Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Identification of a Vav2-dependent mechanism for GDNF/Ret control of mesolimbic DAT trafficking

Abstract

Dopamine (DA) homeostasis is essential for a variety of brain activities. Dopamine transporter (DAT)-mediated DA reuptake is one of the most critical mechanisms for normal DA homeostasis. However, the molecular mechanisms underlying the regulation of DAT activity in the brain remain poorly understood. Here we show that the Rho-family guanine nucleotide exchange factor protein Vav2 is required for DAT cell surface expression and transporter activity modulated by glial cell line–derived neurotrophic factor (GDNF) and its cognate receptor Ret. Mice deficient in either Vav2 or Ret displayed elevated DAT activity, which was accompanied by an increase in intracellular DA selectively in the nucleus accumbens. Vav2−/− mice exposed to cocaine showed reduced DAT activity and diminished behavioral cocaine response. Our data demonstrate that Vav2 is a determinant of DAT trafficking in vivo and contributes to the maintenance of DA homeostasis in limbic DA neuron terminals.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Vav2 is a VTA-enriched protein required for DA homeostasis in mesolimbic neuron terminals.
Figure 2: Enhanced surface expression and DAT activity in Vav2/ mice.
Figure 3: Vav2 is a substrate of Ret receptor tyrosine kinase contributing to the regulation of DAT trafficking and function.
Figure 4: GDNF/Ret/Vav2 signaling modulates DAT activity.
Figure 5: Ret interacts with DAT.
Figure 6: Vav2 is required for DAT trafficking in the context of cocaine.
Figure 7: Altered cocaine-induced behavioral sensitization in Vav2−/− mice.

Similar content being viewed by others

References

  1. Jones, S.R. et al. Profound neuronal plasticity in response to inactivation of the dopamine transporter. Proc. Natl. Acad. Sci. USA 95, 4029–4034 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Loder, M.K. & Melikian, H.E. The dopamine transporter constitutively internalizes and recycles in a protein kinase C-regulated manner in stably transfected PC12 cell lines. J. Biol. Chem. 278, 22168–22174 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Gabriel, L.R. et al. Dopamine transporter endocytic trafficking in striatal dopaminergic neurons: differential dependence on dynamin and the actin cytoskeleton. J. Neurosci. 33, 17836–17846 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Torres, G.E. et al. Functional interaction between monoamine plasma membrane transporters and the synaptic PDZ domain-containing protein PICK1. Neuron 30, 121–134 (2001).

    Article  CAS  PubMed  Google Scholar 

  5. Carneiro, A.M. et al. The multiple LIM domain-containing adaptor protein Hic-5 synaptically colocalizes and interacts with the dopamine transporter. J. Neurosci. 22, 7045–7054 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fog, J.U. et al. Calmodulin kinase II interacts with the dopamine transporter C terminus to regulate amphetamine-induced reverse transport. Neuron 51, 417–429 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Cremona, M.L. et al. Flotillin-1 is essential for PKC-triggered endocytosis and membrane microdomain localization of DAT. Nat. Neurosci. 14, 469–477 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Navaroli, D.M. et al. The plasma membrane-associated GTPase Rin interacts with the dopamine transporter and is required for protein kinase C-regulated dopamine transporter trafficking. J. Neurosci. 31, 13758–13770 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Foster, J.D. & Vaughan, R.A. Palmitoylation controls dopamine transporter kinetics, degradation, and protein kinase C-dependent regulation. J. Biol. Chem. 286, 5175–5186 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Cowan, C.W. et al. Vav family GEFs link activated Ephs to endocytosis and axon guidance. Neuron 46, 205–217 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Hale, C.F. et al. Essential role for vav guanine nucleotide exchange factors in brain-derived neurotrophic factor-induced dendritic spine growth and synapse plasticity. J. Neurosci. 31, 12426–12436 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Schuebel, K.E. et al. Isolation and characterization of murine vav2, a member of the vav family of proto-oncogenes. Oncogene 13, 363–371 (1996).

    CAS  PubMed  Google Scholar 

  13. Movilla, N. & Bustelo, X.R. Biological and regulatory properties of Vav-3, a new member of the Vav family of oncoproteins. Mol. Cell. Biol. 19, 7870–7885 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Doody, G.M. et al. Signal transduction through Vav-2 participates in humoral immune responses and B cell maturation. Nat. Immunol. 2, 542–547 (2001).

    Article  CAS  PubMed  Google Scholar 

  15. Quevedo, C., Sauzeau, V., Menacho-Marquez, M., Castro-Castro, A. & Bustelo, X.R. Vav3-deficient mice exhibit a transient delay in cerebellar development. Mol. Biol. Cell 21, 1125–1139 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bustelo, X.R., Ledbetter, J.A. & Barbacid, M. Product of vav proto-oncogene defines a new class of tyrosine protein kinase substrates. Nature 356, 68–71 (1992).

    Article  CAS  PubMed  Google Scholar 

  17. Bustelo, X.R. Regulatory and signaling properties of the Vav family. Mol. Cell. Biol. 20, 1461–1477 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sauzeau, V., Jerkic, M., Lopez-Novoa, J.M. & Bustelo, X.R. Loss of Vav2 proto-oncogene causes tachycardia and cardiovascular disease in mice. Mol. Biol. Cell 18, 943–952 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sauzeau, V. et al. Vav3 proto-oncogene deficiency leads to sympathetic hyperactivity and cardiovascular dysfunction. Nat. Med. 12, 841–845 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Matsushita, N. et al. Dynamics of tyrosine hydroxylase promoter activity during midbrain dopaminergic neuron development. J. Neurochem. 82, 295–304 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Best, J.A., Nijhout, H.F. & Reed, M.C. Homeostatic mechanisms in dopamine synthesis and release: a mathematical model. Theor. Biol. Med. Model. 6, 21 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Kodama, A. et al. Involvement of an SHP-2-Rho small G protein pathway in hepatocyte growth factor/scatter factor-induced cell scattering. Mol. Biol. Cell 11, 2565–2575 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Vaughan, R.A., Huff, R.A., Uhl, G.R. & Kuhar, M.J. Protein kinase C-mediated phosphorylation and functional regulation of dopamine transporters in striatal synaptosomes. J. Biol. Chem. 272, 15541–15546 (1997).

    Article  CAS  PubMed  Google Scholar 

  24. Foster, J., Cervinski, M., Gorentla, B. & Vaughan, R. Regulation of the dopamine transporter by phosphorylation. in Neurotransmitter Transporters 197–214 (Springer, 2006).

  25. Melikian, H.E. & Buckley, K.M. Membrane trafficking regulates the activity of the human dopamine transporter. J. Neurosci. 19, 7699–7710 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Melikian, H.E. Neurotransmitter transporter trafficking: endocytosis, recycling, and regulation. Pharmacol. Ther. 104, 17–27 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Daniels, G.M. & Amara, S.G. Regulated trafficking of the human dopamine transporter. Clathrin-mediated internalization and lysosomal degradation in response to phorbol esters. J. Biol. Chem. 274, 35794–35801 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Foster, J.D. et al. Dopamine transporter phosphorylation site threonine 53 regulates substrate reuptake and amphetamine-stimulated efflux. J. Biol. Chem. 287, 29702–29712 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Littrell, O.M. et al. Enhanced dopamine transporter activity in middle-aged Gdnf heterozygous mice. Neurobiol. Aging 33, 427. e1–427.e14 (2012).

    Article  PubMed  CAS  Google Scholar 

  30. Airaksinen, M.S. & Saarma, M. The GDNF family: signaling, biological functions and therapeutic value. Nat. Rev. Neurosci. 3, 383–394 (2002).

