Transcriptional Programming of Human Mechanosensory Neuron Subtypes from Pluripotent Stem Cells

Alec R Nickolls, Michelle M Lee, David F Espinoza, Marcin Szczot, Ruby M Lam, Qi Wang, Jeanette Beers, Jizhong Zou, Minh Q Nguyen, Hans J Solinski, Aisha A AlJanahi, Kory R Johnson, Michael E Ward, Alexander T Chesler, Carsten G Bönnemann, Alec R Nickolls, Michelle M Lee, David F Espinoza, Marcin Szczot, Ruby M Lam, Qi Wang, Jeanette Beers, Jizhong Zou, Minh Q Nguyen, Hans J Solinski, Aisha A AlJanahi, Kory R Johnson, Michael E Ward, Alexander T Chesler, Carsten G Bönnemann

Abstract

Efficient and homogeneous in vitro generation of peripheral sensory neurons may provide a framework for novel drug screening platforms and disease models of touch and pain. We discover that, by overexpressing NGN2 and BRN3A, human pluripotent stem cells can be transcriptionally programmed to differentiate into a surprisingly uniform culture of cold- and mechano-sensing neurons. Although such a neuronal subtype is not found in mice, we identify molecular evidence for its existence in human sensory ganglia. Combining NGN2 and BRN3A programming with neural crest patterning, we produce two additional populations of sensory neurons, including a specialized touch receptor neuron subtype. Finally, we apply this system to model a rare inherited sensory disorder of touch and proprioception caused by inactivating mutations in PIEZO2. Together, these findings establish an approach to specify distinct sensory neuron subtypes in vitro, underscoring the utility of stem cell technology to capture human-specific features of physiology and disease.

Keywords: DRG; PIEZO2; TRPM8; differentiation; human; iPSC; mechanosensation; neural crest; neuron; sensory.

Conflict of interest statement

Declaration of Interests The authors declare no competing interests.

Published by Elsevier Inc.

Figures

Figure 1.. NGN2-BRN3A Programming of Human iPSCs…
Figure 1.. NGN2-BRN3A Programming of Human iPSCs Efficiently Yields Induced Sensory Neurons
(A) Protocol for sensory neuron induction using NGN2-BRN3A-engineered iPSCs. Phase-contrast images were captured on days 2, 4, 8, and 14 after doxycycline addition. Neurons induced with NGN2 alone are shown on the bottom for comparison. (B) Phase-contrast and immunocytochemistry images of day 21 neurons induced by NGN2-BRN3A and NGN2 only. (C) Immunocytochemistry of day 21 NGN2-BRN3A neurons to detect proteins found in sensory neurons. (D) Quantification of percent staining NeuN (78.9% ± 2.6%), BRN3A/NeuN (82.0% ± 1.7%), and ISL1/NeuN (90.1% ± 1.0%). For NeuN stains, n = 6 independent coverslips were used and were split for co-staining into n = 3 coverslips for BRN3A and n = 3 for ISL1. At least 200 cells were counted per stain. Values are expressed as mean ± SEM. Scale bars, 100 μm. Δ, medium change; 1/2Δ, half medium change; NTF, neurotrophic factor; Y-27632, ROCK inhibitor. See also Figure S1 and Videos S1 and S2.
Figure 2.. iSNs and Human DRG Neurons…
Figure 2.. iSNs and Human DRG Neurons Co-express TRPM8 and PIEZO2
(A) Heatmap of RNA sequencing results on iPSCs, iSNs, and adult human donor DRG (hDRG). Each column represents an individual sample’s normalized log2 transcripts per million (TPM). The hDRG is a pooled RNA sample from 21 individuals. (B) RNA in situ hybridization on iSNs for transcripts of neurotrophin and sensory receptors. Each small fluorescent punctum roughly indicates one transcript. TUBB3 encodes a general neuron class of microtubule, βIII-tubulin. Scale bars, 10 μm. (C) Quantification of in situ hybridization in iSNs using a cutoff criteria of ≥ 5 puncta per transcript for positive cells. TRPM8+ (17.9% ± 1.5%), PIEZO2+ (14.0% ± 4.6%), TRPM8+/PIEZO2+ (56.0% ± 6.3%), and TUBB3+ only (11.5% ± 3.4%). 199 cells were counted across n = 5 coverslips. (D) RNA in situ hybridization for TRPM8 and PIEZO2 in human DRG (hDRG). Labeled neurons are shown in wide-field view (left) with dotted, dashed, and solid circles indicating TRPM8+, PIEZO2+, and TRPM8+/PIEZO2+ neurons, respectively. Zoomed images of single neurons (right), individually marked by numbers in the wide-field view. Scale bars, 25 μm. (E) Quantification of human DRG in situ hybridization. TRPM8+ (12.8% ± 4.4%), PIEZO2+ (59.7% ± 6.8%), and TRPM8+/PIEZO2+ (27.4% ± 6.9%). 622 positive cells were counted across n = 4 DRG donors. All values are expressed as mean ± SEM. See also Figure S2 and Table S1.
Figure 3.. iSNs Detect Cold Temperature and…
Figure 3.. iSNs Detect Cold Temperature and Mechanical Force
(A) Fluo-4 fluorescence images recording iSNs at 25°C and 4°C. The TRPM8 inhibitor RQ-00203078 was incubated at 10 μM for 5 min before resuming recording. A 5-min room temperature incubation was used in between each 4°C treatment. Scale bar, 500 μm. (B) Representative fluorescence traces of calcium imaging trials, with 5 cells shown per trial. Values are graphed as the change in fluorescence intensity from baseline and divided by the baseline (ΔF/F0). 100 μM AITC, 10 μM capsaicin, 500 μM menthol, and 50 μM α,β-methylene-ATP (α,β-meATP) were used. (C) Scatter dot plots of peak ΔF/F0 of individual cells after application of temperature or chemicals. Each cell is normalized to a percentage of its own KCl response at the end of the recording period. For each condition, ≥ 80 cells were analyzed. Post hoc comparisons of each treatment to vehicle with one-way ANOVA and Dunnett’s correction for multiple comparisons. p values: 50°C, p = 0.99; 4°C pre, p = 0.0001; 4°C RQ, p = 0.5135; 4°C post, p = 0.001; AITC, p = 0.1379; capsaicin, p = 0.5242; menthol, p = 0.0001; α,β-meATP, p = 0.0001. ****p < 0.0001. (D) Example whole-cell current-clamp recording of iSNs in response to depolarizing currents. A total of n = 4 cells were recorded without tetrodotoxin (TTX) andn = 5 cells with 1 μM TTX. (E) Mechanical stimulation of iSNs in whole-cell voltage-clamp mode. In the phase-contrast image, the recording pipette is on the left, and the stimulator probe is on the right. Scale bar, 20 μm. In the recording, the top trace indicates micrometer steps of the stimulator probe, whereas the bottom trace shows whole-cell currents. (F) Quantification of the current amplitude at each probe indentation depth. A total of n = 10 cells were recorded, with 10/10 cells displaying a peak mechanically evoked current above 50 pA. Values are expressed as mean ± SEM. See also Videos S3 and S4.
Figure 4.. Varied Induction of NGN2-BRN3A in…
Figure 4.. Varied Induction of NGN2-BRN3A in Neural Crest Yields Divergent Sensory Neuron Populations
(A) Protocol and phase-contrast images of neural crest differentiation with two strategies for sensory neuron induction (NC-iSN1 and NC-iSN2). Day 0 depicts neurectodermal spheroid formation in a microwell plate. Day 12 shows neural crest migration from an individual spheroid. Scale bars, 100 μm. (B) Quantification of cell soma area based on phase-contrast images across three differentiation experiments of iSNs (n = 301 cells), NC-iSN1s (n = 228 cells), and NC-iSN2s (n = 332 cells). Values are expressed as mean ± SEM. Post hoc comparisons with Kruskal-Wallis test and Dunn’s correction for multiple comparisons, ****p in situ hybridization experiments on NC-iSN1s and NC-iSN2s. Scale bars, 25 μm. (D) Representative mechanical stimulation of NC-iSN2s in whole-cell voltage-clamp. A total of n = 6 cells were recorded. (E) RNA sequencing heatmap expression results of selected genes grouped by known sensory neuron subtype. Each heatmap column depicts the average log2 TPM of three independent samples. Δ, medium change; 1/2Δ, half medium change; C-LTMR, C fiber LTMR; LTMR, low-threshold mechanoreceptor; NC, neural crest; NP, nonpeptidergic nociceptor; PEP, peptidergic nociceptor; Proprio., proprioceptor. See also Figure S3.
