript:

The Neural Cell Adhesion Molecule NCAM Is an Alternative Signaling Receptor for GDNF Family Ligands  Gustavo Paratcha, Fernanda Ledda, Carlos F. Ibáñez  Cell  Volume 113, Issue 7, Pages 867-879 (June 2003) DOI: 10.1016/S0092-8674(03)00435-5

Figure 1 GDNF Interacts Directly with NCAM in Neuronal and Glial Cells (A) Affinity labeling of RN33B cells with 125I-GDNF, followed by chemical crosslinking and immunoprecipitation (IP) with the indicated antibodies. The membrane shown in the middle panel was reprobed (IB) with L1 antibodies. Cold GDNF was used at 100-fold excess. (B) Affinity labeling of Schwann cells and embryonic hippocampal and cortical neurons with 125I-GDNF, followed by immunoprecipitation with NCAMICD or RET antibodies. Complexes containing p140NCAM and PSA-NCAM are indicated. Cell 2003 113, 867-879DOI: (10.1016/S0092-8674(03)00435-5)

Figure 2 GDNF Stimulates NCAM-Associated Fyn Kinase Activity in Neuronal and Glial Cells (A) In vitro kinase assays on NCAM immunoprecipitates after GDNF stimulation of RN33B and Schwann cells. Purified lipid rafts were used for RN33B cells. Phosphorylated products, including endogenous substrates (solid arrows) and exogenously added enolase (open arrows), are indicated. Reprobings with NCAMICD antibodies are shown below each panel. (B) Following NCAM immunoprecipitation and in vitro kinase assay, samples were eluted and reimmunoprecipitated with Fyn antibodies. Phosphorylated Fyn is indicated (arrows). Supernatants were reimmunoprecipitated with NCAMICD antibodies followed by NCAMICD immunoblotting (bottom). (C) Activation of NCAM-associated Fyn in cortical neurons. In vitro kinase assay was performed on NCAM immunoprecipitates from cortical neurons treated with GDNF (100 ng/ml) and then reimmunoprecipitated with Fyn antibodies. NCAMICD reprobing shown below. (D) Activation of Fyn (left) and phosphorylation of Erk (right) in cortical neurons from wild-type and Ret knockout mice stimulated for 10 min with GDNF (100 ng/ml). Immunoprecipitation of Fyn was followed by in vitro kinase assay and autoradiography. Fyn reprobing is shown below. Erk phosphorylation was detected with anti-phospho-Erk antibodies (P-Erk) and controlled by reprobing with anti-AKT antibodies. Cell 2003 113, 867-879DOI: (10.1016/S0092-8674(03)00435-5)

Figure 3 Reconstitution of High-Affinity GDNF Binding to NCAM and Activation of Fyn in Heterologous Cells Requires Coexpression of GFRα1 Receptors (A) Affinity labeling of COS cells transfected with p140NCAM and different GFRα receptors. All panels show NCAM immunoprecipitates from cell lysates after chemical crosslinking with indicated ligands. NCAMICD reprobing is shown below. (B) Displacement of 125I-GDNF binding to p140NCAM by soluble NCAMECD (used at 100- and 200-fold molar excess). Conditioned medium from naive 293 cells was used as control. (C) Saturation (top) and Scatchard (bottom) plots of 125I-GDNF binding to p140NCAM in the presence or absence of GFRα1 in transfected COS cells. Binding is expressed in arbitrary phosphorimager units (p units) obtained after phosphorimaging scanning of 125I-GDNF-labeled p140NCAM complexes. (D) Activation of Fyn in COS cells transfected with Fyn, p140NCAM, and GFRα1 after GDNF stimulation (100 ng/ml). Immunoprecipitation of Fyn (top) was followed by in vitro kinase assay and autoradiography. Fyn reprobing is shown below. The average increase at 15 min in four independent experiments was 1.9-fold ± 0.2. Lower panels show double immunoprecipitation/in vitro kinase assays and reprobings with NCAM and Fyn antibodies. Cell 2003 113, 867-879DOI: (10.1016/S0092-8674(03)00435-5)

