1
|
Koike T, Sato T, Oe S, Hayashi S, Seki-Omura R, Nakano Y, Iwashita H, Sato Y, Hirahara Y, Kitada M. CD34 distribution in C-fiber low threshold mechanoreceptors in the mouse dorsal root ganglion and spinal cord. Brain Res 2024; 1846:149227. [PMID: 39255903 DOI: 10.1016/j.brainres.2024.149227] [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: 05/01/2024] [Revised: 08/15/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
CD34 is a well-known cell marker of hematopoietic stem/ progenitor cells, endothelial cells, and fibrocytes. In the peripheral nervous system, a certain type of primary sensory neuron C-fiber low threshold mechanoreceptors (C-LTMRs) are reported to express CD34 mRNA. Here, we investigated the distribution of CD34 protein among putative C-LTMRs (pC-LTMR) using pC-LTMR markers such as VGLUT3 and TH in the dorsal root ganglion (DRG) and spinal cord. CD34 was frequently observed in DRG neurons double-positive for VGLUT3 and TH and single-positive for VGLUT3 in C8 and L4 levels, however, in C4 and L1 levels most of CD34-positive DRG neurons were demonstrated to be double-positive for VGLUT3 and TH. As for the termination, CD34-positive DRG neurons terminated in the ventral part of inner lamina II (lamina IIiv). At C4 and L1 levels of the dorsal horn, CD34 was observed in the entire region of lamina IIiv, however, in C8 and L4 levels of the dorsal horn CD34 was not detected in the medial part of lamina IIiv, which receives neural inputs from DRG neurons that innervate palm or sole skin. These results indicate that CD34 is expressed in pC-LTMRs and suggest that CD34 may play a role in providing C-LTMRs with a specific sensation by maintaining neural circuits.
Collapse
Affiliation(s)
- Taro Koike
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan.
| | - Tenga Sato
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Souichi Oe
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Shinichi Hayashi
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Ryohei Seki-Omura
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Yosuke Nakano
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Hikaru Iwashita
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Yuki Sato
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Yukie Hirahara
- Faculty of Nursing, Kansai Medical University, Shinmachi 2-2-2, Hirakata, Osaka, Japan
| | - Masaaki Kitada
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan.
| |
Collapse
|
2
|
Desbois M, Grill B. Molecular regulation of axon termination in mechanosensory neurons. Development 2024; 151:dev202945. [PMID: 39268828 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
|
3
|
Guo J, Li L, Chen F, Fu M, Cheng C, Wang M, Hu J, Pei L, Sun J. Forces Bless You: Mechanosensitive Piezo Channels in Gastrointestinal Physiology and Pathology. Biomolecules 2024; 14:804. [PMID: 39062518 PMCID: PMC11274378 DOI: 10.3390/biom14070804] [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: 05/22/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
The gastrointestinal (GI) tract is an organ actively involved in mechanical processes, where it detects forces via a mechanosensation mechanism. Mechanosensation relies on specialized cells termed mechanoreceptors, which convert mechanical forces into electrochemical signals via mechanosensors. The mechanosensitive Piezo1 and Piezo2 are widely expressed in various mechanosensitive cells that respond to GI mechanical forces by altering transmembrane ionic currents, such as epithelial cells, enterochromaffin cells, and intrinsic and extrinsic enteric neurons. This review highlights recent research advances on mechanosensitive Piezo channels in GI physiology and pathology. Specifically, the latest insights on the role of Piezo channels in the intestinal barrier, GI motility, and intestinal mechanosensation are summarized. Additionally, an overview of Piezo channels in the pathogenesis of GI disorders, including irritable bowel syndrome, inflammatory bowel disease, and GI cancers, is provided. Overall, the presence of mechanosensitive Piezo channels offers a promising new perspective for the treatment of various GI disorders.
Collapse
Affiliation(s)
- Jing Guo
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Li Li
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Feiyi Chen
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Minhan Fu
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Cheng Cheng
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Meizi Wang
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Jun Hu
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Lixia Pei
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Jianhua Sun
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| |
Collapse
|
4
|
Taub DG, Woolf CJ. Age-dependent small fiber neuropathy: Mechanistic insights from animal models. Exp Neurol 2024; 377:114811. [PMID: 38723859 PMCID: PMC11131160 DOI: 10.1016/j.expneurol.2024.114811] [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: 01/26/2024] [Revised: 04/07/2024] [Accepted: 05/05/2024] [Indexed: 05/28/2024]
Abstract
Small fiber neuropathy (SFN) is a common and debilitating disease in which the terminals of small diameter sensory axons degenerate, producing sensory loss, and in many patients neuropathic pain. While a substantial number of cases are attributable to diabetes, almost 50% are idiopathic. An underappreciated aspect of the disease is its late onset in most patients. Animal models of human genetic mutations that produce SFN also display age-dependent phenotypes suggesting that aging is an important contributor to the risk of development of the disease. In this review we define how particular sensory neurons are affected in SFN and discuss how aging may drive the disease. We also evaluate how animal models of SFN can define disease mechanisms that will provide insight into early risk detection and suggest novel therapeutic interventions.
Collapse
Affiliation(s)
- Daniel G Taub
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| | - Clifford J Woolf
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Ke J, Lu WC, Jing HY, Qian S, Moon SW, Cui GF, Qian WX, Che XJ, Zhang Q, Lai SS, Zhang L, Zhu YJ, Xie JD, Huang TW. Functional dissection of parabrachial substrates in processing nociceptive information. Zool Res 2024; 45:633-647. [PMID: 38766746 PMCID: PMC11188607 DOI: 10.24272/j.issn.2095-8137.2023.412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 05/22/2024] Open
Abstract
Painful stimuli elicit first-line reflexive defensive reactions and, in many cases, also evoke second-line recuperative behaviors, the latter of which reflects the sensing of tissue damage and the alleviation of suffering. The lateral parabrachial nucleus (lPBN), composed of external- (elPBN), dorsal- (dlPBN), and central/superior-subnuclei (jointly referred to as slPBN), receives sensory inputs from spinal projection neurons and plays important roles in processing affective information from external threats and body integrity disruption. However, the organizational rules of lPBN neurons that provoke diverse behaviors in response to different painful stimuli from cutaneous and deep tissues remain unclear. In this study, we used region-specific neuronal depletion or silencing approaches combined with a battery of behavioral assays to show that slPBN neurons expressing substance P receptor ( NK1R) (lPBN NK1R) are crucial for driving pain-associated self-care behaviors evoked by sustained noxious thermal and mechanical stimuli applied to skin or bone/muscle, while elPBN neurons are dispensable for driving such reactions. Notably, lPBN NK1R neurons are specifically required for forming sustained somatic pain-induced negative teaching signals and aversive memory but are not necessary for fear-learning or escape behaviors elicited by external threats. Lastly, both lPBN NK1R and elPBN neurons contribute to chemical irritant-induced nocifensive reactions. Our results reveal the functional organization of parabrachial substrates that drive distinct behavioral outcomes in response to sustained pain versus external danger under physiological conditions.
Collapse
Affiliation(s)
- Jin Ke
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei-Cheng Lu
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Hai-Yang Jing
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Shen Qian
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Sun-Wook Moon
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Guang-Fu Cui
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Wei-Xin Qian
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiao-Jing Che
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhang
- Department of Anesthesiology, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Shi-Shi Lai
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, China
| | - Ling Zhang
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ying-Jie Zhu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China. E-mail:
| | - Jing-Dun Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China. E-mail:
| | - Tian-Wen Huang
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China. E-mail:
| |
Collapse
|
6
|
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: 0] [Impact Index Per Article: 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
|
7
|
Tong C, Moayedi Y, Lumpkin EA. Merkel cells and keratinocytes in oral mucosa are activated by mechanical stimulation. Physiol Rep 2024; 12:e15826. [PMID: 38246872 PMCID: PMC10800296 DOI: 10.14814/phy2.15826] [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: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 01/23/2024] Open
Abstract
The detection of mechanical qualities of foodstuffs is essential for nutrient acquisition, evaluation of food freshness, and bolus formation during mastication. However, the mechanisms through which mechanosensitive cells in the oral cavity transmit mechanical information from the periphery to the brain are not well defined. We hypothesized Merkel cells, which are epithelial mechanoreceptors and important for pressure and texture sensing in the skin, can be mechanically activated in the oral cavity. Using live-cell calcium imaging, we recorded Merkel cell activity in ex vivo gingival and palatal preparations from mice in response to mechanical stimulation. Merkel cells responded with distinct temporal patterns and activation thresholds in a region-specific manner, with Merkel cells in the hard palate having a higher mean activation threshold than those in the gingiva. Unexpectedly, we found that oral keratinocytes were also activated by mechanical stimulation, even in the absence of Merkel cells. This indicates that mechanical stimulation of oral mucosa independently activates at least two subpopulations of epithelial cells. Finally, we found that oral Merkel cells contribute to preference for consuming oily emulsion. To our knowledge, these data represent the first functional study of Merkel-cell physiology and its role in flavor detection in the oral cavity.
Collapse
Affiliation(s)
- Chi‐Kun Tong
- Department of Physiology and Cellular BiophysicsColumbia University Medical CenterNew YorkNew YorkUSA
| | - Yalda Moayedi
- Department of Physiology and Cellular BiophysicsColumbia University Medical CenterNew YorkNew YorkUSA
- Present address:
Departments of Neurology and Otolaryngology‐Head and Neck SurgeryColumbia UniversityNew YorkNYUSA
| | - Ellen A. Lumpkin
- Department of Physiology and Cellular BiophysicsColumbia University Medical CenterNew YorkNew YorkUSA
- Department of DermatologyColumbia University Medical CenterNew YorkNew YorkUSA
- Present address:
Department of Molecular and Cell BiologyHelen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyCAUSA
| |
Collapse
|
8
|
Villarino NW, Hamed YMF, Ghosh B, Dubin AE, Lewis AH, Odem MA, Loud MC, Wang Y, Servin-Vences MR, Patapoutian A, Marshall KL. Labeling PIEZO2 activity in the peripheral nervous system. Neuron 2023; 111:2488-2501.e8. [PMID: 37321223 PMCID: PMC10527906 DOI: 10.1016/j.neuron.2023.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 03/24/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Sensory neurons detect mechanical forces from both the environment and internal organs to regulate physiology. PIEZO2 is a mechanosensory ion channel critical for touch, proprioception, and bladder stretch sensation, yet its broad expression in sensory neurons suggests it has undiscovered physiological roles. To fully understand mechanosensory physiology, we must know where and when PIEZO2-expressing neurons detect force. The fluorescent styryl dye FM 1-43 was previously shown to label sensory neurons. Surprisingly, we find that the vast majority of FM 1-43 somatosensory neuron labeling in mice in vivo is dependent on PIEZO2 activity within the peripheral nerve endings. We illustrate the potential of FM 1-43 by using it to identify novel PIEZO2-expressing urethral neurons that are engaged by urination. These data reveal that FM 1-43 is a functional probe for mechanosensitivity via PIEZO2 activation in vivo and will facilitate the characterization of known and novel mechanosensory processes in multiple organ systems.
