1
|
Santiago C, Siegrist J, Africawala N, Handler A, Tasnim A, Anjum R, Turecek J, Lehnert BP, Renauld S, Choi J, Nolan-Tamariz M, Iskols M, Magee AR, Paradis S, Sharma N, Ginty DD. Activity-dependent development of the body's touch receptors. Neuron 2025:S0896-6273(25)00298-3. [PMID: 40381613 DOI: 10.1016/j.neuron.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/12/2025] [Accepted: 04/16/2025] [Indexed: 05/20/2025]
Abstract
We report a role for activity in the development of the primary sensory neurons that detect touch. Genetic deletion of Piezo2, the principal mechanosensitive ion channel in somatosensory neurons, caused profound changes in the formation of mechanosensory end-organ structures. Peripheral-nervous-system-specific deletion of the voltage-gated sodium channel Nav1.6 (Scn8a), which resulted in altered electrophysiological responses to mechanical stimuli, also disrupted somatosensory neuron morphologies, supporting a role for neuronal activity in end-organ formation. Single-cell RNA sequencing of Piezo2 mutants revealed changes in gene expression in sensory neurons activated by light mechanical forces, whereas other neuronal classes were minimally affected, and genetic deletion of Piezo2-dependent genes partially reproduced the defects in mechanosensory neuron structures observed in Piezo2 mutants. These findings indicate that mechanically evoked neuronal activity acts early in life to shape the maturation of mechanosensory end-organs that underlie our sense of gentle touch.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Julianna Siegrist
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nusrat Africawala
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brendan P Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sophia Renauld
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jinheon Choi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra R Magee
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Nikhil Sharma
- Department of Biochemistry and Molecular Biophysics and Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Geng D, Li Y, Yang B, Zhang L, Gu H, Zhang T, Zhao Z, Liu H, Cui Q, Zheng R, Cao P, Zhang F. Cholecystokinin neurons in the spinal trigeminal nucleus interpolaris regulate mechanically evoked predatory hunting in male mice. Nat Commun 2025; 16:2544. [PMID: 40087271 PMCID: PMC11909130 DOI: 10.1038/s41467-025-57771-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Predatory hunting plays a critical role in animal survival. Motion-related vibrissal somatosensory signaling is essential for prey detection and hunting in mice. However, little is known about the neural circuits that convert vibrissal somatosensory cues to trigger predatory hunting. Here, we report that mechanical force onto the vibrissal area of the male mice is a key stimulus for predatory hunting. Mechanically evoked predatory hunting was abrogated by the chemogenetic inactivation of cholecystokinin-positive (Cck+) neurons in the spinal trigeminal nucleus interpolaris (Sp5I). The Cck+ Sp5I neurons responded to the intensity of mechanical stimulus and sent neural signals to the superior colliculus that were relevant to stereotypical predatory hunting motor actions. Synaptic inactivation of the projections from Cck+ Sp5I neurons to the superior colliculus impaired mechanically evoked predatory attacks. Together, these data reveal a spinal trigeminal nucleus neural circuit that is specifically engaged in translating vibrissal somatosensory cues to provoke predatory hunting.
Collapse
Affiliation(s)
- Dandan Geng
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Yaning Li
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Bo Yang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Li Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Huating Gu
- National Institute of Biological Sciences, Beijing, China
| | - Tianyun Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Zijie Zhao
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Hui Liu
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Qingzhuo Cui
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Rong Zheng
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Peng Cao
- National Institute of Biological Sciences, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| | - Fan Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of Vascular Biology of Hebei Province; Hebei Medical University, Shijiazhuang, China.
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
3
|
Tran EL, Stuedemann SA, Ridlon M, Link OD, Keil Stietz KP, Crawford LK. Genetic tools that target mechanoreceptors produce reliable labeling of bladder afferents and altered mechanosensation. Am J Physiol Renal Physiol 2025; 328:F360-F374. [PMID: 39611874 DOI: 10.1152/ajprenal.00151.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/28/2024] [Accepted: 11/15/2024] [Indexed: 02/25/2025] Open
Abstract
Mechanosensitive neurons are important sensors of bladder distention, but their role in urologic disease remains unclear. Our current knowledge about how disease alters bladder sensation comes from studies that focus primarily on peptidergic nociceptors, leaving our understanding of neuropeptide-negative mechanoreceptors incomplete. In this study, we found that a substantial proportion of neurofilament heavy (NFH)-positive A-fibers innervating the bladder was calcitonin gene-related peptide (CGRP)-negative, potentially representing uncharacterized mechanoreceptors. We then identified two genetic strategies that label mechanoreceptors in mouse skin and confirmed that they likewise label bladder afferents. Cre-mediated tdTomato reporter expression driven by tropomyosin receptor kinase B (TrkB), which labels Aδ mechanoreceptors in the skin, successfully labeled bladder nerve terminals. The majority of TrkB bladder afferents were CGRP-negative and NFH-positive, with more characteristic staining patterns seen at the level of the cell body. The Ret proto-oncogene (Ret) also produced robust labeling of bladder afferents, where colocalization with CGRP and NFH was consistent with multiple afferent subtypes. Because TrkB labeling was more specific for putative mechanoreceptors, we directly tested the role of TrkB neurons in bladder mechanosensation in vivo. Using an intersectional genetic strategy, we selectively ablated TrkB afferents and measured bladder responses to mechanical distention using anesthetized cystometry. Compared with controls, mice with ablated TrkB afferents required higher distention pressure to elicit voids. Interestingly, after ablation, distention also increased the frequency of nonvoiding contractions, a poorly understood phenotype of several urologic diseases. These genetic strategies comprise critical new tools to advance the study of mechanoreceptors in bladder function and urologic disease pathophysiology.NEW & NOTEWORTHY Most mechanosensitive afferents do not express markers of peptidergic nociceptors and therefore remain largely overlooked in studies of bladder dysfunction and disease. TrkB-mediated labeling of putative Aδ mechanoreceptors emerged as a valuable tool for the study of neuropeptide-negative bladder afferents with a confirmed role in bladder mechanosensation. Targeted neuronal ablation likewise validated an intersectional genetic strategy that can now directly test the role of TrkB mechanoreceptors in bladder physiology and disease.
Collapse
Affiliation(s)
- Emily L Tran
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - Sara A Stuedemann
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - Monica Ridlon
- Department of Comparative Biosciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - Olivia D Link
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - Kimberly P Keil Stietz
- Department of Comparative Biosciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - LaTasha K Crawford
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| |
Collapse
|
4
|
Semizoglou E, Lo Re L, Middleton SJ, Perez‐Sanchez J, Tufarelli T, Bennett DL, Chisholm KI. In vivo calcium imaging reveals directional sensitivity of C-low threshold mechanoreceptors. J Physiol 2025; 603:895-908. [PMID: 39810695 PMCID: PMC11826069 DOI: 10.1113/jp286631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
C-low threshold mechanoreceptors (C-LTMRs) in animals (termed C-tactile (CT) fibres in humans) are a subgroup of C-fibre primary afferents, which innervate hairy skin and respond to low-threshold punctate indentations and brush stimuli. These afferents respond to gentle touch stimuli and are implicated in mediating pleasant/affective touch. These afferents have traditionally been studied using low-throughput, technically challenging approaches, including microneurography in humans and teased fibre electrophysiology in other mammals. Here we suggest a new approach to studying genetically labelled C-LTMRs using in vivo calcium imaging. We used an automated rotating brush stimulus and von Frey filaments, applied to the hairy skin of anaesthetized mice to mirror light and affective touch. Simultaneously we visualized changes in C-LTMR activity and confirmed that these neurons are sensitive to low-threshold punctate mechanical stimuli and brush stimuli with a strong preference for slow brushing speeds. We also reveal that C-LMTRs are directionally sensitive, showing more activity when brushed against the natural orientation of the hair. We present in vivo calcium imaging of genetically labelled C-LTMRs as a useful approach that can reveal new aspects of C-LTMR physiology. KEY POINTS: C-low threshold mechanoreceptors are sensitive to the directionality of a brush stimulus, being preferentially activated by brushing against the grain of the hair, compared with brushing with the grain of the hair. This is surprising as brushing against the grain of the hair is considered less pleasant. In vivo calcium imaging is a useful approach to the study of C-low threshold mechanoreceptors. While viral transfection, using systemic AAV9, is effective in labelling most sensory neuron populations in the dorsal root ganglion, it fails to label C-low threshold mechanoreceptors.
Collapse
Affiliation(s)
- Evangelia Semizoglou
- Department of NeurologyBrigham & Women's Hospital, Harvard Medical SchoolBostonUSA
| | - Laure Lo Re
- Tafalgie TherapeuticsCampus de LuminyMarseilleFrance
| | | | | | - Tommaso Tufarelli
- School of Mathematical SciencesThe University of NottinghamUniversity ParkNottinghamUK
| | - David L. Bennett
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Kim I. Chisholm
- School of Life SciencesThe University of NottinghamQueen's Medical CentreNottinghamUK
| |
Collapse
|
5
|
Yu S, Zhang X, Sun YG. Peripheral and central innervation pattern of mechanosensory neurons in the trigeminal ganglion. Neuroscience 2025; 565:558-566. [PMID: 39643235 DOI: 10.1016/j.neuroscience.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
The trigeminal ganglion (TG) comprises primary sensory neurons responsible for orofacial sensations, subsequently projecting to the trigeminal nuclei in the brainstem. However, the circuit basis of nasal mechanosensation is not well characterized. Here we elucidate the anatomical organization of both peripheral and central projections of the TG. We found that the non-peptidergic nociceptor, MAS-related G protein-coupled receptor member D positive (MrgprD+) neurons in the TG densely innervate the nasal mucosa, whereas the low-threshold mechanoreceptors subtypes rarely innervate the nasal mucosa. We also identified the central projection pattern of the mechanosensory neurons in TG. The tyrosine kinase receptor C positive (TrkC+) neurons, tyrosine kinase receptor B positive (TrkB+) and tyrosine hydroxylase positive (TH+) neurons project to multiple subregions of brainstem trigeminal complex and solitary nucleus. In contrast, the MrgprD+ neurons only densely project to outer edge of Sp5C. In addition, we further determined the ascending pathway of the TG neurons. Taken together, our study demonstrates the peripheral and central projection pattern of mechanosensory neurons in the TG, which provides a basis for the future functional studies.
Collapse
Affiliation(s)
- Su Yu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xinyan Zhang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan-Gang Sun
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
6
|
Gerhardt B, Alfken J, Reichmann J, Salditt T, Brecht M. Three-dimensional architecture and linearized mapping of vibrissa follicle afferents. Nat Commun 2025; 16:499. [PMID: 39779697 PMCID: PMC11711312 DOI: 10.1038/s41467-024-55468-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Understanding vibrissal transduction has advanced by serial sectioning and identified afferent recordings, but afferent mapping onto the complex, encapsulated follicle remains unclear. Here, we reveal male rat C2 vibrissa follicle innervation through synchrotron X-ray phase contrast tomograms. Morphological analysis identified 5% superficial, ~32 % unmyelinated and 63% myelinated deep vibrissal nerve axons. Myelinated afferents consist of each one third Merkel and club-like, and one sixth Ruffini-like and lanceolate endings. Unsupervised clustering of afferent properties aligns with classic morphological categories and revealed previously unrecognized club-like afferent subtypes distinct in axon diameter and Ranvier internode distance. Myelination and axon diameters indicate a proximal-to-distal axon-velocity gradient along the follicle. Axons innervate preferentially dorso-caudally to the vibrissa, presumably to sample contacts from vibrissa protraction. Afferents organize in axon-arms innervating discrete angular territories. The radial axon-arm arrangement around the vibrissa maps into a linear representation of axon-arm bands in the nerve. Such follicle linearization presumably instructs downstream linear brainstem barrelettes. Synchrotron imaging provides a synopsis of afferents and mechanotransductory machinery.
Collapse
Affiliation(s)
- Ben Gerhardt
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jette Alfken
- Institut für Röntgenphysik, Universität Göttingen, Göttingen, Germany
| | - Jakob Reichmann
- Institut für Röntgenphysik, Universität Göttingen, Göttingen, Germany
| | - Tim Salditt
- Institut für Röntgenphysik, Universität Göttingen, Göttingen, Germany
| | - Michael Brecht
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
- NeuroCure Cluster of Excellence, Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
7
|
Jang K, Garraway SM. TrkB Agonist (7,8-DHF)-Induced Responses in Dorsal Root Ganglia Neurons Are Decreased after Spinal Cord Injury: Implication for Peripheral Pain Mechanisms. eNeuro 2025; 12:ENEURO.0219-24.2024. [PMID: 39753357 PMCID: PMC11728855 DOI: 10.1523/eneuro.0219-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 11/08/2024] [Accepted: 12/04/2024] [Indexed: 01/15/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrkB) are known to contribute to both protective and pronociceptive processes. However, their contribution to neuropathic pain after spinal cord injury (SCI) needs further investigation. In a recent study utilizing TrkBF616A mice, it was shown that systemic pharmacogenetic inhibition of TrkB signaling with 1NM-PP1 (1NMP) immediately after SCI delayed the onset of pain hypersensitivity, implicating maladaptive TrkB signaling in pain after SCI. To examine potential neural mechanisms underlying the behavioral outcome, patch-clamp recording was performed in small-diameter dissociated thoracic (T) dorsal root ganglia (DRG) neurons to evaluate TrkB signaling in uninjured mice and after T10 contusion SCI. Bath-applied 7,8-dihydroxyflavone (7,8-DHF), a selective TrkB agonist, induced a robust inward current in neurons from uninjured mice, which was attenuated by 1NMP treatment. SCI also decreased 7,8-DHF-induced current while increasing the latency to its peak amplitude. Western blot revealed a concomitant decrease in TrkB expression in DRGs adjacent to the spinal lesion. Analyses of cellular and membrane properties showed that SCI increased neuronal excitability, evident by an increase in resting membrane potential and the number of spiking neurons. However, SCI did not increase spontaneous firing in DRG neurons. These results suggest that SCI induced changes in TrkB activation in DRG neurons even though these alterations are likely not contributing to pain hypersensitivity by nociceptor hyperexcitability. Overall, this reveals complex interactions involving TrkB signaling and provides an opportunity to investigate other, presumably peripheral, mechanisms by which TrkB contributes to pain hypersensitivity after SCI.
Collapse
Affiliation(s)
- Kyeongran Jang
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, Georgia 30322
| | - Sandra M Garraway
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
8
|
Ohman LC, Huang T, Unwin VA, Singh A, Walters B, Whiddon ZD, Krimm RF. Deciphering Peripheral Taste Neuron Diversity: Using Genetic Identity to Bridge Taste Bud Innervation Patterns and Functional Responses. J Neurosci 2024; 44:e0583242024. [PMID: 39379155 PMCID: PMC11561867 DOI: 10.1523/jneurosci.0583-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/19/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
Peripheral taste neurons exhibit functional, genetic, and morphological diversity, yet understanding how or if these attributes combine into taste neuron types remains unclear. In this study, we used male and female mice to relate taste bud innervation patterns to the function of a subset of proenkephalin-expressing (Penk+) taste neurons. We found that taste arbors (the portion of the axon within the taste bud) stemming from Penk+ neurons displayed diverse branching patterns and lacked stereotypical endings. The range in complexity observed for individual taste arbors from Penk+ neurons mirrored the entire population, suggesting that taste arbor morphologies are not primarily regulated by the neuron type. Notably, the distinguishing feature of arbors from Penk+ neurons was their propensity to come within 110 nm (in apposition with) different types of taste-transducing cells within the taste bud. This finding is contrary to the expectation of genetically defined taste neuron types that functionally represent a single stimulus. Consistently, further investigation of Penk+ neuron function revealed that they are more likely to respond to innately aversive stimuli-sour, bitter, and high salt concentrations-as compared with the full taste population. Penk+ neurons are less likely to respond to nonaversive stimuli-sucrose, umami, and low salt-compared with the full population. Our data support the presence of a genetically defined neuron type in the geniculate ganglion that is responsive to innately aversive stimuli. This implies that genetic expression might categorize peripheral taste neurons into hedonic groups, rather than simply identifying neurons that respond to a single stimulus.
Collapse
Affiliation(s)
- Lisa C Ohman
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Tao Huang
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Victori A Unwin
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Aditi Singh
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Brittany Walters
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Zachary D Whiddon
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| | - Robin F Krimm
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202
| |
Collapse
|
9
|
O’Brien J, Niehaus P, Chang K, Remark J, Barrett J, Dasgupta A, Adenegan M, Salimian M, Kevas Y, Chandrasekaran K, Kristian T, Chellappan R, Rubin S, Kiemen A, Lu CPJ, Russell JW, Ho CY. Skin keratinocyte-derived SIRT1 and BDNF modulate mechanical allodynia in mouse models of diabetic neuropathy. Brain 2024; 147:3471-3486. [PMID: 38554393 PMCID: PMC11449144 DOI: 10.1093/brain/awae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
Diabetic neuropathy is a debilitating disorder characterized by spontaneous and mechanical allodynia. The role of skin mechanoreceptors in the development of mechanical allodynia is unclear. We discovered that mice with diabetic neuropathy had decreased sirtuin 1 (SIRT1) deacetylase activity in foot skin, leading to reduced expression of brain-derived neurotrophic factor (BDNF) and subsequent loss of innervation in Meissner corpuscles, a mechanoreceptor expressing the BDNF receptor TrkB. When SIRT1 was depleted from skin, the mechanical allodynia worsened in diabetic neuropathy mice, likely due to retrograde degeneration of the Meissner-corpuscle innervating Aβ axons and aberrant formation of Meissner corpuscles which may have increased the mechanosensitivity. The same phenomenon was also noted in skin-keratinocyte specific BDNF knockout mice. Furthermore, overexpression of SIRT1 in skin induced Meissner corpuscle reinnervation and regeneration, resulting in significant improvement of diabetic mechanical allodynia. Overall, the findings suggested that skin-derived SIRT1 and BDNF function in the same pathway in skin sensory apparatus regeneration and highlighted the potential of developing topical SIRT1-activating compounds as a novel treatment for diabetic mechanical allodynia.
Collapse
Affiliation(s)
- Jennifer O’Brien
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Peter Niehaus
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, National Taiwan University, Taipei, 100, Taiwan
| | - Juliana Remark
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Joy Barrett
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Abhishikta Dasgupta
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Morayo Adenegan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21021, USA
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | - Samuel Rubin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemistry, College of William and Mary, Williamsburg, VA 23187, USA
| | - Ashley Kiemen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Catherine Pei-Ju Lu
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - James W Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Cheng-Ying Ho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
10
|
Desbois M, Grill B. Molecular regulation of axon termination in mechanosensory neurons. Development 2024; 151:dev202945. [PMID: 39268828 PMCID: PMC11698068 DOI: 10.1242/dev.202945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Spatially and temporally accurate termination of axon outgrowth, a process called axon termination, is required for efficient, precise nervous system construction and wiring. The mechanosensory neurons that sense low-threshold mechanical stimulation or gentle touch have proven exceptionally valuable for studying axon termination over the past 40 years. In this Review, we discuss progress made in deciphering the molecular and genetic mechanisms that govern axon termination in touch receptor neurons. Findings across model organisms, including Caenorhabditis elegans, Drosophila, zebrafish and mice, have revealed that complex signaling is required for termination with conserved principles and players beginning to surface. A key emerging theme is that axon termination is mediated by complex signaling networks that include ubiquitin ligase signaling hubs, kinase cascades, transcription factors, guidance/adhesion receptors and growth factors. Here, we begin a discussion about how these signaling networks could represent termination codes that trigger cessation of axon outgrowth in different species and types of mechanosensory neurons.
Collapse
Affiliation(s)
- Muriel Desbois
- School of Life Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98101, USA
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98101, USA
| |
Collapse
|
11
|
Bhuiyan SA, Xu M, Yang L, Semizoglou E, Bhatia P, Pantaleo KI, Tochitsky I, Jain A, Erdogan B, Blair S, Cat V, Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Green U, McIlvried LA, Copits BA, Bertels Z, Del Rosario JS, Widman AJ, Slivicki RA, Yi J, Sharif-Naeini R, Woolf CJ, Lennerz JK, Whited JL, Price TJ, Gereau RW, Renthal W. Harmonized cross-species cell atlases of trigeminal and dorsal root ganglia. SCIENCE ADVANCES 2024; 10:eadj9173. [PMID: 38905344 PMCID: PMC11804847 DOI: 10.1126/sciadv.adj9173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Sensory neurons in the dorsal root ganglion (DRG) and trigeminal ganglion (TG) are specialized to detect and transduce diverse environmental stimuli to the central nervous system. Single-cell RNA sequencing has provided insights into the diversity of sensory ganglia cell types in rodents, nonhuman primates, and humans, but it remains difficult to compare cell types across studies and species. We thus constructed harmonized atlases of the DRG and TG that describe and facilitate comparison of 18 neuronal and 11 non-neuronal cell types across six species and 31 datasets. We then performed single-cell/nucleus RNA sequencing of DRG from both human and the highly regenerative axolotl and found that the harmonized atlas also improves cell type annotation, particularly of sparse neuronal subtypes. We observed that the transcriptomes of sensory neuron subtypes are broadly similar across vertebrates, but the expression of functionally important neuropeptides and channels can vary notably. The resources presented here can guide future studies in comparative transcriptomics, simplify cell-type nomenclature differences across studies, and help prioritize targets for future analgesic development.
Collapse
Affiliation(s)
- Shamsuddin A. Bhuiyan
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mengyi Xu
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Alan Edwards Center for Research on Pain and Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Lite Yang
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Evangelia Semizoglou
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Parth Bhatia
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katerina I. Pantaleo
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ivan Tochitsky
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Aakanksha Jain
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Burcu Erdogan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Steven Blair
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Victor Cat
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Juliet M. Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Ursula Green
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114, USA
| | - Lisa A. McIlvried
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Bryan A. Copits
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Zachariah Bertels
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - John S. Del Rosario
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Allie J. Widman
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Richard A. Slivicki
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jiwon Yi
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Reza Sharif-Naeini
- Alan Edwards Center for Research on Pain and Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Clifford J. Woolf
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Jochen K. Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114, USA
| | - Jessica L. Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - William Renthal
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Qi L, Iskols M, Greenberg RS, Xiao JY, Handler A, Liberles SD, Ginty DD. Krause corpuscles are genital vibrotactile sensors for sexual behaviours. Nature 2024; 630:926-934. [PMID: 38898273 PMCID: PMC11208142 DOI: 10.1038/s41586-024-07528-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 05/07/2024] [Indexed: 06/21/2024]
Abstract
Krause corpuscles, which were discovered in the 1850s, are specialized sensory structures found within the genitalia and other mucocutaneous tissues1-4. The physiological properties and functions of Krause corpuscles have remained unclear since their discovery. Here we report the anatomical and physiological properties of Krause corpuscles of the mouse clitoris and penis and their roles in sexual behaviour. We observed a high density of Krause corpuscles in the clitoris compared with the penis. Using mouse genetic tools, we identified two distinct somatosensory neuron subtypes that innervate Krause corpuscles of both the clitoris and penis and project to a unique sensory terminal region of the spinal cord. In vivo electrophysiology and calcium imaging experiments showed that both Krause corpuscle afferent types are A-fibre rapid-adapting low-threshold mechanoreceptors, optimally tuned to dynamic, light-touch and mechanical vibrations (40-80 Hz) applied to the clitoris or penis. Functionally, selective optogenetic activation of Krause corpuscle afferent terminals evoked penile erection in male mice and vaginal contraction in female mice, while genetic ablation of Krause corpuscles impaired intromission and ejaculation of males and reduced sexual receptivity of females. Thus, Krause corpuscles of the clitoris and penis are highly sensitive mechanical vibration detectors that mediate sexually dimorphic mating behaviours.
Collapse
Affiliation(s)
- Lijun Qi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Rachel S Greenberg
- Department of Cell Biology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Jia Yin Xiao
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Stephen D Liberles
- Department of Cell Biology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Zhang L, Nagel M, Olson WP, Chesler AT, O'Connor DH. Trigeminal innervation and tactile responses in mouse tongue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.17.553449. [PMID: 37645855 PMCID: PMC10462066 DOI: 10.1101/2023.08.17.553449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The mammalian tongue is richly innervated with somatosensory, gustatory and motor fibers. These form the basis of many ethologically important functions such as eating, speaking and social grooming. Despite its high tactile acuity and sensitivity, the neural basis of tongue mechanosensation remains largely mysterious. Here we explored the organization of mechanosensory afferents in the tongue and found that each lingual papilla is innervated by Piezo2 + trigeminal neurons. Notably, each fungiform papilla contained highly specialized ring-like sensory neuron terminations that asymmetrically circumscribe the taste buds. Myelinated lingual afferents in the mouse lingual papillae did not form corpuscular sensory end organs but rather had only free nerve endings. In vivo single-unit recordings from the trigeminal ganglion revealed lingual low-threshold mechanoreceptors (LTMRs) with conduction velocities in the Aδ range or above and distinct adaptation properties ranging from intermediately adapting (IA) to rapidly adapting (RA). IA units were sensitive to both static indentation and stroking, while RA units had a preference for tangential forces applied by stroking. Lingual LTMRs were not directly responsive to rapid cooling or chemicals that can induce astringent or numbing sensations. Sparse labeling of lingual afferents in the tongue revealed distinct terminal morphologies and innervation patterns in fungiform and filiform papillae. Together, our results indicate that fungiform papillae are mechanosensory structures, while suggesting a simple model that links the functional and anatomical properties of tactile sensory neurons in the tongue.
Collapse
|
14
|
Jeon SM, Pradeep A, Chang D, McDonough L, Chen Y, Latremoliere A, Crawford LK, Caterina MJ. Skin Reinnervation by Collateral Sprouting Following Spared Nerve Injury in Mice. J Neurosci 2024; 44:e1494232024. [PMID: 38471780 PMCID: PMC11007315 DOI: 10.1523/jneurosci.1494-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/15/2024] [Accepted: 02/03/2024] [Indexed: 03/14/2024] Open
Abstract
Following peripheral nerve injury, denervated tissues can be reinnervated via regeneration of injured neurons or collateral sprouting of neighboring uninjured afferents into denervated territory. While there has been substantial focus on mechanisms underlying regeneration, collateral sprouting has received less attention. Here, we used immunohistochemistry and genetic neuronal labeling to define the subtype specificity of sprouting-mediated reinnervation of plantar hindpaw skin in the mouse spared nerve injury (SNI) model, in which productive regeneration cannot occur. Following initial loss of cutaneous afferents in the tibial nerve territory, we observed progressive centripetal reinnervation by multiple subtypes of neighboring uninjured fibers into denervated glabrous and hairy plantar skin of male mice. In addition to dermal reinnervation, CGRP-expressing peptidergic fibers slowly but continuously repopulated denervated epidermis, Interestingly, GFRα2-expressing nonpeptidergic fibers exhibited a transient burst of epidermal reinnervation, followed by a trend towards regression. Presumptive sympathetic nerve fibers also sprouted into denervated territory, as did a population of myelinated TrkC lineage fibers, though the latter did so inefficiently. Conversely, rapidly adapting Aβ fiber and C fiber low threshold mechanoreceptor (LTMR) subtypes failed to exhibit convincing sprouting up to 8 weeks after nerve injury in males or females. Optogenetics and behavioral assays in male mice further demonstrated the functionality of collaterally sprouted fibers in hairy plantar skin with restoration of punctate mechanosensation without hypersensitivity. Our findings advance understanding of differential collateral sprouting among sensory neuron subpopulations and may guide strategies to promote the progression of sensory recovery or limit maladaptive sensory phenomena after peripheral nerve injury.
Collapse
Affiliation(s)
- Sang-Min Jeon
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Aishwarya Pradeep
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Dennis Chang
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Leah McDonough
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Yijia Chen
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Alban Latremoliere
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - LaTasha K Crawford
- Department of Pathological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin 53706
| | - Michael J Caterina
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
15
|
Gautam M, Yamada A, Yamada AI, Wu Q, Kridsada K, Ling J, Yu H, Dong P, Ma M, Gu J, Luo W. Distinct local and global functions of mouse Aβ low-threshold mechanoreceptors in mechanical nociception. Nat Commun 2024; 15:2911. [PMID: 38575590 PMCID: PMC10995180 DOI: 10.1038/s41467-024-47245-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024] Open
Abstract
The roles of Aβ low-threshold mechanoreceptors (LTMRs) in transmitting mechanical hyperalgesia and in alleviating chronic pain have been of great interest but remain contentious. Here we utilized intersectional genetic tools, optogenetics, and high-speed imaging to specifically examine functions of SplitCre labeled mouse Aβ-LTMRs in this regard. Genetic ablation of SplitCre-Aβ-LTMRs increased mechanical nociception but not thermosensation in both acute and chronic inflammatory pain conditions, indicating a modality-specific role in gating mechanical nociception. Local optogenetic activation of SplitCre-Aβ-LTMRs triggered nociception after tissue inflammation, whereas their broad activation at the dorsal column still alleviated mechanical hypersensitivity of chronic inflammation. Taking all data into consideration, we propose a model, in which Aβ-LTMRs play distinctive local and global roles in transmitting or alleviating mechanical hyperalgesia of chronic pain, respectively. Our model suggests a strategy of global activation plus local inhibition of Aβ-LTMRs for treating mechanical hyperalgesia.
Collapse
Affiliation(s)
- Mayank Gautam
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Akihiro Yamada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ayaka I Yamada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Qinxue Wu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kim Kridsada
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jennifer Ling
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Huasheng Yu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Peter Dong
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Minghong Ma
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jianguo Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
16
|
Qi L, Iskols M, Shi D, Reddy P, Walker C, Lezgiyeva K, Voisin T, Pawlak M, Kuchroo VK, Chiu IM, Ginty DD, Sharma N. A mouse DRG genetic toolkit reveals morphological and physiological diversity of somatosensory neuron subtypes. Cell 2024; 187:1508-1526.e16. [PMID: 38442711 PMCID: PMC10947841 DOI: 10.1016/j.cell.2024.02.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 11/12/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024]
Abstract
Dorsal root ganglia (DRG) somatosensory neurons detect mechanical, thermal, and chemical stimuli acting on the body. Achieving a holistic view of how different DRG neuron subtypes relay neural signals from the periphery to the CNS has been challenging with existing tools. Here, we develop and curate a mouse genetic toolkit that allows for interrogating the properties and functions of distinct cutaneous targeting DRG neuron subtypes. These tools have enabled a broad morphological analysis, which revealed distinct cutaneous axon arborization areas and branching patterns of the transcriptionally distinct DRG neuron subtypes. Moreover, in vivo physiological analysis revealed that each subtype has a distinct threshold and range of responses to mechanical and/or thermal stimuli. These findings support a model in which morphologically and physiologically distinct cutaneous DRG sensory neuron subtypes tile mechanical and thermal stimulus space to collectively encode a wide range of natural stimuli.
Collapse
Affiliation(s)
- Lijun Qi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - David Shi
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Pranav Reddy
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Christopher Walker
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Karina Lezgiyeva
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Tiphaine Voisin
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Mathias Pawlak
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Vijay K Kuchroo
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| | - Nikhil Sharma
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
17
|
O’Brien J, Niehaus P, Chang K, Remark J, Barrett J, Dasgupta A, Adenegan M, Salimian M, Kevas Y, Chandrasekaran K, Kristian T, Chellappan R, Rubin S, Kiemen A, Lu CPJ, Russell JW, Ho CY. Skin keratinocyte-derived SIRT1 and BDNF modulate mechanical allodynia in mouse models of diabetic neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.24.523981. [PMID: 36747753 PMCID: PMC9900813 DOI: 10.1101/2023.01.24.523981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Diabetic neuropathy is a debilitating disorder characterized by spontaneous and mechanical pain. The role of skin mechanoreceptors in the development of mechanical pain (allodynia) is unclear. We discovered that mice with diabetic neuropathy had decreased sirtuin 1 (SIRT1) deacetylase activity in foot skin, leading to reduced expression of brain-derived neurotrophic factor (BDNF) and subsequent loss of innervation in Meissner corpuscles, a mechanoreceptor expressing the BDNF receptor TrkB. When SIRT1 was depleted from skin, the mechanical allodynia worsened in diabetic neuropathy mice, likely due to retrograde degeneration of the Meissner-corpuscle innervating Aβ axons and aberrant formation of Meissner corpuscles which may have increased the mechanosensitivity. The same phenomenon was also noted in skin BDNF knockout mice. Furthermore, overexpression of SIRT1 in skin induced Meissner corpuscle reinnervation and regeneration, resulting in significant improvement of diabetic mechanical allodynia. Overall, the findings suggested that skin-derived SIRT1 and BDNF function in the same pathway in skin sensory apparatus regeneration and highlighted the potential of developing topical SIRT1-activating compounds as a novel treatment for diabetic mechanical allodynia.
Collapse
Affiliation(s)
- Jennifer O’Brien
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Peter Niehaus
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Pathology, National Taiwan University, Taipei, 100, Taiwan
| | - Juliana Remark
- Hansj rg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Joy Barrett
- Hansj rg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Abhishikta Dasgupta
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Morayo Adenegan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Mohammad Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD, 21201, USA
| | - Tibor Kristian
- Baltimore Veterans Affairs Medical Center, Baltimore, MD, 21201, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL, 35233, USA
| | - Samuel Rubin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Chemistry, College of William and Mary, Williamsburg, VA, 23187, USA
| | - Ashley Kiemen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Catherine Pei-Ju Lu
- Hansj rg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - James W. Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD, 21201, USA
| | - Cheng-Ying Ho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
18
|
Chakrabarti S, Klich JD, Khallaf MA, Hulme AJ, Sánchez-Carranza O, Baran ZM, Rossi A, Huang ATL, Pohl T, Fleischer R, Fürst C, Hammes A, Bégay V, Hörnberg H, Finol-Urdaneta RK, Poole K, Dottori M, Lewin GR. Touch sensation requires the mechanically gated ion channel ELKIN1. Science 2024; 383:992-998. [PMID: 38422143 DOI: 10.1126/science.adl0495] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/26/2024] [Indexed: 03/02/2024]
Abstract
Touch perception is enabled by mechanically activated ion channels, the opening of which excites cutaneous sensory endings to initiate sensation. In this study, we identify ELKIN1 as an ion channel likely gated by mechanical force, necessary for normal touch sensitivity in mice. Touch insensitivity in Elkin1-/- mice was caused by a loss of mechanically activated currents (MA currents) in around half of all sensory neurons activated by light touch (low-threshold mechanoreceptors). Reintroduction of Elkin1 into sensory neurons from Elkin1-/- mice restored MA currents. Additionally, small interfering RNA-mediated knockdown of ELKIN1 from induced human sensory neurons substantially reduced indentation-induced MA currents, supporting a conserved role for ELKIN1 in human touch. Our data identify ELKIN1 as a core component of touch transduction in mice and potentially in humans.
Collapse
Affiliation(s)
- Sampurna Chakrabarti
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Jasmin D Klich
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Mohammed A Khallaf
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- Department of Zoology and Entomology, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Amy J Hulme
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Oscar Sánchez-Carranza
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Zuzanna M Baran
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- Molecular and Cellular Basis of Behavior, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Alice Rossi
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Angela Tzu-Lun Huang
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Tobias Pohl
- Molecular and Cellular Basis of Behavior, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Raluca Fleischer
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Carina Fürst
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- Molecular Pathways in Cortical Development, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Annette Hammes
- Molecular Pathways in Cortical Development, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Valérie Bégay
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
| | - Hanna Hörnberg
- Molecular and Cellular Basis of Behavior, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- NeuroCure Cluster of Excellence, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Rocio K Finol-Urdaneta
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Kate Poole
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mirella Dottori
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Gary R Lewin
- Molecular Physiology of Somatic Sensation Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin-Buch, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- German Center for Mental Health (DZPG), partner site Berlin, 10117 Berlin, Germany
| |
Collapse
|
19
|
Tasnim A, Alkislar I, Hakim R, Turecek J, Abdelaziz A, Orefice LL, Ginty DD. The developmental timing of spinal touch processing alterations predicts behavioral changes in genetic mouse models of autism spectrum disorders. Nat Neurosci 2024; 27:484-496. [PMID: 38233682 PMCID: PMC10917678 DOI: 10.1038/s41593-023-01552-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024]
Abstract
Altered somatosensory reactivity is frequently observed among individuals with autism spectrum disorders (ASDs). Here, we report that although multiple mouse models of ASD exhibit aberrant somatosensory behaviors in adulthood, some models exhibit altered tactile reactivity as early as embryonic development, whereas in others, altered reactivity emerges later in life. Additionally, tactile overreactivity during neonatal development is associated with anxiety-like behaviors and social behavior deficits in adulthood, whereas tactile overreactivity that emerges later in life is not. The locus of circuit disruption dictates the timing of aberrant tactile behaviors, as altered feedback or presynaptic inhibition of peripheral mechanosensory neurons leads to abnormal tactile reactivity during neonatal development, whereas disruptions in feedforward inhibition in the spinal cord lead to touch reactivity alterations that manifest later in life. Thus, the developmental timing of aberrant touch processing can predict the manifestation of ASD-associated behaviors in mouse models, and differential timing of sensory disturbance onset may contribute to phenotypic diversity across individuals with ASD.
Collapse
Affiliation(s)
- Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Ilayda Alkislar
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Richard Hakim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Amira Abdelaziz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Lauren L Orefice
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Zhang B, Chen T. Local and systemic mechanisms that control the hair follicle stem cell niche. Nat Rev Mol Cell Biol 2024; 25:87-100. [PMID: 37903969 DOI: 10.1038/s41580-023-00662-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 11/01/2023]
Abstract
Hair follicles are essential appendages of the mammalian skin, as hair performs vital functions of protection, thermoregulation and sensation. Hair follicles harbour exceptional regenerative abilities as they contain multiple somatic stem cell populations such as hair follicle stem cells (HFSCs) and melanocyte stem cells. Surrounding the stem cells and their progeny, diverse groups of cells and extracellular matrix proteins are organized to form a microenvironment (called 'niche') that serves to promote and maintain the optimal functioning of these stem cell populations. Recent studies have shed light on the intricate nature of the HFSC niche and its crucial role in regulating hair follicle regeneration. In this Review, we describe how the niche serves as a signalling hub, communicating, deciphering and integrating both local signals within the skin and systemic inputs from the body and environment to modulate HFSC activity. We delve into the recent advancements in identifying the cellular and molecular nature of the niche, providing a holistic perspective on its essential functions in hair follicle morphogenesis, regeneration and ageing.
Collapse
Affiliation(s)
- Bing Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Ting Chen
- National Institute of Biological Sciences, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
21
|
Niu C, Yue X, An JJ, Bass R, Xu H, Xu B. Genetic Dissection of BDNF and TrkB Expression in Glial Cells. Biomolecules 2024; 14:91. [PMID: 38254691 PMCID: PMC10813193 DOI: 10.3390/biom14010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase receptor B (TrkB) are widely expressed in the central nervous system. It is well documented that neurons express BDNF and full-length TrkB (TrkB.FL) as well as a lower level of truncated TrkB (TrkB.T). However, there are conflicting reports regarding the expression of BDNF and TrkB in glial cells, particularly microglia. In this study, we employed a sensitive and reliable genetic method to characterize the expression of BDNF and TrkB in glial cells in the mouse brain. We utilized three Cre mouse strains in which Cre recombinase is expressed in the same cells as BDNF, TrkB.FL, or all TrkB isoforms, and crossed them to Cre-dependent reporter mice to label BDNF- or TrkB-expressing cells with soma-localized EGFP. We performed immunohistochemistry with glial cell markers to examine the expression of BDNF and TrkB in microglia, astrocytes, and oligodendrocytes. Surprisingly, we found no BDNF- or TrkB-expressing microglia in examined CNS regions, including the somatomotor cortex, hippocampal CA1, and spinal cord. Consistent with previous studies, most astrocytes only express TrkB.T in the hippocampus of adult brains. Moreover, there are a small number of astrocytes and oligodendrocytes that express BDNF in the hippocampus, the function of which is to be determined. We also found that oligodendrocyte precursor cells, but not mature oligodendrocytes, express both TrkB.FL and TrkB.T in the hippocampus of adult mice. These results not only clarify the expression of BDNF and TrkB in glial cells but also open opportunities to investigate previously unidentified roles of BDNF and TrkB in astrocytes and oligodendrocytes.
Collapse
Affiliation(s)
- Changran Niu
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Xinpei Yue
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Juan Ji An
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
| | - Robert Bass
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Haifei Xu
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
| | - Baoji Xu
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458, USA
| |
Collapse
|
22
|
Jang K, Garraway SM. A review of dorsal root ganglia and primary sensory neuron plasticity mediating inflammatory and chronic neuropathic pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 15:100151. [PMID: 38314104 PMCID: PMC10837099 DOI: 10.1016/j.ynpai.2024.100151] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
Pain is a sensory state resulting from complex integration of peripheral nociceptive inputs and central processing. Pain consists of adaptive pain that is acute and beneficial for healing and maladaptive pain that is often persistent and pathological. Pain is indeed heterogeneous, and can be expressed as nociceptive, inflammatory, or neuropathic in nature. Neuropathic pain is an example of maladaptive pain that occurs after spinal cord injury (SCI), which triggers a wide range of neural plasticity. The nociceptive processing that underlies pain hypersensitivity is well-studied in the spinal cord. However, recent investigations show maladaptive plasticity that leads to pain, including neuropathic pain after SCI, also exists at peripheral sites, such as the dorsal root ganglia (DRG), which contains the cell bodies of sensory neurons. This review discusses the important role DRGs play in nociceptive processing that underlies inflammatory and neuropathic pain. Specifically, it highlights nociceptor hyperexcitability as critical to increased pain states. Furthermore, it reviews prior literature on glutamate and glutamate receptors, voltage-gated sodium channels (VGSC), and brain-derived neurotrophic factor (BDNF) signaling in the DRG as important contributors to inflammatory and neuropathic pain. We previously reviewed BDNF's role as a bidirectional neuromodulator of spinal plasticity. Here, we shift focus to the periphery and discuss BDNF-TrkB expression on nociceptors, non-nociceptor sensory neurons, and non-neuronal cells in the periphery as a potential contributor to induction and persistence of pain after SCI. Overall, this review presents a comprehensive evaluation of large bodies of work that individually focus on pain, DRG, BDNF, and SCI, to understand their interaction in nociceptive processing.
Collapse
Affiliation(s)
- Kyeongran Jang
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Sandra M. Garraway
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
23
|
Koutsioumpa C, Santiago C, Jacobs K, Lehnert BP, Barrera V, Hutchinson JN, Schmelyun D, Lehoczky JA, Paul DL, Ginty DD. Skin-type-dependent development of murine mechanosensory neurons. Dev Cell 2023; 58:2032-2047.e6. [PMID: 37607547 PMCID: PMC10615785 DOI: 10.1016/j.devcel.2023.07.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/26/2023] [Accepted: 07/27/2023] [Indexed: 08/24/2023]
Abstract
Mechanosensory neurons innervating the skin underlie our sense of touch. Fast-conducting, rapidly adapting mechanoreceptors innervating glabrous (non-hairy) skin form Meissner corpuscles, while in hairy skin, they associate with hair follicles, forming longitudinal lanceolate endings. How mechanoreceptors develop axonal endings appropriate for their skin targets is unknown. We report that mechanoreceptor morphologies across different skin regions are indistinguishable during early development but diverge post-natally, in parallel with skin maturation. Neurons terminating along the glabrous and hairy skin border exhibit hybrid morphologies, forming both Meissner corpuscles and lanceolate endings. Additionally, molecular profiles of neonatal glabrous and hairy skin-innervating neurons largely overlap. In mouse mutants with ectopic glabrous skin, mechanosensory neurons form end-organs appropriate for the altered skin type. Finally, BMP5 and BMP7 are enriched in glabrous skin, and signaling through type I bone morphogenetic protein (BMP) receptors in neurons is critical for Meissner corpuscle morphology. Thus, mechanoreceptor morphogenesis is flexibly instructed by target tissues.
Collapse
Affiliation(s)
- Charalampia Koutsioumpa
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Kiani Jacobs
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Brendan P Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Victor Barrera
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - John N Hutchinson
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Dhane Schmelyun
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Jessica A Lehoczky
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David L Paul
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Handler A, Zhang Q, Pang S, Nguyen TM, Iskols M, Nolan-Tamariz M, Cattel S, Plumb R, Sanchez B, Ashjian K, Shotland A, Brown B, Kabeer M, Turecek J, DeLisle MM, Rankin G, Xiang W, Pavarino EC, Africawala N, Santiago C, Lee WCA, Xu CS, Ginty DD. Three-dimensional reconstructions of mechanosensory end organs suggest a unifying mechanism underlying dynamic, light touch. Neuron 2023; 111:3211-3229.e9. [PMID: 37725982 PMCID: PMC10773061 DOI: 10.1016/j.neuron.2023.08.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 09/21/2023]
Abstract
Across mammalian skin, structurally complex and diverse mechanosensory end organs respond to mechanical stimuli and enable our perception of dynamic, light touch. How forces act on morphologically dissimilar mechanosensory end organs of the skin to gate the requisite mechanotransduction channel Piezo2 and excite mechanosensory neurons is not understood. Here, we report high-resolution reconstructions of the hair follicle lanceolate complex, Meissner corpuscle, and Pacinian corpuscle and the subcellular distribution of Piezo2 within them. Across all three end organs, Piezo2 is restricted to the sensory axon membrane, including axon protrusions that extend from the axon body. These protrusions, which are numerous and elaborate extensively within the end organs, tether the axon to resident non-neuronal cells via adherens junctions. These findings support a unified model for dynamic touch in which mechanical stimuli stretch hundreds to thousands of axon protrusions across an end organ, opening proximal, axonal Piezo2 channels and exciting the neuron.
Collapse
Affiliation(s)
- Annie Handler
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Qiyu Zhang
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Song Pang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Tri M Nguyen
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Stuart Cattel
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Rebecca Plumb
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Brianna Sanchez
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Karyl Ashjian
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Aria Shotland
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Bartianna Brown
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Madiha Kabeer
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Josef Turecek
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michelle M DeLisle
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Genelle Rankin
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Wangchu Xiang
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Elisa C Pavarino
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Nusrat Africawala
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Celine Santiago
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Wei-Chung Allen Lee
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - C Shan Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Santiago C, Sharma N, Africawala N, Siegrist J, Handler A, Tasnim A, Anjum R, Turecek J, Lehnert BP, Renauld S, Nolan-Tamariz M, Iskols M, Magee AR, Paradis S, Ginty DD. Activity-dependent development of the body's touch receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.23.559109. [PMID: 37790437 PMCID: PMC10542488 DOI: 10.1101/2023.09.23.559109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
We report a role for activity in the development of the primary sensory neurons that detect touch. Genetic deletion of Piezo2, the principal mechanosensitive ion channel in somatosensory neurons, caused profound changes in the formation of mechanosensory end organ structures and altered somatosensory neuron central targeting. Single cell RNA sequencing of Piezo2 conditional mutants revealed changes in gene expression in the sensory neurons activated by light mechanical forces, whereas other neuronal classes were less affected. To further test the role of activity in mechanosensory end organ development, we genetically deleted the voltage-gated sodium channel Nav1.6 (Scn8a) in somatosensory neurons throughout development and found that Scn8a mutants also have disrupted somatosensory neuron morphologies and altered electrophysiological responses to mechanical stimuli. Together, these findings indicate that mechanically evoked neuronal activity acts early in life to shape the maturation of the mechanosensory end organs that underlie our sense of gentle touch.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Nikhil Sharma
- Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Nusrat Africawala
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Julianna Siegrist
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA, 02453, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Brendan P. Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Sophia Renauld
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexandra R. Magee
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA, 02453, USA
| | - David D. Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
- Lead Contact
| |
Collapse
|
26
|
Jeon SM, Pradeep A, Chang D, McDonough L, Chen Y, Latremoliere A, Crawford LK, Caterina MJ. SKIN REINNERVATION BY COLLATERAL SPROUTING FOLLOWING SPARED NERVE INJURY IN MICE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.557420. [PMID: 37745384 PMCID: PMC10515828 DOI: 10.1101/2023.09.12.557420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Following peripheral nerve injury, denervated tissues can be reinnervated via regeneration of injured neurons or via collateral sprouting of neighboring uninjured afferents into the denervated territory. While there has been substantial focus on mechanisms underlying regeneration, collateral sprouting has received relatively less attention. In this study, we used immunohistochemistry and genetic neuronal labeling to define the subtype specificity of sprouting-mediated reinnervation of plantar hind paw skin in the mouse spared nerve injury (SNI) model, in which productive regeneration cannot occur. Following an initial loss of cutaneous afferents in the tibial nerve territory, we observed progressive centripetal reinnervation by multiple subtypes of neighboring uninjured fibers into denervated glabrous and hairy plantar skin. In addition to dermal reinnervation, CGRP-expressing peptidergic fibers slowly but continuously repopulated the denervated epidermis, Interestingly, GFRα2-expressing nonpeptidergic fibers exhibited a transient burst of epidermal reinnervation, followed by trend towards regression. Presumptive sympathetic nerve fibers also sprouted into the denervated territory, as did a population of myelinated TrkC lineage fibers, though the latter did so less efficiently. Conversely, rapidly adapting Aβ fiber and C fiber low threshold mechanoreceptor (LTMR) subtypes failed to exhibit convincing collateral sprouting up to 8 weeks after nerve injury. Optogenetics and behavioral assays further demonstrated the functionality of collaterally sprouted fibers in hairy plantar skin with restoration of punctate mechanosensation without hypersensitivity. Our findings advance understanding of differential collateral sprouting among sensory neuron subpopulations and may guide strategies to promote the progression of sensory recovery or limit maladaptive sensory phenomena after peripheral nerve injury. Significance Statement Following nerve injury, whereas one mechanism for tissue reinnervation is regeneration of injured neurons, another, less well studied mechanism is collateral sprouting of nearby uninjured neurons. In this study, we examined collateral sprouting in denervated mouse skin and showed that it involves some, but not all neuronal subtypes. Despite such heterogeneity, a significant degree of restoration of punctate mechanical sensitivity is achieved. These findings highlight the diversity of collateral sprouting among peripheral neuron subtypes and reveal important differences between pre- and post-denervation skin that might be appealing targets for therapeutic correction to enhance functional recovery from denervation and prevent unwanted sensory phenomena such as pain or numbness.
Collapse
|
27
|
Wolfson RL, Abdelaziz A, Rankin G, Kushner S, Qi L, Mazor O, Choi S, Sharma N, Ginty DD. DRG afferents that mediate physiologic and pathologic mechanosensation from the distal colon. Cell 2023; 186:3368-3385.e18. [PMID: 37541195 PMCID: PMC10440726 DOI: 10.1016/j.cell.2023.07.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 04/23/2023] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
The properties of dorsal root ganglia (DRG) neurons that innervate the distal colon are poorly defined, hindering our understanding of their roles in normal physiology and gastrointestinal (GI) disease. Here, we report genetically defined subsets of colon-innervating DRG neurons with diverse morphologic and physiologic properties. Four colon-innervating DRG neuron populations are mechanosensitive and exhibit distinct force thresholds to colon distension. The highest threshold population, selectively labeled using Bmpr1b genetic tools, is necessary and sufficient for behavioral responses to high colon distension, which is partly mediated by the mechanosensory ion channel Piezo2. This Aδ-HTMR population mediates behavioral over-reactivity to colon distension caused by inflammation in a model of inflammatory bowel disease. Thus, like cutaneous DRG mechanoreceptor populations, colon-innervating mechanoreceptors exhibit distinct anatomical and physiological properties and tile force threshold space, and genetically defined colon-innervating HTMRs mediate pathophysiological responses to colon distension, revealing a target population for therapeutic intervention.
Collapse
Affiliation(s)
- Rachel L Wolfson
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Amira Abdelaziz
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Genelle Rankin
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Sarah Kushner
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Lijun Qi
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Ofer Mazor
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Seungwon Choi
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Nikhil Sharma
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Department of Systems Biology, Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY 10032, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
28
|
Whiddon ZD, Marshall JB, Alston DC, McGee AW, Krimm RF. Rapid structural remodeling of peripheral taste neurons is independent of taste cell turnover. PLoS Biol 2023; 21:e3002271. [PMID: 37651406 PMCID: PMC10499261 DOI: 10.1371/journal.pbio.3002271] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 09/13/2023] [Accepted: 07/22/2023] [Indexed: 09/02/2023] Open
Abstract
Taste bud cells are constantly replaced in taste buds as old cells die and new cells migrate into the bud. The perception of taste relies on new taste bud cells integrating with existing neural circuitry, yet how these new cells connect with a taste ganglion neuron is unknown. Do taste ganglion neurons remodel to accommodate taste bud cell renewal? If so, how much of the structure of taste axons is fixed and how much remodels? Here, we measured the motility and branching of individual taste arbors (the portion of the axon innervating taste buds) in mice over time with two-photon in vivo microscopy. Terminal branches of taste arbors continuously and rapidly remodel within the taste bud. This remodeling is faster than predicted by taste bud cell renewal, with terminal branches added and lost concurrently. Surprisingly, blocking entry of new taste bud cells with chemotherapeutic agents revealed that remodeling of the terminal branches on taste arbors does not rely on the renewal of taste bud cells. Although terminal branch remodeling was fast and intrinsically controlled, no new arbors were added to taste buds, and few were lost over 100 days. Taste ganglion neurons maintain a stable number of arbors that are each capable of high-speed remodeling. We propose that terminal branch plasticity permits arbors to locate new taste bud cells, while stability of arbor number supports constancy in the degree of connectivity and function for each neuron over time.
Collapse
Affiliation(s)
- Zachary D. Whiddon
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Jaleia B. Marshall
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - David C. Alston
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Aaron W. McGee
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Robin F. Krimm
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| |
Collapse
|
29
|
Niu C, Yue X, An JJ, Xu H, Xu B. Genetic dissection of BDNF and TrkB expression in glial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549007. [PMID: 37503044 PMCID: PMC10370033 DOI: 10.1101/2023.07.14.549007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase receptor B (TrkB) are widely expressed in the central nervous system. It is well documented that neurons express BDNF and full-length TrkB (TrkB.FL), and a lower level of truncated TrkB (TrkB.T). With conflicting results, glial cells also have been reported to express BDNF and TrkB. In the current study, we employed a more sensitive and reliable genetic method to characterize the expression of BDNF and TrkB in glial cells in the mouse brain. We utilized three Cre mouse strains in which Cre recombinase is expressed in the same cells as BDNF, TrkB.FL, or all TrkB isoforms, and crossed them to Cre-dependent EGFP reporter mice to label BDNF- or TrkB- expressing cells. We performed immunohistochemistry with glial cell markers to examine the expression of BDNF and TrkB in microglia, astrocytes, and oligodendrocytes. Surprisingly, we found no BDNF- or TrkB- expressing microglia in the brain and spinal cord. Consistent with previous studies, most astrocytes only express TrkB.T in the adult brain. Moreover, there are a small number of astrocytes and oligodendrocytes that express BDNF, the function of which is to be determined. We also found that oligodendrocyte precursor cells, but not mature oligodendrocytes, express both TrkB.FL and TrkB.T in the adult brain. These results not only clarify the expression of BDNF and TrkB in glial cells, but also open opportunities to investigate previously unidentified roles of BDNF and TrkB in glial cells.
Collapse
|
30
|
Bhuiyan SA, Xu M, Yang L, Semizoglou E, Bhatia P, Pantaleo KI, Tochitsky I, Jain A, Erdogan B, Blair S, Cat V, Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Green U, McIlvried LA, Copits BA, Bertels Z, Del Rosario JS, Widman AJ, Slivicki RA, Yi J, Woolf CJ, Lennerz JK, Whited JL, Price TJ, Gereau RW, Renthal W. Harmonized cross-species cell atlases of trigeminal and dorsal root ganglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547740. [PMID: 37461736 PMCID: PMC10350076 DOI: 10.1101/2023.07.04.547740] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Peripheral sensory neurons in the dorsal root ganglion (DRG) and trigeminal ganglion (TG) are specialized to detect and transduce diverse environmental stimuli including touch, temperature, and pain to the central nervous system. Recent advances in single-cell RNA-sequencing (scRNA-seq) have provided new insights into the diversity of sensory ganglia cell types in rodents, non-human primates, and humans, but it remains difficult to compare transcriptomically defined cell types across studies and species. Here, we built cross-species harmonized atlases of DRG and TG cell types that describe 18 neuronal and 11 non-neuronal cell types across 6 species and 19 studies. We then demonstrate the utility of this harmonized reference atlas by using it to annotate newly profiled DRG nuclei/cells from both human and the highly regenerative axolotl. We observe that the transcriptomic profiles of sensory neuron subtypes are broadly similar across vertebrates, but the expression of functionally important neuropeptides and channels can vary notably. The new resources and data presented here can guide future studies in comparative transcriptomics, simplify cell type nomenclature differences across studies, and help prioritize targets for future pain therapy development.
Collapse
Affiliation(s)
- Shamsuddin A Bhuiyan
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mengyi Xu
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Alan Edwards Center for Research on Pain and Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Lite Yang
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Evangelia Semizoglou
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Parth Bhatia
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katerina I Pantaleo
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ivan Tochitsky
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children's Hospital and Harvard Medical School, 3 Blackfan Cir. Boston, MA 02115
| | - Aakanksha Jain
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children's Hospital and Harvard Medical School, 3 Blackfan Cir. Boston, MA 02115
| | - Burcu Erdogan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, 02138
| | - Steven Blair
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, 02138
| | - Victor Cat
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, 02138
| | - Juliet M Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080
| | - Ursula Green
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114
| | - Lisa A McIlvried
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Bryan A Copits
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Zachariah Bertels
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - John S Del Rosario
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Allie J Widman
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Richard A Slivicki
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Jiwon Yi
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children's Hospital and Harvard Medical School, 3 Blackfan Cir. Boston, MA 02115
| | - Jochen K Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114
| | - Jessica L Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, 02138
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080
| | - Robert W Gereau
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - William Renthal
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
31
|
Esvald EE, Tuvikene J, Kiir CS, Avarlaid A, Tamberg L, Sirp A, Shubina A, Cabrera-Cabrera F, Pihlak A, Koppel I, Palm K, Timmusk T. Revisiting the expression of BDNF and its receptors in mammalian development. Front Mol Neurosci 2023; 16:1182499. [PMID: 37426074 PMCID: PMC10325033 DOI: 10.3389/fnmol.2023.1182499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/22/2023] [Indexed: 07/11/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) promotes the survival and functioning of neurons in the central nervous system and contributes to proper functioning of many non-neural tissues. Although the regulation and role of BDNF have been extensively studied, a rigorous analysis of the expression dynamics of BDNF and its receptors TrkB and p75NTR is lacking. Here, we have analyzed more than 3,600 samples from 18 published RNA sequencing datasets, and used over 17,000 samples from GTEx, and ~ 180 samples from BrainSpan database, to describe the expression of BDNF in the developing mammalian neural and non-neural tissues. We show evolutionarily conserved dynamics and expression patterns of BDNF mRNA and non-conserved alternative 5' exon usage. Finally, we also show increasing BDNF protein levels during murine brain development and BDNF protein expression in several non-neural tissues. In parallel, we describe the spatiotemporal expression pattern of BDNF receptors TrkB and p75NTR in both murines and humans. Collectively, our in-depth analysis of the expression of BDNF and its receptors gives insight into the regulation and signaling of BDNF in the whole organism throughout life.
Collapse
Affiliation(s)
- Eli-Eelika Esvald
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
| | - Jürgen Tuvikene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
- dxlabs LLC, Tallinn, Estonia
| | - Carl Sander Kiir
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Annela Avarlaid
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Laura Tamberg
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Alex Sirp
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Anastassia Shubina
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | | | - Indrek Koppel
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | - Tõnis Timmusk
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Protobios LLC, Tallinn, Estonia
| |
Collapse
|
32
|
Gautam M, Yamada A, Yamada A, Wu Q, Kridsada K, Ling J, Yu H, Dong P, Ma M, Gu J, Luo W. Distinct Local and Global Functions of Aβ Low-Threshold Mechanoreceptors in Mechanical Pain Transmission. RESEARCH SQUARE 2023:rs.3.rs-2939309. [PMID: 37398333 PMCID: PMC10312941 DOI: 10.21203/rs.3.rs-2939309/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The roles of Aβ low-threshold mechanoreceptors (LTMRs) in transmitting mechanical hyperalgesia and in alleviating chronic pain have been of great interest but remain contentious. Here we utilized intersectional genetic tools, optogenetics, and high-speed imaging to specifically examine functions of SplitCre labeled Aβ-LTMRs in this regard. Genetic ablation of SplitCre-Aβ-LTMRs increased mechanical pain but not thermosensation in both acute and chronic inflammatory pain conditions, indicating their modality-specific role in gating mechanical pain transmission. Local optogenetic activation of SplitCre-Aβ-LTMRs triggered nociception after tissue inflammation, whereas their broad activation at the dorsal column still alleviated mechanical hypersensitivity of chronic inflammation. Taking all data into consideration, we propose a new model, in which Aβ-LTMRs play distinctive local and global roles in transmitting and alleviating mechanical hyperalgesia of chronic pain, respectively. Our model suggests a new strategy of global activation plus local inhibition of Aβ-LTMRs for treating mechanical hyperalgesia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Minghong Ma
- University of Pennsylvania School of Medicine
| | | | | |
Collapse
|
33
|
Qi L, Iskols M, Handler A, Ginty DD. Krause corpuscles of the genitalia are vibrotactile sensors required for normal sexual behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.14.545006. [PMID: 37398085 PMCID: PMC10312780 DOI: 10.1101/2023.06.14.545006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Krause corpuscles, first discovered in the 1850s, are enigmatic sensory structures with unknown physiological properties and functions found within the genitalia and other mucocutaneous tissues. Here, we identified two distinct somatosensory neuron subtypes that innervate Krause corpuscles of the mouse penis and clitoris and project to a unique sensory terminal region of the spinal cord. Using in vivo electrophysiology and calcium imaging, we found that both Krause corpuscle afferent types are A-fiber rapid-adapting low-threshold mechanoreceptors, optimally tuned to dynamic, light touch and mechanical vibrations (40-80 Hz) applied to the clitoris or penis. Optogenetic activation of male Krause corpuscle afferent terminals evoked penile erection, while genetic ablation of Krause corpuscles impaired intromission and ejaculation of males as well as reduced sexual receptivity of females. Thus, Krause corpuscles, which are particularly dense in the clitoris, are vibrotactile sensors crucial for normal sexual behavior.
Collapse
Affiliation(s)
- Lijun Qi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - David D. Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| |
Collapse
|
34
|
Meltzer S, Boulanger KC, Chirila AM, Osei-Asante E, DeLisle M, Zhang Q, Kalish BT, Tasnim A, Huey EL, Fuller LC, Flaherty EK, Maniatis T, Garrett AM, Weiner JA, Ginty DD. γ-Protocadherins control synapse formation and peripheral branching of touch sensory neurons. Neuron 2023; 111:1776-1794.e10. [PMID: 37028432 PMCID: PMC10365546 DOI: 10.1016/j.neuron.2023.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 01/20/2023] [Accepted: 03/07/2023] [Indexed: 04/09/2023]
Abstract
Light touch sensation begins with activation of low-threshold mechanoreceptor (LTMR) endings in the skin and propagation of their signals to the spinal cord and brainstem. We found that the clustered protocadherin gamma (Pcdhg) gene locus, which encodes 22 cell-surface homophilic binding proteins, is required in somatosensory neurons for normal behavioral reactivity to a range of tactile stimuli. Developmentally, distinct Pcdhg isoforms mediate LTMR synapse formation through neuron-neuron interactions and peripheral axonal branching through neuron-glia interactions. The Pcdhgc3 isoform mediates homophilic interactions between sensory axons and spinal cord neurons to promote synapse formation in vivo and is sufficient to induce postsynaptic specializations in vitro. Moreover, loss of Pcdhgs and somatosensory synaptic inputs to the dorsal horn leads to fewer corticospinal synapses on dorsal horn neurons. These findings reveal essential roles for Pcdhg isoform diversity in somatosensory neuron synapse formation, peripheral axonal branching, and stepwise assembly of central mechanosensory circuitry.
Collapse
Affiliation(s)
- Shan Meltzer
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Katelyn C Boulanger
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Anda M Chirila
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Emmanuella Osei-Asante
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michelle DeLisle
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Qiyu Zhang
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Brian T Kalish
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Erica L Huey
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Leah C Fuller
- Department of Biology and Iowa Neuroscience Institute, University of Iowa, 143 Biology Building, Iowa City, IA 52242, USA
| | - Erin K Flaherty
- Department of Biochemistry and Molecular Biophysics, Zuckerman Institute of Mind Brain and Behavior, Columbia University, New York, NY 10032, USA
| | - Tom Maniatis
- Department of Biochemistry and Molecular Biophysics, Zuckerman Institute of Mind Brain and Behavior, Columbia University, New York, NY 10032, USA
| | - Andrew M Garrett
- Department of Pharmacology and Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield St. 7322 Scott Hall, Detroit, MI 48201, USA
| | - Joshua A Weiner
- Department of Biology and Iowa Neuroscience Institute, University of Iowa, 143 Biology Building, Iowa City, IA 52242, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Kupari J, Ernfors P. Molecular taxonomy of nociceptors and pruriceptors. Pain 2023; 164:1245-1257. [PMID: 36718807 PMCID: PMC10184562 DOI: 10.1097/j.pain.0000000000002831] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/10/2022] [Accepted: 11/21/2022] [Indexed: 02/01/2023]
Affiliation(s)
- Jussi Kupari
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Wang K, Cai B, Song Y, Chen Y, Zhang X. Somatosensory neuron types and their neural networks as revealed via single-cell transcriptomics. Trends Neurosci 2023:S0166-2236(23)00130-3. [PMID: 37268541 DOI: 10.1016/j.tins.2023.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 06/04/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) has allowed profiling cell types of the dorsal root ganglia (DRG) and their transcriptional states in physiology and chronic pain. However, the evaluation criteria used in previous studies to classify DRG neurons varied, which presents difficulties in determining the various types of DRG neurons. In this review, we aim to integrate findings from previous transcriptomic studies of the DRG. We first briefly introduce the history of DRG-neuron cell-type profiling, and discuss the advantages and disadvantages of different scRNA-seq methods. We then examine the classification of DRG neurons based on single-cell profiling under physiological and pathological conditions. Finally, we propose further studies on the somatosensory system at the molecular, cellular, and neural network levels.
Collapse
Affiliation(s)
- Kaikai Wang
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Bing Cai
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Yurang Song
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Yan Chen
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China; Xuhui Central Hospital, Shanghai, 200031, China
| | - Xu Zhang
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; SIMR Joint Lab of Drug Innovation, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China; Xuhui Central Hospital, Shanghai, 200031, China.
| |
Collapse
|
37
|
Gautam M, Yamada A, Yamada AI, Wu Q, Kridsada K, Ling J, Yu H, Dong P, Ma M, Gu J, Luo W. Distinct Local and Global Functions of Aβ Low-Threshold Mechanoreceptors in Mechanical Pain Transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.540962. [PMID: 37293085 PMCID: PMC10245756 DOI: 10.1101/2023.05.16.540962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The roles of Aβ low-threshold mechanoreceptors (LTMRs) in transmitting mechanical hyperalgesia and in alleviating chronic pain have been of great interest but remain contentious. Here we utilized intersectional genetic tools, optogenetics, and high-speed imaging to specifically examine functions of Split Cre labeled Aβ-LTMRs in this regard. Genetic ablation of Split Cre -Aβ-LTMRs increased mechanical pain but not thermosensation in both acute and chronic inflammatory pain conditions, indicating their modality-specific role in gating mechanical pain transmission. Local optogenetic activation of Split Cre -Aβ-LTMRs triggered nociception after tissue inflammation, whereas their broad activation at the dorsal column still alleviated mechanical hypersensitivity of chronic inflammation. Taking all data into consideration, we propose a new model, in which Aβ-LTMRs play distinctive local and global roles in transmitting and alleviating mechanical hyperalgesia of chronic pain, respectively. Our model suggests a new strategy of global activation plus local inhibition of Aβ-LTMRs for treating mechanical hyperalgesia.
Collapse
|
38
|
Ohman L, Hanbali L, Krimm R. Taste arbor structural variability analyzed across taste regions. J Comp Neurol 2023; 531:743-758. [PMID: 36740741 PMCID: PMC10082444 DOI: 10.1002/cne.25459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 01/04/2023] [Accepted: 01/13/2023] [Indexed: 02/07/2023]
Abstract
Taste ganglion neurons are functionally and molecularly diverse, but until recently morphological diversity was completely unexplored. Specifically, taste arbors (the portion of the neuron within the taste bud) vary in structure, but the reason for this variability is unclear. Here, we analyzed structural variability in taste arbors to determine which factors determine their morphological diversity. To characterize arbor morphology and its relationship to taste bud cells capable of transducing taste stimuli (taste-transducing cell) number and type, we utilized sparse cell genetic labeling of taste ganglion neurons in combination with whole-mount immunohistochemistry. Reconstruction of 151 taste arbors revealed variation in arbor size, complexity, and symmetry. Overall, taste arbors exist on a continuum of complexity, cannot be categorized into discrete morphological groups, and do not have stereotyped endings. Arbor size/complexity was not related to the size of the taste bud in which it was located or the type of taste-transducing cell contacted (membranes within 180 nm). Instead, arbors could be broadly categorized into three groups: large asymmetrical arbors contacting many taste-transducing cells, small symmetrical arbors contacting one or two taste-transducing cells, and unbranched arbors. Neurons with multiple arbors had arbors in more than one of these categories, indicating that this variability is not an intrinsic feature of neuron type. Instead, we speculate that arbor structure is determined primarily by nerve fiber remodeling in response to cell turnover and that large asymmetrical arbors represent a particularly plastic state.
Collapse
Affiliation(s)
- Lisa Ohman
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Lama Hanbali
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Robin Krimm
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| |
Collapse
|
39
|
Pomaville MB, Wright KM. Follicle-innervating Aδ-low threshold mechanoreceptive neurons form receptive fields through homotypic competition. Neural Dev 2023; 18:2. [PMID: 37106422 PMCID: PMC10134579 DOI: 10.1186/s13064-023-00170-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
The mammalian somatosensory system is comprised of multiple neuronal populations that form specialized, highly organized sensory endings in the skin. The organization of somatosensory endings is essential to their functions, yet the mechanisms which regulate this organization remain unclear. Using a combination of genetic and molecular labeling approaches, we examined the development of mouse hair follicle-innervating low-threshold mechanoreceptors (LTMRs) and explored competition for innervation targets as a mechanism involved in the patterning of their receptive fields. We show that follicle innervating neurons are present in the skin at birth and that LTMR receptive fields gradually add follicle-innervating endings during the first two postnatal weeks. Using a constitutive Bax knockout to increase the number of neurons in adult animals, we show that two LTMR subtypes have differential responses to an increase in neuronal population size: Aδ-LTMR neurons shrink their receptive fields to accommodate the increased number of neurons innervating the skin, while C-LTMR neurons do not. Our findings suggest that competition for hair follicles to innervate plays a role in the patterning and organization of follicle-innervating LTMR neurons.
Collapse
Affiliation(s)
- Matthew B Pomaville
- Vollum Institute, Oregon Health and Science University, Portland, OR, 97239, USA
- Department of Cell, Developmental, and Cancer Biology, Cell and Developmental Biology Graduate Program, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
40
|
Hovhannisyan AH, Lindquist K, Belugin S, Mecklenburg J, Ibrahim T, Tram M, Corey T, Salmon A, Ruparel S, Ruparel S, Akopian A. Sensory innervation of masseter, temporal and lateral pterygoid muscles in common marmosets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528062. [PMID: 36798270 PMCID: PMC9934658 DOI: 10.1101/2023.02.10.528062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Myogenous temporomandibular disorders (TMDM) is associated with an increased responsiveness of nerves innervating the masseter (MM), temporal (TM), medial pterygoid (MPM) and lateral pterygoid muscles (LPM). This study aimed to examine sensory nerve types innervating MM, TM and LPM of adult non-human primate - common marmosets. Sensory nerves are localized in specific regions of these muscles. Pgp9.5, marker for all nerves, and NFH, a marker for A-fibers, showed that masticatory muscles were predominantly innervated with A-fibers. The proportion of C- to A-fibers was highest in LPM, and minimal (6-8%) in MM. All C-fibers (pgp9.5+/NFH-) observed in masticatory muscles were peptidergic (CGRP+) and lacked mrgprD, trpV1 and CHRNA3, a silent nociceptive marker. All fibers in masticatory muscles were labeled with GFAP+, a myelin sheath marker. There were substantially more peptidergic A-fibers (CGRP+/NFH+) in TM and LPM compared to MM. Almost all A-fibers in MM expressed trkC, with some of them having trkB and parvalbumin. In contrast, a lesser number of TM and LPM nerves expressed trkC, and lacked trkB. Tyrosine hydroxylase antibodies, which did not label TG, highlighted sympathetic fibers around blood vessels of the masticatory muscles. Overall, masticatory muscle types of marmosets have distinct and different innervation patterns.
Collapse
|
41
|
Shimizu N, Saito T, Wada N, Hashimoto M, Shimizu T, Kwon J, Cho KJ, Saito M, Karnup S, de Groat WC, Yoshimura N. Molecular Mechanisms of Neurogenic Lower Urinary Tract Dysfunction after Spinal Cord Injury. Int J Mol Sci 2023; 24:7885. [PMID: 37175592 PMCID: PMC10177842 DOI: 10.3390/ijms24097885] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
This article provides a synopsis of current progress made in fundamental studies of lower urinary tract dysfunction (LUTD) after spinal cord injury (SCI) above the sacral level. Animal models of SCI allowed us to examine the effects of SCI on the micturition control and the underlying neurophysiological processes of SCI-induced LUTD. Urine storage and elimination are the two primary functions of the LUT, which are governed by complicated regulatory mechanisms in the central and peripheral nervous systems. These neural systems control the action of two functional units in the LUT: the urinary bladder and an outlet consisting of the bladder neck, urethral sphincters, and pelvic-floor striated muscles. During the storage phase, the outlet is closed, and the bladder is inactive to maintain a low intravenous pressure and continence. In contrast, during the voiding phase, the outlet relaxes, and the bladder contracts to facilitate adequate urine flow and bladder emptying. SCI disrupts the normal reflex circuits that regulate co-ordinated bladder and urethral sphincter function, leading to involuntary and inefficient voiding. Following SCI, a spinal micturition reflex pathway develops to induce an overactive bladder condition following the initial areflexic phase. In addition, without proper bladder-urethral-sphincter coordination after SCI, the bladder is not emptied as effectively as in the normal condition. Previous studies using animal models of SCI have shown that hyperexcitability of C-fiber bladder afferent pathways is a fundamental pathophysiological mechanism, inducing neurogenic LUTD, especially detrusor overactivity during the storage phase. SCI also induces neurogenic LUTD during the voiding phase, known as detrusor sphincter dyssynergia, likely due to hyperexcitability of Aδ-fiber bladder afferent pathways rather than C-fiber afferents. The molecular mechanisms underlying SCI-induced LUTD are multifactorial; previous studies have identified significant changes in the expression of various molecules in the peripheral organs and afferent nerves projecting to the spinal cord, including growth factors, ion channels, receptors and neurotransmitters. These findings in animal models of SCI and neurogenic LUTD should increase our understanding of pathophysiological mechanisms of LUTD after SCI for the future development of novel therapies for SCI patients with LUTD.
Collapse
Affiliation(s)
- Nobutaka Shimizu
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
- Pelvic Floor Center, Kochi Medical School, Kochi University, Nankoku 783-8505, Japan
| | - Tetsuichi Saito
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Naoki Wada
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Mamoru Hashimoto
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Takahiro Shimizu
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku 783-8505, Japan
| | - Joonbeom Kwon
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Kang Jun Cho
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku 783-8505, Japan
| | - Sergei Karnup
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - William C. de Groat
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Naoki Yoshimura
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (N.S.)
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
42
|
Qi L, Iskols M, Shi D, Reddy P, Walker C, Lezgiyeva K, Voisin T, Pawlak M, Kuchroo VK, Chiu I, Ginty DD, Sharma N. A DRG genetic toolkit reveals molecular, morphological, and functional diversity of somatosensory neuron subtypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.22.537932. [PMID: 37131664 PMCID: PMC10153270 DOI: 10.1101/2023.04.22.537932] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Mechanical and thermal stimuli acting on the skin are detected by morphologically and physiologically distinct sensory neurons of the dorsal root ganglia (DRG). Achieving a holistic view of how this diverse neuronal population relays sensory information from the skin to the central nervous system (CNS) has been challenging with existing tools. Here, we used transcriptomic datasets of the mouse DRG to guide development and curation of a genetic toolkit to interrogate transcriptionally defined DRG neuron subtypes. Morphological analysis revealed unique cutaneous axon arborization areas and branching patterns of each subtype. Physiological analysis showed that subtypes exhibit distinct thresholds and ranges of responses to mechanical and/or thermal stimuli. The somatosensory neuron toolbox thus enables comprehensive phenotyping of most principal sensory neuron subtypes. Moreover, our findings support a population coding scheme in which the activation thresholds of morphologically and physiologically distinct cutaneous DRG neuron subtypes tile multiple dimensions of stimulus space.
Collapse
Affiliation(s)
- Lijun Qi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - David Shi
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Columbia University, New York, NY
| | - Pranav Reddy
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Columbia University, New York, NY
| | - Christopher Walker
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Columbia University, New York, NY
| | - Karina Lezgiyeva
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - Tiphaine Voisin
- Department of Immunology, Harvard Medical School, Boston, MA 02115
| | - Mathias Pawlak
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Isaac Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115
| | - David D. Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - Nikhil Sharma
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Columbia University, New York, NY
| |
Collapse
|
43
|
Sonkodi B. LF Power of HRV Could Be the Piezo2 Activity Level in Baroreceptors with Some Piezo1 Residual Activity Contribution. Int J Mol Sci 2023; 24:ijms24087038. [PMID: 37108199 PMCID: PMC10138994 DOI: 10.3390/ijms24087038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Heart rate variability is a useful measure for monitoring the autonomic nervous system. Heart rate variability measurements have gained significant demand not only in science, but also in the public due to the fairly low price and wide accessibility of the Internet of things. The scientific debate about one of the measures of heart rate variability, i.e., what low-frequency power is reflecting, has been ongoing for decades. Some schools reason that it represents the sympathetic loading, while an even more compelling reasoning is that it measures how the baroreflex modulates the cardiac autonomic outflow. However, the current opinion manuscript proposes that the discovery of the more precise molecular characteristics of baroreceptors, i.e., that the Piezo2 ion channel containing vagal afferents could invoke the baroreflex, may possibly resolve this debate. It is long known that medium- to high-intensity exercise diminishes low-frequency power to almost undetectable values. Moreover, it is also demonstrated that the stretch- and force-gated Piezo2 ion channels are inactivated in a prolonged hyperexcited state in order to prevent pathological hyperexcitation. Accordingly, the current author suggests that the almost undetectable value of low-frequency power at medium- to high-intensity exercise reflects the inactivation of Piezo2 from vagal afferents in the baroreceptors with some Piezo1 residual activity contribution. Consequently, this opinion paper highlights how low-frequency power of the heart rate variability could represent the activity level of Piezo2 in baroreceptors.
Collapse
Affiliation(s)
- Balázs Sonkodi
- Department of Health Sciences and Sport Medicine, Hungarian University of Sports Science, 1123 Budapest, Hungary
| |
Collapse
|
44
|
Handler A, Zhang Q, Pang S, Nguyen TM, Iskols M, Nolan-Tamariz M, Cattel S, Plumb R, Sanchez B, Ashjian K, Shotland A, Brown B, Kabeer M, Turecek J, Rankin G, Xiang W, Pavarino EC, Africawala N, Santiago C, Lee WCA, Shan Xu C, Ginty DD. Three-dimensional reconstructions of mechanosensory end organs suggest a unifying mechanism underlying dynamic, light touch. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533188. [PMID: 36993253 PMCID: PMC10055218 DOI: 10.1101/2023.03.17.533188] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Specialized mechanosensory end organs within mammalian skin-hair follicle-associated lanceolate complexes, Meissner corpuscles, and Pacinian corpuscles-enable our perception of light, dynamic touch 1 . In each of these end organs, fast-conducting mechanically sensitive neurons, called Aβ low-threshold mechanoreceptors (Aβ LTMRs), associate with resident glial cells, known as terminal Schwann cells (TSCs) or lamellar cells, to form complex axon ending structures. Lanceolate-forming and corpuscle-innervating Aβ LTMRs share a low threshold for mechanical activation, a rapidly adapting (RA) response to force indentation, and high sensitivity to dynamic stimuli 1-6 . How mechanical stimuli lead to activation of the requisite mechanotransduction channel Piezo2 7-15 and Aβ RA-LTMR excitation across the morphologically dissimilar mechanosensory end organ structures is not understood. Here, we report the precise subcellular distribution of Piezo2 and high-resolution, isotropic 3D reconstructions of all three end organs formed by Aβ RA-LTMRs determined by large volume enhanced Focused Ion Beam Scanning Electron Microscopy (FIB-SEM) imaging. We found that within each end organ, Piezo2 is enriched along the sensory axon membrane and is minimally or not expressed in TSCs and lamellar cells. We also observed a large number of small cytoplasmic protrusions enriched along the Aβ RA-LTMR axon terminals associated with hair follicles, Meissner corpuscles, and Pacinian corpuscles. These axon protrusions reside within close proximity to axonal Piezo2, occasionally contain the channel, and often form adherens junctions with nearby non-neuronal cells. Our findings support a unified model for Aβ RA-LTMR activation in which axon protrusions anchor Aβ RA-LTMR axon terminals to specialized end organ cells, enabling mechanical stimuli to stretch the axon in hundreds to thousands of sites across an individual end organ and leading to activation of proximal Piezo2 channels and excitation of the neuron.
Collapse
|
45
|
Moayedi Y, Xu S, Obayashi SK, Hoffman BU, Gerling GJ, Lumpkin EA. The cellular basis of mechanosensation in mammalian tongue. Cell Rep 2023; 42:112087. [PMID: 36763499 PMCID: PMC10409885 DOI: 10.1016/j.celrep.2023.112087] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Mechanosensory neurons that innervate the tongue provide essential information to guide feeding, speech, and social grooming. We use in vivo calcium imaging of mouse trigeminal ganglion neurons to identify functional groups of mechanosensory neurons innervating the anterior tongue. These sensory neurons respond to thermal and mechanical stimulation. Analysis of neuronal activity patterns reveal that most mechanosensory trigeminal neurons are tuned to detect moving stimuli across the tongue. Using an unbiased, multilayer hierarchical clustering approach to classify pressure-evoked activity based on temporal response dynamics, we identify five functional classes of mechanosensory neurons with distinct force-response relations and adaptation profiles. These populations are tuned to detect different features of touch. Molecular markers of functionally distinct clusters are identified by analyzing cluster representation in genetically marked neuronal subsets. Collectively, these studies provide a platform for defining the contributions of functionally distinct mechanosensory neurons to oral behaviors crucial for survival in mammals.
Collapse
Affiliation(s)
- Yalda Moayedi
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA; Department of Otolaryngology - Head & Neck Surgery, Columbia University, New York, NY 10032, USA
| | - Shan Xu
- School of Engineering and Applied Science, University of Virginia, Charlottesville, VA 22904, USA
| | - Sophie K Obayashi
- Department of Molecular & Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Benjamin U Hoffman
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Gregory J Gerling
- School of Engineering and Applied Science, University of Virginia, Charlottesville, VA 22904, USA.
| | - Ellen A Lumpkin
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA; Department of Molecular & Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
46
|
Brown TL, Horton EC, Craig EW, Goo CEA, Black EC, Hewitt MN, Yee NG, Fan ET, Raible DW, Rasmussen JP. Dermal appendage-dependent patterning of zebrafish atoh1a+ Merkel cells. eLife 2023; 12:85800. [PMID: 36648063 PMCID: PMC9901935 DOI: 10.7554/elife.85800] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Touch system function requires precise interactions between specialized skin cells and somatosensory axons, as exemplified by the vertebrate mechanosensory Merkel cell-neurite complex. Development and patterning of Merkel cells and associated neurites during skin organogenesis remain poorly understood, partly due to the in utero development of mammalian embryos. Here, we discover Merkel cells in the zebrafish epidermis and identify Atonal homolog 1a (Atoh1a) as a marker of zebrafish Merkel cells. We show that zebrafish Merkel cells derive from basal keratinocytes, express neurosecretory and mechanosensory machinery, extend actin-rich microvilli, and complex with somatosensory axons, all hallmarks of mammalian Merkel cells. Merkel cells populate all major adult skin compartments, with region-specific densities and distribution patterns. In vivo photoconversion reveals that Merkel cells undergo steady loss and replenishment during skin homeostasis. Merkel cells develop concomitant with dermal appendages along the trunk and loss of Ectodysplasin signaling, which prevents dermal appendage formation, reduces Merkel cell density by affecting cell differentiation. By contrast, altering dermal appendage morphology changes the distribution, but not density, of Merkel cells. Overall, our studies provide insights into touch system maturation during skin organogenesis and establish zebrafish as an experimentally accessible in vivo model for the study of Merkel cell biology.
Collapse
Affiliation(s)
- Tanya L Brown
- Department of Biology, University of WashingtonSeattleUnited States
| | - Emma C Horton
- Department of Biology, University of WashingtonSeattleUnited States
| | - Evan W Craig
- Department of Biology, University of WashingtonSeattleUnited States
| | - Camille EA Goo
- Department of Biology, University of WashingtonSeattleUnited States
| | - Erik C Black
- Department of Biology, University of WashingtonSeattleUnited States
- Molecular and Cellular Biology Program, University of WashingtonSeattleUnited States
| | - Madeleine N Hewitt
- Molecular and Cellular Biology Program, University of WashingtonSeattleUnited States
- Department of Biological Structure, University of WashingtonSeattleUnited States
| | - Nathaniel G Yee
- Department of Biology, University of WashingtonSeattleUnited States
| | - Everett T Fan
- Department of Biology, University of WashingtonSeattleUnited States
| | - David W Raible
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Department of Otolaryngology - Head and Neck Surgery, University of WashingtonSeattleUnited States
- Institute for Stem Cell and Regenerative Medicine, University of WashingtonSeattleUnited States
| | - Jeffrey P Rasmussen
- Department of Biology, University of WashingtonSeattleUnited States
- Institute for Stem Cell and Regenerative Medicine, University of WashingtonSeattleUnited States
| |
Collapse
|
47
|
Angular Tuning Properties of Low Threshold Mechanoreceptors in Isolated Rat Whisker Hair Follicles. eNeuro 2022; 9:ENEURO.0175-22.2022. [PMID: 36376066 PMCID: PMC9721307 DOI: 10.1523/eneuro.0175-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/20/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
Angular tuning is preferential sensory response to a directional stimulus and is observed in the whisker tactile system. In whisker hair follicles, there are at least three types of low threshold mechanoreceptors (LTMRs): rapidly adapting (RA), slowly adapting type 1 (SA1), and slowly adapting type 2 (SA2). These LTMRs display angular tuning but their properties remain incompletely studied. Here, we used isolated rat whisker hair follicles and pressure-clamped single-fiber recordings to study angular tuning of these LTMRs. Angular tuning was determined with impulses elicited by ramp-and-hold deflection of whisker hair in 24 directions each at 15° for a total of 360°. We show that RA display impulses during ramp-up, both ramp-up and ramp-down, or ramp-down dynamic phases. Both SA1 and SA2 respond to angular stimuli with slowly adapting impulses in most angles. However, SA1 and SA2 show rapidly adapting responses in other angles. All the three types of LTMRs display strong angular tuning, and there is no significant difference in angular tuning index among them. Population wise, the majority of SA1 are tuned in the caudal direction, a large part of SA2 is tuned in the rostral direction, and RAs are tuned in multiple directions. In the angles showing strong tuning, the three LTMRs respond to increased stimulation amplitudes with increased impulse numbers in a hyperbola relationship, and the responsiveness based on impulse numbers is SA2 > SA1 > RA. Our findings provide new information on angular tuning properties of LTMRs in whisker hair follicles and help to understand directional encoding.
Collapse
|
48
|
A role for axon-glial interactions and Netrin-G1 signaling in the formation of low-threshold mechanoreceptor end organs. Proc Natl Acad Sci U S A 2022; 119:e2210421119. [PMID: 36252008 PMCID: PMC9618144 DOI: 10.1073/pnas.2210421119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Low-threshold mechanoreceptors (LTMRs) and their cutaneous end organs convert light mechanical forces acting on the skin into electrical signals that propagate to the central nervous system. In mouse hairy skin, hair follicle-associated longitudinal lanceolate complexes, which are end organs comprising LTMR axonal endings that intimately associate with terminal Schwann cell (TSC) processes, mediate LTMR responses to hair deflection and skin indentation. Here, we characterized developmental steps leading to the formation of Aβ rapidly adapting (RA)-LTMR and Aδ-LTMR lanceolate complexes. During early postnatal development, Aβ RA-LTMRs and Aδ-LTMRs extend and prune cutaneous axonal branches in close association with nascent TSC processes. Netrin-G1 is expressed in these developing Aβ RA-LTMR and Aδ-LTMR lanceolate endings, and Ntng1 ablation experiments indicate that Netrin-G1 functions in sensory neurons to promote lanceolate ending elaboration around hair follicles. The Netrin-G ligand (NGL-1), encoded by Lrrc4c, is expressed in TSCs, and ablation of Lrrc4c partially phenocopied the lanceolate complex deficits observed in Ntng1 mutants. Moreover, NGL-1-Netrin-G1 signaling is a general mediator of LTMR end organ formation across diverse tissue types demonstrated by the fact that Aβ RA-LTMR endings associated with Meissner corpuscles and Pacinian corpuscles are also compromised in the Ntng1 and Lrrc4c mutant mice. Thus, axon-glia interactions, mediated in part by NGL-1-Netrin-G1 signaling, promote LTMR end organ formation.
Collapse
|
49
|
Gotoh D, Saito T, Karnup S, Morizawa Y, Hori S, Nakai Y, Miyake M, Torimoto K, Fujimoto K, Yoshimura N. Therapeutic effects of a soluble guanylate cyclase activator, BAY 60-2770, on lower urinary tract dysfunction in mice with spinal cord injury. Am J Physiol Renal Physiol 2022; 323:F447-F454. [PMID: 35952343 PMCID: PMC9485004 DOI: 10.1152/ajprenal.00105.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/22/2022] Open
Abstract
We aimed to evaluate the effects of a soluble guanylate cyclase (sGC) activator, BAY 60-2770, on neurogenic lower urinary tract dysfunction in mice with spinal cord injury (SCI). Mice were divided into the following three groups: spinal cord intact (group A), SCI + vehicle (group B), and SCI + BAY 60-2770 (group C). SCI mice underwent Th8-Th9 spinal cord transection and treatment with BAY 60-2770 (10 mg/kg/day) once daily for 2-4 wk after SCI. We evaluated urodynamic parameters using awake cystometry and external urethral sphincter electromyograms (EMG); mRNA levels of mechanosensory channels, nitric oxide (NO)-, ischemia-, and inflammation-related markers in L6-S1 dorsal root ganglia, the urethra, and bladder tissues; and protein levels of cGMP in the urethra at 4 wk after SCI. With awake cystometry, nonvoiding contractions, postvoid residual, and bladder capacity were significantly larger in group B than in group C. Voiding efficiency (VE) was significantly higher in group C than in group B. In external urethral sphincter EMGs, the duration of notch-like reductions in intravesical pressure and reduced EMG activity time were significantly longer in group C than in group B. mRNA expression levels of transient receptor potential ankyrin 1, transient receptor potential vanilloid 1, acid-sensing ion channel (ASIC)1, ASIC2, ASIC3, and Piezo2 in the dorsal root ganglia, and hypoxia-inducible factor-1α, VEGF, and transforming growth factor-β1 in the bladder were significantly higher in group B than in groups A and C. mRNA levels of neuronal NO synthase, endothelial NO synthase, and sGCα1 and protein levels of cGMP in the urethra were significantly lower in group B than in groups A and C. sGC modulation might be useful for the treatment of SCI-related neurogenic lower urinary tract dysfunction.NEW & NOTEWORTHY This is the first report to evaluate the effects of a soluble guanylate cyclase activator, BAY 60-2770, on neurogenic lower urinary tract dysfunction in mice with spinal cord injury.
Collapse
Affiliation(s)
- Daisuke Gotoh
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Urology, Nara Medical University, Kashihara, Japan
| | - Tetsuichi Saito
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sergei Karnup
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yosuke Morizawa
- Department of Urology, Nara Medical University, Kashihara, Japan
| | - Shunta Hori
- Department of Urology, Nara Medical University, Kashihara, Japan
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, Kashihara, Japan
| | - Makito Miyake
- Department of Urology, Nara Medical University, Kashihara, Japan
| | | | | | - Naoki Yoshimura
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
50
|
Martin KK, Noble DJ, Parvin S, Jang K, Garraway SM. Pharmacogenetic inhibition of TrkB signaling in adult mice attenuates mechanical hypersensitivity and improves locomotor function after spinal cord injury. Front Cell Neurosci 2022; 16:987236. [PMID: 36226073 PMCID: PMC9548551 DOI: 10.3389/fncel.2022.987236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) signals through tropomyosin receptor kinase B (TrkB), to exert various types of plasticity. The exact involvement of BDNF and TrkB in neuropathic pain states after spinal cord injury (SCI) remains unresolved. This study utilized transgenic TrkBF616 mice to examine the effect of pharmacogenetic inhibition of TrkB signaling, induced by treatment with 1NM-PP1 (1NMP) in drinking water for 5 days, on formalin-induced inflammatory pain, pain hypersensitivity, and locomotor dysfunction after thoracic spinal contusion. We also examined TrkB, ERK1/2, and pERK1/2 expression in the lumbar spinal cord and trunk skin. The results showed that formalin-induced pain responses were robustly attenuated in 1NMP-treated mice. Weekly assessment of tactile sensitivity with the von Frey test showed that treatment with 1NMP immediately after SCI blocked the development of mechanical hypersensitivity up to 4 weeks post-SCI. Contrastingly, when treatment started 2 weeks after SCI, 1NMP reversibly and partially attenuated hind-paw hypersensitivity. Locomotor scores were significantly improved in the early-treated 1NMP mice compared to late-treated or vehicle-treated SCI mice. 1NMP treatment attenuated SCI-induced increases in TrkB and pERK1/2 levels in the lumbar cord but failed to exert similar effects in the trunk skin. These results suggest that early onset TrkB signaling after SCI contributes to maladaptive plasticity that leads to spinal pain hypersensitivity and impaired locomotor function.
Collapse
Affiliation(s)
| | | | | | | | - Sandra M. Garraway
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|