Figure 4 Complex Formation between GFRα1 and NCAM Inhibits Homophilic NCAM Interactions and NCAM-Mediated Cell Adhesion (A) Coimmunoprecipitation of hemagglutinin (HA)-tagged GFRα1 and NCAM in the surface of transfected COS cells. Biotinylated NCAM and GFRα1 were detected by NCAM immunoprecipitation and probing with neutravidin. Reprobings with anti-HA and NCAM antibodies are shown. (B) Direct physical interaction between p140NCAM and GFRα1 in transfected COS cells analyzed by chemical crosslinking and immunoprecipitation. Monomeric p140NCAM (arrowhead), oligomeric NCAM complexes (solid arrows), and complexes containing both NCAM and GFRα1 (open arrows) are indicated. (C) Chemical crosslinking of radiolabeled PSA-NCAM to COS cells expressing HA-tagged p140NCAM in the presence or absence of different amounts of transfected GFRα1 (in μg plasmid DNA). Reprobing with anti-HA antibodies is shown below. (D) Cell adhesion assay in Jurkat cells transiently transfected with p140NCAM and increasing amounts of GFRα1 (in μg plasmid DNA). Only cell aggregates containing more than five cells were counted (arrows). Results are expressed as number of aggregates per 1000 GFP-positive cells ± SD (n = 4). Similar results were obtained in three independent experiments. Scale bar, 50 μm. (E) Chemical crosslinking of 125I-GDNF to COS cells transfected with p140NCAM and increasing amounts of GFRα1 (in μg plasmid DNA). Quantitative results are expressed as 125I-GDNF binding ± SD (n = 3) relative to p140NCAM alone and normalized to the levels of p140NCAM in each lane as determined by immunoblotting. Cell 2003 113, 867-879DOI: (10.1016/S0092-8674(03)00435-5)

Figure 5 GDNF Promotes Schwann Cell Migration via NCAM but Independently of RET (A) Migration of Schwann cells (S-100 staining) from newborn rat sciatic nerve explants plated on naive (control) or GDNF-expressing FR3T3 fibroblasts (GDNF). Scale bar, 200 μm. The results correspond to the averages ± SEM of the means of four independent experiments, each including measurements from 5 to 12 different explants. *p ≤ 0.001. (B) Migration of Schwann cells from newborn mouse sciatic nerve explants isolated from Ret knockout mice. Shown is the mean of at least four different explants ± SD from three animals in each condition. *p ≤ 0.001. (C) Migration of Schwann cells from newborn mouse sciatic nerve explants isolated from Ncam knockout mice. Shown is the mean of at least five different explants ± SD from three animals in each condition. Cell 2003 113, 867-879DOI: (10.1016/S0092-8674(03)00435-5)

Figure 6 GDNF Promotes Axonal Growth in Hippocampal and Cortical Neurons via Fyn and NCAM but Independently of RET (A) Representative photomicrographs of embryonic rat hippocampal and cortical neurons grown on PDL or GDNF (β III tubulin staining). Scale bar, 50 μm. Results are mean ± SEM from a representative experiment performed in triplicate. *p < 0.001 versus PDL or GDNF+PP2 (ANOVA). Similar results were obtained in four independent experiments. (B) Blockade of GDNF outgrowth activity by NCAMECD antibodies. Scale bar, 50 μm. *p ≤ 0.001 (ANOVA). Four independent experiments were performed with similar results. (C) Axonal outgrowth in hippocampal neurons from wild-type and Ret knockout mice. Scale bar, 25 μm. Similar results were obtained in three independent experiments. (D) Axonal outgrowth in cortical and hippocampal neurons from wild-type and Ncam knockout mice. *p < 0.01, compared to wild-type (Student's t test). Similar results were obtained in three independent experiments. (E) Neurite outgrowth of hippocampal neurons from Ncam heterozygous and knockout mice grown on BDNF. The length of the complete dendritic arbor was measured in these experiments. Cell 2003 113, 867-879DOI: (10.1016/S0092-8674(03)00435-5)

Figure 7 GFRα1 and NCAM in the Development of the Rostral Migratory Stream (A) Expression of PSA-NCAM and GFRα1 in sagittal sections of the RMS of adult mice. The inset shows prominent RET staining in cells of the adult facial motor nucleus. Arrowheads delineate the RMS stained with GFRα1 antibodies. Abbreviations: cc, corpus callosum; rms, rostral migratory stream; st, striatum; v, ventricle. Scale bars, 100 μm. (B and C) Expression of PSA-NCAM and GFRα1 in the RMS of newborn mice. Arrows indicate the developing RMS. Boxed areas are shown at higher magnification in (C). Scale bar, 200 μm. (D) Morphometric analysis of the RMS in newborn wild-type, Gfra1, and Ret mutant mice. The region measured in equivalent sections of wild-type and mutant animals is indicated in diagram form. The scatter graphs show measurements of RMS width for each individual animal normalized to the mean value in wild-type mice. Bars indicate the mean value in each group. *p = 0.0136, Student's t test. Cell 2003 113, 867-879DOI: (10.1016/S0092-8674(03)00435-5)