Collapse
Affiliation(s)
- Nicholas W Villarino
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yasmeen M F Hamed
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Development, Disease Models, and Therapeutics, Baylor College of Medicine, Houston, TX 77030
| | - Britya Ghosh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Adrienne E Dubin
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Amanda H Lewis
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Max A Odem
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meaghan C Loud
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yu Wang
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - M Rocio Servin-Vences
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ardem Patapoutian
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Kara L Marshall
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
9
|
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: 8] [Impact Index Per Article: 8.0] [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
|
10
|
Mathiesen BK, Miyakoshi LM, Cederroth CR, Tserga E, Versteegh C, Bork PAR, Hauglund NL, Gomolka RS, Mori Y, Edvall NK, Rouse S, Møllgård K, Holt JR, Nedergaard M, Canlon B. Delivery of gene therapy through a cerebrospinal fluid conduit to rescue hearing in adult mice. Sci Transl Med 2023; 15:eabq3916. [PMID: 37379370 DOI: 10.1126/scitranslmed.abq3916] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/09/2023] [Indexed: 06/30/2023]
Abstract
Inner ear gene therapy has recently effectively restored hearing in neonatal mice, but it is complicated in adulthood by the structural inaccessibility of the cochlea, which is embedded within the temporal bone. Alternative delivery routes may advance auditory research and also prove useful when translated to humans with progressive genetic-mediated hearing loss. Cerebrospinal fluid flow via the glymphatic system is emerging as a new approach for brain-wide drug delivery in rodents as well as humans. The cerebrospinal fluid and the fluid of the inner ear are connected via a bony channel called the cochlear aqueduct, but previous studies have not explored the possibility of delivering gene therapy via the cerebrospinal fluid to restore hearing in adult deaf mice. Here, we showed that the cochlear aqueduct in mice exhibits lymphatic-like characteristics. In vivo time-lapse magnetic resonance imaging, computed tomography, and optical fluorescence microscopy showed that large-particle tracers injected into the cerebrospinal fluid reached the inner ear by dispersive transport via the cochlear aqueduct in adult mice. A single intracisternal injection of adeno-associated virus carrying solute carrier family 17, member 8 (Slc17A8), which encodes vesicular glutamate transporter-3 (VGLUT3), rescued hearing in adult deaf Slc17A8-/- mice by restoring VGLUT3 protein expression in inner hair cells, with minimal ectopic expression in the brain and none in the liver. Our findings demonstrate that cerebrospinal fluid transport comprises an accessible route for gene delivery to the adult inner ear and may represent an important step toward using gene therapy to restore hearing in humans.
Collapse
Affiliation(s)
- Barbara K Mathiesen
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Denmark
| | - Leo M Miyakoshi
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Denmark
| | - Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, Biomedicum, 171 65 Stockholm, Sweden
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Evangelia Tserga
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, Biomedicum, 171 65 Stockholm, Sweden
| | - Corstiaen Versteegh
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, Biomedicum, 171 65 Stockholm, Sweden
| | - Peter A R Bork
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Denmark
| | - Natalie L Hauglund
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Denmark
| | - Ryszard Stefan Gomolka
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Denmark
| | - Yuki Mori
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Denmark
| | - Niklas K Edvall
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, Biomedicum, 171 65 Stockholm, Sweden
| | - Stephanie Rouse
- Department of Otolaryngology and Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen; Copenhagen, 2200, Denmark
| | - Jeffrey R Holt
- Department of Otolaryngology and Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Denmark
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center; Rochester, NY 14642, USA
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, Biomedicum, 171 65 Stockholm, Sweden
| |
Collapse
|
11
|
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
|
12
|
Huo J, Du F, Duan K, Yin G, Liu X, Ma Q, Dong D, Sun M, Hao M, Su D, Huang T, Ke J, Lai S, Zhang Z, Guo C, Sun Y, Cheng L. Identification of brain-to-spinal circuits controlling the laterality and duration of mechanical allodynia in mice. Cell Rep 2023; 42:112300. [PMID: 36952340 DOI: 10.1016/j.celrep.2023.112300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 12/22/2022] [Accepted: 03/07/2023] [Indexed: 03/24/2023] Open
Abstract
Mechanical allodynia (MA) represents one prevalent symptom of chronic pain. Previously we and others have identified spinal and brain circuits that transmit or modulate the initial establishment of MA. However, brain-derived descending pathways that control the laterality and duration of MA are still poorly understood. Here we report that the contralateral brain-to-spinal circuits, from Oprm1 neurons in the lateral parabrachial nucleus (lPBNOprm1), via Pdyn neurons in the dorsal medial regions of hypothalamus (dmHPdyn), to the spinal dorsal horn (SDH), act to prevent nerve injury from inducing contralateral MA and reduce the duration of bilateral MA induced by capsaicin. Ablating/silencing dmH-projecting lPBNOprm1 neurons or SDH-projecting dmHPdyn neurons, deleting Dyn peptide from dmH, or blocking spinal κ-opioid receptors all led to long-lasting bilateral MA. Conversely, activation of dmHPdyn neurons or their axonal terminals in SDH can suppress sustained bilateral MA induced by lPBN lesion.
Collapse
Affiliation(s)
- Jiantao Huo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Feng Du
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kaifang Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guangjuan Yin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xi Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Quan Ma
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dong Dong
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mengge Sun
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mei Hao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dongmei Su
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tianwen Huang
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Jin Ke
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Shishi Lai
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Zhi Zhang
- Division of Life Sciences and Medicine, CAS Key Laboratory of Brain Function and Diseases, University of Science and Technology of China, Hefei 230027, China
| | - Chao Guo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuanjie Sun
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Longzhen Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
13
|
Deng T, Jovanovic VM, Tristan CA, Weber C, Chu PH, Inman J, Ryu S, Jethmalani Y, Ferreira de Sousa J, Ormanoglu P, Twumasi P, Sen C, Shim J, Jayakar S, Bear Zhang HX, Jo S, Yu W, Voss TC, Simeonov A, Bean BP, Woolf CJ, Singeç I. Scalable generation of sensory neurons from human pluripotent stem cells. Stem Cell Reports 2023; 18:1030-1047. [PMID: 37044067 PMCID: PMC10147831 DOI: 10.1016/j.stemcr.2023.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 04/14/2023] Open
Abstract
Development of new non-addictive analgesics requires advanced strategies to differentiate human pluripotent stem cells (hPSCs) into relevant cell types. Following principles of developmental biology and translational applicability, here we developed an efficient stepwise differentiation method for peptidergic and non-peptidergic nociceptors. By modulating specific cell signaling pathways, hPSCs were first converted into SOX10+ neural crest, followed by differentiation into sensory neurons. Detailed characterization, including ultrastructural analysis, confirmed that the hPSC-derived nociceptors displayed cellular and molecular features comparable to native dorsal root ganglion (DRG) neurons, and expressed high-threshold primary sensory neuron markers, transcription factors, neuropeptides, and over 150 ion channels and receptors relevant for pain research and axonal growth/regeneration studies (e.g., TRPV1, NAV1.7, NAV1.8, TAC1, CALCA, GAP43, DPYSL2, NMNAT2). Moreover, after confirming robust functional activities and differential response to noxious stimuli and specific drugs, a robotic cell culture system was employed to produce large quantities of human sensory neurons, which can be used to develop nociceptor-selective analgesics.
Collapse
Affiliation(s)
- Tao Deng
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Vukasin M Jovanovic
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Carlos A Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Claire Weber
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pei-Hsuan Chu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Jason Inman
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Yogita Jethmalani
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Juliana Ferreira de Sousa
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Prisca Twumasi
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Chaitali Sen
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Jaehoon Shim
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Selwyn Jayakar
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Weifeng Yu
- Sophion Bioscience, North Brunswick, NJ 08902, USA
| | - Ty C Voss
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA.
| |
Collapse
|
14
|
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: 9] [Impact Index Per Article: 9.0] [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
|
15
|
Elias LJ, Succi IK, Schaffler MD, Foster W, Gradwell MA, Bohic M, Fushiki A, Upadhyay A, Ejoh LL, Schwark R, Frazer R, Bistis B, Burke JE, Saltz V, Boyce JE, Jhumka A, Costa RM, Abraira VE, Abdus-Saboor I. Touch neurons underlying dopaminergic pleasurable touch and sexual receptivity. Cell 2023; 186:577-590.e16. [PMID: 36693373 PMCID: PMC9898224 DOI: 10.1016/j.cell.2022.12.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 10/21/2022] [Accepted: 12/20/2022] [Indexed: 01/24/2023]
Abstract
Pleasurable touch is paramount during social behavior, including sexual encounters. However, the identity and precise role of sensory neurons that transduce sexual touch remain unknown. A population of sensory neurons labeled by developmental expression of the G protein-coupled receptor Mrgprb4 detects mechanical stimulation in mice. Here, we study the social relevance of Mrgprb4-lineage neurons and reveal that these neurons are required for sexual receptivity and sufficient to induce dopamine release in the brain. Even in social isolation, optogenetic stimulation of Mrgprb4-lineage neurons through the back skin is sufficient to induce a conditioned place preference and a striking dorsiflexion resembling the lordotic copulatory posture. In the absence of Mrgprb4-lineage neurons, female mice no longer find male mounts rewarding: sexual receptivity is supplanted by aggression and a coincident decline in dopamine release in the nucleus accumbens. Together, these findings establish that Mrgprb4-lineage neurons initiate a skin-to-brain circuit encoding the rewarding quality of social touch.
Collapse
Affiliation(s)
- Leah J Elias
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Isabella K Succi
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Melanie D Schaffler
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - William Foster
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Mark A Gradwell
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Akira Fushiki
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Aman Upadhyay
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Lindsay L Ejoh
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Schwark
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Rachel Frazer
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Brittany Bistis
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Jessica E Burke
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Victoria Saltz
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Jared E Boyce
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Anissa Jhumka
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Rui M Costa
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Victoria E Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA.
| |
Collapse
|
16
|
Noble DJ, Dongmo R, Parvin S, Martin KK, Garraway SM. C-low threshold mechanoreceptor activation becomes sufficient to trigger affective pain in spinal cord-injured mice in association with increased respiratory rates. Front Integr Neurosci 2022; 16:1081172. [PMID: 36619238 PMCID: PMC9811591 DOI: 10.3389/fnint.2022.1081172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
The mechanisms of neuropathic pain after spinal cord injury (SCI) are not fully understood. In addition to the plasticity that occurs within the injured spinal cord, peripheral processes, such as hyperactivity of primary nociceptors, are critical to the expression of pain after SCI. In adult rats, truncal stimulation within the tuning range of C-low threshold mechanoreceptors (C-LTMRs) contributes to pain hypersensitivity and elevates respiratory rates (RRs) after SCI. This suggests that C-LTMRs, which normally encode pleasant, affiliative touch, undergo plasticity to transmit pain sensation following injury. Because tyrosine hydroxylase (TH) expression is a specific marker of C-LTMRs, in the periphery, here we used TH-Cre adult mice to investigate more specifically the involvement of C-LTMRs in at-level pain after thoracic contusion SCI. Using a modified light-dark chamber conditioned place aversion (CPA) paradigm, we assessed chamber preferences and transitions between chambers at baseline, and in response to mechanical and optogenetic stimulation of C-LTMRs. In parallel, at baseline and select post-surgical timepoints, mice underwent non-contact RR recordings and von Frey assessment of mechanical hypersensitivity. The results showed that SCI mice avoided the chamber associated with C-LTMR stimulation, an effect that was more pronounced with optical stimulation. They also displayed elevated RRs at rest and during CPA training sessions. Importantly, these changes were restricted to chronic post-surgery timepoints, when hindpaw mechanical hypersensitivity was also evident. Together, these results suggest that C-LTMR afferent plasticity, coexisting with potentially facilitatory changes in breathing, drives at-level affective pain following SCI in adult mice.
Collapse
|
17
|
Tlx3 controls the development of C-low threshold mechanoreceptors. Neuroreport 2022; 33:617-622. [DOI: 10.1097/wnr.0000000000001824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
18
|
Cao Z, Huang C, Lu F, Jiang X, Hu Y, Cao C, Liu Z. Meis1 Regulates Nociceptor Development and Behavioral Response to Tactile Stimuli. Front Mol Neurosci 2022; 15:901466. [PMID: 35875660 PMCID: PMC9301487 DOI: 10.3389/fnmol.2022.901466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Nociceptors in the dorsal root ganglia (DRG) and trigeminal ganglia (TG) are necessary for transmitting pain and itch signals. However, the molecular mechanism regulating nociceptor development remains largely unknown. This study identifies that the transcription factor Meis1 is generally expressed in two groups of sensory neurons in the developing DRG. During prenatal and neonatal stages, approximately 2/3 of Meis1+ neurons are Runx1+ nociceptors, while 1/3 of Meis1+ neurons are NF200+ myelinated neurons. At postnatal stages, Meis1 expression in nociceptors is gradually reduced. Here, we constructed a Meis1 conditional knockout mouse line to selectively delete Meis1 in Nav1.8 lineage nociceptors. Microarray analyses showed that differentially expressed genes in the Meis1 mutant DRG were enriched in pathways related to sensory perception of pain and nervous system development. In addition, Meis1 regulates the expression of some marker genes of Nppb+ neurons and C-LTMRs. Furthermore, Meis1 mutant mice exhibit behavioral deficits in response to light mechanical pain, static touch and chemical itch. Therefore, this study reveals that Meis1 is required to regulate the development of nociceptors.
Collapse
Affiliation(s)
- Zheng Cao
- Beijing Institute of Biotechnology, Beijing, China.,School of Biological Engineering and Food Science, Hubei University of Technology, Wuhan, China
| | - Chengcheng Huang
- Beijing Institute of Biotechnology, Beijing, China.,General Hospital of Central Theater Command, Wuhan, China
| | - Fumin Lu
- Beijing Institute of Biotechnology, Beijing, China
| | - Xuequan Jiang
- Beijing Institute of Biotechnology, Beijing, China.,School of Biological Engineering and Food Science, Hubei University of Technology, Wuhan, China
| | - Yong Hu
- Beijing Institute of Biotechnology, Beijing, China
| | - Cheng Cao
- Beijing Institute of Biotechnology, Beijing, China
| | - Zijing Liu
- Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
19
|
Bridging skin, brain, and behavior to understand pleasurable social touch. Curr Opin Neurobiol 2022; 73:102527. [PMID: 35453001 DOI: 10.1016/j.conb.2022.102527] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023]
Abstract
Social touch-the affiliative skin-to-skin contact between individuals-can rapidly evoke emotions of comfort, pleasure, or calm, and is essential for mental and physical well-being. Physical isolation from social support can be devastating. During the COVID-19 pandemic, we observed a global increase in suicidal ideation, anxiety, domestic violence, and worsening of pre-existing physical conditions, alerting society to our need to understand the neurobiology of social touch and how it promotes normal health. Gaining a mechanistic understanding of how sensory neuron stimulation induces pleasure, calm, and analgesia may reveal untapped therapeutic targets in the periphery for treatment of anxiety and depression, as well as social disorders and traumas in which social touch becomes aversive. Bridging the gap between stimulation in the skin and positive affect in the brain-especially during naturally occurring social touch behaviors-remains a challenge to the field. However, with advances in mouse genetics, behavioral quantification, and brain imaging approaches to measure neuronal firing and neurochemical release, completing this mechanistic picture may be on the horizon. Here, we summarize some exciting new findings about social touch in mammals, emphasizing both the peripheral and central nervous systems, with attempts to bridge the gap between external stimulation and internal representations in the brain.
Collapse
|
20
|
Ma Q. A functional subdivision within the somatosensory system and its implications for pain research. Neuron 2022; 110:749-769. [PMID: 35016037 PMCID: PMC8897275 DOI: 10.1016/j.neuron.2021.12.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/07/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022]
Abstract
Somatosensory afferents are traditionally classified by soma size, myelination, and their response specificity to external and internal stimuli. Here, we propose the functional subdivision of the nociceptive somatosensory system into two branches. The exteroceptive branch detects external threats and drives reflexive-defensive reactions to prevent or limit injury. The interoceptive branch senses the disruption of body integrity, produces tonic pain with strong aversive emotional components, and drives self-caring responses toward to the injured region to reduce suffering. The central thesis behind this functional subdivision comes from a reflection on the dilemma faced by the pain research field, namely, the use of reflexive-defensive behaviors as surrogate assays for interoceptive tonic pain. The interpretation of these assays is now being challenged by the discovery of distinct but interwoven circuits that drive exteroceptive versus interoceptive types of behaviors, with the conflation of these two components contributing partially to the poor translation of therapies from preclinical studies.
Collapse
Affiliation(s)
- Qiufu Ma
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Larsson M, Nagi SS. Role of C-tactile fibers in pain modulation: animal and human perspectives. Curr Opin Behav Sci 2022. [DOI: 10.1016/j.cobeha.2021.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
22
|
|
23
|
Fazekas CL, Szabó A, Török B, Bánrévi K, Correia P, Chaves T, Daumas S, Zelena D. A New Player in the Hippocampus: A Review on VGLUT3+ Neurons and Their Role in the Regulation of Hippocampal Activity and Behaviour. Int J Mol Sci 2022; 23:790. [PMID: 35054976 PMCID: PMC8775679 DOI: 10.3390/ijms23020790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 01/05/2023] Open
Abstract
Glutamate is the most abundant excitatory amino acid in the central nervous system. Neurons using glutamate as a neurotransmitter can be characterised by vesicular glutamate transporters (VGLUTs). Among the three subtypes, VGLUT3 is unique, co-localising with other "classical" neurotransmitters, such as the inhibitory GABA. Glutamate, manipulated by VGLUT3, can modulate the packaging as well as the release of other neurotransmitters and serve as a retrograde signal through its release from the somata and dendrites. Its contribution to sensory processes (including seeing, hearing, and mechanosensation) is well characterised. However, its involvement in learning and memory can only be assumed based on its prominent hippocampal presence. Although VGLUT3-expressing neurons are detectable in the hippocampus, most of the hippocampal VGLUT3 positivity can be found on nerve terminals, presumably coming from the median raphe. This hippocampal glutamatergic network plays a pivotal role in several important processes (e.g., learning and memory, emotions, epilepsy, cardiovascular regulation). Indirect information from anatomical studies and KO mice strains suggests the contribution of local VGLUT3-positive hippocampal neurons as well as afferentations in these events. However, further studies making use of more specific tools (e.g., Cre-mice, opto- and chemogenetics) are needed to confirm these assumptions.
Collapse
Affiliation(s)
- Csilla Lea Fazekas
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
- Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS) INSERM, Sorbonne Université, CNRS, 75005 Paris, France;
| | - Adrienn Szabó
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Bibiána Török
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Krisztina Bánrévi
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
| | - Pedro Correia
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Tiago Chaves
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Stéphanie Daumas
- Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS) INSERM, Sorbonne Université, CNRS, 75005 Paris, France;
| | - Dóra Zelena
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (C.L.F.); (A.S.); (B.T.); (K.B.); (P.C.); (T.C.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
24
|
Chakrabarti R, Jaime Tobón LM, Slitin L, Redondo Canales M, Hoch G, Slashcheva M, Fritsch E, Bodensiek K, Özçete ÖD, Gültas M, Michanski S, Opazo F, Neef J, Pangrsic T, Moser T, Wichmann C. Optogenetics and electron tomography for structure-function analysis of cochlear ribbon synapses. eLife 2022; 11:79494. [PMID: 36562477 PMCID: PMC9908081 DOI: 10.7554/elife.79494] [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: 04/14/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Ribbon synapses of cochlear inner hair cells (IHCs) are specialized to indefatigably transmit sound information at high rates. To understand the underlying mechanisms, structure-function analysis of the active zone (AZ) of these synapses is essential. Previous electron microscopy studies of synaptic vesicle (SV) dynamics at the IHC AZ used potassium stimulation, which limited the temporal resolution to minutes. Here, we established optogenetic IHC stimulation followed by quick freezing within milliseconds and electron tomography to study the ultrastructure of functional synapse states with good temporal resolution in mice. We characterized optogenetic IHC stimulation by patch-clamp recordings from IHCs and postsynaptic boutons revealing robust IHC depolarization and neurotransmitter release. Ultrastructurally, the number of docked SVs increased upon short (17-25 ms) and long (48-76 ms) light stimulation paradigms. We did not observe enlarged SVs or other morphological correlates of homotypic fusion events. Our results indicate a rapid recruitment of SVs to the docked state upon stimulation and suggest that univesicular release prevails as the quantal mechanism of exocytosis at IHC ribbon synapses.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Lina María Jaime Tobón
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Loujin Slitin
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Magdalena Redondo Canales
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Gerhard Hoch
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Marina Slashcheva
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of GöttingenGöttingenGermany
| | - Elisabeth Fritsch
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of GöttingenGöttingenGermany
| | - Kai Bodensiek
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany
| | - Özge Demet Özçete
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Mehmet Gültas
- Faculty of Agriculture, South Westphalia University of Applied SciencesSoestGermany
| | - Susann Michanski
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Felipe Opazo
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,NanoTag Biotechnologies GmbHGöttingenGermany,Institute of Neuro- and Sensory Physiology, University Medical Center GöttingenGöttingenGermany
| | - Jakob Neef
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Tina Pangrsic
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany,Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany
| | - Tobias Moser
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany
| |
Collapse
|
25
|
Parpaite T, Brosse L, Séjourné N, Laur A, Mechioukhi Y, Delmas P, Coste B. Patch-seq of mouse DRG neurons reveals candidate genes for specific mechanosensory functions. Cell Rep 2021; 37:109914. [PMID: 34731626 PMCID: PMC8578708 DOI: 10.1016/j.celrep.2021.109914] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/16/2021] [Accepted: 10/09/2021] [Indexed: 12/13/2022] Open
Abstract
A variety of mechanosensory neurons are involved in touch, proprioception, and pain. Many molecular components of the mechanotransduction machinery subserving these sensory modalities remain to be discovered. Here, we combine recordings of mechanosensitive (MS) currents in mechanosensory neurons with single-cell RNA sequencing. Transcriptional profiles are mapped onto previously identified sensory neuron types to identify cell-type correlates between datasets. Correlation of current signatures with single-cell transcriptomes provides a one-to-one correspondence between mechanoelectric properties and transcriptomically defined neuronal populations. Moreover, a gene-expression differential comparison provides a set of candidate genes for mechanotransduction complexes. Piezo2 is expectedly found to be enriched in rapidly adapting MS current-expressing neurons, whereas Tmem120a and Tmem150c, thought to mediate slow-type MS currents, are uniformly expressed in all mechanosensory neuron subtypes. Further knockdown experiments disqualify them as mediating MS currents in sensory neurons. This dataset constitutes an open resource to explore further the cell-type-specific determinants of mechanosensory properties.
Collapse
Affiliation(s)
- Thibaud Parpaite
- Aix Marseille Université, CNRS, LNC-UMR 7291, 13344 Marseille, France
| | - Lucie Brosse
- Aix Marseille Université, CNRS, LNC-UMR 7291, 13344 Marseille, France
| | - Nina Séjourné
- Aix Marseille Université, CNRS, LNC-UMR 7291, 13344 Marseille, France
| | - Amandine Laur
- Aix Marseille Université, CNRS, LNC-UMR 7291, 13344 Marseille, France
| | | | - Patrick Delmas
- Aix Marseille Université, CNRS, LNC-UMR 7291, 13344 Marseille, France
| | - Bertrand Coste
- Aix Marseille Université, CNRS, LNC-UMR 7291, 13344 Marseille, France.
| |
Collapse
|
26
|
Middleton SJ, Perez-Sanchez J, Dawes JM. The structure of sensory afferent compartments in health and disease. J Anat 2021; 241:1186-1210. [PMID: 34528255 PMCID: PMC9558153 DOI: 10.1111/joa.13544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/12/2021] [Accepted: 08/30/2021] [Indexed: 12/20/2022] Open
Abstract
Primary sensory neurons are a heterogeneous population of cells able to respond to both innocuous and noxious stimuli. Like most neurons they are highly compartmentalised, allowing them to detect, convey and transfer sensory information. These compartments include specialised sensory endings in the skin, the nodes of Ranvier in myelinated axons, the cell soma and their central terminals in the spinal cord. In this review, we will highlight the importance of these compartments to primary afferent function, describe how these structures are compromised following nerve damage and how this relates to neuropathic pain.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - John M Dawes
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
27
|
Leveraging VGLUT3 Functions to Untangle Brain Dysfunctions. Trends Pharmacol Sci 2021; 42:475-490. [PMID: 33775453 DOI: 10.1016/j.tips.2021.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 11/21/2022]
Abstract
Vesicular glutamate transporters (VGLUTs) were long thought to be specific markers of glutamatergic excitatory transmission. The discovery, two decades ago, of the atypical VGLUT3 has thoroughly modified this oversimplified view. VGLUT3 is strategically expressed in discrete populations of glutamatergic, cholinergic, serotonergic, and even GABAergic neurons. Recent reports show the subtle, but critical, implications of VGLUT3-dependent glutamate co-transmission and its roles in the regulation of diverse brain functions and dysfunctions. Progress in the neuropharmacology of VGLUT3 could lead to decisive breakthroughs in the treatment of Parkinson's disease (PD), addiction, eating disorders, anxiety, presbycusis, or pain. This review summarizes recent findings on VGLUT3 and its vesicular underpinnings as well as on possible ways to target this atypical transporter for future therapeutic strategies.
Collapse
|
28
|
Sakamoto T, Ishio Y, Ishida Y, Mogi K, Kikusui T. Low maternal licking/grooming stimulation increases pain sensitivity in male mouse offspring. Exp Anim 2021; 70:13-21. [PMID: 32741955 PMCID: PMC7887629 DOI: 10.1538/expanim.20-0030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
Deprivation of maternal care has been associated with higher pain sensitivity in offspring. In the present study, we hypothesized that the maternal licking/grooming behavior was an important factor for the development of the pain regulatory system. To test this hypothesis, we used male F2 offspring of early-weaned (EW) F1 mother mice that exhibit lower frequency of licking/grooming behavior. The formalin test revealed that F2 offspring of EW F1 dams showed significantly higher pain behavior than F2 offspring of normally-weaned (NW) F1 dams. We found that the mRNA levels of transient receptor potential vanilloid 1 (TRPV1), a nociceptor, were higher in the lumbosacral dorsal root ganglion (DRG) of F2 offspring of EW F1 dams than those of F2 offspring of NW F1 dams, suggesting that the higher pain sensitivity may be attributed to low licking/grooming, which may result in developmental changes in nociceptive neurons. In the DRG, mRNA levels of Mas-related G-protein coupled receptor B4 (MrgprB4), a marker of sensory neurons that detect gentle stroking, was also up-regulated in the F2 offspring of EW F1 dams. Considering that gentle touch alleviates pain, Mrgprb4 up-regulation may reflect a compensatory change. The present findings indicate important implications of maternal licking/grooming behavior in the development of the pain regulatory system.
Collapse
Affiliation(s)
- Takashi Sakamoto
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Yukino Ishio
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Yuiko Ishida
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Kazutaka Mogi
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| | - Takefumi Kikusui
- Department of Animal Science and Biotechnology, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa 252-5201, Japan
| |
Collapse
|
29
|
Watkins RH, Dione M, Ackerley R, Backlund Wasling H, Wessberg J, Löken LS. Evidence for sparse C-tactile afferent innervation of glabrous human hand skin. J Neurophysiol 2020; 125:232-237. [PMID: 33296618 DOI: 10.1152/jn.00587.2020] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
C-tactile (CT) afferents were long-believed to be lacking in humans, but these were subsequently shown to densely innervate the face and arm skin, and to a lesser extent the leg. Their firing frequency to stroking touch at different velocities has been correlated with ratings of tactile pleasantness. CT afferents were thought to be absent in human glabrous skin; however, tactile pleasantness can be perceived across the whole body, including glabrous hand skin. We used microneurography to investigate mechanoreceptive afferents in the glabrous skin of the human hand, during median and radial nerve recordings. We describe CTs found in the glabrous skin, with characteristics comparable with those in hairy arm skin, and detail recordings from three such afferents. CTs were infrequently encountered in the glabrous skin and we estimate that the ratio of recorded CTs relative to myelinated mechanoreceptors (1:80) corresponds to an absolute innervation density of around seven times lower than in hairy skin. This sparse innervation sheds light on discrepancies between psychophysical findings of touch perception on glabrous skin and hairy skin, although the role of these CT afferents in the glabrous skin remains subject to future work.NEW & NOTEWORTHY Human touch is encoded by low-threshold mechanoreceptors, including myelinated Aβ afferents and unmyelinated C-tactile (CT) afferents. CTs are abundant in hairy skin and are thought to code gentle, stroking touch that signals positive affective interactions. CTs have never been described in human glabrous skin, yet we show evidence of their existence on the hand, albeit at a relatively low density. Glabrous skin CTs may provide modulatory reinforcement of gentle tactile interactions during touch using the hands.
