An ultrafast system for signaling mechanical pain in human skin

Saad S Nagi, Andrew G Marshall, Adarsh Makdani, Ewa Jarocka, Jaquette Liljencrantz, Mikael Ridderström, Sumaiya Shaikh, Francis O'Neill, Dimah Saade, Sandra Donkervoort, A Reghan Foley, Jan Minde, Mats Trulsson, Jonathan Cole, Carsten G Bönnemann, Alexander T Chesler, M Catherine Bushnell, Francis McGlone, Håkan Olausson, Saad S Nagi, Andrew G Marshall, Adarsh Makdani, Ewa Jarocka, Jaquette Liljencrantz, Mikael Ridderström, Sumaiya Shaikh, Francis O'Neill, Dimah Saade, Sandra Donkervoort, A Reghan Foley, Jan Minde, Mats Trulsson, Jonathan Cole, Carsten G Bönnemann, Alexander T Chesler, M Catherine Bushnell, Francis McGlone, Håkan Olausson

Abstract

The canonical view is that touch is signaled by fast-conducting, thickly myelinated afferents, whereas pain is signaled by slow-conducting, thinly myelinated ("fast" pain) or unmyelinated ("slow" pain) afferents. While other mammals have thickly myelinated afferents signaling pain (ultrafast nociceptors), these have not been demonstrated in humans. Here, we performed single-unit axonal recordings (microneurography) from cutaneous mechanoreceptive afferents in healthy participants. We identified A-fiber high-threshold mechanoreceptors (A-HTMRs) that were insensitive to gentle touch, encoded noxious skin indentations, and displayed conduction velocities similar to A-fiber low-threshold mechanoreceptors. Intraneural electrical stimulation of single ultrafast A-HTMRs evoked painful percepts. Testing in patients with selective deafferentation revealed impaired pain judgments to graded mechanical stimuli only when thickly myelinated fibers were absent. This function was preserved in patients with a loss-of-function mutation in mechanotransduction channel PIEZO2. These findings demonstrate that human mechanical pain does not require PIEZO2 and can be signaled by fast-conducting, thickly myelinated afferents.

Figures

Fig. 1. Humans are equipped with high-threshold…
Fig. 1. Humans are equipped with high-threshold and very fast conducting primary afferents.
(A) Location of myelinated HTMR receptive fields from recordings in the peroneal and radial nerves. Each red dot represents the location of an individual A-HTMR (n = 18). The pattern of receptive field spots, mapped with a Semmes-Weinstein monofilament, is shown for two A-HTMRs (marked by arrows; top, radial; bottom, peroneal). Receptive field spots were redrawn on photographic images so they can easily be seen. The horizontal lines represent the medial-lateral dimension, and the vertical lines represent the proximal-distal dimension. The average size of an A-HTMR receptive field, mapped using a filament force six times higher than that for an A-LTMR, was 26.8 mm2 (±6.9; n = 9). This was significantly smaller than the receptive field of field afferents (100.5 ± 7.2 mm2; n = 51; P < 0.0001, Dunnett’s test) but was not different from that of SA1 afferents (41.6 ± 7.5 mm2; n = 17; P > 0.05, Dunnett’s test). (B) Brush responses of an A-HTMR and a field afferent. Using a soft or a coarse brush, the skin area centered on the receptive field of the recorded afferent was gently stroked at 3 cm/s. The field afferent responded vigorously to soft brush stroking. The A-HTMR did not respond to soft brush stroking, but it did respond to coarse brush stroking. The mechanical threshold of this A-HTMR was 4 mN, and the conduction velocity was 52 m/s. It responded to pinching, and its receptive field moved with skin translocation. Freq, frequency. (C) Mechanical threshold distribution of HTMRs and LTMRs in the recorded sample. For RA1 afferents, the preferred stimulus is hair movement, so monofilament thresholds were not measured. For A-HTMR, the median mechanical threshold was 10.0 mN (Q, 5.5–20.0; n = 18). This was significantly higher than the mechanical thresholds of all tested A-LTMR types (at least P < 0.001 for all individual comparisons, Dunn’s test) but was not different from C-HTMRs (10.0 mN; Q, 10.0–27.0; n = 5). (D) Spike activity of an A-HTMR to electrical and mechanical stimulations of the receptive field. Individual electrically and mechanically evoked spikes were superimposed on an expanded time scale to show that the electrically stimulated spike (used for latency measurement) was from the same unit as the one that was mechanically probed at the receptive field. (E) Conduction velocities of HTMRs and LTMRs to surface electrical stimulation (and monofilament tapping in case of one HTMR, conducting at 30 m/s). The data show individual and average (±SEM) conduction velocities of single afferents from peroneal (circles) and radial (diamonds) nerves. Conduction velocities of peroneal A-HTMRs (33.5 ± 2.1; n = 13) were statistically indistinguishable from peroneal A-LTMRs [SA1: 39.8 ± 2.3, n = 10; SA2: 38.6 ± 4.0, n = 4; RA1: 36.8 ± 2.8, n = 6; field: 34.3 ± 1.3, n = 18; F(4,46) = 1.70; P = 0.17, one-way analysis of variance (ANOVA)]. All three peroneal C-HTMRs were conducting at 0.9 m/s. In comparison to the peroneal nerve, conduction velocities of A-fiber types were faster in the radial nerve as expected (A-HTMR: 54.5 ± 2.4, n = 4; SA1: 56.8 m/s, n = 1; SA2: 53.0 ± 3.3, n = 3; RA1: 48.7 ± 1.6, n = 3; field: 47.3 ± 0.2, n = 2) (46). Both radial C-HTMRs were conducting at 1.1 m/s. Conduction velocity of RA2 afferents was not measured. (F) Slowly adapting properties of an A-HTMR at higher indentation forces. Spike activity of a field afferent and an A-HTMR during monofilament stimulation at three different forces, applied using electronic filaments with force feedback. Compared to the field afferent, the A-HTMR showed a sustained response at a lower indentation force (see also fig. S1).
Fig. 2. Neural discharge of A-HTMRs and…
Fig. 2. Neural discharge of A-HTMRs and perception of mechanical pain in response to punctate forces.
(A) Peak discharge rates of human A-HTMRs for monofilament stimulation. The data show individual and average (±SEM) responses of nine A-HTMRs (≥30 m/s) to 5-s monofilament stimulation at eight different indentation forces. A significant linear fit was displayed (R2 = 0.6981, P = 0.0098), and the response at the highest indentation force (3000 mN) was significantly higher than responses to all weaker indentation forces (at least P < 0.05, Dunnett’s test). Individual units are color-coded. (B) Psychophysical data for pain intensity to graded monofilament stimulation. Psychophysical pain ratings were collected from 16 healthy participants (dorsal foot: 8 participants, 12 trial sets; dorsal toe: 8 participants, 9 trial sets) from the microneurography sample. The monofilament force had a significant effect on psychophysical pain ratings [F(8,171) = 29.96; P < 0.0001, two-way ANOVA], with a significant pain of ~14 of 100 emerging at 260 mN, and higher pain ratings with increasing forces (Tukey’s multiple comparisons test). No difference in pain ratings was found between the foot and toe stimulation sites [F(1,171) = 0.1733; P = 0.6777, two-way ANOVA]; hence, the data were pooled for a subsequent analysis. (C) Psychophysical pain ratings as a function of neural discharge in A-HTMRs. A comparison of the average peak discharge rates of nine A-HTMRs and average psychophysical pain ratings for skin indentations (eight forces, 4 to 3000 mN) revealed a significant positive correlation (Pearson r = 0.8944, R2 = 0.8, P = 0.0027). (D) Number of spikes produced by monofilament stimulation in human A-HTMRs. Individual and average (±SEM) responses of nine A-HTMRs to 5-s monofilament stimulation at eight different indentation forces. A significant linear fit was displayed (R2 = 0.6361, P = 0.0177). Individual units are color-coded. (E) Spike activity of an A-HTMR during the first 0.5 s of monofilament stimulation (with force feedback). Top: Neural discharge rates. Middle: Spike markers and indentation force markers. Bottom: Neural recording. The first 0.5 s was selected as the onset period (dynamic phase) of monofilament stimulation (total duration, 5 s). (F) Mean discharge rates of human A-HTMRs for monofilament onset. Individual and average (±SEM) responses of nine A-HTMRs to the onset period (0.5 s) of monofilament stimulation at eight different indentation forces. A significant linear fit was displayed (R2 = 0.6784, P = 0.0120). Individual units are color-coded.
Fig. 3. Mechanical pain sensitivity is dependent…
Fig. 3. Mechanical pain sensitivity is dependent on Aβ afferents but not PIEZO2 stretch-gated ion channels.
(A) Psychophysical pain ratings to graded monofilament stimulation of the peroneal foot for two patients with Aβ deafferentation, two patients with HSAN-V and six healthy participants (pooled means ± SEM and individual data comprising mean of triplicates). A comparison of the pain ratings between patients with HSAN-V and healthy participants revealed no effect of condition—healthy versus HSAN-V [F(1,197) = 1.372; P = 0.2428, two-way ANOVA]. In contrast, a comparison of patients with HSAN-V and healthy participants, on one hand, and patients with Aβ deafferentation, on the other hand, revealed a significant effect of condition—healthy versus Aβ deafferentation [F(1,198) = 15.72, P = 0.0001] and HSAN-V versus Aβ deafferentation [F(1,89) = 13.01, P = 0.0005]. ABD, Aβ deafferentation. (B) Psychophysical pain ratings to graded monofilament stimulation of the radial forearm for two patients with PIEZO2LOF, in addition to average pain ratings from four healthy participants (pooled means ± SEM and individual data comprising mean of triplicates). A comparison of the pain ratings between patients with PIEZO2LOF and four healthy participants revealed slightly higher ratings in the patients with PIEZO2LOF compared to healthy participants [F(1,144) = 4.079; P = 0.0453, two-way ANOVA]. The two patients with Aβ deafferentation reported no pain to graded monofilament stimulation of the forearm.

References

    1. Lloyd D. P. C., Neuron patterns controlling transmission of ipsilateral hind limb reflexes in cat. J. Neurophysiol. 6, 293–315 (1943).
    1. Adrian E. D., Zotterman Y., The impulses produced by sensory nerve endings: Part 3. Impulses set up by touch and pressure. J. Physiol. 61, 465–483 (1926).
    1. Adrian E. D., The impulses produced by sensory nerve-endings: Part 4. Impulses from pain receptors. J. Physiol. 62, 33–51 (1926).
    1. Torebjörk H. E., Hallin R. G., Perceptual changes accompanying controlled preferential blocking of A and C fibre responses in intact human skin nerves. Exp. Brain Res. 16, 321–332 (1973).
    1. Mackenzie R. A., Burke D., Skuse N. F., Lethlean A. K., Fibre function and perception during cutaneous nerve block. J. Neurol. Neurosurg. Psychiatry 38, 865–873 (1975).
    1. Ochoa J., Torebjörk E., Sensations evoked by intraneural microstimulation of C nociceptor fibres in human skin nerves. J. Physiol. 415, 583–599 (1989).
    1. Bromm B., Treede R. D., Nerve fibre discharges, cerebral potentials and sensations induced by CO2 laser stimulation. Hum. Neurobiol. 3, 33–40 (1984).
    1. Löken L. S., Wessberg J., Morrison I., McGlone F., Olausson H., Coding of pleasant touch by unmyelinated afferents in humans. Nat. Neurosci. 12, 547–548 (2009).
    1. Rolke R., Baron R., Maier C., Tölle T. R., Treede R. D., Beyer A., Binder A., Birbaumer N., Birklein F., Bötefür I. C., Braune S., Flor H., Huge V., Klug R., Landwehrmeyer G. B., Magerl W., Maihöfner C., Rolko C., Schaub C., Scherens A., Sprenger T., Valet M., Wasserka B., Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): Standardized protocol and reference values. Pain 123, 231–243 (2006).
    1. Djouhri L., Lawson S. N., Aβ-fiber nociceptive primary afferent neurons: A review of incidence and properties in relation to other afferent A-fiber neurons in mammals. Brain Res. Rev. 46, 131–145 (2004).
    1. Koltzenburg M., Stucky C. L., Lewin G. R., Receptive properties of mouse sensory neurons innervating hairy skin. J. Neurophysiol. 78, 1841–1850 (1997).
    1. Treede R.-D., Meyer R. A., Campbell J. N., Myelinated mechanically insensitive afferents from monkey hairy skin: Heat-response properties. J. Neurophysiol. 80, 1082–1093 (1998).
    1. Burgess P. R., Perl E. R., Myelinated afferent fibres responding specifically to noxious stimulation of the skin. J. Physiol. 190, 541–562 (1967).
    1. Willer J. C., Boureau F., Albe-Fessard D., Role of large diameter cutaneous afferents in transmission of nociceptive messages: Electrophysiological study in man. Brain Res. 152, 358–364 (1978).
    1. Willer J.-C., Albe-Fessard D., Further studies on the role of afferent input from relatively large diameter fibers in transmission of nociceptive messages in humans. Brain Res. 278, 318–321 (1983).
    1. Bai L., Lehnert B. P., Liu J., Neubarth N. L., Dickendesher T. L., Nwe P. H., Cassidy C., Woodbury C. J., Ginty D. D., Genetic identification of an expansive mechanoreceptor sensitive to skin stroking. Cell 163, 1783–1795 (2015).
    1. H. Merskey, N. Bogduk, Classification of Chronic Pain. Descriptions of Chronic Pain Syndromes and Definitions of Pain Terms (IASP Press, 1994).
    1. Ranade S. S., Woo S.-H., Dubin A. E., Moshourab R. A., Wetzel C., Petrus M., Mathur J., Begay V., Coste B., Mainquist J., Wilson A. J., Francisco A. G., Reddy K., Qiu Z., Wood J. N., Lewin G. R., Patapoutian A., Piezo2 is the major transducer of mechanical forces for touch sensation in mice. Nature 516, 121–125 (2014).
    1. Woo S.-H., Lukacs V., de Nooij J. C., Zaytseva D., Criddle C. R., Francisco A., Jessell T. M., Wilkinson K. A., Patapoutian A., Piezo2 is the principal mechanotransduction channel for proprioception. Nat. Neurosci. 18, 1756–1762 (2015).
    1. Chesler A. T., Szczot M., Bharucha-Goebel D., Čeko M., Donkervoort S., Laubacher C., Hayes L. H., Alter K., Zampieri C., Stanley C., Innes A. M., Mah J. K., Grosmann C. M., Bradley N., Nguyen D., Foley A. R., Le Pichon C. E., Bönnemann C. G., The role of PIEZO2 in human mechanosensation. N. Engl. J. Med. 375, 1355–1364 (2016).
    1. Murthy S. E., Loud M. C., Daou I., Marshall K. L., Schwaller F., Kühnemund J., Francisco A. G., Keenan W. T., Dubin A. E., Lewin G. R., Patapoutian A., The mechanosensitive ion channel Piezo2 mediates sensitivity to mechanical pain in mice. Sci. Transl. Med. 10, eaat9897 (2018).
    1. Vallbo Å. B., Hagbarth K.-E., Activity from skin mechanoreceptors recorded percutaneously in awake human subjects. Exp. Neurol. 21, 270–289 (1968).
    1. Vallbo Å. B., Olausson H., Wessberg J., Kakuda N., Receptive field characteristics of tactile units with myelinated afferents in hairy skin of human subjects. J. Physiol. 483, 783–795 (1995).
    1. Serra J., Campero M., Ochoa J., Bostock H., Activity-dependent slowing of conduction differentiates functional subtypes of C fibres innervating human skin. J. Physiol. 515, 799–811 (1999).
    1. Watkins R. H., Wessberg J., Wasling H. B., Dunham J. P., Olausson H., Johnson R. D., Ackerley R., Optimal delineation of single C-tactile and C-nociceptive afferents in humans using latency slowing. J. Neurophysiol. 117, 1608–1614 (2017).
    1. Ackerley R., Wiklund Fernstrom K., Backlund Wasling H., Watkins R. H., Johnson R. D., Vallbo Å. B., Wessberg J., Differential effects of radiant and mechanically applied thermal stimuli on human C-tactile afferent firing patterns. J. Neurophysiol. 120, 1885–1892 (2018).
    1. Ochoa J., Torebjörk H., Sensation evoked by intraneural microstimulation of single mechanoreceptor units innervating the human hand. J. Physiol. 342, 633–654 (1983).
    1. Vallbo Å. B., Olsson K. A., Westberg K. G., Clark F. J., Microstimulation of single tactile afferents from the human hand. Sensory attributes related to unit type and properties of receptive fields. Brain 107, 727–749 (1984).
    1. Olausson H., Cole J., Rylander K., McGlone F., Lamarre Y., Wallin B. G., Krämer H., Wessberg J., Elam M., Bushnell M. C., Vallbo Å., Functional role of unmyelinated tactile afferents in human hairy skin: Sympathetic response and perceptual localization. Exp. Brain Res. 184, 135–140 (2008).
    1. Cole J. D., Merton W. L., Barrett G., Katifi H. A., Treede R. D., Evoked potentials in a subject with a large-fibre sensory neuropathy below the neck. Can. J. Physiol. Pharmacol. 73, 234–245 (1995).
    1. Cole J., Bushnell M. C., McGlone F., Elam M., Lamarre Y., Vallbo Å., Olausson H., Unmyelinated tactile afferents underpin detection of low-force monofilaments. Muscle Nerve 34, 105–107 (2006).
    1. Minde J., Svensson O., Holmberg M., Solders G., Toolanen G., Orthopedic aspects of familial insensitivity to pain due to a novel nerve growth factor beta mutation. Acta Orthop. 77, 198–202 (2006).
    1. Minde J., Andersson T., Fulford M., Aguirre M., Nennesmo I., Remahl I. N., Svensson O., Holmberg M., Toolanen G., Solders G., A novel NGFB point mutation: A phenotype study of heterozygous patients. J. Neurol. Neurosurg. Psychiatry 80, 188–195 (2009).
    1. Morrison I., Löken L. S., Minde J., Wessberg J., Perini I., Nennesmo I., Olausson H., Reduced C-afferent fibre density affects perceived pleasantness and empathy for touch. Brain 134, 1116–1126 (2011).
    1. Szczot M., Liljencrantz J., Ghitani N., Barik A., Lam R., Thompson J. H., Bharucha-Goebel D., Saade D., Necaise A., Donkervoort S., Foley A. R., Gordon T., Case L., Bushnell M. C., Bönnemann C. G., Chesler A. T., PIEZO2 mediates injury-induced tactile pain in mice and humans. Sci. Transl. Med. 10, eaat9892 (2018).
    1. Perl E. R., Myelinated afferent fibres innervating the primate skin and their response to noxious stimuli. J. Physiol. 197, 593–615 (1968).
    1. Vallbo Å. B., Olausson H., Wessberg J., Unmyelinated afferents constitute a second system coding tactile stimuli of the human hairy skin. J. Neurophysiol. 81, 2753–2763 (1999).
    1. Lawson S. N., Phenotype and function of somatic primary afferent nociceptive neurones with C-, Aδ- or Aα/β-fibres. Exp. Physiol. 87, 239–244 (2002).
    1. Adriaensen H., Gybels J., Handwerker H. O., Van Hees J., Response properties of thin myelinated (A-delta) fibers in human skin nerves. J. Neurophysiol. 49, 111–122 (1983).
    1. Ghitani N., Barik A., Szczot M., Thompson J. H., Li C., Le Pichon C. E., Krashes M. J., Chesler A. T., Specialized mechanosensory nociceptors mediating rapid responses to hair pull. Neuron 95, 944–954.e4 (2017).
    1. Arcourt A., Gorham L., Dhandapani R., Prato V., Taberner F. J., Wende H., Gangadharan V., Birchmeier C., Heppenstall P. A., Lechner S. G., Touch receptor-derived sensory information alleviates acute pain signaling and fine-tunes nociceptive reflex coordination. Neuron 93, 179–193 (2017).
    1. Garell P. C., McGillis S. L., Greenspan J. D., Mechanical response properties of nociceptors innervating feline hairy skin. J. Neurophysiol. 75, 1177–1189 (1996).
    1. Slugg R. M., Meyer R. A., Campbell J. N., Response of cutaneous A- and C-fiber nociceptors in the monkey to controlled-force stimuli. J. Neurophysiol. 83, 2179–2191 (2000).
    1. Krarup C., Trojaborg W., Compound sensory action potentials evoked by tactile and by electrical stimulation in normal median and sural nerves. Muscle Nerve 17, 733–740 (1994).
    1. Kakuda N., Conduction velocity of low-threshold mechanoreceptive afferent fibers in the glabrous and hairy skin of human hands measured with microneurography and spike-triggered averaging. Neurosci. Res. 15, 179–188 (1992).
    1. Macefield G., Gandevia S. C., Burke D., Conduction velocities of muscle and cutaneous afferents in the upper and lower limbs of human subjects. Brain 112, 1519–1532 (1989).
    1. Simone D. A., Marchettini P., Caputi G., Ochoa J. L., Identification of muscle afferents subserving sensation of deep pain in humans. J. Neurophysiol. 72, 883–889 (1994).
    1. Inui K., Tran T. D., Hoshiyama M., Kakigi R., Preferential stimulation of Aδ fibers by intra-epidermal needle electrode in humans. Pain 96, 247–252 (2002).
    1. Tran T. D., Lam K., Hoshiyama M., Kakigi R., A new method for measuring the conduction velocities of Aβ-, Aδ- and C-fibers following electric and CO2 laser stimulation in humans. Neurosci. Lett. 301, 187–190 (2001).
    1. Iannetti G. D., Baumgärtner U., Tracey I., Treede R. D., Magerl W., Pinprick-evoked brain potentials: A novel tool to assess central sensitization of nociceptive pathways in humans. J. Neurophysiol. 110, 1107–1116 (2013).
    1. Melzack R., Wall P. D., Pain mechanisms: A new theory. Science 150, 971–978 (1965).
    1. Richardson R., Arbit J., Siqueira E., Zagar R., Transcutaneous electrical neurostimulation in functional pain. Spine 6, 185–188 (1981).
    1. Woodbury C. J., Kullmann F. A., McIlwrath S. L., Koerber H. R., Identity of myelinated cutaneous sensory neurons projecting to nocireceptive laminae following nerve injury in adult mice. J. Comp. Neurol. 508, 500–509 (2008).
    1. Edin B. B., Quantitative analysis of static strain sensitivity in human mechanoreceptors from hairy skin. J. Neurophysiol. 67, 1105–1113 (1992).
    1. Trulsson M., Francis S. T., Kelly E. F., Westling G., Bowtell R., McGlone F., Cortical responses to single mechanoreceptive afferent microstimulation revealed with fMRI. Neuroimage 13, 613–622 (2001).
    1. Panchuelo R. M. S., Ackerley R., Glover P. M., Bowtell R. W., Wessberg J., Francis S. T., McGlone F., Mapping quantal touch using 7 Tesla functional magnetic resonance imaging and single-unit intraneural microstimulation. eLife 5, e12812 (2016).
    1. Sterman A. B., Schaumburg H. H., Asbury A. K., The acute sensory neuronopathy syndrome: A distinct clinical entity. Ann. Neurol. 7, 354–358 (1980).
    1. Herman W. H., Pop-Busui R., Braffett B. H., Martin C. L., Cleary P. A., Albers J. W., Feldman E. L.; DCCT/EDIC Research Group , Use of the Michigan neuropathy screening instrument as a measure of distal symmetrical peripheral neuropathy in type 1 diabetes: Results from the diabetes control and complications trial/epidemiology of diabetes interventions and complications. Diabet. Med. 29, 937–944 (2012).
    1. Edin B. B., Bäckström P. A., Bäckström L. O., Single unit retrieval in microneurography: A microprocessor-based device controlled by an operator. J. Neurosci. Methods 24, 137–144 (1988).

Source: PubMed

3
Subscribe