Skip to main content

Genetic Labeling of Synapses

  • Protocol
  • First Online:
Neural Tracing Methods

Part of the book series: Neuromethods ((NM,volume 92))

  • 2113 Accesses

Abstract

A major challenge in neuroscience is to unravel how the synaptic contacts between neurons give rise to brain circuits. A number of techniques have been developed to visualize the synaptic organization of neurons. In this chapter, we focus on genetic methods to mark specific types of synapses so that synaptic sites can be visualized throughout the entire dendritic or axonal arbor of single neurons. Genetic synaptic labeling can be achieved by cell-type-specific viral or transgenic delivery of synaptic proteins tagged by fluorescent proteins. Sparse genetic labeling of neurons permits semiautomated quantification of the distribution and densities of selected types of synapses in segregated domains of the axonal and dendritic trees. These approaches can reduce the complexity and ambiguity of attributing synaptic sites to unravel principles of the synaptic organization of identified neuronal types in the circuit.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 89.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 119.00
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Price JL, Powell TP (1970) The synaptology of the granule cells of the olfactory bulb. J Cell Sci 7:125–155

    CAS  PubMed  Google Scholar 

  2. Woolf TB, Shepherd GM, Greer CA (1991) Serial reconstructions of granule cell spines in the mammalian olfactory bulb. Synapse (NY) 7:181–192

    Article  CAS  Google Scholar 

  3. Briggman KL, Helmstaedter M, Denk W (2011) Wiring specificity in the direction-selectivity circuit of the retina. Nature 471:183–188

    Article  CAS  PubMed  Google Scholar 

  4. White JG, Southgate E, Thomson JN et al (1986) The structure of the nervous system of the nematode Caenorhabditis elegans. Phil Trans R Soc Lond B Biol Sci 314:1–340

    Article  CAS  Google Scholar 

  5. Mishchenko Y, Hu T, Spacek J et al (2010) Ultrastructural analysis of hippocampal neuropil from the connectomics perspective. Neuron 67:1009–1020

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Kelsch W, Lin C-W, Lois C (2008) Sequential development of synapses in dendritic domains during adult neurogenesis. Proc Natl Acad Sci U S A 105:16803–16808

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Livneh Y, Feinstein N, Klein M et al (2009) Sensory input enhances synaptogenesis of adult-born neurons. J Neurosci 29:86–97

    Article  CAS  PubMed  Google Scholar 

  8. Meyer MP, Smith SJ (2006) Evidence from in vivo imaging that synaptogenesis guides the growth and branching of axonal arbors by two distinct mechanisms. J Neurosci 26:3604–3614

    Article  CAS  PubMed  Google Scholar 

  9. Niell CM, Meyer MP, Smith SJ (2004) In vivo imaging of synapse formation on a growing dendritic arbor. Nat Neurosci 7:254–260

    Article  CAS  PubMed  Google Scholar 

  10. Ebihara T, Kawabata I, Usui S et al (2003) Synchronized formation and remodeling of postsynaptic densities: long-term visualization of hippocampal neurons expressing postsynaptic density proteins tagged with green fluorescent protein. J Neurosci 23:2170–2181

    CAS  PubMed  Google Scholar 

  11. Gray NW, Weimer RM, Bureau I et al (2006) Rapid redistribution of synaptic PSD-95 in the neocortex in vivo. PLoS Biol 4:e370

    Article  PubMed Central  PubMed  Google Scholar 

  12. Sheng M (2001) Molecular organization of the postsynaptic specialization. Proc Natl Acad Sci U S A 98:7058–7061

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Washbourne P, Bennett JE, McAllister AK (2002) Rapid recruitment of NMDA receptor transport packets to nascent synapses. Nat Neurosci 5:751–759

    CAS  PubMed  Google Scholar 

  14. Südhof TC, Jahn R (1991) Proteins of synaptic vesicles involved in exocytosis and membrane recycling. Neuron 6:665–677

    Article  PubMed  Google Scholar 

  15. Shu F, Ohno K, Wang T et al (2001) Developmental changes in PSD-95 and Narp mRNAs in the rat olfactory bulb. Dev Brain Res 132:91–95

    Article  CAS  Google Scholar 

  16. Sassoé-Pognetto M, Utvik JK, Camoletto P et al (2003) Organization of postsynaptic density proteins and glutamate receptors in axodendritic and dendrodendritic synapses of the rat olfactory bulb. J Comp Neurol 463:237–248

    Article  PubMed  Google Scholar 

  17. El-Husseini AE, Schnell E, Chetkovich DM et al (2000) PSD-95 involvement in maturation of excitatory synapses. Science (NY) 290:1364–1368

    CAS  Google Scholar 

  18. Kelsch W, Li Z, Eliava M et al (2012) GluN2B-containing NMDA receptors promote wiring of adult-born neurons into olfactory bulb circuits. J Neurosci 32:12603–12611

    Article  CAS  PubMed  Google Scholar 

  19. Chen JL, Villa KL, Cha JW et al (2012) Clustered dynamics of inhibitory synapses and dendritic spines in the adult neocortex. Neuron 74:361–373

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Chen L, Wang H, Vicini S et al (2000) The γ-aminobutyric acid type A (GABAA) receptor-associated protein (GABARAP) promotes GABAA receptor clustering and modulates the channel kinetics. Proc Natl Acad Sci 97:11557–11562

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Jacob TC, Bogdanov YD, Magnus C et al (2005) Gephyrin regulates the cell surface dynamics of synaptic GABAA receptors. J Neurosci 25:10469–10478

    Article  CAS  PubMed  Google Scholar 

  22. Pennuto M, Bonanomi D, Benfenati F et al (2003) Synaptophysin I controls the targeting of VAMP2/Synaptobrevin II to synaptic vesicles. Mol Biol Cell 14:4909–4919

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Valtorta F, Pennuto M, Bonanomi D et al (2004) Synaptophysin: leading actor or walk-on role in synaptic vesicle exocytosis? BioEssays News Rev Mol Cell Dev Biol 26:445–453

    Article  CAS  Google Scholar 

  24. Nakata T, Terada S, Hirokawa N (1998) Visualization of the dynamics of synaptic vesicle and plasma membrane proteins in living axons. J Cell Biol 140:659–674

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Li Z, Murthy VN (2001) Visualizing postendocytic traffic of synaptic vesicles at hippocampal synapses. Neuron 31:593–605

    Article  CAS  PubMed  Google Scholar 

  26. Kaether C, Skehel P, Dotti CG (2000) Axonal membrane proteins are transported in distinct carriers: a two-color video microscopy study in cultured hippocampal neurons. Mol Biol Cell 11:1213–1224

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Bergmann M, Lahr G, Mayerhofer A et al (1991) Expression of synaptophysin during the prenatal development of the rat spinal cord: correlation with basic differentiation processes of neurons. Neuroscience 42:569–582

    Article  CAS  PubMed  Google Scholar 

  28. Wickersham IR, Lyon DC, Barnard RJO et al (2007) Monosynaptic restriction of transsynaptic tracing from single, genetically targeted neurons. Neuron 53:639–647

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Wickersham IR, Feinberg EH (2012) New technologies for imaging synaptic partners. Curr Opin Neurobiol 22:121–127

    Article  CAS  PubMed  Google Scholar 

  30. Scott CA, Rossiter JP, Andrew RD et al (2008) Structural abnormalities in neurons are sufficient to explain the clinical disease and fatal outcome of experimental rabies in yellow fluorescent protein-expressing transgenic mice. J Virol 82:513–521

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  31. Marshel JH, Mori T, Nielsen KJ et al (2010) Targeting single neuronal networks for gene expression and cell labeling in vivo. Neuron 67:562–574

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  32. Feinberg EH, Vanhoven MK, Bendesky A et al (2008) GFP reconstitution across synaptic partners (GRASP) defines cell contacts and synapses in living nervous systems. Neuron 57:353–363

    Article  CAS  PubMed  Google Scholar 

  33. Cabantous S, Terwilliger TC, Waldo GS (2005) Protein tagging and detection with engineered self-assembling fragments of green fluorescent protein. Nat Biotechnol 23:102–107

    Article  CAS  PubMed  Google Scholar 

  34. Park J, Knezevich PL, Wung W et al (2011) A conserved juxtacrine signal regulates synaptic partner recognition in Caenorhabditis elegans. Neural Dev 6:28

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  35. Kim J, Zhao T, Petralia RS et al (2012) mGRASP enables mapping mammalian synaptic connectivity with light microscopy. Nat Methods 9:96–102

    Article  CAS  Google Scholar 

  36. Yamagata M, Sanes JR (2012) Transgenic strategy for identifying synaptic connections in mice by fluorescence complementation (GRASP). Front Mol Neurosci 5:18

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  37. Micheva KD, Smith SJ (2007) Array tomography: a new tool for imaging the molecular architecture and ultrastructure of neural circuits. Neuron 55:25–36

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  38. Toni N, Teng EM, Bushong EA et al (2007) Synapse formation on neurons born in the adult hippocampus. Nat Neurosci 10:727

    Article  CAS  PubMed  Google Scholar 

  39. Briggman KL, Denk W (2006) Towards neural circuit reconstruction with volume electron microscopy techniques. Curr Opin Neurobiol 16:562–570

    Article  CAS  PubMed  Google Scholar 

  40. Sanes JR (1989) Analysing cell lineage with a recombinant retrovirus. Trends Neurosci 12:21–28

    Article  CAS  PubMed  Google Scholar 

  41. Livneh Y, Mizrahi A (2012) Experience-dependent plasticity of mature adult-born neurons. Nat Neurosci 15:26–28

    Article  CAS  Google Scholar 

  42. Scheiffele P (2003) Cell-cell signaling during synapse formation in the CNS. Annu Rev Neurosci 26:485–508

    Article  CAS  PubMed  Google Scholar 

  43. Helmchen F, Denk W (2005) Deep tissue two-photon microscopy. Nat Methods 2:932–940

    Article  CAS  PubMed  Google Scholar 

  44. Mizrahi A, Crowley JC, Shtoyerman E et al (2004) High-resolution in vivo imaging of hippocampal dendrites and spines. J Neurosci 24:3147–3151

    Article  CAS  PubMed  Google Scholar 

  45. Flusberg BA, Cocker ED, Piyawattanametha W et al (2005) Fiber-optic fluorescence imaging. Nat Methods 2:941–950

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Galimberti I, Gogolla N, Alberi S et al (2006) Long-term rearrangements of hippocampal mossy fiber terminal connectivity in the adult regulated by experience. Neuron 50:749–763

    Article  CAS  PubMed  Google Scholar 

  47. Berdichevsky Y, Sabolek H, Levine JB et al (2009) Microfluidics and multielectrode array-compatible organotypic slice culture method. J Neurosci Methods 178:59–64

    Article  PubMed Central  PubMed  Google Scholar 

  48. Kirov SA, Petrak LJ, Fiala JC et al (2004) Dendritic spines disappear with chilling but proliferate excessively upon rewarming of mature hippocampus. Neuroscience 127:69–80

    Article  CAS  PubMed  Google Scholar 

  49. Donohue DE, Ascoli GA (2011) Automated reconstruction of neuronal morphology: an overview. Brain Res Rev 67:94–102

    Article  PubMed Central  PubMed  Google Scholar 

  50. Brown KM, Barrionuevo G, Canty AJ et al (2011) The DIADEM data sets: representative light microscopy images of neuronal morphology to advance automation of digital reconstructions. Neuroinformatics 9:143–157

    Article  PubMed  Google Scholar 

  51. Hanrahan O, Harris J, Egan C (2011) Advanced microscopy: laser scanning confocal microscopy. In: O’Driscoll L (ed) Gene expression profiling. Humana, Totowa, NJ, pp 169–180

    Chapter  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carlos Lois .

Editor information

Editors and Affiliations

Appendices

Appendix: Gene Delivery of Genetic Synaptic Markers with Retroviruses and Quantification of Synapses

This appendix describes some specific procedures to deliver retroviruses carrying genetically encoded synaptic markers into the brain of rodents. General details about production and titration of lentiviral and retroviral vectors can be found elsewhere.

Injection of Viruses into the Brain

Viral prep: It is critical that the viral suspension is very clean. During the preparation of the viruses, there will be some cellular debris that can be strongly autofluorescent. To eliminate this debris it is useful to centrifuge the viral prep with a 20 % sucrose cushion.

Stereotaxic injection: It is critical to minimize the damage to the brain during injection. In particular, bleeding associated with the injection will cause very high levels of autofluorescence that will make quantification of synapses very difficult. To minimize damage it is useful to use thin borosilicate pipettes pulled to an outer diameter of approx. 15–20 μm. It is not advisable to inject through metallic needles as this will cause severe tissue damage on the injection site. Similarly, it is advisable to inject the viral prep slowly, at a rate of approx. 1 μl over 5 min. Rapid injection can severely damage and distort the tissue. Regarding the timing of imaging after injection, for lentiviral vectors, the expression of the transgene peaks as early as 3 days, but there will likely be some acute damage in the injection area at this early time. Thus, it is advisable to wait at least a week so that the autofluorescence due to damage is resolved before perfusion of the animal.

Acquisition and Analysis of Genetically Labeled Synapses

This section describes the procedure that we optimized to visualize the synaptic organization of single genetically labeled neurons, which can be easily modified for individual experimental needs. The procedure is divided into three main steps, and technical issues are highlighted that are critical in our experience: preparation of the tissue (“Preparation of Tissue”), image acquisition by confocal microscopy (“Image Acquisition”), and semiautomated image analysis (“Quantification of Synaptic Clusters”).

3.1 Preparation of Tissue

  1. 1.

    The protocol is described for small rodents, but can be easily adapted to other species. Animals are transcardially perfused initially with phosphate buffered saline (PBS, 1×) for 10–15 s, followed by 4 % paraformaldehyde (PFA) for 3–5 min. The animal should become rigid within the first 30 s to 1 min of perfusion with PFA. It is optimal to use an overdose of an injectable anesthetic drug (such as Ketamin/Xylazin) and to start perfusion when the heart is still beating. PBS should be infused at a pressure such that the liver becomes pale within 10–15 s and clear PBS flows out of the right atrium after this period. It is equally important to perfuse with relatively low pressure, because at high perfusion pressure, the small capillaries in the brain will break and perfusion will not be homogeneous throughout the brain. PBS should be set to pH 7.0–7.4 and pre-warmed to 32–37 °C to prevent contraction of smaller blood vessels in the brain. Following PBS, perfusion should be immediately switched to room-temperature PFA. Incorrect perfusion leads to delayed fixation with PFA, which results in beaded structures of dendrites and loss of genetically labeled synaptic clusters. After perfusion is complete, the brains are extracted from the skull and post-fixed in 4 % PFA overnight at 4 °C.

  2. 2.

    After preparation of floating sections with a vibratome (e.g., 50 μm sections), tissue can be incubated (overnight at 4 °C) with primary antibody raised against the fluorescent protein tagged to the synaptic marker. The following day sections are rinsed in PBS and stained with a secondary fluorescent antibody for two hours at room temperature. Sections are then washed with PBS and mounted with an aqueous mounting medium that preserved fluorescent molecules. This procedure allows for the visualization of the neuronal tree and to attribute synaptic clusters to a neuron and specific dendritic domains. Blocking solutions (PBS containing 1 % bovine serum albumin or related serum proteins) for antibody incubation usually contain a detergent, i.e., Triton X-100, to permeabilize the tissue. We keep the procedure and times as constant as possible to avoid introducing additional variability, i.e., by differentially affecting the brightness of the fluorescence of the synaptic clusters.