Collapse
Affiliation(s)
- Roger Holmes Watkins
- LNC (Laboratoire de Neurosciences Cognitives (UMR 7291), Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Marseille, France.,Department of Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Mariama Dione
- LNC (Laboratoire de Neurosciences Cognitives (UMR 7291), Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Marseille, France.,Department of Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Rochelle Ackerley
- LNC (Laboratoire de Neurosciences Cognitives (UMR 7291), Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université, Marseille, France.,Department of Physiology, University of Gothenburg, Gothenburg, Sweden
| | | | - Johan Wessberg
- Department of Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Line S Löken
- Department of Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
30
|
Vermeiren S, Bellefroid EJ, Desiderio S. Vertebrate Sensory Ganglia: Common and Divergent Features of the Transcriptional Programs Generating Their Functional Specialization. Front Cell Dev Biol 2020; 8:587699. [PMID: 33195244 PMCID: PMC7649826 DOI: 10.3389/fcell.2020.587699] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Sensory fibers of the peripheral nervous system carry sensation from specific sense structures or use different tissues and organs as receptive fields, and convey this information to the central nervous system. In the head of vertebrates, each cranial sensory ganglia and associated nerves perform specific functions. Sensory ganglia are composed of different types of specialized neurons in which two broad categories can be distinguished, somatosensory neurons relaying all sensations that are felt and visceral sensory neurons sensing the internal milieu and controlling body homeostasis. While in the trunk somatosensory neurons composing the dorsal root ganglia are derived exclusively from neural crest cells, somato- and visceral sensory neurons of cranial sensory ganglia have a dual origin, with contributions from both neural crest and placodes. As most studies on sensory neurogenesis have focused on dorsal root ganglia, our understanding of the molecular mechanisms underlying the embryonic development of the different cranial sensory ganglia remains today rudimentary. However, using single-cell RNA sequencing, recent studies have made significant advances in the characterization of the neuronal diversity of most sensory ganglia. Here we summarize the general anatomy, function and neuronal diversity of cranial sensory ganglia. We then provide an overview of our current knowledge of the transcriptional networks controlling neurogenesis and neuronal diversification in the developing sensory system, focusing on cranial sensory ganglia, highlighting specific aspects of their development and comparing it to that of trunk sensory ganglia.
Collapse
Affiliation(s)
- Simon Vermeiren
- ULB Neuroscience Institute, Université Libre de Bruxelles, Gosselies, Belgium
| | - Eric J Bellefroid
- ULB Neuroscience Institute, Université Libre de Bruxelles, Gosselies, Belgium
| | - Simon Desiderio
- Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier, France
| |
Collapse
|
31
|
Low-Threshold Mechanosensitive VGLUT3-Lineage Sensory Neurons Mediate Spinal Inhibition of Itch by Touch. J Neurosci 2020; 40:7688-7701. [PMID: 32895292 DOI: 10.1523/jneurosci.0091-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 07/07/2020] [Accepted: 07/20/2020] [Indexed: 11/21/2022] Open
Abstract
Innocuous mechanical stimuli, such as rubbing or stroking the skin, relieve itch through the activation of low-threshold mechanoreceptors. However, the mechanisms behind this inhibition remain unknown. We presently investigated whether stroking the skin reduces the responses of superficial dorsal horn neurons to pruritogens in male C57BL/6J mice. Single-unit recordings revealed that neuronal responses to chloroquine were enhanced during skin stroking, and this was followed by suppression of firing below baseline levels after the termination of stroking. Most of these neurons additionally responded to capsaicin. Stroking did not suppress neuronal responses to capsaicin, indicating state-dependent inhibition. Vesicular glutamate transporter 3 (VGLUT3)-lineage sensory nerves compose a subset of low-threshold mechanoreceptors. Stroking-related inhibition of neuronal responses to chloroquine was diminished by optogenetic inhibition of VGLUT3-lineage sensory nerves in male and female Vglut3-cre/NpHR-EYFP mice. Conversely, in male and female Vglut3-cre/ChR2-EYFP mice, optogenetic stimulation of VGLUT3-lineage sensory nerves inhibited firing responses of spinal neurons to pruritogens after the termination of stimulation. This inhibition was nearly abolished by spinal delivery of the κ-opioid receptor antagonist nor-binaltorphimine dihydrochloride, but not the neuropeptide Y receptor Y1 antagonist BMS193885. Optogenetic stimulation of VGLUT3-lineage sensory nerves inhibited pruritogen-evoked scratching without affecting mechanical and thermal pain behaviors. Therefore, VGLUT3-lineage sensory nerves appear to mediate inhibition of itch by tactile stimuli.SIGNIFICANCE STATEMENT Rubbing or stroking the skin is known to relieve itch. We investigated the mechanisms behind touch-evoked inhibition of itch in mice. Stroking the skin reduced the activity of itch-responsive spinal neurons. Optogenetic inhibition of VGLUT3-lineage sensory nerves diminished stroking-evoked inhibition, and optogenetic stimulation of VGLUT3-lineage nerves inhibited pruritogen-evoked firing. Together, our results provide a mechanistic understanding of touch-evoked inhibition of itch.
Collapse
|
32
|
Leiguarda C, McCarthy CJ, Casadei M, Lundgren KH, Coronel MF, Trigosso-Venario H, Seal RP, Seroogy KB, Brumovsky PR. Transcript Expression of Vesicular Glutamate Transporters in Rat Dorsal Root Ganglion and Spinal Cord Neurons: Impact of Spinal Blockade during Hindpaw Inflammation. ACS Chem Neurosci 2020; 11:2602-2614. [PMID: 32697906 DOI: 10.1021/acschemneuro.0c00272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Studies in mouse, and to a lesser extent in rat, have revealed the neuroanatomical distribution of vesicular glutamate transporters (VGLUTs) and begun exposing the critical role of VGLUT2 and VGLUT3 in pain transmission. In the present study in rat, we used specific riboprobes to characterize the transcript expression of all three VGLUTs in lumbar dorsal root ganglia (DRGs) and in the thoracolumbar, lumbar, and sacral spinal cord. We show for the first time in rat a very discrete VGLUT3 expression in DRGs and in deep layers of the dorsal horn. We confirm the abundant expression of VGLUT2, in both DRGs and the spinal cord, including presumable motorneurons in the latter. As expected, VGLUT1 was present in many DRG neuron profiles, and in the spinal cord it was mostly localized to neurons in the dorsal nucleus of Clarke. In rats with a 10 day long hindpaw inflammation, increased spinal expression of VGLUT2 transcript was detected by qRT-PCR, and intrathecal administration of the nonselective VGLUT inhibitor Chicago Sky Blue 6B resulted in reduced mechanical and thermal allodynia for up to 24 h. In conclusion, our results provide a collective characterization of VGLUTs in rat DRGs and the spinal cord, demonstrate increased spinal expression of VGLUT2 during chronic peripheral inflammation, and support the use of spinal VGLUT blockade as a strategy for attenuating inflammatory pain.
Collapse
Affiliation(s)
- Candelaria Leiguarda
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Austral, Derqui, Pilar B1629AHJ, Buenos Aires, Argentina
| | - Carly J. McCarthy
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Austral, Derqui, Pilar B1629AHJ, Buenos Aires, Argentina
| | - Mailin Casadei
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Austral, Derqui, Pilar B1629AHJ, Buenos Aires, Argentina
| | - Kerstin H. Lundgren
- Department of Neurology, University of Cincinnati, Cincinnati, Ohio 45267, United States
| | - María Florencia Coronel
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Austral, Derqui, Pilar B1629AHJ, Buenos Aires, Argentina
| | - Harry Trigosso-Venario
- Hospital Universitario Austral, Austral University, Pilar B1629AHJ, Buenos Aires, Argentina
| | - Rebecca P. Seal
- Pittsburgh Center for Pain Research, Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Kim B. Seroogy
- Department of Neurology, University of Cincinnati, Cincinnati, Ohio 45267, United States
| | - Pablo R. Brumovsky
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Austral, Derqui, Pilar B1629AHJ, Buenos Aires, Argentina
| |
Collapse
|
33
|
Harding EK, Fung SW, Bonin RP. Insights Into Spinal Dorsal Horn Circuit Function and Dysfunction Using Optical Approaches. Front Neural Circuits 2020; 14:31. [PMID: 32595458 PMCID: PMC7303281 DOI: 10.3389/fncir.2020.00031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022] Open
Abstract
Somatosensation encompasses a variety of essential modalities including touch, pressure, proprioception, temperature, pain, and itch. These peripheral sensations are crucial for all types of behaviors, ranging from social interaction to danger avoidance. Somatosensory information is transmitted from primary afferent fibers in the periphery into the central nervous system via the dorsal horn of the spinal cord. The dorsal horn functions as an intermediary processing center for this information, comprising a complex network of excitatory and inhibitory interneurons as well as projection neurons that transmit the processed somatosensory information from the spinal cord to the brain. It is now known that there can be dysfunction within this spinal cord circuitry in pathological pain conditions and that these perturbations contribute to the development and maintenance of pathological pain. However, the complex and heterogeneous network of the spinal dorsal horn has hampered efforts to further elucidate its role in somatosensory processing. Emerging optical techniques promise to illuminate the underlying organization and function of the dorsal horn and provide insights into the role of spinal cord sensory processing in shaping the behavioral response to somatosensory input that we ultimately observe. This review article will focus on recent advances in optogenetics and fluorescence imaging techniques in the spinal cord, encompassing findings from both in vivo and in vitro preparations. We will also discuss the current limitations and difficulties of employing these techniques to interrogate the spinal cord and current practices and approaches to overcome these challenges.
Collapse
Affiliation(s)
- Erika K Harding
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.,Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
| | - Samuel Wanchi Fung
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Robert P Bonin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.,University of Toronto Centre for the Study of Pain, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
34
|
Hill RZ, Bautista DM. Getting in Touch with Mechanical Pain Mechanisms. Trends Neurosci 2020; 43:311-325. [DOI: 10.1016/j.tins.2020.03.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/14/2020] [Accepted: 03/04/2020] [Indexed: 01/10/2023]
|
35
|
Neurochemical and Ultrastructural Characterization of Unmyelinated Non-peptidergic C-Nociceptors and C-Low Threshold Mechanoreceptors Projecting to Lamina II of the Mouse Spinal Cord. Cell Mol Neurobiol 2020; 41:247-262. [PMID: 32306148 DOI: 10.1007/s10571-020-00847-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
C-nociceptors (C-Ncs) and non-nociceptive C-low threshold mechanoreceptors (C-LTMRs) are two subpopulations of small unmyelinated non-peptidergic C-type neurons of the dorsal root ganglia (DRGs) with central projections displaying a specific pattern of termination in the spinal cord dorsal horn. Although these two subpopulations exist in several animals, remarkable neurochemical differences occur between mammals, particularly rat/humans from one side and mouse from the other. Mouse is widely investigated by transcriptomics. Therefore, we here studied the immunocytochemistry of murine C-type DRG neurons and their central terminals in spinal lamina II at light and electron microscopic levels. We used a panel of markers for peptidergic (CGRP), non-peptidergic (IB4), nociceptive (TRPV1), non-nociceptive (VGLUT3) C-type neurons and two strains of transgenic mice: the TAFA4Venus knock-in mouse to localize the TAFA4+ C-LTMRs, and a genetically engineered ginip mouse that allows an inducible and tissue-specific ablation of the DRG neurons expressing GINIP, a key modulator of GABABR-mediated analgesia. We confirmed that IB4 and TAFA4 did not coexist in small non-peptidergic C-type DRG neurons and separately tagged the C-Ncs and the C-LTMRs. We then showed that TRPV1 was expressed in only about 7% of the IB4+ non-peptidergic C-Ncs and their type Ia glomerular terminals within lamina II. Notably, the selective ablation of GINIP did not affect these neurons, whereas it reduced IB4 labeling in the medial part of lamina II and the density of C-LTMRs glomerular terminals to about one half throughout the entire lamina. We discuss the significance of these findings for interspecies differences and functional relevance.
Collapse
|
36
|
Qi L, Yin G, Zhang Y, Tao Y, Wu X, Gronostajski RM, Qiu M, Liu Y. Nuclear Factor I/A Controls A-fiber Nociceptor Development. Neurosci Bull 2020; 36:685-695. [PMID: 32221845 PMCID: PMC7340684 DOI: 10.1007/s12264-020-00486-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
Noxious mechanical information is transmitted through molecularly distinct nociceptors, with pinprick-evoked sharp sensitivity via A-fiber nociceptors marked by developmental expression of the neuropeptide Y receptor 2 (Npy2r) and von Frey filament-evoked punctate pressure information via unmyelinated C fiber nociceptors marked by MrgprD. However, the molecular programs controlling their development are only beginning to be understood. Here we demonstrate that Npy2r-expressing sensory neurons are in fact divided into two groups, based on transient or persistent Npy2r expression. Npy2r-transient neurons are myelinated, likely including A-fiber nociceptors, whereas Npy2r-persistent ones belong to unmyelinated pruriceptors that co-express Nppb. We then showed that the transcription factors NFIA and Runx1 are necessary for the development of Npy2r-transient A-fiber nociceptors and MrgprD+ C-fiber nociceptors, respectively. Behaviorally, mice with conditional knockout of Nfia, but not Runx1 showed a marked attenuation of pinprick-evoked nocifensive responses. Our studies therefore identify a transcription factor controlling the development of myelinated nociceptors.
Collapse
Affiliation(s)
- Lu Qi
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.,Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Guangjuan Yin
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Yongchao Zhang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Yeqi Tao
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Xiaohua Wu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Mengsheng Qiu
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China. .,Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310036, China.
| | - Yang Liu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310036, China.
| |
Collapse
|
37
|
Chen Z, Yuan M, Ma Z, Wen J, Wang X, Zhao M, Liu J, Zhang X, Zhao S, Guo L. Significance of piezo‐type mechanosensitive ion channel component 2 in premature ejaculation: An animal study. Andrology 2020; 8:1347-1359. [PMID: 32100938 DOI: 10.1111/andr.12779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 02/09/2020] [Accepted: 02/22/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Zhenghao Chen
- Institution of Urology The Second Hospital of Shandong University Jinan China
| | - Mingzhen Yuan
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China
| | - Zhen Ma
- School of Medicine Shandong University Jinan China
| | - Jiliang Wen
- School of Medicine Shandong University Jinan China
| | | | | | - Jiaxin Liu
- The First Affiliated Hospital of Zhejiang University Hangzhou China
| | - Xiulin Zhang
- Institution of Urology The Second Hospital of Shandong University Jinan China
| | - Shengtian Zhao
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China
| | - Liqiang Guo
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China
- School of Medicine Shandong University Jinan China
| |
Collapse
|
38
|
Mevel R, Draper JE, Lie-A-Ling M, Kouskoff V, Lacaud G. RUNX transcription factors: orchestrators of development. Development 2019; 146:dev148296. [PMID: 31488508 DOI: 10.1242/dev.148296] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RUNX transcription factors orchestrate many different aspects of biology, including basic cellular and developmental processes, stem cell biology and tumorigenesis. In this Primer, we introduce the molecular hallmarks of the three mammalian RUNX genes, RUNX1, RUNX2 and RUNX3, and discuss the regulation of their activities and their mechanisms of action. We then review their crucial roles in the specification and maintenance of a wide array of tissues during embryonic development and adult homeostasis.
Collapse
Affiliation(s)
- Renaud Mevel
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK
| | - Julia E Draper
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK
| | - Michael Lie-A-Ling
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK
| | - Valerie Kouskoff
- Division of Developmental Biology & Medicine, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Georges Lacaud
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield SK10 4TG, UK
| |
Collapse
|
39
|
Gong J, Liu J, Ronan EA, He F, Cai W, Fatima M, Zhang W, Lee H, Li Z, Kim GH, Pipe KP, Duan B, Liu J, Xu XZS. A Cold-Sensing Receptor Encoded by a Glutamate Receptor Gene. Cell 2019; 178:1375-1386.e11. [PMID: 31474366 DOI: 10.1016/j.cell.2019.07.034] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/31/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022]
Abstract
In search of the molecular identities of cold-sensing receptors, we carried out an unbiased genetic screen for cold-sensing mutants in C. elegans and isolated a mutant allele of glr-3 gene that encodes a kainate-type glutamate receptor. While glutamate receptors are best known to transmit chemical synaptic signals in the CNS, we show that GLR-3 senses cold in the peripheral sensory neuron ASER to trigger cold-avoidance behavior. GLR-3 transmits cold signals via G protein signaling independently of its glutamate-gated channel function, suggesting GLR-3 as a metabotropic cold receptor. The vertebrate GLR-3 homolog GluK2 from zebrafish, mouse, and human can all function as a cold receptor in heterologous systems. Mouse DRG sensory neurons express GluK2, and GluK2 knockdown in these neurons suppresses their sensitivity to cold but not cool temperatures. Our study identifies an evolutionarily conserved cold receptor, revealing that a central chemical receptor unexpectedly functions as a thermal receptor in the periphery.
Collapse
Affiliation(s)
- Jianke Gong
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, and International Research Center for Sensory Biology and Technology of MOST, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jinzhi Liu
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, and International Research Center for Sensory Biology and Technology of MOST, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth A Ronan
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Feiteng He
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, and International Research Center for Sensory Biology and Technology of MOST, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wei Cai
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mahar Fatima
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wenyuan Zhang
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, and International Research Center for Sensory Biology and Technology of MOST, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hankyu Lee
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhaoyu Li
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gun-Ho Kim
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, 50 UNIST-gil, South Korea
| | - Kevin P Pipe
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bo Duan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianfeng Liu
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, and International Research Center for Sensory Biology and Technology of MOST, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.
| | - X Z Shawn Xu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
40
|
Gu JG. Molecular Mechanisms of the Sense of Touch: An Overview of Mechanical Transduction and Transmission in Merkel Discs of Whisker Hair Follicles and Some Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1099:1-12. [PMID: 30306510 DOI: 10.1007/978-981-13-1756-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The Merkel disc is a main type of tactile end organs for sensing gentle touch and is essential for sophisticated sensory tasks including social interaction, environmental exploration, and tactile discrimination. Recent studies have shown that Merkel cells are primary sites of mechanotransduction using Piezo2 channels as a molecular transducer in Merkel discs. Furthermore, tactile stimuli trigger serotonin release from Merkel cells to excite their associated whisker Aβ-afferent endings and transmit tactile signals. The tactile transduction and transmission at Merkel discs may have important clinical implications in sensory dysfunctions such as the loss of tactile sensitivity and tactile allodynia seen in patients who have diabetes and inflammatory diseases and undergo chemotherapy.
Collapse
Affiliation(s)
- Jianguo G Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
41
|
Identification of a Spinal Circuit for Mechanical and Persistent Spontaneous Itch. Neuron 2019; 103:1135-1149.e6. [PMID: 31324538 DOI: 10.1016/j.neuron.2019.06.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/17/2019] [Accepted: 06/20/2019] [Indexed: 12/12/2022]
Abstract
Lightly stroking the lips or gently poking some skin regions can evoke mechanical itch in healthy human subjects. Sensitization of mechanical itch and persistent spontaneous itch are intractable symptoms in chronic itch patients. However, the underlying neural circuits are not well defined. We identified a subpopulation of excitatory interneurons expressing Urocortin 3::Cre (Ucn3+) in the dorsal spinal cord as a central node in the pathway that transmits acute mechanical itch and mechanical itch sensitization as well as persistent spontaneous itch under chronic itch conditions. This population receives peripheral inputs from Toll-like receptor 5-positive (TLR5+) Aβ low-threshold mechanoreceptors and is directly innervated by inhibitory interneurons expressing neuropeptide Y::Cre (NPY+) in the dorsal spinal cord. Reduced synaptic inhibition and increased intrinsic excitability of Ucn3+ neurons lead to chronic itch sensitization. Our study sheds new light on the neural basis of chronic itch and unveils novel avenues for developing mechanism-specific therapeutic advancements.