Figure 5.. Single-Cell RNA Sequencing of Neural…
Figure 5.. Single-Cell RNA Sequencing of Neural Crest-Derived iSNs
(A) Merged UMAP plot representing individual cells from a single differentiation of both NC-iSN1 (22,804 cells) and NC-iSN2 (15,004 cells). (B) Expression UMAP plots indicating cells in the upper 90% of the population distribution with mapped reads to specific genes, based on unique molecular identifier counts.
Figure 6.. PIEZO2 LOF iSNs Are Insensitive…
Figure 6.. PIEZO2LOF iSNs Are Insensitive to Mechanical Stimuli
(A) Representative immunocytochemistry from at least three differentiations of control and patient iSNs. Scale bar, 50 μm. (B) qRT-PCR of sensory-related genes in day 21 iSNs, normalized to the housekeeping gene RPLP0 and expressed as −ΔCt for color coding. Each column represents an independent sample, three samples per cell line. Human DRG (hDRG) total RNA was used as a positive control, and WTC11 iPSCs were used as a negative control. Text color denotes gene category: orange, pluripotency; magenta, pan-sensory; purple, LTMR; blue, proprioceptor; green, nociceptor and/or thermoreceptor. (C) Whole-cell voltage-clamp recordings of iSNs during mechanical stimulation. Traces are examples from n = 10 neurons for each cell line. (D) Quantification of mechanically activated current peak amplitudes in n = 10 cells per cell line. Values are expressed as mean ± SEM. Post hoc comparisons with Kruskal-Wallis test and Dunn’s correction for multiple comparisons. *p F/F0 of individual cells after exposure to 500 μM menthol, normalized to KCl response. For each cell line, ≥80 cells were analyzed. In post hoc comparisons between each sample with one-way ANOVA and Dunnett’s correction for multiple comparisons, no comparison was significantly different (p > 0.05). See also Figures S4–S7 and Table S2.

References

    1. Adriaensen H, Gybels J, Handwerker HO, and Van Hees J (1983). Response properties of thin myelinated (A-delta) fibers in human skin nerves. J. Neurophysiol 49, 111–122.
    1. Airaksinen MS, Koltzenburg M, Lewin GR, Masu Y, Helbig C, Wolf E, Brem G, Toyka KV, Thoenen H, and Meyer M (1996). Specific subtypes of cutaneous mechanoreceptors require neurotrophin-3 following peripheral target innervation. Neuron 16, 287–295.
    1. Alshawaf AJ, Viventi S, Qiu W, D’Abaco G, Nayagam B, Erlichster M, Chana G, Everall I, Ivanusic J, Skafidas E, and Dottori M (2018). Phenotypic and Functional Characterization of Peripheral Sensory Neurons derived from Human Embryonic Stem Cells. Sci. Rep 8, 603.
    1. Arikawa K, Molday LL, Molday RS, and Williams DS (1992). Localization of peripherin/rds in the disk membranes of cone and rod photoreceptors: relationship to disk membrane morphogenesis and retinal degeneration. J. Cell Biol 116, 659–667.
    1. Bai L, Lehnert BP, Liu J, Neubarth NL, Dickendesher TL, Nwe PH, Cassidy C, Woodbury CJ, and Ginty DD (2015). Genetic Identification of an Expansive Mechanoreceptor Sensitive to Skin Stroking. Cell 163, 1783–1795.
    1. Bajpai R, Chen DA, Rada-Iglesias A, Zhang J, Xiong Y, Helms J, Chang CP, Zhao Y, Swigut T, and Wysocka J (2010). CHD7 cooperates with PBAF to control multipotent neural crest formation. Nature 463, 958–962.
    1. Bandell M, Story GM, Hwang SW, Viswanath V, Eid SR, Petrus MJ, Earley TJ, and Patapoutian A (2004). Noxious cold ion channel TRPA1 is activated by pungent compounds and bradykinin. Neuron 41, 849–857.
    1. Beers J, Linask KL, Chen JA, Siniscalchi LI, Lin Y, Zheng W, Rao M, and Chen G (2015). A cost-effective and efficient reprogramming platform for large-scale production of integration-free human induced pluripotent stem cells in chemically defined culture. Sci. Rep. 5, 11319.
    1. Bel-Vialar S, Itasaki N, and Krumlauf R (2002). Initiating Hox gene expression: in the early chick neural tube differential sensitivity to FGF and RA signaling subdivides the HoxB genes in two distinct groups. Development 129, 5103–5115.
    1. Benchetrit H, Jaber M, Zayat V, Sebban S, Pushett A, Makedonski K, Zakheim Z, Radwan A, Maoz N, Lasry R, et al. (2019). Direct Induction of the Three Pre-implantation Blastocyst Cell Types from Fibroblasts. Cell Stem Cell 24, 983–994.e7.
    1. Blanchard JW, Eade KT, Szücs A, Lo Sardo V, Tsunemoto RK, Williams D, Sanna PP, and Baldwin KK (2015). Selective conversion of fibroblasts into peripheral sensory neurons. Nat. Neurosci 18, 25–35.
    1. Bourane S, Garces A, Venteo S, Pattyn A, Hubert T, Fichard A, Puech S, Boukhaddaoui H, Baudet C, Takahashi S, et al. (2009). Low-threshold mechanoreceptor subtypes selectively express MafA and are specified by Ret signaling. Neuron 64, 857–870.
    1. Butler A, Hoffman P, Smibert P, Papalexi E, and Satija R (2018). Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol 36, 411–420.
    1. Cahusac PMB, and Noyce R (2007). A pharmacological study of slowly adapting mechanoreceptors responsive to cold thermal stimulation. Neuroscience 148, 489–500.
    1. Carroll P, Lewin GR, Koltzenburg M, Toyka KV, and Thoenen H (1998). A role for BDNF in mechanosensation. Nat. Neurosci 1, 42–46.
    1. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, and Julius D (1997). The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 389, 816–824.
    1. Cerbini T, Funahashi R, Luo Y, Liu C, Park K, Rao M, Malik N, and Zou J (2015). Transcription activator-like effector nuclease (TALEN)-mediated CLYBL targeting enables enhanced transgene expression and one-step generation of dual reporter human induced pluripotent stem cell (iPSC) and neural stem cell (NSC) lines. PLoS ONE 10, e0116032.
    1. Chambers SM, Qi Y, Mica Y, Lee G, Zhang X-J, Niu L, Bilsland J, Cao L, Stevens E, Whiting P, et al. (2012). Combined small-molecule inhibition accelerates developmental timing and converts human pluripotent stem cells into nociceptors. Nat. Biotechnol 30, 715–720.
    1. Chesler AT, Szczot M, Bharucha-Goebel D, Čeko M, Donkervoort S, Laubacher C, Hayes LH, Alter K, Zampieri C, Stanley C, et al. (2016). The Role of PIEZO2 in Human Mechanosensation. N. Engl. J. Med 375, 1355–1364.
    1. Chiu IM, Barrett LB, Williams EK, Strochlic DE, Lee S, Weyer AD, Lou S, Bryman GS, Roberson DP, Ghasemlou N, et al. (2014). Transcriptional profiling at whole population and single cell levels reveals somatosensory neuron molecular diversity. eLife 3, e04660.
    1. Cook SP, Vulchanova L, Hargreaves KM, Elde R, and McCleskey EW (1997). Distinct ATP receptors on pain-sensing and stretch-sensing neurons. Nature 387, 505–508.
    1. Das AT, Tenenbaum L, and Berkhout B (2016). Tet-On Systems For Doxycycline-inducible Gene Expression. Curr. Gene Ther 16, 156–167.
    1. Davidson S, Copits BA, Zhang J, Page G, Ghetti A, and Gereau RW (2014). Human sensory neurons: Membrane properties and sensitization by inflammatory mediators. Pain 155, 1861–1870.
    1. Del Mármol JI, Touhara KK, Croft G, and MacKinnon R (2018). Piezo1 forms a slowly-inactivating mechanosensory channel in mouse embryonic stem cells. eLife 7, e33149.
    1. Dhaka A, Earley TJ, Watson J, and Patapoutian A (2008). Visualizing cold spots: TRPM8-expressing sensory neurons and their projections. J. Neurosci. 28, 566–575.
    1. Dib-Hajj SD, Cummins TR, Black JA, and Waxman SG (2010). Sodium channels in normal and pathological pain. Annu. Rev. Neurosci 33, 325–347.
    1. Dussor G, and Cao YQ (2016). TRPM8 and Migraine. Headache 56, 1406–1417.
    1. Dykes IM, Tempest L, Lee S-I, and Turner EE (2011). Brn3a and Islet1 act epistatically to regulate the gene expression program of sensory differentiation. J. Neurosci 31, 9789–9799.
    1. Eijkelkamp N, Linley JE, Torres JM, Bee L, Dickenson AH, Gringhuis M, Minett MS, Hong GS, Lee E, Oh U, et al. (2013). A role for Piezo2 in EPAC1-dependent mechanical allodynia. Nat. Commun 4, 1682.
    1. Fang X, Djouhri L, McMullan S, Berry C, Okuse K, Waxman SG, and Lawson SN (2005). trkA is expressed in nociceptive neurons and influences electrophysiological properties via Nav1.8 expression in rapidly conducting nociceptors. J. Neurosci 25, 4868–4878.
    1. Fariñas I, Wilkinson GA, Backus C, Reichardt LF, and Patapoutian A (1998). Characterization of neurotrophin and Trk receptor functions in developing sensory ganglia: direct NT-3 activation of TrkB neurons in vivo. Neuron 21, 325–334.
    1. Fernandopulle MS, Prestil R, Grunseich C, Wang C, Gan L, and Ward ME (2018). Transcription Factor-Mediated Differentiation of Human iPSCs into Neurons. Curr. Protoc. Cell Biol. 79, e51.
    1. Fornaro M, Lee JM, Raimondo S, Nicolino S, Geuna S, and Giacobini-Robecchi M (2008). Neuronal intermediate filament expression in rat dorsal root ganglia sensory neurons: an in vivo and in vitro study. Neuroscience 153, 1153–1163.
    1. Genç B, Ulupinar E, and Erzurumlu RS (2005). Differential Trk expression in explant and dissociated trigeminal ganglion cell cultures. J. Neurobiol 64, 145–156.
    1. Hensel H, and Zotterman Y (1951). The response of mechanoreceptors to thermal stimulation. J. Physiol 115, 16–24.
    1. Holford LC, Case P, and Lawson SN (1994). Substance P, neurofilament, peripherin and SSEA4 immunocytochemistry of human dorsal root ganglion neurons obtained from post-mortem tissue: a quantitative morphometric analysis. J. Neurocytol 23, 577–589.
    1. Iggo A, and Muir AR (1969). The structure and function of a slowly adapting touch corpuscle in hairy skin. J. Physiol 200, 763–796.
    1. Iggo A, and Ogawa H (1977). Correlative physiological and morphological studies of rapidly adapting mechanoreceptors in cat’s glabrous skin. J. Physiol 266, 275–296.
    1. Inoue K, Tsuda M, and Koizumi S (2003). ATP induced three types of pain behaviors, including allodynia. Drug Dev. Res 59, 56–63.
    1. Janssens A, Gees M, Toth BI, Ghosh D, Mulier M, Vennekens R, Vriens J, Talavera K, and Voets T (2016). Definition of two agonist types at the mammalian cold-activated channel TRPM8. eLife 5, e17240.
    1. Jones I, Yelhekar TD, Wiberg R, Kingham PJ, Johansson S, Wiberg M, and Carlsson L (2018). Development and validation of an in vitro model system to study peripheral sensory neuron development and injury. Sci. Rep 8, 15961.
    1. Jordt SE, Bautista DM, Chuang HH, McKemy DD, Zygmunt PM, Hoögestä tt ED, Meng ID, and Julius D (2004). Mustard oils and cannabinoids excite sensory nerve fibres through the TRP channel ANKTM1. Nature 427, 260–265.
    1. Knowlton WM, Palkar R, Lippoldt EK, McCoy DD, Baluch F, Chen J, and McKemy DD (2013). A sensory-labeled line for cold: TRPM8-expressing sensory neurons define the cellular basis for cold, cold pain, and cooling-mediated analgesia. J. Neurosci 33, 2837–2848.
    1. Kobayashi K, Fukuoka T, Obata K, Yamanaka H, Dai Y, Tokunaga A, and Noguchi K (2005). Distinct expression of TRPM8, TRPA1, and TRPV1 mRNAs in rat primary afferent neurons with adelta/c-fibers and colocalization with trk receptors. J. Comp. Neurol 493, 596–606.
    1. Kumazawa T, and Perl ER (1977). Primate cutaneous sensory units with unmyelinated (C) afferent fibers. J. Neurophysiol 40, 1325–1338.
    1. Lallemend F, and Ernfors P (2012). Molecular interactions underlying the specification of sensory neurons. Trends Neurosci. 35, 373–381.
    1. Levanon D, Bettoun D, Harris-cerruti C, Woolf E, Negreanu V, Eilam R, Bernstein Y, Goldenberg D, Xiao C, Fliegauf M, et al. (2002). The Runx3 transcription factor regulates development and survival of TrkC dorsal root ganglia neurons. EMBO J. 21, 3454–3463.
    1. Li L, Rutlin M, Abraira VE, Cassidy C, Kus L, Gong S, Jankowski MP, Luo W, Heintz N, Koerber HR, et al. (2011). The functional organization of cutaneous low-threshold mechanosensory neurons. Cell 147, 1615–1627.
    1. Liu Z, Chen O, Wall JBJ, Zheng M, Zhou Y, Wang L, Ruth Vaseghi H, Qian L, and Liu J (2017). Systematic comparison of 2A peptides for cloning multi-genes in a polycistronic vector. Sci. Rep 7, 2193.
    1. Lo AWI, Sprung CN, Fouladi B, Pedram M, Sabatier L, Ricoul M, Reynolds GE, and Murnane JP (2002). Chromosome instability as a result of double-strand breaks near telomeres in mouse embryonic stem cells. Mol. Cell. Biol 22, 4836–4850.
    1. Luo W, Enomoto H, Rice FL, Milbrandt J, and Ginty DD (2009). Molecular identification of rapidly adapting mechanoreceptors and their developmental dependence on ret signaling. Neuron 64, 841–856.
    1. Ma Q, Fode C, Guillemot F, and Anderson DJ (1999). Neurogenin1 and neurogenin2 control two distinct waves of neurogenesis in developing dorsal root ganglia. Genes Dev. 13, 1717–1728.
    1. Marmigère F, and Ernfors P (2007). Specification and connectivity of neuronal subtypes in the sensory lineage. Nat. Rev. Neurosci. 8, 114–127.
    1. Mazzoni EO, Mahony S, Closser M, Morrison CA, Nedelec S, Williams DJ, An D, Gifford DK, and Wichterle H (2013). Synergistic binding of transcription factors to cell-specific enhancers programs motor neuron identity. Nat. Neurosci 16, 1219–1227.
    1. McKemy DD, Neuhausser WM, and Julius D (2002). Identification of a cold receptor reveals a general role for TRP channels in thermosensation. Nature 416, 52–58.
    1. Miyaoka Y, Chan AH, Judge LM, Yoo J, Huang M, Nguyen TD, Lizarraga PP, So PL, and Conklin BR (2014). Isolation of single-base genomeedited human iPS cells without antibiotic selection. Nat. Methods 11, 291–293.
    1. Moehring F, Halder P, Seal RP, and Stucky CL (2018). Uncovering the Cells and Circuits of Touch in Normal and Pathological Settings. Neuron 100, 349–360.
    1. Mu X, Silos-Santiago I, Carroll SL, and Snider WD (1993). Neurotrophin receptor genes are expressed in distinct patterns in developing dorsal root ganglia. J. Neurosci. 13, 4029–4041.
    1. Murthy SE, Loud MC, Daou I, Marshall KL, Schwaller F, Kühnemund J, Francisco AG, Keenan WT, Dubin AE, Lewin GR, et al. (2018). The mechanosensitive ion channel Piezo2 mediates sensitivity to mechanical pain in mice. Sci. Transl. Med 10, eaat9897.
    1. Nguyen MQ, Wu Y, Bonilla LS, von Buchholtz LJ, and Ryba NJP (2017). Diversity amongst trigeminal neurons revealed by high throughput single cell sequencing. PLoS ONE 12, e0185543.
    1. Peier AM, Moqrich A, Hergarden AC, Reeve AJ, Andersson DA, Story GM, Earley TJ, Dragoni I, McIntyre P, Bevan S, and Patapoutian A (2002). A TRP channel that senses cold stimuli and menthol. Cell 108, 705–715.
    1. Ranade SS, Woo SH, Dubin AE, Moshourab RA, Wetzel C, Petrus M, Mathur J, Bégay V, Coste B, Mainquist J, et al. (2014). Piezo2 is the major transducer of mechanical forces for touch sensation in mice. Nature 516, 121–125.
    1. Rodriguez-Tébar A, and Rohrer H (1991). Retinoic acid induces NGF-dependent survival response and high-affinity NGF receptors in immature chick sympathetic neurons. Development 112, 813–820.
    1. Romero LO, Massey AE, Mata-Daboin AD, Sierra-Valdez FJ, Chauhan SC, Cordero-Morales JF, and Vásquez V (2019). Dietary fatty acids finetune Piezo1 mechanical response. Nat. Commun. 10, 1200.
    1. Rostock C, Schrenk-Siemens K, Pohle J, and Siemens J (2018). Human vs. mouse nociceptors—similarities and differences. Neuroscience 387, 13–27.
    1. Schrenk-Siemens K, Wende H, Prato V, Song K, Rostock C, Loewer A, Utikal J, Lewin GR, Lechner SG, and Siemens J (2015). PIEZO2 is required for mechanotransduction in human stem cell-derived touch receptors. Nat. Neurosci 18, 10–16.
    1. Schwartzentruber J, Foskolou S, Kilpinen H, Rodrigues J, Alasoo K, Knights AJ, Patel M, Goncalves A, Ferreira R, Benn CL, et al.; HIPSCI Consortium (2018). Molecular and functional variation in iPSC-derived sensory neurons. Nat. Genet 50, 54–61.
    1. Sharp CJ, Reeve AJ, Collins SD, Martindale JC, Summerfield SG, Sargent BS, Bate ST, and Chessell IP (2006). Investigation into the role of P2X(3)/P2X(2/3) receptors in neuropathic pain following chronic constriction injury in the rat: an electrophysiological study. Br. J. Pharmacol. 148, 845–852.
    1. Sumino R, and Dubner R (1981). Response characteristics of specific thermoreceptive afferents innervating monkey facial skin and their relationship to human thermal sensitivity. Brain Res. Rev 3, 105–122.
    1. Szczot M, Pogorzala LA, Solinski HJ, Young L, Yee P, Le Pichon CE, Chesler AT, and Hoon MA (2017). Cell-Type-Specific Splicing of Piezo2 Regulates Mechanotransduction. Cell Rep. 21, 2760–2771.
    1. Szczot M, Liljencrantz J, Ghitani N, Barik A, Lam R, Thompson JH, Bharucha-Goebel D, Saade D, Necaise A, Donkervoort S, et al. (2018). PIEZO2 mediates injury-induced tactile pain in mice and humans. Sci. Transl. Med. 10, eaat9892.
    1. Takashima Y, Daniels RL, Knowlton W, Teng J, Liman ER, and McKemy DD (2007). Diversity in the neural circuitry of cold sensing revealed by genetic axonal labeling of transient receptor potential melastatin 8 neurons. J. Neurosci 27, 14147–14157.
    1. Tapper DN (1965). Stimulus-response relationships in the cutaneous slowlyadapting mechanoreceptor in hairy skin of the cat. Exp. Neurol 13, 364–385.
    1. Thaler J, Harrison K, Sharma K, Lettieri K, Kehrl J, and Pfaff SL (1999). Active suppression of interneuron programs within developing motor neurons revealed by analysis of homeodomain factor HB9. Neuron 23, 675–687.
    1. Usoskin D, Furlan A, Islam S, Abdo H, Lönnerberg P, Lou D, HjerlingLeffler J, Haeggström J, Kharchenko O, Kharchenko PV, et al. (2015). Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nat. Neurosci 18, 145–153.
    1. Vang H, Chung G, Kim HY, Park S-B, Jung SJ, Kim J-S, and Oh SB (2012). Neurochemical properties of dental primary afferent neurons. Exp. Neurobiol 21, 68–74.
    1. Velasco S, Ibrahim MM, Kakumanu A, Garipler G, Aydin B, Al-Sayegh MA, Hirsekorn A, Abdul-Rahman F, Satija R, Ohler U, et al. (2017). A Multi-step Transcriptional and Chromatin State Cascade Underlies Motor Neuron Programming from Embryonic Stem Cells. Cell Stem Cell 20, 205–217.e8.
    1. Wainger BJ, Buttermore ED, Oliveira JT, Mellin C, Lee S, Saber WA, Wang AJ, Ichida JK, Chiu IM, Barrett L, et al. (2015). Modeling pain in vitro using nociceptor neurons reprogrammed from fibroblasts. Nat. Neurosci 18, 17–24.
    1. Wang C, Ward ME, Chen R, Liu K, Tracy TE, Chen X, Xie M, Sohn PD, Ludwig C, Meyer-Franke A, et al. (2017). Scalable Production of iPSC-Derived Human Neurons to Identify Tau-Lowering Compounds by High-Content Screening. Stem Cell Reports 9, 1221–1233.
    1. Wapinski OL, Vierbuchen T, Qu K, Lee QY, Chanda S, Fuentes DR, Giresi PG, Ng YH, Marro S, Neff NF, et al. (2013). Hierarchical mechanisms for direct reprogramming of fibroblasts to neurons. Cell 155, 621–635.
    1. Weber EH (1851). Tastsinn und Gemeingefühl (Leipzig: Wilhelm Engelmann; ).
    1. Wende H, Lechner SG, Cheret C, Bourane S, Kolanczyk ME, Pattyn A, Reuter K, Munier FL, Carroll P, Lewin GR, and Birchmeier C (2012). The Transcription Factor c-Maf Controls Touch Receptor Development and Function. Science 335, 1373–1376.
    1. Woo S-H, Lukacs V, de Nooij JC, Zaytseva D, Criddle CR, Francisco A, Jessell TM, Wilkinson KA, and Patapoutian A (2015). Piezo2 is the principal mechanotransduction channel for proprioception. Nat. Neurosci 18, 1756–1762.
    1. Yang N, Chanda S, Marro S, Ng YH, Janas JA, Haag D, Ang CE, Tang Y, Flores Q, Mall M, et al. (2017). Generation of pure GABAergic neurons by transcription factor programming. Nat. Methods 14, 621–628.
    1. Zhang Y, Pak C, Han Y, Ahlenius H, Zhang Z, Chanda S, Marro S, Patzke C, Acuna C, Covy J, et al. (2013). Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78, 785–798.
    1. Zhang XL, Lee KY, Priest BT, Belfer I, and Gold MS (2015). Inflammatory mediator-induced modulation of GABAA currents in human sensory neurons. Neuroscience 310, 401–409.

Source: PubMed

3
Subscribe