    Article  CAS  PubMed  Google Scholar 

  31. Vaughan, R.A., Huff, R.A., Uhl, G.R. & Kuhar, M.J. Protein kinase C-mediated phosphorylation and functional regulation of dopamine transporters in striatal synaptosomes. J. Biol. Chem. 272, 15541–15546 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Coulpier, M., Anders, J. & Ibanez, C.F. Coordinated activation of autophosphorylation sites in the RET receptor tyrosine kinase: importance of tyrosine 1062 for GDNF mediated neuronal differentiation and survival. J. Biol. Chem. 277, 1991–1999 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Melamed, I., Patel, H., Brodie, C. & Gelfand, E.W. Activation of Vav and Ras through the nerve growth factor and B cell receptors by different kinases. Cell. Immunol. 191, 83–89 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Little, K.Y., Elmer, L.W., Zhong, H., Scheys, J.O. & Zhang, L. Cocaine induction of dopamine transporter trafficking to the plasma membrane. Mol. Pharmacol. 61, 436–445 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Daws, L.C. et al. Cocaine increases dopamine uptake and cell surface expression of dopamine transporters. Biochem. Biophys. Res. Commun. 290, 1545–1550 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Jain, S. et al. RET is dispensable for maintenance of midbrain dopaminergic neurons in adult mice. J. Neurosci. 26, 11230–11238 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hunter, S.G. et al. Essential role of Vav family guanine nucleotide exchange factors in EphA receptor-mediated angiogenesis. Mol. Cell. Biol. 26, 4830–4842 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Encinas, M. et al. c-Src is required for glial cell line-derived neurotrophic factor (GDNF) family ligand-mediated neuronal survival via a phosphatidylinositol-3 kinase (PI-3K)-dependent pathway. J. Neurosci. 21, 1464–1472 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Adkins, E.M. et al. Membrane mobility and microdomain association of the dopamine transporter studied with fluorescence correlation spectroscopy and fluorescence recovery after photobleaching. Biochemistry 46, 10484–10497 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Shamah, S.M. et al. EphA receptors regulate growth cone dynamics through the novel guanine nucleotide exchange factor ephexin. Cell 105, 233–244 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Crits-Christoph, P. et al. Dopamine transporter levels in cocaine dependent subjects. Drug Alcohol Depend. 98, 70–76 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mandt, B.H. & Zahniser, N.R. Low and high cocaine locomotor responding male Sprague-Dawley rats differ in rapid cocaine-induced regulation of striatal dopamine transporter function. Neuropharmacology 58, 605–612 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Cai, L. et al. Ethanol-induced neurodegeneration in NRSF/REST neuronal conditional knockout mice. Neuroscience 181, 196–205 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Zhou, Q., Li, J., Wang, H., Yin, Y. & Zhou, J. Identification of nigral dopaminergic neuron-enriched genes in adult rats. Neurobiol. Aging 32, 313–326 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Shu, F. et al. Functional characterization of human PFTK1 as a cyclin-dependent kinase. Proc. Natl. Acad. Sci. USA 104, 9248–9253 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Di Cesare, S. The Guanine Nucleotide Exchanger Vav2 Interacts with c-ErbB-2 and Induces Alveolar Morphogenesis of Mammary Epithelial Cells. PhD Thesis, der Humboldt-Universität zu Berlin (2001).

  47. Shao, W. et al. Suppression of neuroinflammation by astrocytic dopamine D2 receptors via αB-crystallin. Nature 494, 90–94 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Tamás, P., Solti, Z. & Buday, L. Membrane-targeting is critical for the phosphorylation of Vav2 by activated EGF receptor. Cell. Signal. 13, 475–481 (2001).

    Article  PubMed  Google Scholar 

  49. Sindrilaru, A. et al. Wound healing defect of Vav3−/− mice due to impaired β2-integrin-dependent macrophage phagocytosis of apoptotic neutrophils. Blood 113, 5266–5276 (2009).

    Article  CAS  PubMed  Google Scholar 

  50. Fetchko, M. & Stagljar, I. Application of the split-ubiquitin membrane yeast two-hybrid system to investigate membrane protein interactions. Methods 32, 349–362 (2004).