3.2 Image Acquisition

Neurons expressing synaptic marker proteins can be conveniently imagined using confocal laser scanning microscopy. In most experimental conditions, it is advantageous to image sections that are sparsely labeled, where individual neurons are clearly separated from each other. In this case it is easy to analyze the full dendritic arbor of a single neuron without having to deal with neurites that could belong to neighboring cells. Confocal stacks are acquired at high magnification (with a 60–63× oil immersion objective) to efficiently capture emitted light from the clustered fluorescent proteins. The pixel size should be sufficiently small to obtain high intensity of all the pixels that are grouped in individual synaptic clusters, and to easily distinguish them from the occasionally observed noise that may result in random isolated pixels with high intensity. As most synaptic clusters have a size around 1 μm, we found a cluster size between 0.2 × 0.2 and 0.3 × 0.3 μm2 most reliable for subsequent analysis. Laser excitation intensities should be set to levels that result in little or no obvious bleaching of the clusters. This can be easily tested by imaging the same neuron twice in the same day and comparing the clusters among the two images. A reference section containing neurons with good synaptic cluster intensity should be used to guarantee comparable acquisition conditions over time. The sensitivity of the photomultipliers (PMT) should be set to a level that clusters just saturate but low enough that individual clusters do not become confluent due to overexposure. Similarly, the pinhole size should be kept in the recommended range [51] for the chosen magnification. Once the settings are initially defined with a test sample, the conditions should be kept constant throughout the different imaging session. Upon acquisition of confocal stacks, maximum density projections are prepared for further image analysis. Two-dimensional projections are generally used for analysis, as current version of most image processing software cannot handle 3D data for quantification.

3.3 Quantification of Synaptic Clusters

Analysis of densities and distribution of genetically labeled synapses can be semiautomated. Fully automated analysis is currently limited by the still challenging task for computers to reconstruct neuronal trees due to overlap of labeled neurons and discontinuities in the processes deriving from histological processing and incomplete filling with fluorescent proteins. Thus, reconstruction of processes has to be performed individually or at least be supervised.

Contrary to reconstruction of dendritic trees, analysis of clusters can be fully automatized provided the original image quality has a good signal-to-noise ratio. Signal-to-noise ratio for these experiments means high intensity of fluorescence in the synaptic cluster and low levels of autofluorescent background outside of synaptic sites. Similarly, it is important that there is a low level of fluorescence originating from diffusely distributed XFP in the cells’ processes outside the synapses. Another potential source of “contamination” with artifactual autofluorescent clusters can be due to lipofuscin granules observed in some brain structures and species. The appearance of these autofluorescent granules is difficult to predict. For example, we have found them in mouse olfactory bulb and dentate gyrus, but not in the rat olfactory bulb or mouse neocortex. These autofluorescent artifacts can be easily diagnosed as they are excited by all wavelengths. In contrast, real genetic synaptic markers containing XFPs can only be detected at a specific wavelength (e.g., 550 nm for GFP). In addition, these autofluorescent granules can usually be excluded from analysis as they are mostly present in cell bodies.

Given these considerations the analysis is relatively straightforward using different analysis software packages. We will describe the different steps of analysis and particularly refer to the ImageJ-based MacBiophotonics software (www.macbiophotonics.ca/). Similar procedures can be performed in Metamorph software from Molecular Probes.

Steps:

  1. 1.

    Open maximum density projection (File>Open).

  2. 2.

    Define pixel size for subsequent distance measurements (Analyze>Set scale).

  3. 3.

    Split color channels (Image>Color>Split channels).

  4. 4.

    Choose the channel that displays the fluorescent synaptic clusters.

  5. 5.

    Set inclusive threshold so that only clusters are included (Image>Adjust>Threshold). The threshold value should be kept constant throughout the analysis. Therefore, it proves useful to use a reference as described in the acquisition part to set the threshold.

  6. 6.

    Draw a contour using the freehand selection tool to define a region of interest to measure a specific dendritic domain and exclude neighboring neurons.

  7. 7.

    Perform region measurement (Analyze>Analyze particles). Desired parameter data can be set in the results window (Analyze>Set Measurements) and copied to a data sheet of a given statistics program.

Measure the length of the neuronal processes in the region of interest using the freehand line tool and Analyze>Measure.

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer Science+Business Media New York

About this protocol

Cite this protocol

Lois, C., Kelsch, W. (2015). Genetic Labeling of Synapses. In: Arenkiel, B. (eds) Neural Tracing Methods. Neuromethods, vol 92. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-1963-5_11

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-1963-5_11

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-1962-8

  • Online ISBN: 978-1-4939-1963-5

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics