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 cellular node for motor control pathways

Abstract

The rich behavioral repertoire of animals is encoded in the CNS as a set of motorneuron activation patterns, also called 'motor synergies'. However, the neurons that orchestrate these motor programs as well as their cellular properties and connectivity are poorly understood. Here we identify a population of molecularly defined motor synergy encoder (MSE) neurons in the mouse spinal cord that may represent a central node in neural pathways for voluntary and reflexive movement. This population receives direct inputs from the motor cortex and sensory pathways and, in turn, has monosynaptic outputs to spinal motorneurons. Optical stimulation of MSE neurons drove reliable patterns of activity in multiple motor groups, and we found that the evoked motor patterns varied on the basis of the rostrocaudal location of the stimulated MSE. We speculate that these neurons comprise a cellular network for encoding coordinated motor output programs.

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: Labeling of first-order spinal neurons targeting gastrocnemius motorneurons.
Figure 2: Motorneuron responses to optical stimulation of medial deep dorsal horn premotor neurons or of nonspecific ventral interneurons.
Figure 3: Molecular markers Tcfap2β and Satb1/2 identify medial deep dorsal horn neurons.
Figure 4: Molecular markers Tcfap2β and Satb1/2 identify medial deep dorsal horn premotor neurons.
Figure 5: MSE neurons receive sensory and corticospinal inputs.
Figure 6: Rostral-caudal position of MSE neurons maps to distinct functional outputs.

Similar content being viewed by others

References

  1. Tresch, M.C., Saltiel, P., d'Avella, A. & Bizzi, E. Coordination and localization in spinal motor systems. Brain Res. Rev. 40, 66–79 (2002).

    Article  PubMed  Google Scholar 

  2. Bizzi, E., Cheung, V.C., d'Avella, A., Saltiel, P. & Tresch, M. Combining modules for movement. Brain Res. Brain Res. Rev. 57, 125–133 (2008).

    Article  CAS  Google Scholar 

  3. Duysens, J., De Groote, F. & Jonkers, I. The flexion synergy, mother of all synergies and father of new models of gait. Front. Comput. Neurosci. 7, 14 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Sherrington, C.S. The Integrative Action of the Nervous System (C. Scribner's Sons, 1906).

  5. Lundberg, A. Supraspinal control of transmission in reflex paths to motoneurones and primary afferents. Prog. Brain Res. 12, 197–221 (1964).

    Article  CAS  PubMed  Google Scholar 

  6. Leyton, S. & Sherrington, C.S. Observations on the excitable cortex of the chimpanzee, orang-utan and gorilla. Q. J. Exp. Physiol. 11, 135–222 (1917).

    Article  Google Scholar 

  7. Graziano, M.S., Taylor, C.S. & Moore, T. Complex movements evoked by microstimulation of precentral cortex. Neuron 34, 841–851 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. McHanwell, S. & Biscoe, T.J. The localization of motoneurons supplying the hindlimb muscles of the mouse. Phil. Trans. R. Soc. Lond. B 293, 477–508 (1981).

    Article  CAS  Google Scholar 

  9. Nicolopoulos-Stournaras, S. & Iles, J.F. Motor neuron columns in the lumbar spinal cord of the rat. J. Comp. Neurol. 217, 75–85 (1983).

    Article  CAS  PubMed  Google Scholar 

  10. de Leon, R., Hodgson, J.A., Roy, R.R. & Edgerton, V.R. Extensor- and flexor-like modulation within motor pools of the rat hindlimb during treadmill locomotion and swimming. Brain Res. 654, 241–250 (1994).

    Article  CAS  PubMed  Google Scholar 

  11. Wickersham, I.R., Finke, S., Conzelmann, K.K. & Callaway, E.M. Retrograde neuronal tracing with a deletion-mutant rabies virus. Nat. Methods 4, 47–49 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Stepien, A.E., Tripodi, M. & Arber, S. Monosynaptic rabies virus reveals premotor network organization and synaptic specificity of cholinergic partition cells. Neuron 68, 456–472 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Coulon, P., Bras, H. & Vinay, L. Characterization of last-order premotor interneurons by transneuronal tracing with rabies virus in the neonatal mouse spinal cord. J. Comp. Neurol. 519, 3470–3487 (2011).

    Article  PubMed  Google Scholar 

  14. Ruigrok, T.J., Pijpers, A., Goedknegt-Sabel, E. & Coulon, P. Multiple cerebellar zones are involved in the control of individual muscles: a retrograde transneuronal tracing study with rabies virus in the rat. Eur. J. Neurosci. 28, 181–200 (2008).

    Article  PubMed  Google Scholar 

  15. Scheibel, M.E. & Scheibel, A.B. Terminal axonal patterns in cat spinal cord. II. The dorsal horn. Brain Res. 9, 32–58 (1968).

    Article  CAS  PubMed  Google Scholar 

  16. Sherrington, C.S. & Laslett, E.E. Observations on some spinal reflexes and the interconnection of spinal segments. J. Physiol. (Lond.) 29, 58–96 (1903).

    Article  CAS  Google Scholar 

  17. Sengul, G., Puchalski, R.B. & Watson, C. Cytoarchitecture of the spinal cord of the postnatal (P4) mouse. Anat. Rec. 295, 837–845 (2012).

    Article  Google Scholar 

  18. Harrison, P.J., Jankowska, E. & Zytnicki, D. Lamina VIII interneurones interposed in crossed reflex pathways in the cat. J. Physiol. 371, 147–166 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hongo, T., Kitazawa, S., Ohki, Y. & Xi, M.C. Functional identification of last-order interneurones of skin reflex pathways in the cat forelimb segments. Brain Res. 505, 167–170 (1989).

    Article  CAS  PubMed  Google Scholar 

  20. Puskar, Z. & Antal, M. Localization of last-order premotor interneurons in the lumbar spinal cord of rats. J. Comp. Neurol. 389, 377–389 (1997).

    Article  CAS  PubMed  Google Scholar 

  21. Asante, C.O. & Martin, J.H. Differential joint-specific corticospinal tract projections within the cervical enlargement. PLoS ONE 8, e74454 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hongo, T., Kitazawa, S., Ohki, Y., Sasaki, M. & Xi, M.C. A physiological and morphological study of premotor interneurones in the cutaneous reflex pathways in cats. Brain Res. 505, 163–166 (1989).

    Article  CAS  PubMed  Google Scholar 

  23. Tresch, M.C. & Bizzi, E. Responses to spinal microstimulation in the chronically spinalized rat and their relationship to spinal systems activated by low threshold cutaneous stimulation. Exp. Brain Res. 129, 401–416 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Osakada, F. et al. New rabies virus variants for monitoring and manipulating activity and gene expression in defined neural circuits. Neuron 71, 617–631 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Clarke, R.W. & Harris, J. The organization of motor responses to noxious stimuli. Brain Res. Brain Res. Rev. 46, 163–172 (2004).

    Article  PubMed  Google Scholar 

  26. Pierrot-Deseilligny, E. & Burke, D. The Circuitry of the Human Spinal Cord. 350–352 (Cambridge University Press, 2012).

  27. Schouenborg, J. & Kalliomaki, J. Functional organization of the nociceptive withdrawal reflexes. I. Activation of hindlimb muscles in the rat. Exp. Brain Res. 83, 67–78 (1990).

    Article  CAS  PubMed  Google Scholar 

  28. Gong, S. et al. A gene expression atlas of the central nervous system based on bacterial artificial chromosomes. Nature 425, 917–925 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Henry, A.M. & Hohmann, J.G. High-resolution gene expression atlases for adult and developing mouse brain and spinal cord. Mammalian Genome 23, 539–549 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. Wildner, H. et al. Genome-wide expression analysis of Ptf1a- and Ascl1-deficient mice reveals new markers for distinct dorsal horn interneuron populations contributing to nociceptive reflex plasticity. J. Neurosci. 33, 7299–7307 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Britanova, O., Akopov, S., Lukyanov, S., Gruss, P. & Tarabykin, V. Novel transcription factor Satb2 interacts with matrix attachment region DNA elements in a tissue-specific manner and demonstrates cell-type-dependent expression in the developing mouse CNS. Eur. J. Neurosci. 21, 658–668 (2005).

    Article  PubMed  Google Scholar 

  32. Alaynick, W.A., Jessell, T.M. & Pfaff, S.L. Snapshot: spinal cord development. Cell. 146, 178 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Harrison, T.C., Ayling, O.G. & Murphy, T.H. Distinct cortical circuit mechanisms for complex forelimb movement and motor map topography. Neuron 74, 397–409 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Ramanathan, D., Conner, J.M. & Tuszynski, M.H. A form of motor cortical plasticity that correlates with recovery of function after brain injury. Proc. Natl. Acad. Sci. USA 103, 11370–11375 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ishizuka, N., Mannen, H., Hongo, T. & Sasaki, S. Trajectory of group Ia afferent fibers stained with horseradish peroxidase in the lumbosacral spinal cord of the cat: three dimensional reconstructions from serial sections. J. Comp. Neurol. 186, 189–211 (1979).

    Article  CAS  PubMed  Google Scholar 

  36. Scheibel, M.E. & Scheibel, A.B. Terminal patterns in cat spinal cord. 3. Primary afferent collaterals. Brain Res. 13, 417–443 (1969).

    Article  CAS  PubMed  Google Scholar 

  37. Gianino, S. et al. Postnatal growth of corticospinal axons in the spinal cord of developing mice. Brain Res. Dev. Brain Res. 112, 189–204 (1999).

    Article  CAS  PubMed  Google Scholar 

  38. Curfs, M.H., Gribnau, A.A. & Dederen, P.J. Selective elimination of transient corticospinal projections in the rat cervical spinal cord gray matter. Brain Res. Dev. Brain Res. 78, 182–190 (1994).

    Article  CAS  PubMed  Google Scholar 

  39. Martin, J.H. Differential spinal projections from the forelimb areas of the rostral and caudal subregions of primary motor cortex in the cat. Exp. Brain Res. 108, 191–205 (1996).

    Article  CAS  PubMed  Google Scholar 

  40. Schouenborg, J., Weng, H.R., Kalliomaki, J. & Holmberg, H. A survey of spinal dorsal horn neurones encoding the spatial organization of withdrawal reflexes in the rat. Exp. Brain Res. 106, 19–27 (1995).

    Article  CAS  PubMed  Google Scholar 

  41. Saltiel, P., Tresch, M.C. & Bizzi, E. Spinal cord modular organization and rhythm generation: an NMDA iontophoretic study in the frog. J. Neurophysiol. 80, 2323–2339 (1998).

    Article  CAS  PubMed  Google Scholar 

  42. Goulding, M. Circuits controlling vertebrate locomotion: moving in a new direction. Nat. Rev. Neurosci. 10, 507–518 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bareyre, F.M., Kerschensteiner, M., Misgeld, T. & Sanes, J.R. Transgenic labeling of the corticospinal tract for monitoring axonal responses to spinal cord injury. Nat. Med. 11, 1355–1360 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Liang, H., Paxinos, G. & Watson, C. The red nucleus and the rubrospinal projection in the mouse. Brain Struct. Funct. 217, 221–232 (2012).

    Article  PubMed  Google Scholar 

  45. Brown, L.T. Rubrospinal projections in the rat. J. Comp. Neurol. 154, 169–187 (1974).

    Article  CAS  PubMed  Google Scholar 

  46. Antal, M. et al. The termination pattern and postsynaptic targets of rubrospinal fibers in the rat spinal cord: a light and electron microscopic study. J. Comp. Neurol. 325, 22–37 (1992).

    Article  CAS  PubMed  Google Scholar 

  47. Lundberg, A. Multisensory control of spinal reflex pathways. Prog. Brain Res. 50, 11–28 (1979).

    Article  CAS  PubMed  Google Scholar 

  48. Schomburg, E.D. Spinal sensorimotor systems and their supraspinal control. Neurosci. Res. 7, 265–340 (1990).

    Article  CAS  PubMed  Google Scholar 

  49. Barthelemy, D., Leblond, H., Provencher, J. & Rossignol, S. Nonlocomotor and locomotor hindlimb responses evoked by electrical microstimulation of the lumbar cord in spinalized cats. J. Neurophysiol. 96, 3273–3292 (2006).

    Article  PubMed  Google Scholar 

  50. Mushahwar, V.K., Aoyagi, Y., Stein, R.B. & Prochazka, A. Movements generated by intraspinal microstimulation in the intermediate gray matter of the anesthetized, decerebrate, and spinal cat. Can. J. Physiol. Pharmacol. 82, 702–714 (2004).

    Article  CAS  PubMed  Google Scholar 

  51. Wickersham, I.R., Sullivan, H.A. & Seung, H.S. Production of glycoprotein-deleted rabies viruses for monosynaptic tracing and high-level gene expression in neurons. Nat. Protoc. 5, 595–606 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Joshi, P.S. et al. Bhlhb5 regulates the postmitotic acquisition of area identities in layers II–V of the developing neocortex. Neuron 60, 258–272 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cheng, L. et al. Tlx3 and Tlx1 are post-mitotic selector genes determining glutamatergic over GABAergic cell fates. Nat. Neurosci. 7, 510–517 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge the generosity and advice of M. Goulding, R. Edgerton, A. Engmann, E. Callaway, J. Young, F. Osakada and F. Stam, P. Joshi and S. McConnell for instruction in cortical injections, and Q. Ma (Harvard Medical School, in situ probes) and C. Birchmeier (Max Delbruck Center for Molecular Medicine, Tlx3 antibody) for providing reagents. M. Sternfeld, M. Hayashi, K. Lettieri and Y. Bogoch provided support and advice. We thank R. Levine for his helpful reading of the manuscript. A.J.L. was supported by George E. Hewitt Foundation for Medical Research and Christopher and Dana Reeve Foundation. C.A.H. was supported by a US National Research Service Award fellowship from US National Institutes of Health NINDS. K.L.H. is supported as a National Science Foundation Graduate Research Fellow. S.L.P. is supported as a Howard Hughes Medical Institute investigator and as a Benjamin H. Lewis chair in neuroscience. This research was supported by the National Institute of Neurological Disorders and Stroke (grant R37NS037116), the Marshall Foundation and the Christopher and Dana Reeve Foundation.

Author information

Authors and Affiliations

Authors

Contributions

A.J.L. and S.L.P. conceived of the study. A.J.L., C.A.H. and S.L.P. designed the study and prepared the manuscript. A.J.L., C.A.H. and K.L.H. carried out the experiments. S.P.D. performed the statistical analysis. J.M.M. contributed to histology experiments. A.J.L. and T.H.P. prepared the rabies virus.

Corresponding author

Correspondence to Samuel L Pfaff.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of cellular morphology of MSE column neurons.

(a) Transverse 50 μm sections of a spinal cord labeled with RabΔG from the gastrocnemius muscle, showing gastrocnemius motorneurons and pre-gastrocnemius neurons from different rostral (top) to caudal (bottom) levels. The lateral propriospinal white matter tracts and the cornu-commisuralis of Marie (CCM) white matter in the dorsal funiculus are indicated. These white matter tracts are known to contain axons of intersegmentally projecting propriospinal interneurons. MSE column neurons can be seen projecting axons into both white matter tracts. (b) Examples of typical cellular morphology of MSE column neurons.

Supplementary Figure 2 Medial deep dorsal horn premotor distribution for diverse muscles.

Distribution of spinal neurons pre-synaptic to the wrist extensor, tibialis anterior, hamstrings, and quadriceps motorneurons, each shown at the level of peak motorneurons for that muscle, together with immunolabeling of Tcfap2β (red) and Satb1/2 (white). Images are projected confocal stacks. Scale bar is 250 μm.

Supplementary Figure 3 Principal component analysis of reliability of evoked motor responses.

(a) Principal component significance plot from principal component analysis on the combined data set of MSE and non-specific ventral location summary metrics (see Methods). The first component (PC1) is the only component identified as significant relative to the significance threshold (dashed red line). (b) PC1 loading pattern represents a reliability signature of low latency to the first motorneuron spike, low standard deviation in the latency to the first motorneuron spike, a high response fraction among a set of ten trials, high similarity in the pattern of motorneuron spikes, and a high fraction of dual root responses with negligible contribution of rostral-caudal location. (c) PC1 association scores (mean and standard error) for MSE evoked L5 (red), MSE evoked L2 (purple), non-specific ventral evoked L5 (orange), and non-specific ventral evoked L2 (blue) responses. A positive association score to PC1 corresponds to greater reliability. (d) Examples of hemisected transverse sections for a transynaptic MSE-focused optical stimulation (left) and a non-specific ventral interneuron stimulation (right). The blue bar represents the dorsal-ventral position of light exposure. MSE and intracord experiments had comparable numbers of ChR2-expressing neurons. Transynaptic (MSE) experiments were only considered if the spinal cords met a minimum criteria for efficiency (at least 15 interneurons in a single peak 50 μm section), and the range was 15-30 interneurons in a peak section in the transynaptic ChR2 experiments. In comparison, intracord injections had an average of 35 ± 10 interneuron per section.

Supplementary Figure 4 MSE neurons target functionally related motor pools.

Individual MSE neurons that directly contact two motor pools were identified using RabΔG:GFP that was injected into the medial gastrocnemius and RabΔG:Cherry was injected into the lateral gastrocnemius or quadriceps at P0. Spinal cords were analyzed at P8. The locations (a-c) and high magnification views (d-f) of individual cells that were directly pre-synaptic to the medial gastrocnemius muscle (green) and either the lateral gastrocnemius (red) (a,d) or the quadriceps (red) (b,c,e,f) are shown, as projected confocal images. Double pre-motor MSE are yellow. We found that 13/389 (n=6 cords) pre-medial gastrocnemius neurons were yellow following lateral gastrocnemius or quadriceps injections. We did not observe any yellow cells following injections of the antagonistic pair of medial gastrocnemius and tibialis anterior (0/278), suggesting that yellow double premotor neurons are specifically associated with functionally co-recruited muscles. Scale bars are 250 μm in a-c and 25 μm in d-f.

Supplementary Figure 5 Interconnectivity among MSE cells.

Projected confocal images of sections from P8 spinal cords with pre-medial gastrocnemius (medial GS-RabΔG, green) and pre-lateral gastrocnemius (lateral GS-RabΔG, cyan) labeling. Excitatory (vGlut2, red) and inhibitory (Gad65 or GlyT2, red) synaptic contacts between medial GS MSE and lateral GS MSE neurons are highlighted with arrowheads. Middle and right panels are single optical sections.

Supplementary Figure 6 Heterogeneity of neurotransmitter expression among MSE neurons.

(a-c) In situ hybridization in P10 spinal cords against excitatory vGlut2 (a), inhibitory Gad65 (b), and inhibitory Gad67 (c) in black, with immunolabeling against Tcfap2β (green). (d-e) Immunolabeling in P2 spinal cords against Satb1/2 (red), and inhibitory Pax2 (green) or excitatory Tlx3 (green).

Supplementary Figure 7 Proprioceptive sensory and cortical contacts onto single MSE cells.

(a) Three-dimensional reconstruction of proprioceptive (PV-syn-Tomato) colocalized terminals (red) onto a RabΔG labeled pre-gastrocnemius MSE neuron (white). (b) Three-dimensional reconstruction of cortical (Ctx-syn-Tomato) colocalized terminals (red) onto a RabΔG labeled pre-TA MSE neuron (white), using Emx1Cre::synaptophysin-tomato. Neurons in a and b express Satb1/2, not shown. Scale bars are 10 μm.

Supplementary Figure 8 Reliability and specificity of distinct motor outputs evoked by optical stimulation of MSE neurons.

L5 (red) and L2 (purple) ventral root responses following 10 trials of optical stimulation of pre-gastrocnemius MSE at L3/L4 (left panel, corresponding to Fig. 6e) and at L2 (right panel, corresponding to Fig. 6d). Note the consistency of the evoked motor patterns. Sets of ten individual traces are shown in each column.

Supplementary Figure 9 Conduction latency within the MSE white matter.

(a) Schematic of experimental setup. Electrodes were placed in the cornu-commisuralis of Marie in the deep dorsal funiculus in densities of RabΔG-labeled MSE axons. (b) Optical stimulation at the site of the rostral electrode (L2) evoked spikes in the local white matter (latency 11.6 ms) preceding those at the caudal site by 1.3 ms. Optical stimulation at the caudal site (c) reversed this relationship. Electrodes were separated by 1.7 mm. The mean conduction delay in n=4 spinal cords was 0.96 ms/mm.

Supplementary Figure 10 MSE cells are a central node in motor control networks.

L2 (left) and L5 (right) lumbar spinal cord segments are depicted with the L5 MSE network (orange). This population receives direct corticospinal (purple) and proprioceptive information (red), and indirect inputs from nociceptive sensory pathways (pink). Outputs include motor pools in multiple segments, such as quadriceps (blue, Q) and gastrocnemius (green, GS).

Supplementary information

Supplementary Text and Figures

Supplementary Table 1 and Supplementary Figures 1–10 (PDF 40866 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Levine, A., Hinckley, C., Hilde, K. et al. Identification of a cellular node for motor control pathways. Nat Neurosci 17, 586–593 (2014). https://doi.org/10.1038/nn.3675

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3675

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