    Article  CAS  PubMed  Google Scholar 

  51. Khanna, N., Fang, Y., Yoon, M.S. & Chen, J. XPLN is an endogenous inhibitor of mTORC2. Proc. Natl. Acad. Sci. USA 110, 15979–15984 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hallett, P.J., Collins, T.L., Standaert, D.G. & Dunah, A.W. Biochemical fractionation of brain tissue for studies of receptor distribution and trafficking. Curr. Protoc. Neurosci. Ch. 1, Unit 1.16 (2008).

  53. Guo, H. et al. Apomorphine induces trophic factors that support fetal rat mesencephalic dopaminergic neurons in cultures. Eur. J. Neurosci. 16, 1861–1870 (2002).

    Article  PubMed  Google Scholar 

  54. Zhao-Shea, R. et al. Dopamine D2-receptor activation elicits akinesia, rigidity, catalepsy, and tremor in mice expressing hypersensitive α4 nicotinic receptors via a cholinergic-dependent mechanism. FASEB J. 24, 49–57 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Paxinos, G. & Franklin, K.B.J. The Mouse Brain in Stereotaxic Coordinates (Academic, San Diego, 2001).

  56. Fernandez-Zapico, M.E. et al. Ectopic expression of VAV1 reveals an unexpected role in pancreatic cancer tumorigenesis. Cancer Cell 7, 39–49 (2005).

    Article  CAS  PubMed  Google Scholar 

  57. Ventura, A. et al. Cre-lox-regulated conditional RNA interference from transgenes. Proc. Natl. Acad. Sci. USA 101, 10380–10385 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Auricchio, A., Hildinger, M., O'Connor, E., Gao, G.P. & Wilson, J.M. Isolation of highly infectious and pure adeno-associated virus type 2 vectors with a single-step gravity-flow column. Hum. Gene Ther. 12, 71–76 (2001).

    Article  CAS  PubMed  Google Scholar 

  59. Gehrke, B. J. et al. The effect of neurotoxic doses of methamphetamine on methamphetamine-conditioned place preference in rats. Psychopharmacology (Berl.). 166, 249–257 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Dai, J.X. et al. Enhanced contextual fear memory in central serotonin-deficient mice. Proc. Natl. Acad. Sci. USA 105, 11981–11986 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pothion, S., Bizot, J.C., Trovero, F. & Belzung, C. Strain differences in sucrose preference and in the consequences of unpredictable chronic mild stress. Behav. Brain Res. 155, 135–146 (2004).

    Article  PubMed  Google Scholar 

  62. Zhu, X. et al. MSC p43 required for axonal development in motor neurons. Proc. Natl. Acad. Sci. USA 106, 15944–15949 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Willott, J.F. et al. Acoustic startle and prepulse inhibition in 40 inbred strains of mice. Behav. Neurosci. 117, 716–727 (2003).

    Article  PubMed  Google Scholar 

  64. Garfield, A.S. et al. A neural basis for melanocortin-4 receptor-regulated appetite. Nat. Neurosci. 18, 863–871 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank X.C. Zhen (Soochow University) for providing the DAT cDNA, C. He (Second Military Medical University, China) for human RET cDNA, H.L. Hu and Z.L. Qiu (Institute of Neuroscience, Chinese Academy of Sciences) for the AAV virus constructs, L.P. Cheng (Institute of Neuroscience, Chinese Academy of Sciences) for the Ret mutant mice and the Riken BioResource Center for the TH-GFP transgenic mice. We also thank the Optical Imaging Center of the Institute of Neuroscience, Chinese Academy of Sciences, for technical support in confocal microscopy and the Behavioral Core of ION for providing assistance. This work was supported by grants from the National Key Basic Research Program of China (2015CB553500, 2011CB504102), Natural Science Foundation of China (31123002, 31321091 and 30621130075), Beijing Institute for Brain Disorders (BIBD-PXM2013_014226_07_000084) and Strategic Priority Research Program (B) of the Chinese Academy of Sciences (XDB01020300). X.R.B.'s work has been funded by the Spanish Ministry of Economy and Competitiveness (SAF2009-07172, SAF2012-3171, RD06/0020/0001 and RD12/0036/0002), the Castilla-León Autonomous Government (CSI101U13) and Spanish Association of Science Communication.

