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:

Purinergic receptors in the carotid body as a new drug target for controlling hypertension

Abstract

In view of the high proportion of individuals with resistance to antihypertensive medication and/or poor compliance or tolerance of this medication, new drugs to treat hypertension are urgently needed. Here we show that peripheral chemoreceptors generate aberrant signaling that contributes to high blood pressure in hypertension. We discovered that purinergic receptor P2X3 (P2rx3, also known as P2x3) mRNA expression is upregulated substantially in chemoreceptive petrosal sensory neurons in rats with hypertension. These neurons generate both tonic drive and hyperreflexia in hypertensive (but not normotensive) rats, and both phenomena are normalized by the blockade of P2X3 receptors. Antagonism of P2X3 receptors also reduces arterial pressure and basal sympathetic activity and normalizes carotid body hyperreflexia in conscious rats with hypertension; no effect was observed in rats without hypertension. We verified P2X3 receptor expression in human carotid bodies and observed hyperactivity of carotid bodies in individuals with hypertension. These data support the identification of the P2X3 receptor as a potential new target for the control of human hypertension.

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: Overactive peripheral chemoreceptors in spontaneously hypertensive (SH) rats.
Figure 2: P2X3-receptor-mediated hyperreflexia and tonicity of chemoreceptive petrosal neurons in spontaneously hypertensive (SH) rats are associated with the upregulation of P2x3 receptor mRNA.
Figure 3: Upregulation of P2X3-receptor protein in the carotid body of SH rats and sensitization of chemoreceptive petrosal neurons to ATP in SH rats.
Figure 4: P2X3-receptor antagonism lowers arterial pressure in conscious SH rats.
Figure 5: The antihypertensive action of P2X3-receptor antagonism is associated with a reduction in sympathetic activity in SH rats in situ and in vivo.
Figure 6: P2X3-receptor expression in carotid bodies from cadavers of individuals with a medical history of hypertension, and aberrant tone generation in carotid bodies from individuals with hypertension.

Similar content being viewed by others

References

  1. Go, A.S. et al. Executive summary: heart disease and stroke statistics—2014 update: a report from the American Heart Association. Circulation 129, 399–410 (2014).

    Article  PubMed  Google Scholar 

  2. Lloyd, A., Schmieder, C. & Marchant, N. Financial and health costs of uncontrolled blood pressure in the United Kingdom. Pharmacoeconomics 21 (Suppl. 1), 33–41 (2003).

    Article  PubMed  Google Scholar 

  3. Prospective Studies Collaboration. Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 360, 1903–1913 (2002).

  4. Kearney, P.M. et al. Global burden of hypertension: analysis of worldwide data. Lancet 365, 217–223 (2005).

    Article  PubMed  Google Scholar 

  5. Carey, R.M. Resistant hypertension. Hypertension 61, 746–750 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Plump, A. Accelerating the pulse of cardiovascular R&D. Nat. Rev. Drug Discov. 9, 823–824 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Brown, M.J. Aliskiren. Circulation 118, 773–784 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Krum, H. et al. Catheter-based renal sympathetic denervation for resistant hypertension: a multicentre safety and proof-of-principle cohort study. Lancet 373, 1275–1281 (2009).

    Article  PubMed  Google Scholar 

  9. Heusser, K. et al. Carotid baroreceptor stimulation, sympathetic activity, baroreflex function, and blood pressure in hypertensive patients. Hypertension 55, 619–626 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Patel, N.K. et al. Deep brain stimulation relieves refractory hypertension. Neurology 76, 405–407 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Burchell, A.E., Lobo, M.D., Sulke, N., Sobotka, P.A. & Paton, J.F. Arteriovenous anastomosis: is this the way to control hypertension? Hypertension 64, 6–12 (2014).

    Article  CAS  PubMed  Google Scholar 

  12. Lobo, M.D. et al. Central arteriovenous anastomosis for the treatment of patients with uncontrolled hypertension (the ROX CONTROL HTN study): a randomised controlled trial. Lancet 385, 1634–1641 (2015).

    Article  PubMed  Google Scholar 

  13. Narkiewicz, K. et al. Contribution of tonic chemoreflex activation to sympathetic activity and blood pressure in patients with obstructive sleep apnea. Circulation 97, 943–945 (1998).

    Article  CAS  PubMed  Google Scholar 

  14. Tan, Z.Y. et al. Chemoreceptor hypersensitivity, sympathetic excitation, and overexpression of ASIC and TASK channels before the onset of hypertension in SHR. Circ. Res. 106, 536–545 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Przybylski, J. Do arterial chemoreceptors play a role in the pathogenesis of hypertension? Med. Hypotheses 7, 127–131 (1981).

    Article  CAS  PubMed  Google Scholar 

  16. Somers, V.K., Mark, A.L. & Abboud, F.M. Potentiation of sympathetic nerve responses to hypoxia in borderline hypertensive subjects. Hypertension 11, 608–612 (1988).

    Article  CAS  PubMed  Google Scholar 

  17. Trzebski, A., Tafil, M., Zoltowski, M. & Przybylski, J. Increased sensitivity of the arterial chemoreceptor drive in young men with mild hypertension. Cardiovasc. Res. 16, 163–172 (1982).

    Article  CAS  PubMed  Google Scholar 

  18. Siński, M. et al. Tonic activity of carotid body chemoreceptors contributes to the increased sympathetic drive in essential hypertension. Hypertens. Res. 35, 487–491 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Abdala, A.P. et al. Hypertension is critically dependent on the carotid body input in the spontaneously hypertensive rat. J. Physiol. (Lond.) 590, 4269–4277 (2012).

    Article  CAS  Google Scholar 

  20. McBryde, F.D. et al. The carotid body as a putative therapeutic target for the treatment of neurogenic hypertension. Nat. Commun. 4, 2395 (2013).

    Article  PubMed  Google Scholar 

  21. Paton, J.F. et al. The carotid body as a therapeutic target for the treatment of sympathetically mediated diseases. Hypertension 61, 5–13 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Nakayama, K. Surgical removal of the carotid body for bronchial asthma. Dis. Chest 40, 595–604 (1961).

    Article  CAS  PubMed  Google Scholar 

  23. Yuan, G. et al. Protein kinase G-regulated production of H2S governs oxygen sensing. Sci. Signal. 8, ra37 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Prabhakar, N.R. & Peers, C. Gasotransmitter regulation of ion channels: a key step in O2 sensing by the carotid body. Physiology (Bethesda) 29, 49–57 (2014).

    CAS  Google Scholar 

  25. Buckler, K.J. TASK channels in arterial chemoreceptors and their role in oxygen and acid sensing. Pflugers Arch. 467, 1013–1025 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Evans, A.M., Peers, C., Wyatt, C.N., Kumar, P. & Hardie, D.G. Ion channel regulation by the LKB1-AMPK signalling pathway: the key to carotid body activation by hypoxia and metabolic homeostasis at the whole body level. Adv. Exp. Med. Biol. 758, 81–90 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Schultz, H.D., Marcus, N.J. & Del Rio, R. Mechanisms of carotid body chemoreflex dysfunction during heart failure. Exp. Physiol. 100, 124–129 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Ford, A.P. et al. P2X3 receptors and sensitization of autonomic reflexes. Auton. Neurosci. 191, 16–24 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Abdulqawi, R. et al. P2X3 receptor antagonist (AF-219) in refractory chronic cough: a randomised, double-blind, placebo-controlled phase 2 study. Lancet 385, 1198–1205 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Zhang, M., Zhong, H., Vollmer, C. & Nurse, C.A. Co-release of ATP and ACh mediates hypoxic signalling at rat carotid body chemoreceptors. J. Physiol. (Lond.) 525, 143–158 (2000).

    Article  CAS  Google Scholar 

  31. Varas, R., Alcayaga, J. & Iturriaga, R. ACh and ATP mediate excitatory transmission in cat carotid identified chemoreceptor units in vitro. Brain Res. 988, 154–163 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Zapata, P. Is ATP a suitable co-transmitter in carotid body arterial chemoreceptors? Respir. Physiol. Neurobiol. 157, 106–115 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Burnstock, G. Purines and sensory nerves. Handb. Exp. Pharmacol. 194, 333–392 (2009).

    Article  CAS  Google Scholar 

  34. Icekson, G., Dominguez, C.V., Dedios, V.P., Arroyo, J. & Alcayaga, J. Petrosal ganglion responses to acetylcholine and ATP are enhanced by chronic normobaric hypoxia in the rabbit. Respir. Physiol. Neurobiol. 189, 624–631 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Kåhlin, J. et al. The human carotid body releases acetylcholine, ATP and cytokines during hypoxia. Exp. Physiol. 99, 1089–1098 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Prasad, M. et al. Expression of P2X2 and P2X3 receptor subunits in rat carotid body afferent neurones: role in chemosensory signalling. J. Physiol. (Lond.) 537, 667–677 (2001).

    Article  CAS  Google Scholar 

  37. Rong, W. et al. Pivotal role of nucleotide P2X2 receptor subunit of the ATP-gated ion channel mediating ventilatory responses to hypoxia. J. Neurosci. 23, 11315–11321 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Welsh, M.J., Heistad, D.D. & Abboud, F.M. Depression of ventilation by dopamine in man. Evidence for an effect on the chemoreceptor reflex. J. Clin. Invest. 61, 708–713 (1978).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cardenas, H. & Zapata, P. Dopamine-induced ventilatory depression in the rat, mediated by carotid nerve afferents. Neurosci. Lett. 24, 29–33 (1981).

    Article  CAS  PubMed  Google Scholar 

  40. Gever, J.R. et al. AF-353, a novel, potent and orally bioavailable P2X3/P2X2/3 receptor antagonist. Br. J. Pharmacol. 160, 1387–1398 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Clarke, J.A., Daly, M.D. & Ead, H.W. Vascular analysis of the carotid body in the spontaneously hypertensive rat. Adv. Exp. Med. Biol. 337, 3–8 (1993).

    Article  CAS  PubMed  Google Scholar 

  42. Nurse, C.A. Synaptic and paracrine mechanisms at carotid body arterial chemoreceptors. J. Physiol. (Lond.) 592, 3419–3426 (2014).

    Article  CAS  Google Scholar 

  43. Fan, J. et al. Interleukin-6 increases intracellular Ca2+ concentration and induces catecholamine secretion in rat carotid body glomus cells. J. Neurosci. Res. 87, 2757–2762 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Ford, A.P. & Undem, B.J. The therapeutic promise of ATP antagonism at P2X3 receptors in respiratory and urological disorders. Front. Cell. Neurosci. 7, 267 (2013).

    PubMed  PubMed Central  Google Scholar 

  45. Daly, D.M. et al. Age-related changes in afferent pathways and urothelial function in the male mouse bladder. J. Physiol. (Lond.) 592, 537–549 (2014).

    Article  CAS  Google Scholar 

  46. Ford, A.P. & Cockayne, D.A. ATP and P2X purinoceptors in urinary tract disorders. Handb. Exp. Pharmacol. 202, 485–526 (2011).

    Article  CAS  Google Scholar 

  47. Adriaensen, D., Brouns, I. & Timmermans, J.P. Sensory input to the central nervous system from the lungs and airways: A prominent role for purinergic signalling via P2X2/3 receptors. Auton. Neurosci. 191, 39–47 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Burnstock, G. Purinergic signalling in the gastrointestinal tract and related organs in health and disease. Purinergic Signal. 10, 3–50 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Deiteren, A. et al. P2X3 receptors mediate visceral hypersensitivity during acute chemically-induced colitis and in the post-inflammatory phase via different mechanisms of sensitization. PLoS One 10, e0123810 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Li, J., Xing, J. & Lu, J. Nerve growth factor, muscle afferent receptors and autonomic responsiveness with femoral artery occlusion. J. Mod. Physiol. Res 1, 1–18 (2014).

    PubMed  PubMed Central  Google Scholar 

  51. Hansen, R.R. et al. Chronic administration of the selective P2X3, P2X2/3 receptor antagonist, A-317491, transiently attenuates cancer-induced bone pain in mice. Eur. J. Pharmacol. 688, 27–34 (2012).

    Article  CAS  PubMed  Google Scholar 

  52. Liu, M. et al. Coexpression of P2X(3) and P2X(2) receptor subunits in varying amounts generates heterogeneous populations of P2X receptors that evoke a spectrum of agonist responses comparable to that seen in sensory neurons. J. Pharmacol. Exp. Ther. 296, 1043–1050 (2001).

    CAS  PubMed  Google Scholar 

  53. Gerevich, Z. et al. Dual effect of acid pH on purinergic P2X3 receptors depends on the histidine 206 residue. J. Biol. Chem. 282, 33949–33957 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Reichling, D.B. & Levine, J.D. Critical role of nociceptor plasticity in chronic pain. Trends Neurosci. 32, 611–618 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Schiavuzzo, J.G. et al. Muscle hyperalgesia induced by peripheral P2X3 receptors is modulated by inflammatory mediators. Neuroscience 285, 24–33 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. Zoccal, D.B. et al. Increased sympathetic outflow in juvenile rats submitted to chronic intermittent hypoxia correlates with enhanced expiratory activity. J. Physiol. (Lond.) 586, 3253–3265 (2008).

    Article  CAS  Google Scholar 

  57. Moraes, D.J., Machado, B.H. & Paton, J.F. Specific respiratory neuron types have increased excitability that drive presympathetic neurones in neurogenic hypertension. Hypertension 63, 1309–1318 (2014).

    Article  CAS  PubMed  Google Scholar 

  58. Somers, V.K., Mark, A.L. & Abboud, F.M. Interaction of baroreceptor and chemoreceptor reflex control of sympathetic nerve activity in normal humans. J. Clin. Invest. 87, 1953–1957 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Comroe, J.H. Jr. The functions of the lung. Harvey Lect. 48, 110–143 (1952-1953).

    PubMed  Google Scholar 

  60. Paton, J.F.R. A working heart-brainstem preparation of the mouse. J. Neurosci. Methods 65, 63–68 (1996).

    Article  CAS  PubMed  Google Scholar 

  61. Moraes, D.J. et al. Electrophysiological properties of rostral ventrolateral medulla presympathetic neurons modulated by the respiratory network in rats. J. Neurosci. 33, 19223–19237 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Waynforth, H.B. & Flecknell, P. Experimental and surgical techniques in the rat. 2nd edn.382 (Academic Press, London, 1992).

  63. Stickland, M.K. et al. Carotid chemoreceptor modulation of blood flow during exercise in healthy humans. J. Physiol. (Lond.) 589, 6219–6230 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We wish to thank J.-C. Isner (School of Biological Sciences, University of Bristol) for his expertise on software used for analyzing aspects of some of the in vivo cardiovascular data. Technical support from P. Chappell (mechanical workshop) and D. Carr (electronic workshop) is appreciated. The research support of the British Heart Foundation RG/12/6/29670 (J.F.R.P.), Afferent Pharmaceuticals (A.P.F. & J.F.R.P.) and the James Tudor Foundation (J.F.R.P.) are acknowledged. This research was supported by the National Institute for Health Research (NIHR) Biomedical Research Unit in Cardiovascular Disease at the University Hospitals Bristol National Health Service Foundation Trust and the University of Bristol (A.K.N. & J.F.R.P.). Studies in situ were supported by grants from 'Fundação de Amparo à Pesquisa do Estado de São Paulo' FAPESP Thematic Project 2013/06077-5 (B.H.M.) and research grant 2013/10484-5 (D.J.A.M.). The University of Bristol's Wolfson Bioimaging Facility BBSRC Alert 13 capital grant BB/L014181/1 is acknowledged. This project also received funding from the Marie Curie International Research Staff Exchange Scheme within the 7th European Community Framework Program under grant agreement no. 612280. This article presents independent research funded by the National Institute for Health Research (NIHR). The views expressed are those of the author(s) and not necessarily those of the National Health Service, the NIHR or the Department of Health.

Author information

Authors and Affiliations

Authors

Contributions

W.P. conducted all in vivo radiotelemetry blood pressure studies and the rat and human immunocytochemistry and western blotting; this also included data analysis and figure and manuscript preparation. D.J.A.M. performed all in situ rat nerve and petrosal neuron whole-cell recording studies; this also included data analysis and figure preparation. M.P.d.S. performed the single-neuron PCR study. L.E.K.R. with A.K.N. carried out the diagnosis and recruitment of humans with hypertension, and L.E.K.R. with E.C.H. performed and analyzed data from the dopamine-infusion study. B.H.M. supported all in situ studies and assisted in experimental design, data analysis and manuscript preparation. F.D.M. conducted the in vivo radiotelemetry study for recording renal sympathetic nerve activity; this also included data analysis and figure preparation. A.P.A. performed some of the first immunohistochemistry on human carotid bodies. A.P.F. provided the P2X3-receptor antagonists, carried out the pharmacokinetic analysis and assisted in drug trial design and manuscript preparation and revision. J.F.R.P. orchestrated the design of the project, provided supervision with data acquisition and analysis, wrote the manuscript and revised it, and raised the funding.

Corresponding author

Correspondence to Julian F R Paton.

Ethics declarations

Competing interests

A.P.F. is Chief Scientific Officer for Afferent Pharmaceuticals. The other authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 (PDF 2047 kb)

Supplementary Dataset 1

Supplementary Source Data (XLSX 110 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pijacka, W., Moraes, D., Ratcliffe, L. et al. Purinergic receptors in the carotid body as a new drug target for controlling hypertension. Nat Med 22, 1151–1159 (2016). https://doi.org/10.1038/nm.4173

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4173

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research