Collapse
|
42
|
Tetrodotoxin-Sensitive Sodium Channels Mediate Action Potential Firing and Excitability in Menthol-Sensitive Vglut3-Lineage Sensory Neurons. J Neurosci 2019; 39:7086-7101. [PMID: 31300524 DOI: 10.1523/jneurosci.2817-18.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 06/04/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022] Open
Abstract
Small-diameter vesicular glutamate transporter 3-lineage (Vglut3lineage) dorsal root ganglion (DRG) neurons play an important role in mechanosensation and thermal hypersensitivity; however, little is known about their intrinsic electrical properties. We therefore set out to investigate mechanisms of excitability within this population. Calcium microfluorimetry analysis of male and female mouse DRG neurons demonstrated that the cooling compound menthol selectively activates a subset of Vglut3lineage neurons. Whole-cell recordings showed that small-diameter Vglut3lineage DRG neurons fire menthol-evoked action potentials and exhibited robust, transient receptor potential melastatin 8 (TRPM8)-dependent discharges at room temperature. This heightened excitability was confirmed by current-clamp and action potential phase-plot analyses, which showed menthol-sensitive Vglut3lineage neurons to have more depolarized membrane potentials, lower firing thresholds, and higher evoked firing frequencies compared with menthol-insensitive Vglut3lineage neurons. A biophysical analysis revealed voltage-gated sodium channel (NaV) currents in menthol-sensitive Vglut3lineage neurons were resistant to entry into slow inactivation compared with menthol-insensitive neurons. Multiplex in situ hybridization showed similar distributions of tetrodotoxin (TTX)-sensitive NaV transcripts between TRPM8-positive and -negative Vglut3lineage neurons; however, NaV1.8 transcripts, which encode TTX-resistant channels, were more prevalent in TRPM8-negative neurons. Conversely, pharmacological analyses identified distinct functional contributions of NaV subunits, with NaV1.1 driving firing in menthol-sensitive neurons, whereas other small-diameter Vglut3lineage neurons rely primarily on TTX-resistant NaV channels. Additionally, when NaV1.1 channels were blocked, the remaining NaV current readily entered into slow inactivation in menthol-sensitive Vglut3lineage neurons. Thus, these data demonstrate that TTX-sensitive NaVs drive action potential firing in menthol-sensitive sensory neurons and contribute to their heightened excitability.SIGNIFICANCE STATEMENT Somatosensory neurons encode various sensory modalities including thermoreception, mechanoreception, nociception, and itch. This report identifies a previously unknown requirement for tetrodotoxin-sensitive sodium channels in action potential firing in a discrete subpopulation of small-diameter sensory neurons that are activated by the cooling agent menthol. Together, our results provide a mechanistic understanding of factors that control intrinsic excitability in functionally distinct subsets of peripheral neurons. Furthermore, as menthol has been used for centuries as an analgesic and anti-pruritic, these findings support the viability of NaV1.1 as a therapeutic target for sensory disorders.
Collapse
|
43
|
Noble DJ, Martin KK, Parvin S, Garraway SM. Spontaneous and Stimulus-Evoked Respiratory Rate Elevation Corresponds to Development of Allodynia in Spinal Cord-Injured Rats. J Neurotrauma 2019; 36:1909-1922. [PMID: 30489202 DOI: 10.1089/neu.2018.5936] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Respiratory complications frequently accompany spinal cord injury (SCI) and slowed breathing has been shown to mitigate pain sensitivity. It is possible that elevated respiratory rates (RRs) signal the emergence of chronic pain after SCI. We previously validated the use of remote electric field sensors to noninvasively track breathing in freely behaving rodents. Here, we examined spontaneous (resting) and stimulus-evoked RRs as potential indices of mechanical hypersensitivity following SCI. Adult male Long-Evans rats received a lower thoracic hemisection or contusion SCI, or sham surgery, and underwent weekly assessments of mechanical and thermal sensitivity using the von Frey and Hargreaves tests, respectively. Resting RRs were recorded with remote sensors prior to nociception assays as well as 1 day post-surgery. Evoked RRs were quantified weekly in response to at-level mechanical stimulation provided by a small brush at various stimulation speeds, including those corresponding to the distinct tuning properties of a sub-population of cutaneous afferents known as C-low threshold mechanoreceptors. SCI rats developed mechanical hypersensitivity, which peaked 2-3 weeks after SCI. Compared with at baseline, hemisection SCI rats showed significantly heightened resting RRs at 1 day and 7 days post-injury, and the latter predicted development of pain hypersensitivity. In contusion SCI rats, resting RR increases were less substantial but occurred at all weekly time-points. Increases in brush-evoked RR coincided with full expression of hypersensitivity at 14 (hemisection) or 21 (contusion) days after SCI, and these effects were restricted to the lowest brush speeds. Our results support the possibility that early changes in RR may convey pain information in rats.
Collapse
Affiliation(s)
- Donald J Noble
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Karmarcha K Martin
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Shangrila Parvin
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
44
|
Synaptic Organization of VGLUT3 Expressing Low-Threshold Mechanosensitive C Fiber Terminals in the Rodent Spinal Cord. eNeuro 2019; 6:eN-NWR-0007-19. [PMID: 30783617 PMCID: PMC6378328 DOI: 10.1523/eneuro.0007-19.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 11/21/2022] Open
Abstract
Low-threshold mechanosensitive C fibers (C-LTMRs) that express the vesicular glutamate transporter VGLUT3 are thought to signal affective touch, and may also play a role in mechanical allodynia. However, the nature of the central termination of C-LTMRs in the dorsal horn remains largely unexplored. Here, we used light and electron microscopy in combination with VGLUT3 immunolabeling as a marker of C-LTMR terminations to investigate this issue. VGLUT3+ C-LTMRs formed central terminals of Type II glomeruli in the inner part of lamina II of the dorsal horn, often establishing multiple asymmetric synapses with postsynaptic dendrites but also participating in synaptic configurations with presynaptic axons and dendrites. Unexpectedly, essentially all VGLUT3+ C-LTMR terminals showed substantial VGLUT1 expression in the rat, whereas such terminals in mice lacked VGLUT1. Most VGLUT3+ C-LTMR terminals exhibited weak-to-moderate VGLUT2 expression. Further, C-LTMR terminals formed numerous synapses with excitatory protein kinase Cγ (PKCγ) interneurons and inhibitory parvalbumin neurons, whereas synapses with calretinin neurons were scarce. C-LTMR terminals rarely if ever established synapses with neurokinin 1 receptor (NK1R)-possessing dendrites traversing lamina II. Thus, VGLUT3+ C-LTMR terminals appear to largely correspond to neurofilament-lacking central terminals of Type II glomeruli in inner lamina II and can thus be identified at the ultrastructural level by morphological criteria. The participation of C-LTMR terminals in Type II glomeruli involving diverse populations of interneuron indicates highly complex modes of integration of C-LTMR mediated signaling in the dorsal horn. Furthermore, differences in VGLUT1 expression indicate distinct species differences in synaptic physiology of C-LTMR terminals.
Collapse
|
45
|
Huang T, Lin SH, Malewicz NM, Zhang Y, Zhang Y, Goulding M, LaMotte RH, Ma Q. Identifying the pathways required for coping behaviours associated with sustained pain. Nature 2018; 565:86-90. [PMID: 30532001 PMCID: PMC6461409 DOI: 10.1038/s41586-018-0793-8] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 11/05/2018] [Indexed: 01/17/2023]
Abstract
Animals and humans display two types of responses to noxious stimuli. The first includes reflexive-defensive responses to prevent or limit injury. A well-known example is the quick withdrawal of one’s hand touching a hot object. When the first-line response fails to prevent tissue damage (e.g., a finger is burnt), the resulting pain invokes a second-line coping response, such as licking the injured area to soothe suffering. However, the underlying neural circuits driving these two strings of behaviors remain poorly understood. Here we show that in mice, spinal neurons marked by coexpression of TુCre and Lbx1Flpo, called Tac1Lbx1, drive pain-related coping responses. Ablation of Tac1Lbx1 neurons led to loss of persistent licking and conditioned aversion evoked by stimuli that produce sustained pain in humans, including skin pinching and burn injury, without affecting all tested reflexive-defensive reactions. This selective indifference to sustained pain resembles the phenotype seen in humans with lesions of medial thalamic nuclei1–3. Consistently, spinal Tac1 lineage neurons are connected to medial thalamic nuclei, via direct projections and indirect routes through the superior lateral parabrachial nuclei. Furthermore, the anatomical and functional segregation observed at the spinal levels is applied to primary sensory neurons. For example, in response to noxious mechanical stimuli, Mrgprd+ and TRPV1+ nociceptors are required to elicit reflexive and coping responses, respectively. Our studies therefore reveal a fundamental subdivision within the cutaneous somatosensory system. The implications for translational success from preclinical pain studies will be discussed.
Collapse
Affiliation(s)
- Tianwen Huang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Shing-Hong Lin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Nathalie M Malewicz
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yan Zhang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine; Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China.,Cell Electrophysiology Laboratory, Wannan Medical College, Wuhu, China
| | - Ying Zhang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Martyn Goulding
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Robert H LaMotte
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Qiufu Ma
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA. .,Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Demethylation of G-Protein-Coupled Receptor 151 Promoter Facilitates the Binding of Krüppel-Like Factor 5 and Enhances Neuropathic Pain after Nerve Injury in Mice. J Neurosci 2018; 38:10535-10551. [PMID: 30373770 DOI: 10.1523/jneurosci.0702-18.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 09/13/2018] [Accepted: 10/22/2018] [Indexed: 01/27/2023] Open
Abstract
G-protein-coupled receptors are considered to be cell-surface sensors of extracellular signals, thereby having a crucial role in signal transduction and being the most fruitful targets for drug discovery. G-protein-coupled receptor 151 (GPR151) was reported to be expressed specifically in the habenular area. Here we report the expression and the epigenetic regulation of GRP151 in the spinal cord after spinal nerve ligation (SNL) and the contribution of GPR151 to neuropathic pain in male mice. SNL dramatically increased GPR151 expression in spinal neurons. GPR151 mutation or spinal inhibition by shRNA alleviated SNL-induced mechanical allodynia and heat hyperalgesia. Interestingly, the CpG island in the GPR151 gene promoter region was demethylated, the expression of DNA methyltransferase 3b (DNMT3b) was decreased, and the binding of DNMT3b with GPR151 promoter was reduced after SNL. Overexpression of DNMT3b in the spinal cord decreased GPR151 expression and attenuated SNL-induced neuropathic pain. Furthermore, Krüppel-like factor 5 (KLF5), a transcriptional factor of the KLF family, was upregulated in spinal neurons, and the binding affinity of KLF5 with GPR151 promoter was increased after SNL. Inhibition of KLF5 reduced GPR151 expression and attenuated SNL-induced pain hypersensitivity. Further mRNA microarray analysis revealed that mutation of GPR151 reduced the expression of a variety of pain-related genes in response to SNL, especially mitogen-activated protein kinase (MAPK) signaling pathway-associated genes. This study reveals that GPR151, increased by DNA demethylation and the enhanced interaction with KLF5, contributes to the maintenance of neuropathic pain via increasing MAPK pathway-related gene expression.SIGNIFICANCE STATEMENT G-protein-coupled receptors (GPCRs) are targets of various clinically approved drugs. Here we report that SNL increased GPR151 expression in the spinal cord, and mutation or inhibition of GPR151 alleviated SNL-induced neuropathic pain. In addition, SNL downregulated the expression of DNMT3b, which caused demethylation of GPR151 gene promoter, facilitated the binding of transcriptional factor KLF5 with the GPR151 promoter, and further increased GPR151 expression in spinal neurons. The increased GPR151 may contribute to the pathogenesis of neuropathic pain via activating MAPK signaling and increasing pain-related gene expression. Our study reveals an epigenetic mechanism underlying GPR151 expression and suggests that targeting GPR151 may offer a new strategy for the treatment of neuropathic pain.
Collapse
|
47
|
Zhang FX, Ge SN, Dong YL, Shi J, Feng YP, Li Y, Li YQ, Li JL. Vesicular glutamate transporter isoforms: The essential players in the somatosensory systems. Prog Neurobiol 2018; 171:72-89. [PMID: 30273635 DOI: 10.1016/j.pneurobio.2018.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/28/2018] [Accepted: 09/23/2018] [Indexed: 02/08/2023]
Abstract
In nervous system, glutamate transmission is crucial for centripetal conveyance and cortical perception of sensory signals of different modalities, which necessitates vesicular glutamate transporters 1-3 (VGLUT 1-3), the three homologous membrane-bound protein isoforms, to load glutamate into the presysnaptic vesicles. These VGLUTs, especially VGLUT1 and VGLUT2, selectively label and define functionally distinct neuronal subpopulations at each relay level of the neural hierarchies comprising spinal and trigeminal sensory systems. In this review, by scrutinizing each structure of the organism's fundamental hierarchies including dorsal root/trigeminal ganglia, spinal dorsal horn/trigeminal sensory nuclear complex, somatosensory thalamic nuclei and primary somatosensory cortex, we summarize and characterize in detail within each relay the neuronal clusters expressing distinct VGLUT protein/transcript isoforms, with respect to their regional distribution features (complementary distribution in some structures), axonal terminations/peripheral innervations and physiological functions. Equally important, the distribution pattern and characteristics of VGLUT1/VGLUT2 axon terminals within these structures are also epitomized. Finally, the correlation of a particular VGLUT isoform and its physiological role, disclosed thus far largely via studying the peripheral receptors, is generalized by referring to reports on global and conditioned VGLUT-knockout mice. Also, researches on VGLUTs relating to future direction are tentatively proposed, such as unveiling the elusive differences between distinct VGLUTs in mechanism and/or pharmacokinetics at ionic/molecular level, and developing VGLUT-based pain killers.
Collapse
Affiliation(s)
- Fu-Xing Zhang
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Shun-Nan Ge
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China; Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China
| | - Yu-Lin Dong
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Juan Shi
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Yu-Peng Feng
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Yang Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China
| | - Yun-Qing Li
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, PR China.
| | - Jin-Lian Li
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
48
|
Ackerley R, Wiklund Fernström K, Backlund Wasling H, Watkins RH, Johnson RD, Vallbo Å, Wessberg J. Differential effects of radiant and mechanically applied thermal stimuli on human C-tactile afferent firing patterns. J Neurophysiol 2018; 120:1885-1892. [PMID: 30044679 DOI: 10.1152/jn.00940.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
C-tactile (CT) afferents respond to gentle tactile stimulation, but only a handful of studies in humans and animals have investigated whether their firing is modified by temperature. We describe the effects of radiant thermal stimuli, and of stationary and very slowly moving mechanothermal stimuli, on CT afferent responses. We find that CT afferents are primarily mechanoreceptors, as they fired little during radiant thermal stimuli, but they exhibited different patterns of firing during combined mechano-cool stimulation compared with warming. CTs fired optimally to gentle, very slowly moving, or stationary mechanothermal stimuli delivered at neutral temperature (~32°C, normal skin temperature), but they responded with fewer spikes (median 67% decrease) and at significantly lower rates (47% decrease) during warm (~42°C) tactile stimuli. During cool tactile stimuli (~18°C), their mean instantaneous firing frequency significantly decreased by 35%, but they often fired a barrage of afterdischarge spikes at a low frequency (~5 Hz) that outlasted the mechanical stimulus. These effects were observed under a variety of stimulus conditions, including during stationary and slowly moving touch (0.1 cm/s), and we complemented these tactile approaches using a combined electrical-thermal stimulation experiment where we found a suppression of spiking during warming. Overall, CT afferents are exquisitely sensitive to tactile events, and we show that their firing is modulated with touch temperatures above and below neutral skin temperature. Warm touch consistently decreased their propensity to fire, whereas cool touch produced lower firing rates but afterdischarge spiking. NEW & NOTEWORTHY C-tactile (CT) afferents are thought to underpin pleasant touch, and previous work has shown that they respond optimally to a slow caress delivered at typical (neutral) skin temperature. Here, we show that, although CTs are primarily mechanoreceptive afferents, they are modified by temperature: warm touch decreases their firing, whereas cool touch produces lower firing rates but long-lasting spiking, frequently seen as afterdischarges. This has implications for the encoding of affective sensory events in human skin.
Collapse
Affiliation(s)
- Rochelle Ackerley
- Aix Marseille Université, CNRS, LNSC (Laboratoire de Neurosciences Sensorielles et Cognitives - UMR 7260), Marseille , France.,Department of Physiology, University of Gothenburg , Gothenburg , Sweden
| | | | | | - Roger H Watkins
- Department of Physiology, University of Gothenburg , Gothenburg , Sweden
| | - Richard D Johnson
- Department of Physiology, University of Gothenburg , Gothenburg , Sweden.,Department of Physiological Sciences, University of Florida College of Veterinary Medicine , Gainesville, Florida
| | - Åke Vallbo
- Department of Physiology, University of Gothenburg , Gothenburg , Sweden
| | - Johan Wessberg
- Department of Physiology, University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
49
|
Anatomical and functional dichotomy of ocular itch and pain. Nat Med 2018; 24:1268-1276. [PMID: 29988128 PMCID: PMC6093777 DOI: 10.1038/s41591-018-0083-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 05/02/2018] [Indexed: 12/16/2022]
Abstract
Itch and pain are refractory symptoms of many ocular conditions. Ocular itch is generated mainly in the conjunctiva, and is absent from the cornea. In contrast, most ocular pain arises from the cornea. However, the underlying mechanisms remain unknown. Using genetic axonal tracing approaches, we discovered distinct sensory innervation patterns between the conjunctiva and cornea. Further genetic and functional analyses in rodent models demonstrate that a subset of conjunctival-selective sensory fibers marked by MrgprA3 expression, rather than corneal sensory fibers, mediates ocular itch. Importantly, the actions of both histamine and non-histamine pruritogens converge onto this unique subset of conjunctiva sensory fibers, and enable them to play a key role in mediating itch associated with allergic conjunctivitis. This is distinct from skin itch in which discrete populations of sensory neurons co-operate to carry itch. Finally, we provide a proof-of-concept that selective silencing of conjunctiva itch-sensing fibers by pruritogen-mediated entry of sodium channel blocker QX-314 is a feasible therapeutic strategy to treat ocular itch in mice. Itch-sensing fibers also innervate the human conjunctiva,and allow pharmacological silence using QX-314.Our results cast new light on the neural mechanisms of ocular itch and open a new avenue for developing therapeutic strategies.
Collapse
|
50
|
Patil MJ, Hovhannisyan AH, Akopian AN. Characteristics of sensory neuronal groups in CGRP-cre-ER reporter mice: Comparison to Nav1.8-cre, TRPV1-cre and TRPV1-GFP mouse lines. PLoS One 2018; 13:e0198601. [PMID: 29864146 PMCID: PMC5986144 DOI: 10.1371/journal.pone.0198601] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/22/2018] [Indexed: 12/19/2022] Open
Abstract
Peptidergic sensory neurons play a critical role in nociceptive pathways. To precisely define the function and plasticity of sensory neurons in detail, new tools such as transgenic mouse models are needed. We employed electrophysiology and immunohistochemistry to characterize in detail dorsal root ganglion (DRG) neurons expressing an inducible CGRPcre-ER (CGRP-cre+); and compared them to DRG neurons expressing Nav1.8cre (Nav1.8-cre+), TRPV1cre (TRPV1-cre+) and TRPV1-GFP (V1-GFP+). Tamoxifen effectively induced CGRPcre-ER production in DRG. ≈87% of CGRPcre-ER-expressing neurons were co-labeled CGRP antibody. Three small and two medium-large-sized (5HT3a+/NPY2R- and NPY2R+) neuronal groups with unique electrophysiological profiles were CGRP-cre+. Nav1.8-cre+ neurons were detected in all CGRP-cre+ groups, as well as in 5 additional neuronal groups: MrgprD+/TRPA1-, MrgprD+/TRPA1+, TRPV1+/CGRP-, vGLUT3+ and ≈30% of trkC+ neurons. Differences between TRPV1cre and Nav1.8cre reporters were that unlike TRPV1-cre+, Nav1.8-cre+ expression was detected in non-nociceptive vGLUT3+ and trkC+ populations. Many TRPV1-cre+ neurons did not respond to capsaicin. In contrast, V1-GFP+ neurons were in 4 groups, each of which was capsaicin-sensitive. Finally, none of the analyzed reporter lines showed cre-recombination in trkB+, calbindin+, 70% of trkC+ or parvalbumin+ neurons, which together encompassed ≈20% of Nav1.8-cre- DRG neurons. The data presented here increases our knowledge of peptidergic sensory neuron characteristics, while showing the efficiency and specificity manipulation of peptidergic neurons by the CGRPcre-ER reporter. We also demonstrate that manipulation of all C- and A-nociceptors is better achieved with TRPV1-cre reporter. Finally, the described approach for detailed characterization of sensory neuronal groups can be applied to a variety of reporter mice.
Collapse
Affiliation(s)
- Mayur J. Patil
- Departments of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Anahit H. Hovhannisyan
- Departments of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Armen N. Akopian
- Departments of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Departments of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|