Author information

Authors and Affiliations

Authors

Contributions

S.Z. conducted most of the biochemical experiments and the data analysis, and C.Z. conducted most of the in vivo experiments and the data analysis. Y.W. conducted some of the in vivo and in vitro experiments. Q.Y. contributed to the in vitro experiments. A.S. and J.W. contributed to mutant and protein interaction analysis. T.W. contributed to the CPP assay. Y.Y. conducted in situ hybridization; Y.Y., Y.L. and J.H. conducted genotyping; X.Z. contributed to in vitro experiments. X.G., G.Z. and X.W. provided advice; X.R.B. provided Vav2/ and Vav3/ mice and Vav2 and 3 constructs; and J.Z. supervised the project and wrote the manuscript.

Corresponding author

Correspondence to Jiawei Zhou.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Vav2, but not Vav3, is preferentially expressed in the VTA of adult mice and is required for the maintenance of dopamine homeostasis.

a, Immunoblotting reveals the Vav2 expression levels in the ventral tagmental area (VTA) is significantly higher than in the substantia nigra (SN) of adult brain. b, Quantitative data of a. (n = 4); t(6)= 6.317, *P = 0.0007. c. Western analysis of brain tissue lysates from adult mouse brain shows that Vav3 is ubiquitously expressed in major brain regions. The experiments were repeated at least twice. d-f, Measurement of DA, 5-HT and their metabolites levels were performed in the olfactory bulb (OB) of Vav2 mutants (d), nucleus accumbens (NAc) (e) and dorsal striatum (dSTR) (f) of Vav3 mutant and wild-type mice. d, n = 3 per group; unpaired two-tailed t-test, t(4) = 0.8303, P = 0.4530 for DA. e, f, n = 10 for WT, n = 8 for Vav3 mutants. unpaired two-tailed t-test. e, t(16) = 1.196, P = 0.2490 for DA. f, t(16) = 0.02254, P = 0.9823 for DA. g-h, Tyrosine hydroxylase immunosignal density was measured in the NAc (g) and dSTR (h) of Vav2 mutant and WT mice. (g, h, n = 4). unpaired two-tailed t-test. g, t(46) = 1.913, P = 0.0620. h, t(60) = 1.108, P = 0.2721. i, Western blotting shows that AAV-mediated overexpression of Vav2 following injection in the VTA or SN, validating the transfection efficiency in vivo. j, [3H]DA uptake assays were performed in the NAc or dSTR synapotosome preparation from Vav3-null and wild-type. (n = 3 mice per group). unpaired two-tailed t-test. t(4) = 0.8844, P = 0.4264 for NAc, t(4) = 0.2033, P = 0.8488 for dSTR. k, Immunoblotting reveals that the levels of Vav2 expression were significantly down-regulated 48, 72 h following transfection with shVav2 #2 in N2a cells. Error bars represent s.e.m. i, k are cropped blot images from their corresponding full-length blots presented in Supplementary Figure 12.

Supplementary Figure 2 DA biosynthesis and degradation in the NAc of Vav2 mutant mice are not significantly altered.

a, Western analysis of VTA tissue lysates from adult Vav2-null mouse brain, in which rabbit monoclonal anti-Vav2 antibody was used, shows the absence of Vav2 protein. The experiments were repeated at least twice. b, Quantitative data of a. There is no significant difference in the levels of TH and VMAT2 protein expression between genotypes. unpaired two-tailed t-test. (n = 3 per group); t(4) = 1.460, P = 0.2182. *P = 0.2182. c-e, Western analysis of phosphor-TH (ser 40) expression in the NAc (c, d) or VTA (c, e) of Vav2 mutant and WT mice. d, e, Quantitative data of (c). unpaired two-tailed t-test. (d, n = 6 mice per group, t(10) = 0.8557, P = 0.4122, e, n = 5 mice per group, t(8) = 0.3065, P = 0.7670). f, The accumulation of l-dopa in the NAc was measured in the animals exposed to NSD-1015, an aromatic acid decarboxylase (AADC) inhibitor, using HPLC. Lower levels of L-dopa were seen in the NAc of Vav2 mutant mice, suggesting that TH enzymatic activity was markedly inhibited. unpaired two-tailed t-test. (n = 5 mice per group); t(8) = 2.400, *P = 0.0432. g, h, qPCR analysis of mRNA expression of MAO-A (g) and MAO-B (h), which are responsible for enzymatic degradation of 5-HT, NE and DA in the VTA and NAc of Vav2 mutant and WT mice. unpaired two-tailed t-test. (n = 3 mice). (g, t(4) = 0.3998, P = 0.7097 for VTA, t(4) = 0.06329, P = 0.9526 for NAc; h, t(4) = 2.946, P = 0.0421 for VTA, t(4) = 2.382, P = 0.0758 for NAc). i, qPCR analysis of MAO-A and Vav2 mRNA expression in MN9D dopaminergic cell line transfected with shVav2 #2. unpaired two-tailed t-test. (n = 3 independent experiments). t(4) = 35.55, *P < 0.0001 for Vav2, t(4) = 0.0, *P > 0.9999 for MAO-A. j, Measurement of enzymatic activities of MAO-A were performed in the VTA and NAc of Vav2 mutant and WT mice. unpaired two-tailed t-test. (n = 3 mice per group). t(4) = 0.1290, P = 0.9036 for VTA, t(4) = 1.202, P =0.2958 for NAc. k, Immunoblotting reveals that the levels of VMAT2 expression were significantly altered in the striatum of Vav2 mutant mice compared with wild-type counterparts. l, Quantitative data shown in k. unpaired two-tailed t-test. (n = 6 mice per group). t(10) = 0.02854, P = 0.9778. m, Full-length gel showing immunoblotting analysis of phosphor-Thr53-DAT expression. Their expression levels were not significantly changed between genotypes in various brain regions. n, Quantitative data shown in l. unpaired two-tailed t-test. (n = 5 mice). t(8) = 1.290, P = 0.2331 for NAc, t(8) = 1.849, P = 0.1016 for dSTR, t(8) = 1.146, P = 0.2847 for VTA. Error bars represent s.e.m. n.s., not significant. k shows cropped blot images from their corresponding full-length blot presented in Supplementary Figure 12.

Supplementary Figure 3 Quantification of total DAT expression in synaptosomal preparations derived from Vav2–/– mice and WT control under various experimental conditions.

a-c, correspond to Fig. 2f-i. (a, n = 4). Two-way ANOVA with Bonferroni's multiple comparisons test. F(1, 12) = 1.067, P > 0.9999 for Nac, P = 0.6297 for dSTR. (b, c, n = 3). One-way ANOVA with Bonferroni's multiple comparisons test. b, F(2, 6) = 0.2230, P = 0.7976 between control and shVav2 #1, P = 0.8972 between control and rescue. c, F(2, 6) = 0.1670, P > 0.9999 between control and RFP-Vav2, P > 0.9999 between control and Y172F. d, corresponds to Fig. 3b. (n = 3). Two-way ANOVA with Bonferroni's multiple comparisons test. F(1, 8) = 0.1616. P > 0.9999 between NAc WT and NAc Ret+/, P > 0.9999 between dSTR WT and dSTR Ret+/. e, f, correspond to Fig. 4e, g. (n = 3). e, unpaired two-tailed t-test. (f, n = 2). Two-way ANOVA with Bonferroni's multiple comparisons test. F(1, 4) = 0.2292, P > 0.9999 between scrambled control and scrambled GDNF, P > 0.9999 between shVav2 control and shVav2 GDNF. g, h, correspond to Fig. 6b, c. (g, n = 2). Two-way ANOVA with Bonferroni's multiple comparisons test. F(1, 4) = 0.5856, P > 0.9999 between WT saline and WT cocaine, P> 0.9999 between Vav2 mutant saline and Vav2 mutant cocaine. (h, n = 4). Two-way ANOVA with Sidak's multiple comparisons test. F(1, 12) = 0.3031. P = 0.9998 between WT saline and WT cocaine, P = 0.9983 between Vav2 mutant saline and Vav2 mutant cocaine. n.s. no significance. The data are expressed as ratios of total DAT to β-actin. Error bars represent s.e.m.

Supplementary Figure 4 Ret+/– mutant mice exhibit no significant alterations in the levels of DA, 5-HT or their metabolites in the olfactory bulb and expression levels of DAT and phospho-Thr53-DAT in the NAc and VTA.

a, The levels of DA, 5-HT and their metabolites were measured in the olfactory bulb of Ret+/- and wild-type mice. The olfactory bulb (OB) is one of brain regions enriched in dopaminergic neurons. unpaired two-tailed t-test. (n=5 mice per group). t(8) = 0.8544. P = 0.4177 for DA. b, Western blotting reveals that the levels of phosphor-Thr53-DAT expression are not significantly altered in the NAc and VTA between genotypes. b shows cropped blot images from their corresponding full-length blots presented in Supplementary Figure 12. c, Quantitative data of b. unpaired two-tailed t-test. (n = 3 mice per group). t(4) = 1.092, P = 0.3364 for NAc, t(4) = 1.672, P = 0.1698 for VTA. Error bars represent s.e.m.

Supplementary Figure 5 Vav2, but not Vav3, strongly binds to Ret.

a, Y2H assays reveal that Vav2 strongly binds to Ret intracellular domain (ICD). In contrast, Vav3 weakly interacts with Ret ICD. b, Co-IP assays show that Ret-flag is immunoprecipitated with Myc-Vav2. c, [3H]DA uptake assays reveal that overexpression of TrkA in HEK293 cells stably expressing YFP-DAT does not significantly alter DAT activity compared to control. (n = 4). One-way ANOVA with Bonferroni's multiple comparisons test: F(2, 9) = 40.37, P < 0.0001; post hoc: t(9) = 6.610, *P = 0.0002. Error bars represent s.e.m. d, Western blotting reveals that treatment of N2a cells with 100 ng/ml GDNF increases the phosphorylation of Ret at Tyr905 site. e, Western blotting reveals that overexpression of Flag-tagged Ret elevates the levels of phosphor-Ret Tyr905 in HEK293T cells. These experiments were repeated at least three times. b, d, e are cropped blot images from their corresponding full-length blots presented in Supplementary Figure 12.

Supplementary Figure 6 Involvement of PKC in the Vav2-mediated DAT activity.

a, shVav2 #2-induced Vav2 knockdown attenuates PMA-induced reduction of DAT activity in N2a cells stably expressing RFP-DAT and GFRa1. Two-way ANOVA with Bonferroni‘s post-hoc test was performed. (n = 10). Main effect of genotype (F(1, 36) = 38.06, P <0.0001), treatment (F(1, 36) = 58.23, P <0.0001) and interaction (F(1, 36) = 4.376, P = 0.0436); post hoc: t(36) = 6.875, P < 0.0001 for Scrambled group, t(36) = 3.917, P = 0.0008 for shVav2 group. *P < 0.05. b, Treatment of N2a cells stably expressing RFP-DAT and GFRa1 with either PMA (1 mm, 30 min) or GDNF (100 ng/ml, 30 min) significantly down-regulate DAT-mediated DA uptake. These effects could not be significantly attenuated by pretreatment with PKC inhibitor GF-109203X (1 mM, 30 min). One way ANOVA with Tukey's multiple comparisons test was performed. (n = 11). F(3, 40) = 28.13, P<0.0001; q(40)=12.58, P < 0.0001 between control and PMA, q(40) = 8.185, P < 0.0001 between Control and GDNF, q(40) = 3.219, P = 0.1208 between GDNF and GDNF+GF. *P <0.05. Error bars represent s.e.m.

Supplementary Figure 7 Characterization of interaction among Vav2, DAT and Ret.

a, Colony growth assay on the SD-His medium showed the DAT-Cub-LexA-VP16 and Ret-NubG were expressed with no self-activation of the histidine reporter gene expression. b, Co-IP assays show that treatment with PP2 does not alter their interaction between Ret and DAT in HEK293 cells. c, Immunoblotting reveals that activation of Ret signaling by overexpression of Ret-Flag in HEK293 cells does not cause significant alteration in the levels of phosphor-Thr53-DAT. These experiments were repeated at least twice. c shows cropped blot images from their corresponding full-length blots presented in Supplementary Figure 12.

Supplementary Figure 8 Altered cocaine-induced behavioral response in Vav2–/– mice.

a, Measurement of extracellular DA concentrations in the NAc of WT and Vav2 mutant mice using in vivo microdialysis combined with HPLC analysis. The mice received single injection of cocaine (20 mg/kg, i.p.) as indicated by an arrow. The effects of cocaine on the dopamine output were presented as relative to the baseline (the average concentration of four consecutive stable samples defined as 100%). (n = 8 for WT, n = 11 for Vav2 mutants). Two-way ANOVA test. F(7, 136) = 0.5922, P = 0.7614. b, The spontaneous activities are recorded during a 24-h period. There is no alteration between genotypes (n = 5 mice for WT, n = 7 mice for mutants). Two-way ANOVA test. (no effect of genotype (F(1, 170) = 0.02575, P = 0.8727) and interaction (F(16, 170) = 1.017, P = 0.4410)). c, d, The locomotor activity of Vav2 mutants administrated with nomifensine (10 mg/kg, i.p) is altered compared with WT mice (n = 13 mice for WT, n = 14 mice for mutants). c, Two-way ANOVA with Bonferroni’s post test. main effect of genotype (F(1, 375) = 9.091, P = 0.0027) and interaction (F(14, 375) = 3.072, P = 0.0002); d, F(1, 50) = 8.137, *P = 0.0003. e, The performance in sucrose preference test is not altered in Vav2 mutants. (n = 9 mice per group). unpaired two-tailed t-test, t(16) = 0.1830, P = 0.8571. f, Recent memory of Vav2 mutant and WT mice is expressed as the percentage of time spent in freezing during contextual fear conditioning test. (n = 16 mice per group). unpaired two-tailed t-test, t(30) = 1.114, P = 0.2739. g, The performance in CPP is not significantly altered in Vav3 mutants (n = 8 mice per group), t(14) = 1.304, P = 0.2132. h, i. The response to acoustic startle stimulation (h) and prepulse inhibition (i) is not significantly altered in Vav2 mutant mice compared with control. (n = 17 mice for WT, n = 19 mice for Vav2 mutant). h, unpaired two-tailed t-test, t(34) = 1.530, P = 0.1354. i, Two-way ANOVA test. (no effect of genotype (F(1, 102) = 0.4790, P = 0.4904) and interaction (F(2, 102) = 0.06487, P = 0.9372)). Error bars represent s.e.m.

Supplementary Figure 9 A proposed model for how Vav2 regulates GDNF/Ret signaling, thereby modulating DAT internalization and DA homeostasis, in the mesolimbic DA neuron terminals of adult mice under physiological conditions.

Our data also suggest that cocaine could influence the trafficking of DAT in a way that intersects with RET/Vav2, given that the conformation of DAT when bound to cocaine disables the DAT/RET interaction, with no effect if Vav2 is absent. Moreover, elevated extracellular DA produced by cocaine acting normally to produce RET dephosphorylation and reduced Vav2 associations, and which is of no significance in the context of a Vav2 knockout.

Supplementary Figure 10 Full-length images of cropped immunoblots presented in Figures 1a,c,e, 2c–f,h,i and 3b,g,h,j.

Supplementary Figure 11 Full-length images of cropped immunoblots presented in Figures 4b,c,e,f, 5b,c,e and 6a,f.

Supplementary Figure 12 Full-length images of cropped immunoblots presented in Supplementary Figures 1i,k, 2k, 4b, 5b,d,e and 7c.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, S., Zhao, C., Wu, Y. et al. Identification of a Vav2-dependent mechanism for GDNF/Ret control of mesolimbic DAT trafficking. Nat Neurosci 18, 1084–1093 (2015). https://doi.org/10.1038/nn.4060

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4060

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing