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:

Neurons in the caudal ventrolateral medulla mediate descending pain control

Abstract

Supraspinal brain regions modify nociceptive signals in response to various stressors including stimuli that elevate pain thresholds. The medulla oblongata has previously been implicated in this type of pain control, but the neurons and molecular circuits involved have remained elusive. Here we identify catecholaminergic neurons in the caudal ventrolateral medulla that are activated by noxious stimuli in mice. Upon activation, these neurons produce bilateral feed-forward inhibition that attenuates nociceptive responses through a pathway involving the locus coeruleus and norepinephrine in the spinal cord. This pathway is sufficient to attenuate injury-induced heat allodynia and is required for counter-stimulus induced analgesia to noxious heat. Our findings define a component of the pain modulatory system that regulates nociceptive responses.

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

Fig. 1: The cVLMTH, a brainstem nucleus activated by capsaicin.
Fig. 2: cVLMTH neurons are activated by noxious stimuli.
Fig. 3: VLMTH neurons potently control nociceptive behavioral responses.
Fig. 4: Optogenetic stimulation of cVLMTH triggers extended suppression of nociceptive responses.
Fig. 5: A descending antinociceptive cVLM–LC–SC circuit.
Fig. 6: cVLMTH neurons form monosynaptic inputs in the LC.
Fig. 7: Antinociceptive and pro-nociceptive responses induced by cVLMTH neurons are mediated via noradrenergic mediated processes.
Fig. 8: The cVLM circuit is required and sufficient for counter-stimulus-induced analgesia.

Similar content being viewed by others

Data availability

All data are deposited in an open-source database at https://doi.org/10.5061/dryad.kkwh70s82.

References

  1. Basbaum, A. I. & Fields, H. L. Endogenous pain control systems: brainstem spinal pathways and endorphin circuitry. Annu. Rev. Neurosci. 7, 309–338 (1984).

    Article  CAS  PubMed  Google Scholar 

  2. Heinricher, M. M., Tavares, I., Leith, J. L. & Lumb, B. M. Descending control of nociception: specificity, recruitment and plasticity. Brain Res. Rev. 60, 214–225 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Basbaum, A. I., Bautista, D. M., Scherrer, G. & Julius, D. Cellular and molecular mechanisms of pain. Cell 139, 267–284 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Alhadeff, A. L. et al. A neural circuit for the suppression of pain by a competing need state. Cell 173, 140–152 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bodnar, R. J., Kelly, D. D., Brutus, M., Mansour, A. & Glusman, M. 2-deoxy-d-glucose-induced decrements in operant and reflex pain thresholds. Pharmacol. Biochem. Behav. 9, 543–549 (1978).

    Article  CAS  PubMed  Google Scholar 

  6. Fields, H. L. Pain modulation: expectation, opioid analgesia and virtual pain. Prog. Brain Res. 122, 245–253 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Reynolds, D. V. Surgery in the rat during electrical analgesia induced by focal brain stimulation. Science 164, 444–445 (1969).

    Article  CAS  PubMed  Google Scholar 

  8. Mayer, D. J. & Liebeskind, J. C. Pain reduction by focal electrical stimulation of the brain: an anatomical and behavioral analysis. Brain Res. 68, 73–93 (1974).

    Article  CAS  PubMed  Google Scholar 

  9. Gebhart, G. F. Descending modulation of pain. Neurosci. Biobehav Rev. 27, 729–737 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Liu, Y. et al. Touch and tactile neuropathic pain sensitivity are set by corticospinal projections. Nature 561, 547–550 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tavares, I. & Lima, D. The caudal ventrolateral medulla as an important inhibitory modulator of pain transmission in the spinal cord. J. Pain. 3, 337–346 (2002).

    Article  PubMed  Google Scholar 

  12. Borsook, D. Pain: the past, present and future. Adv. Drug Deliv. Rev. 55, 931–934 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Tavares, I., Lima, D. & Coimbra, A. The ventrolateral medulla of the rat is connected with the spinal cord dorsal horn by an indirect descending pathway relayed in the A5 noradrenergic cell group. J. Comp. Neurol. 374, 84–95 (1996).

    Article  CAS  PubMed  Google Scholar 

  14. Hickey, L. et al. Optoactivation of locus ceruleus neurons evokes bidirectional changes in thermal nociception in rats. J. Neurosci. 34, 4148–4160 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hirschberg, S., Li, Y., Randall, A., Kremer, E. J. & Pickering, A. E. Functional dichotomy in spinal- versus prefrontal-projecting locus coeruleus modules splits descending noradrenergic analgesia from ascending aversion and anxiety in rats. Elife 6, e29808 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Chandler, D. J. et al. Redefining noradrenergic neuromodulation of behavior: impacts of a modular locus coeruleus architecture. J. Neurosci. 39, 8239–8249 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Schwarz, L. A. et al. Viral-genetic tracing of the input–output organization of a central noradrenaline circuit. Nature 524, 88–92 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bullitt, E. Expression of c-Fos-like protein as a marker for neuronal activity following noxious stimulation in the rat. J. Comp. Neurol. 296, 517–530 (1990).

    Article  CAS  PubMed  Google Scholar 

  19. Lanteri-Minet, M., Weil-Fugazza, J., de Pommery, J. & Menetrey, D. Hindbrain structures involved in pain processing as revealed by the expression of c-Fos and other immediate early gene proteins. Neuroscience 58, 287–298 (1994).

    Article  CAS  PubMed  Google Scholar 

  20. Gebhart, G. F. & Ossipov, M. H. Characterization of inhibition of the spinal nociceptive tail-flick reflex in the rat from the medullary lateral reticular nucleus. J. Neurosci. 6, 701–713 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Janss, A. J. & Gebhart, G. F. Spinal monoaminergic receptors mediate the antinociception produced by glutamate in the medullary lateral reticular nucleus. J. Neurosci. 7, 2862–2873 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Foong, F. W. & Duggan, A. W. Brainstem areas tonically inhibiting dorsal horn neurones: studies with microinjection of the GABA analogue piperidine-4-sulphonic acid. Pain 27, 361–371 (1986).

    Article  CAS  PubMed  Google Scholar 

  23. Hall, J. G., Duggan, A. W., Morton, C. R. & Johnson, S. M. The location of brainstem neurones tonically inhibiting dorsal horn neurones of the cat. Brain Res. 244, 215–222 (1982).

    Article  CAS  PubMed  Google Scholar 

  24. Tavares, I. & Lima, D. Descending projections from the caudal medulla oblongata to the superficial or deep dorsal horn of the rat spinal cord. Exp. Brain Res. 99, 455–463 (1994).

    Article  CAS  PubMed  Google Scholar 

  25. Hokfelt, T., Johansson, O. & Goldstein, M. Chemical anatomy of the brain. Science 225, 1326–1334 (1984).

    Article  CAS  PubMed  Google Scholar 

  26. Madisen, L. et al. Transgenic mice for intersectional targeting of neural sensors and effectors with high specificity and performance. Neuron 85, 942–958 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zingg, B. et al. AAV-mediated anterograde transsynaptic tagging: mapping corticocollicular input-defined neural pathways for defense behaviors. Neuron 93, 33–47 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Fields, H. L., Malick, A. & Burstein, R. Dorsal horn projection targets of ON and OFF cells in the rostral ventromedial medulla. J. Neurophysiol. 74, 1742–1759 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Pop, I. V. et al. Structure of long-range direct and indirect spinocerebellar pathways as well as local spinal circuits mediating proprioception. J. Neurosci. 42, 581–600 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bernard, J. F. & Besson, J. M. The spino(trigemino)pontoamygdaloid pathway: electrophysiological evidence for an involvement in pain processes. J. Neurophysiol. 63, 473–490 (1990).

    Article  CAS  PubMed  Google Scholar 

  31. Woulfe, J. M., Hrycyshyn, A. W. & Flumerfelt, B. A. Collateral axonal projections from the A1 noradrenergic cell group to the paraventricular nucleus and bed nucleus of the stria terminalis in the rat. Exp. Neurol. 102, 121–124 (1988).

    Article  CAS  PubMed  Google Scholar 

  32. Jones, S. L. & Gebhart, G. F. Characterization of coeruleospinal inhibition of the nociceptive tail-flick reflex in the rat: mediation by spinal alpha 2-adrenoceptors. Brain Res. 364, 315–330 (1986).

    Article  CAS  PubMed  Google Scholar 

  33. Westlund, K. N., Zhang, D., Carlton, S. M., Sorkin, L. S. & Willis, W. D. Noradrenergic innervation of somatosensory thalamus and spinal cord. Prog. Brain Res. 88, 77–88 (1991).

    Article  CAS  PubMed  Google Scholar 

  34. Sofia Beas, B. et al. A ventrolateral medulla-midline thalamic circuit for hypoglycemic feeding. Nat. Commun. 11, 6218 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Petreanu, L., Mao, T., Sternson, S. M. & Svoboda, K. The subcellular organization of neocortical excitatory connections. Nature 457, 1142–1145 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bruinstroop, E. et al. Spinal projections of the A5, A6 (locus coeruleus) and A7 noradrenergic cell groups in rats. J. Comp. Neurol. 520, 1985–2001 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kim, E. J., Jacobs, M. W., Ito-Cole, T. & Callaway, E. M. Improved monosynaptic neural circuit tracing using engineered rabies virus glycoproteins. Cell Rep. 15, 692–699 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hunker, A. C. et al. Conditional single vector CRISPR/SaCas9 viruses for efficient mutagenesis in the adult mouse nervous system. Cell Rep. 30, 4303–4316 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dogrul, A., Ossipov, M. H. & Porreca, F. Differential mediation of descending pain facilitation and inhibition by spinal 5HT-3 and 5HT-7 receptors. Brain Res. 1280, 52–59 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Brenchat, A. et al. 5-HT7 receptor activation inhibits mechanical hypersensitivity secondary to capsaicin sensitization in mice. Pain 141, 239–247 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Le Bars, D., Dickenson, A. H. & Besson, J. M. Diffuse noxious inhibitory controls (DNIC). II. Lack of effect on non-convergent neurones, supraspinal involvement and theoretical implications. Pain 6, 305–327 (1979).

    Article  PubMed  Google Scholar 

  42. Le Bars, D., Dickenson, A. H. & Besson, J. M. Diffuse noxious inhibitory controls. Effects on dorsal horn convergent neurones in the rat. Pain 6, 283–304 (1979).

    Article  PubMed  Google Scholar 

  43. Le Bars, D. The whole body receptive field of dorsal horn multireceptive neurones. Brain Res. Brain Res. Rev. 40, 29–44 (2002).

    Article  PubMed  Google Scholar 

  44. Gebhart, G. F., Sandkuhler, J., Thalhammer, J. G. & Zimmermann, M. Inhibition of spinal nociceptive information by stimulation in midbrain of the cat is blocked by lidocaine microinjected in nucleus raphe magnus and medullary reticular formation. J. Neurophysiol. 50, 1446–1459 (1983).

    Article  CAS  PubMed  Google Scholar 

  45. Francois, A. et al. A brainstem-spinal cord inhibitory circuit for mechanical pain modulation by GABA and enkephalins. Neuron 93, 822–839 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhang, Y. et al. Identifying local and descending inputs for primary sensory neurons. J. Clin. Invest. 125, 3782–3794 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Samineni, V. K. et al. Divergent modulation of nociception by glutamatergic and GABAergic neuronal subpopulations in the periaqueductal gray. eNeuro https://doi.org/10.1523/ENEURO.0129-16.2017 (2017).

  48. Scherrer, G. et al. Dissociation of the opioid receptor mechanisms that control mechanical and heat pain. Cell 137, 1148–1159 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cavanaugh, D. J. et al. Distinct subsets of unmyelinated primary sensory fibers mediate behavioral responses to noxious thermal and mechanical stimuli. Proc. Natl Acad. Sci. USA 106, 9075–9080 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mishra, S. K., Tisel, S. M., Orestes, P., Bhangoo, S. K. & Hoon, M. A. TRPV1-lineage neurons are required for thermal sensation. EMBO J. 30, 582–593 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Cui, L. et al. Identification of early RET+ deep dorsal spinal cord interneurons in gating pain. Neuron 91, 1413 (2016).

    Article  CAS  PubMed  Google Scholar 

  52. Peirs, C. et al. Dorsal horn circuits for persistent mechanical pain. Neuron 87, 797–812 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Petitjean, H. et al. Dorsal horn parvalbumin neurons are gate-keepers of touch-evoked pain after Nerve Injury. Cell Rep. 13, 1246–1257 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Duan, B. et al. Identification of spinal circuits transmitting and gating mechanical pain. Cell 159, 1417–1432 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bannister, K. & Dickenson, A. H. What the brain tells the spinal cord. Pain 157, 2148–2151 (2016).

    Article  PubMed  Google Scholar 

  56. Bannister, K., Patel, R., Goncalves, L., Townson, L. & Dickenson, A. H. Diffuse noxious inhibitory controls and nerve injury: restoring an imbalance between descending monoamine inhibitions and facilitations. Pain 156, 1803–1811 (2015).

    Article  CAS  PubMed  Google Scholar 

  57. Fields, H. L. & Heinricher, M. M. Brainstem modulation of nociceptor-driven withdrawal reflexes. Ann. N. Y. Acad. Sci. 563, 34–44 (1989).

    Article  CAS  PubMed  Google Scholar 

  58. Bunemann, M., Bucheler, M. M., Philipp, M., Lohse, M. J. & Hein, L. Activation and deactivation kinetics of alpha 2A- and alpha 2C-adrenergic receptor-activated G-protein-activated inwardly rectifying K+ channel currents. J. Biol. Chem. 276, 47512–47517 (2001).

    Article  CAS  PubMed  Google Scholar 

  59. Klapoetke, N. C. et al. Independent optical excitation of distinct neural populations. Nat. Methods 11, 338–346 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Brenner, D. S., Golden, J. P. & Gereau, R. W. T. A novel behavioral assay for measuring cold sensation in mice. PLoS ONE 7, e39765 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Fischer, K. B., Collins, H. K. & Callaway, E. M. Sources of off-target expression from recombinase-dependent AAV vectors and mitigation with cross-over insensitive ATG-out vectors. Proc. Natl Acad. Sci. USA 116, 27001–27010 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Hoover and E. Henry for their help in collecting preliminary data. This work was supported by the intramural research program of the NIDCR, NIH, project ZIADE000721-20 (to M.A.H.) and National Institute of Mental Health, project ZIAMH002950 (to M.P.).

Author information

Authors and Affiliations

Authors

Contributions

X.G., M.P. and M.A.H. designed the experiments. X.G. and M.A.H. wrote and edited the paper with input from all authors. X.G., Y.Z.Z., J.J.O. and C.C.D.P. performed experiments and analyzed data. Y.Z.Z. performed and analyzed ISH experiments. C.C.D.P. performed and analyzed c-Fos studies. J.J.O. carried out and analyzed slice physiological experiments. All other studies were performed and analyzed by X.G. M.A.H. and M.P. supervised the project.

Corresponding author

Correspondence to Mark A. Hoon.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Gregory Scherrer and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 cVLMTH-neurons are catecholaminergic.

Related to Fig. 1. A. Representative magnified images, from mice treated with capsaicin, of ventral lateral medulla regions where tyrosine hydroxylase immune-positive neurons (magenta) are found. In control mice, intraplantar application of capsaicin to the hind-paw induced cfos expression (green) prominently in TH-positive neurons. By contrast this treatment, in Trpv1 knockout (Trpv1KO) mice resulted in expression of few cfos-positive neurons. Scale bars: 50 µm. B. Quantification of cfos expression in TH-neurons, n = 6 control mice and n = 3 Trpv1KOmice (Trpv1KO ipsilateral 94/304, Trpv1KO contralateral 92/278; WT ipsilateral 341/452, WT contralateral 219/404), data are presented as mean ± SEM. C. Multilabel ISH of cVLMTH neurons revealed that the majority of these neurons express DDC and DBH. Scale bars: 50 µm. D. Multilabel ISH of cVLMTH neurons revealed that the majority of these neurons express monoamine transporter Vmat (Slc18a2) but not adrenaline synthesizing enzyme PNMT. Scale bars: 50 µm. E. Quantification of the expression of DDC and DBH with TH; from 239 TH+-neurons there were 239 TH-DCC+, 237 TH-DBH+-cells, n = 3 mice. And quantification of the expression of Vmat and PNMT with TH; from 239 TH+-neurons there were 239 TH-Vmat+, 0 TH-PNMT+-cells, n = 3 mice, data are presented as mean ± SEM. F. Using spinal cord injection of AAV1-hSyn-Cre in Ai9 reporter mice together with injection of AAVretro-CAG-GFP, labeled spinal cord anterograde neurons (labeled magenta - tdT) and retrograde labeled spinal cord projecting neurons (labeled green - GFP). Section from caudal medulla (approx. −7.8 Bregma) showed scattered spinal cord anterograde neurons in the cVLM (circled), nucleus of the solitary tract (NTS), and inferior olive complex (IO). Concentrated axonal labeling of corticospinal neurons was seen in the pyramid (py). G. Section from the rostral medulla (approx. −5.7 Bregma) showed anterograde and retrograde labeled neurons in the rostral ventral medulla as well as axonal labeling of corticospinal neurons in the pyramid (py). H. Monosynaptic rabies tracing from cVLMTH-neurons did not reveal pre-synaptic neurons in the spinal cord. I. Adjacent to cVLMTH-starter neurons (red- helper virus and blue TH-immuno-stained) in the medulla were retrograde labeled neurons (green -rabies virus). Note, all red helper virus labeled neurons were TH-positive. However, we note that these neurons could potentially constitute artefacts as non-specific labeled neurons have been reported adjacent to the site of helper virus injections61.

Extended Data Fig. 2 Photometry of cVLMTH-neurons to mild somatosensory stimuli.

Related to Fig. 2. A. Heatmap traces from 3 individual animals to tail clip. BE. Averaged intracellular calcium responses, using in vivo fiber photometry, of cVLMTH neurons. Responses to mild mechanical with brush to hind-paw (B), von Frey filament on plantar surface of hind-paw (C), localized heating (Hargreaves test on plantar surface of hind-paw) (D), and cold plate stimulation (E) show, only modest increases in intracellular calcium in cVLMTH neurons; n = 6 mice, data are represented as mean results (blue) ± SEM (grey). Quantification of the AUC for measurements are shown to the right of each data set and showed that compared to baseline responses, GCaMP6s responses were not significantly different, p = 0.67, p = 0.997, p = 0. 40, p = 0.31, respectively for panels B-E, two-sided unpaired t-test. Box chart legend: box is defined by 25th, 75th percentiles, whiskers are determined by maximum and minimum, horizontal line represents the mean.

Extended Data Fig. 3 Control chemogenetic activation and inhibition of cVLMTH-neurons.

Related to Fig. 2A. Injection of AAV22-hSyn-DIO -GFP into TH-CreER mice did not alter CNO evoked behavioral responses in Hargreaves tests, n = 6 mice, p = 0.4031 for left L and p = 0.47, two-sided unpaired t-test, data are presented as mean ± SEM. B. For both male and female mice, withdrawal latencies were significantly increased, in Hargreaves tests, after chemogenetic activation of cVLMTH neurons (CNO administration) compared to saline injected mice (L and R indicate left and right hind-paws respectively), n = 8 male mice, p = 0.0004 for L and p < 0.0001 for R hind-paws; n = 8 female mice, p = 0.0016 for L and t = 7.35, p = 0.0002 for R hind-paws, two-sided paired t-test, data are presented as mean ± SEM. data represent means ± SEM. There were no significant differences in responses between male and female mice, p = 0.63 for L and p = 0.95 for R hind-paws, two-sided unpaired t-testdata are presented as mean ± SEM. C. Hargreaves test responses of mice before and after administration of tamoxifen (to induce translocation of CreERT2 and recombination) were not significantly different, n = 8 mice, p = 0.42 for L and p = 0.089 for R hind-paws, two-sided paired Student T-test, data are presented as mean ± SEM. D. For both male and female mice withdrawal latencies were significantly decreased, in Hargreaves tests, after chemogenetic inhibition of cVLMTH neurons (CNO administration) compared to saline injected mice, n = 3 male, p = 0.041 for L and p = 0.023 for R hind-paws; n = 8 female mice, p = 0.0026 for L and p = 0.0033 for R hind-paws, two-sided paired t-test, data are presented as mean ± SEM. There were no significant differences in responses between male and female mice. p = 0.072 for L and, p = 0.38 for R hind-paws. E. Hargreaves test responses of mice before and after administration of tamoxifen. The were no significant differences between treatment groups, n = 5, p = 0.30 for L and p = 0.078 for R hind-paws, two-sided unpaired t-test, Data are presented as mean ± SEM.

Extended Data Fig. 4 Effects of chemogenetic activation of cVLMTH-neurons on itch, touch, cold, motor co-ordination and body temperature.

Related to Fig. 2. A–F Analysis of behavioral responses in TH-CreER mice injected unilaterally in the cVLM with AAV2-hSyn-DIO-hM3D(Gq)-mCherry and tested in behavioral assays following chemogenetic activation of cVLMTH-neurons (CNO). A. Number of scratching bouts over 30 minutes to intradermal injection of chloroquine (200 µg) in the nape of the neck was not significantly different between treatment groups (±CNO) n = 8 mice, p = 0.76, two-sided paired t-test, data are presented as mean ± SEM. B. Threshold responses to von Frey filament stimulation was not significantly different between treatment groups (±CNO) n = 8 mice, p = 0.095 for L and p = 0.80 for R hind-paws, two-sided paired t-test, data are presented as mean ± SEM. C. Mechanical pinch responses (Randal Selitto method) were not significantly different between treatment groups (±CNO) n = 8 mice, p = 0.47,, two-sided paired t-test, data are presented as mean ± SEM. D. Latencies for withdrawal in plantar reflex responses to cold stimulation were not significantly different between treatment groups (±CNO) n = 8 mice, p = 0.11 for L and p = 0.796 for R hind-paws,, two-sided paired t-test, data are presented as mean ± SEM. E. Motor coordination was not significantly different between treatment groups (±CNO) n = 8 mice, p = 0.43, two-sided paired t-test, data are presented as mean ± SEM. F. Core body temperature measured with a rectal thermal probe was not significantly different between treatment groups (±CNO) n = 5 mice, p = 0.58, two-sided paired t-test, data are presented as mean ± SEM.

Extended Data Fig. 5 Effects of chemogenetic inhibition of cVLMTH-neurons on itch, touch, cold, motor co-ordination and body temperature.

Related to Fig. 2. A–F Analysis of behavioral responses in TH-CreER mice injected unilaterally in the cVLM with AAV2-hSyn-DIO-hM4D(Gi)-mCherry and tested in behavioral assays following chemogenetic inhibition of cVLMTH neurons (CNO). A. Number of scratching bouts over 30 minutes to intradermal injection of chloroquine (200 µg) in the nape of the neck was not significantly different between treatment groups (±CNO) n = 6 mice, p = 0.83, two-sided paired t-test, data are presented as mean ± SEM. B, Threshold responses to von Frey filament stimulation was not significantly different between treatment groups (±CNO) n = 11 mice, p = 0.30 for L and p = 0.55 for R hind-paws, two-sided paired t-test, data are presented as mean ± SEM. C. Mechanical pinch responses (Randal Selitto method) were not significantly different between treatment groups (±CNO) n = 6 mice, p = 0.75, two-sided paired t-test, data are presented as mean ± SEM. D. Latencies for withdrawal in plantar reflex responses to cold stimulation were not significantly different between treatment groups (±CNO) n = 5 mice, p = 0.64 for L and p = 0.08 for R hind-paws,, two-sided paired t-test, data are presented as mean ± SEM. E. Motor coordination was not significantly different between treatment groups (±CNO) n = 5 mice, p = 0.28 for light ON and p = 0.13 for R hind-paws, two-sided paired t-test, data are presented as mean ± SEM. F. Core body temperature measured with a rectal thermal probe was not significantly different between treatment groups (±CNO) n = 6 mice, p = 0.41, two-sided paired t-test, data are presented as mean ± SEM.

Extended Data Fig. 6 Innervation of LC by cVLMTH-neurons.

Related to Fig. 3. AF. Representative images of the pattern of fiber projections from TH-Cre mice injected in the cVLM with AAV9-hSyn-DIO-mCherry-2A-SybGFP, showing synaptic boutons (green) on projecting axons and TH-staining of LC-neurons (blue), n = 3 mice. AC. Representative image of a sagittal section of the hindbrain showing cVLMTH neuron projections to the LC. DF. Magnified view of LC. Scale bars, 500 µm in A-C and 50 µm in DF. 3 V indicates third ventricle.

Extended Data Fig. 7 Innervation of PVT, PVN, BNST, and PAG by cVLMTH-neurons.

Related to Fig. 3. A-G. Representative images of the pattern of fiber projections of TH-Cre mice injected in the cVLM with AAV2-Syn-DIO-TVA-mCherry, showing labeled fibers (magenta), n = 3 mice. A. Representative image of a coronal section of the midbrain showing cVLMTH neuron projections to the PVT and PVN. B. Section through midbrain showing cVLMTH neuron projections to the BNST. C. Section of the hindbrain showing cVLMTH neuron projections to the PAG. D and E. Magnified view of boxed areas in A. F. Magnified view of boxed areas in B. G. Magnified view of boxed areas in C. Scale bars: 1 mm in A-C and 100 µm in D-G. 3 V, LV, D3V, and Aq indicate the third ventricle, lateral ventricle, dorsal third ventricle, and aqueduct respectfully.

Extended Data Fig. 8 cVLMTH-neurons trigger PVT dependent food-seeking but not analgesia.

Related to Figs. 56. A-B. Feeding behavior was measured during optogenetic stimulation of VLM terminals in either the LC or in the pPVT. Food intake was quantified in well-fed mice prior to, during and after light stimulation (30 min, pre-test, stimulation and post-test). The stimulation protocol consisted of 30 mins in which light stimulation alternated between 1 min ‘light ON’ (20 Hz) and 2 min ‘light OFF’ bouts. A. Plot of individual performance on feeding behavior during the pre-test, stimulation and post-test for ChR2 (VLM-ChR2) mice. B. Quantification of feeding behavior during VLM-LC and VLM–pPVT stimulation for ChR2 mice. Total food intake in grams, VLM-LC: Pre-Test, 0.048 ± 0.01; Stimulation, 0.035 ± 0.02; Post-Test, 0.06 ± 0.01, n = 8 mice; F (2,23) = 0.623, one-way ANOVA followed by Tukey’s test. Group comparisons: Pre-Test vs Stimulation, p = 0.811; Post-Test vs Stimulation, p = 0.483. for VLM-PVT: Pre-Test, 0.116 ± 0.07; Stimulation, 0.429 ± 0.11; Post-Test, 0.115 ± 0.05, n = 12 mice; F (2,35) = 6.27, one-way ANOVA followed by Tukey’s test. Group comparisons: Pre-Test vs Stimulation, p = 0.02; Post-Test vs Stimulation, p = 0.04. Box chart legend: box is defined by 25th, 75th percentiles, whiskers are determined by maximum and minimum, horizontal lines represent the mean. C. Hargreaves behavioral responses to optogenetic stimulation of VLM terminals in the PVT of TH-Cre mice injected bilaterally with AAV-FLEX-Chrimson. Stimulation did not significantly alter hind-paw withdrawal latency, n = 6 mice, p = 0.42 and p = 0.28 for L and R respectively, two-sided paired t-test, data are presented as mean ± SEM. DE. cVLMTH neurons project collaterally to the PVT and the LC. Retrograde tracers were injected into the PVT (CTB, magenta) and the LC (Fluoro-Gold (FG) green). D. Quantification of results from three animals, data represent means ± SEM. TH+ neurons, n = 440; TH+CTB+ neurons, n = 161; TH+ G+ neurons, n = 107; TH+CTB+FG+ neurons, n = 55. CTB&FG/CTB only + FG only + CTB&FG = 25.6 ± 9.1 mean ± SEM. E. Representative image of the cVLM showing PVT-projecting cVLMTH neurons (TH+CTB+), LC-projecting cVLMTH neurons (TH+FG+) and PVT/LC projecting cVLMTH neurons (TH+CTB+FG+). F. Glutamate receptors antagonists NBQX and AP5 inhibit cVLMTH-mediated optogenetic responses in the LC, but β-adrenergic receptor antagonist Propranolol did not. Z-scores were calculated by using the mean and standard deviation of baseline fluorescence before optical stimulation. Data are presented as the average intensity peak of jGCaMP7s fluorescence relative to baseline fluorescence. Peak intensity of jGCaMP7s fluorescence for ACSF, and for ACSF with glutamate receptor antagonists, p < 0.0001, n = 42 neurons from three animals, two-sided unpaired t-test, data are presented as mean ± SEM. G. Peak intensity of jGCaMP7s fluorescence for ACSF, for ACSF with beta blocker Propranolol, p = 0.23, n = 36 neurons from three animals, two-sided unpaired t-test, data are presented as mean ± SEM.

Extended Data Fig. 9 Controls for optogenetic activation of terminals of cVLMTH-neurons in the LC.

Related to Fig. 7. A. Experimental paradigm used to examine the influence of spinal cord NA on cVLMTH neuron induced antinociceptive effects. B. Optogenetic activation of cVLMTH neuron fiber terminals in the LC induced increased withdrawal latencies in Hargreaves test which were significantly attenuated by intrathecal administration of yohimbine, n = 8 mice, p = 0.0001 for L and p = 0.017 for R hind-paws respectivelytwo-sided paired t-test, data represent means ± SEM. C. Serotonin 5-hydroytryptamine type 3 (5-HT3) receptor antagonist ondansetron (suggested to be responsible, in part, for descending facilitation of pain from the RVM) had no effect on CNO-induced pronociception elicited by chemogenetic inhibition of cVLMTH neuron, n = 7 mice, p = 0.49 for L and p = 0.17 for R hind-paws. D. Similarly, 5-hydroytryptamine type 7 (5-HT7) receptor agonist, AS19 (thought to be responsible, in part, for the descending inhibition of pain from the RVM) also did not affect CNO-induced pronociception, n = 7 mice, p = 0.59 for L and p = 0.0.20 for R hind-paws,two-sided paired t-test, data are presented as mean ± SEM.

Extended Data Fig. 10 Controls for photometry measurements of pain induced analgesia.

Related to Fig. 8. A. Representative example of calcium responses of cVLMTH neurons, measured using in vivo fiber photometry, from a single mouse to repeated noxious heat stimulation (on a hot plate; temperature ramps indicated above trace) before and after injection of capsaicin counter-stimulus into the fore paw (indicated with red arrow). A 1-hour rest period was included between naïve and counter-stimulus trials. B. Behavioral responses to heat challenge (heat ramp to 55 °C) on a hot plate before and after injection of capsaicin in the forepaw; latency to first lick (left columns). There was a significant difference between trial groups for first lick (±capsaicin), n = 6 mice, p = 0.043, two-sided paired t-test, data are presented as mean ± SEM. C. Averaged in vivo photometry responses of cVLMTH neurons for three trials (averaged) before and after a 1-hour rest period to heat challenges (3 x heat ramp to 55 °C) on a hot plate, showed that averaged responses were not altered to repeated noxious thermal insult or by the 1-hour rest period.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gu, X., Zhang, Y.Z., O’Malley, J.J. et al. Neurons in the caudal ventrolateral medulla mediate descending pain control. Nat Neurosci 26, 594–605 (2023). https://doi.org/10.1038/s41593-023-01268-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-023-01268-w

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