51
|
Shekhtmeyster P, Carey EM, Duarte D, Ngo A, Gao G, Nelson NA, Clark CL, Nimmerjahn A. Multiplex translaminar imaging in the spinal cord of behaving mice. Nat Commun 2023; 14:1427. [PMID: 36944637 PMCID: PMC10030868 DOI: 10.1038/s41467-023-36959-2] [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: 12/23/2021] [Accepted: 02/24/2023] [Indexed: 03/23/2023] Open
Abstract
While the spinal cord is known to play critical roles in sensorimotor processing, including pain-related signaling, corresponding activity patterns in genetically defined cell types across spinal laminae have remained challenging to investigate. Calcium imaging has enabled cellular activity measurements in behaving rodents but is currently limited to superficial regions. Here, using chronically implanted microprisms, we imaged sensory and motor-evoked activity in regions and at speeds inaccessible by other high-resolution imaging techniques. To enable translaminar imaging in freely behaving animals through implanted microprisms, we additionally developed wearable microscopes with custom-compound microlenses. This system addresses multiple challenges of previous wearable microscopes, including their limited working distance, resolution, contrast, and achromatic range. Using this system, we show that dorsal horn astrocytes in behaving mice show sensorimotor program-dependent and lamina-specific calcium excitation. Additionally, we show that tachykinin precursor 1 (Tac1)-expressing neurons exhibit translaminar activity to acute mechanical pain but not locomotion.
Collapse
Affiliation(s)
- Pavel Shekhtmeyster
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Electrical and Computer Engineering Graduate Program, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Erin M Carey
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Daniela Duarte
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Alexander Ngo
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Grace Gao
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Nicholas A Nelson
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Charles L Clark
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
52
|
Qi L, Lin SH, Ma Q. Spinal VGLUT3 lineage neurons drive visceral mechanical allodynia but not sensitized visceromotor reflexes. Neuron 2023; 111:669-681.e5. [PMID: 36584681 DOI: 10.1016/j.neuron.2022.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 09/08/2022] [Accepted: 11/30/2022] [Indexed: 12/30/2022]
Abstract
Visceral pain is among the most prevalent and bothersome forms of chronic pain, but their transmission in the spinal cord is still poorly understood. Here, we conducted focal colorectal distention (fCRD) to drive both visceromotor responses (VMRs) and aversion. We first found that spinal CCK neurons were necessary for noxious fCRD to drive both VMRs and aversion under naive conditions. We next showed that spinal VGLUT3 neurons mediate visceral allodynia, whose ablation caused loss of aversion evoked by low-intensity fCRD in mice with gastrointestinal (GI) inflammation or spinal circuit disinhibition. Importantly, these neurons were dispensable for driving sensitized VMRs under both inflammatory and central disinhibition conditions. Anatomically, a subset of VGLUT3 neurons projected to parabrachial nuclei, whose photoactivation sufficiently generated aversion in mice with GI inflammation, without influencing VMRs. Our studies suggest the presence of different spinal substrates that transmit nociceptive versus affective dimensions of visceral sensory information.
Collapse
Affiliation(s)
- Lu Qi
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shing-Hong Lin
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Qiufu Ma
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
53
|
Time-dependent and selective microglia-mediated removal of spinal synapses in neuropathic pain. Cell Rep 2023; 42:112010. [PMID: 36656715 DOI: 10.1016/j.celrep.2023.112010] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 09/20/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Neuropathic pain is a debilitating condition resulting from damage to the nervous system. Imbalance of spinal excitation and inhibition has been proposed to contribute to neuropathic pain. However, the structural basis of this imbalance remains unknown. Using a preclinical model of neuropathic pain, we show that microglia selectively engulf spinal synapses that are formed by central neurons and spare those of peripheral sensory neurons. Furthermore, we reveal that removal of inhibitory and excitatory synapses exhibits distinct temporal patterns, in which microglia-mediated inhibitory synapse removal precedes excitatory synapse removal. We also find selective and gradual increase in complement depositions on dorsal horn synapses that corresponds to the temporal pattern of microglial synapse pruning activity and type-specific synapse loss. Together, these results define a specific role for microglia in the progression of neuropathic pain pathogenesis and implicate these immune cells in structural remodeling of dorsal horn circuitry.
Collapse
|
54
|
Chopra D, Stern E, Bushell WC, Castle RD. Yoga and pain: A mind-body complex system. FRONTIERS IN PAIN RESEARCH 2023; 4:1075866. [PMID: 36910253 PMCID: PMC9996306 DOI: 10.3389/fpain.2023.1075866] [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: 10/21/2022] [Accepted: 01/05/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction The human body's response to pain is indicative of a complex adaptive system. Therapeutic yoga potentially represents a similar complex adaptive system that could interact with the pain response system with unique benefits. Objectives To determine the viability of yoga as a therapy for pain and whether pain responses and/or yoga practice should be considered complex adaptive systems. Methods Examination through 3 different approaches, including a narrative overview of the evidence on pain responses, yoga, and complex system, followed by a network analysis of associated keywords, followed by a mapping of the functional components of complex systems, pain response, and yoga. Results The narrative overview provided extensive evidence of the unique efficacy of yoga as a pain therapy, as well as articulating the relevance of applying complex systems perspectives to pain and yoga interventions. The network analysis demonstrated patterns connecting pain and yoga, while complex systems topics were the most extensively connected to the studies as a whole. Conclusion All three approaches support considering yoga a complex adaptive system that exhibits unique benefits as a pain management system. These findings have implications for treating chronic, pervasive pain with behavioral medicine as a systemic intervention. Approaching yoga as complex system suggests the need for research of mind-body topics that focuses on long-term systemic changes rather than short-term isolated effects.
Collapse
Affiliation(s)
| | - Eddie Stern
- Vivekananda Yoga University, Los Angeles, CA, United States
| | | | - Ryan D Castle
- Chopra Foundation Institute, Honolulu, HI, United States
| |
Collapse
|
55
|
Yang HY, Wu J, Lu H, Cheng ML, Wang BH, Zhu HL, Liu L, Xie M. Emodin suppresses oxaliplatin-induced neuropathic pain by inhibiting COX2/NF-κB mediated spinal inflammation. J Biochem Mol Toxicol 2023; 37:e23229. [PMID: 36184831 DOI: 10.1002/jbt.23229] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 07/25/2022] [Accepted: 09/16/2022] [Indexed: 01/15/2023]
Abstract
Oxaliplatin (OXA) is a common chemotherapy drug for colorectal, gastric, and pancreatic cancers. The anticancer effect of OXA is often accompanied by neurotoxicity and acute and chronic neuropathy. The symptoms present as paresthesia and pain which adversely affect patients' quality of life. Herein, five consecutive intraperitoneal injections of OXA at a dose of 4 mg/kg were used to mimic chemotherapy. OXA administration induced mechanical allodynia, activated spinal astrocytes, and increased inflammatory response. To develop an effective therapeutic measure for OXA-induced neuropathic pain, emodin was intrathecally injected into OXA rats. Emodin developed an analgesic effect, as demonstrated by a significant increase in the paw withdrawal threshold of OXA rats. Moreover, emodin treatment reduced the pro-inflammatory cytokines (tumor necrosis factor-α and interleukin-1β) which upregulated in OXA rats. Furthermore, autodock data showed four hydrogen bonds were formed between emodin and cyclooxygenase-2 (COX2), and emodin treatment decreased COX2 expression in OXA rats. Cell research further proved that emodin suppressed nuclear factor κB (NF-κB)-mediated inflammatory signal and reactive oxygen species level. Taken together, emodin reduced spinal COX2/NF-κB mediated inflammatory signal and oxidative stress in the spinal cord of OXA rats which consequently relieved OXA-induced neuropathic pain.
Collapse
Affiliation(s)
- He-Yu Yang
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Ji Wu
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Hong Lu
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China
| | - Meng-Lin Cheng
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China
| | - Bang-Hua Wang
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China
| | - Hai-Li Zhu
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China
| | - Ling Liu
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China
| | - Min Xie
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
56
|
Macionis V. Chronic pain and local pain in usually painless conditions including neuroma may be due to compressive proximal neural lesion. FRONTIERS IN PAIN RESEARCH 2023; 4:1037376. [PMID: 36890855 PMCID: PMC9986610 DOI: 10.3389/fpain.2023.1037376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/12/2023] [Indexed: 02/22/2023] Open
Abstract
It has been unexplained why chronic pain does not invariably accompany chronic pain-prone disorders. This question-driven, hypothesis-based article suggests that the reason may be varying occurrence of concomitant peripheral compressive proximal neural lesion (cPNL), e.g., radiculopathy and entrapment plexopathies. Transition of acute to chronic pain may involve development or aggravation of cPNL. Nociceptive hypersensitivity induced and/or maintained by cPNL may be responsible for all types of general chronic pain as well as for pain in isolated tissue conditions that are usually painless, e.g., neuroma, scar, and Dupuytren's fibromatosis. Compressive PNL induces focal neuroinflammation, which can maintain dorsal root ganglion neuron (DRGn) hyperexcitability (i.e., peripheral sensitization) and thus fuel central sensitization (i.e., hyperexcitability of central nociceptive pathways) and a vicious cycle of chronic pain. DRGn hyperexcitability and cPNL may reciprocally maintain each other, because cPNL can result from reflexive myospasm-induced myofascial tension, muscle weakness, and consequent muscle imbalance- and/or pain-provoked compensatory overuse. Because of pain and motor fiber damage, cPNL can worsen the causative musculoskeletal dysfunction, which further accounts for the reciprocity between the latter two factors. Sensitization increases nerve vulnerability and thus catalyzes this cycle. Because of these mechanisms and relatively greater number of neurons involved, cPNL is more likely to maintain DRGn hyperexcitability in comparison to distal neural and non-neural lesions. Compressive PNL is associated with restricted neural mobility. Intermittent (dynamic) nature of cPNL may be essential in chronic pain, because healed (i.e., fibrotic) lesions are physiologically silent and, consequently, cannot provide nociceptive input. Not all patients may be equally susceptible to develop cPNL, because occurrence of cPNL may vary as vary patients' predisposition to musculoskeletal impairment. Sensitization is accompanied by pressure pain threshold decrease and consequent mechanical allodynia and hyperalgesia, which can cause unusual local pain via natural pressure exerted by space occupying lesions or by their examination. Worsening of local pain is similarly explainable. Neuroma pain may be due to cPNL-induced axonal mechanical sensitivity and hypersensitivity of the nociceptive nervi nervorum of the nerve trunk and its stump. Intermittence and symptomatic complexity of cPNL may be the cause of frequent misdiagnosis of chronic pain.
Collapse
|
57
|
Liang Y, Fan Z, Li J, Ma R, Zhang Y, Shi X, Zhu Y, Huang J. GABAergic neurons in the ventral lateral geniculate nucleus and intergeniculate leaflet modulate itch processing in mice. Biochem Biophys Res Commun 2023; 659:72-79. [PMID: 37054505 DOI: 10.1016/j.bbrc.2023.01.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Itch and pain are two closely related sensations that receiving similar encodings at multiple levels. Accumulated evidences suggest that activation of the ventral lateral geniculate nucleus and intergeniculate leaflet (vLGN/IGL)-to-lateral and ventrolateral periaqueductal gray (l/vlPAG) projections mediates the antinociceptive effects of bright light therapy. Clinical study showed that bright light therapy may ameliorate cholestasis-induced pruritus. However, the underlying mechanism and whether this circuit participates in itch modulation remains unclear. In this study, chloroquine and histamine were utilized to induce acute itch models in mice. Neuronal activities in vLGN/IGL nucleus were evaluated with c-fos immunostaining as well as fiber photometry. Optogenetic manipulations were performed to activate or inhibit GABAergic neurons in the vLGN/IGL nucleus. Our results showed that the expressions of c-fos in vLGN/IGL were significantly increased upon both chloroquine- and histamine-induced acute itch stimuli. GABAergic neurons in vLGN/IGL were activated during histamine and chloroquine-induced scratching. Optogenetic activation of the vLGN/IGL GABAergic neurons exerts antipruritic effect, while inhibiting these neurons exerts pruritic effect. Our results provide evidence that GABAergic neurons in vLGN/IGL nucleus might play a crucial role in modulating itch, which may provide clue for application of bright light as an antipruritic treatment in clinic.
Collapse
|
58
|
Shin SM, Itson-Zoske B, Fan F, Gani U, Rahman M, Hogan QH, Yu H. Peripheral sensory neurons and non-neuronal cells express functional Piezo1 channels. Mol Pain 2023; 19:17448069231174315. [PMID: 37247618 PMCID: PMC10240879 DOI: 10.1177/17448069231174315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/31/2023] Open
Abstract
Here, we present evidence showing Piezo1 protein expression in the primary sensory neurons (PSNs) and non-neuronal cells of rat peripheral nervous system. Using a knockdown/knockout validated antibody, we detected Piezo1 immunoreactivity (IR) in ∼60% of PSNs of rat dorsal root ganglia (DRG) with higher IR density in the small- and medium-sized neurons. Piezo1-IR was clearly identified in DRG perineuronal glia, including satellite glial cells (SGCs) and Schwann cells; in sciatic nerve Schwann cells surrounding the axons and cutaneous afferent endings; and in skin epidermal Merkel cells and melanocytes. Neuronal and non-neuronal Piezo1 channels were functional since various cells (dissociated PSNs and SGCs from DRGs, isolated Schwann cells, and primary human melanocytes) exhibited a robust response to Piezo1 agonist Yoda1 by an increase of intracellular Ca2+ concentration ([Ca2+]i). These responses were abolished by non-specific Piezo1 antagonist GsMTx4. Immunoblots showed elevated Piezo1 protein in DRG proximal to peripheral nerve injury-induced painful neuropathy, while PSNs and SGCs from rats with neuropathic pain showed greater Yoda1-evoked elevation of [Ca2+]i and an increased frequency of cells responding to Yoda1, compared to controls. Sciatic nerve application of GsMTx4 alleviated mechanical hypersensitivity induced by Yoda1. Overall, our data show that Piezo1 is widely expressed by the neuronal and non-neuronal cells in the peripheral sensory pathways and that painful nerve injury appeared associated with activation of Piezo1 in PSNs and peripheral glial cells.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fan Fan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Uarda Gani
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mahmudur Rahman
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
59
|
Cao B, Scherrer G, Chen L. Spinal cord retinoic acid receptor signaling gates mechanical hypersensitivity in neuropathic pain. Neuron 2022; 110:4108-4124.e6. [PMID: 36223767 PMCID: PMC9789181 DOI: 10.1016/j.neuron.2022.09.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/27/2022] [Accepted: 09/22/2022] [Indexed: 02/08/2023]
Abstract
Central sensitization caused by spinal disinhibition is a key mechanism of mechanical allodynia in neuropathic pain. However, the molecular mechanisms underlying spinal disinhibition after nerve injury remain unclear. Here, we show in mice that spared nerve injury (SNI), which induces mechanical hypersensitivity and neuropathic pain, triggers homeostatic reduction of inhibitory outputs from dorsal horn parvalbumin-positive (PV+) interneurons onto both primary afferent terminals and excitatory interneurons. The reduction in inhibitory outputs drives hyperactivation of the spinal cord nociceptive pathway, causing mechanical hypersensitivity. We identified the retinoic acid receptor RARα, a central regulator of homeostatic plasticity, as the key molecular mediator for this synaptic disinhibition. Deletion of RARα in spinal PV+ neurons or application of an RARα antagonist in the spinal cord prevented the development of SNI-induced mechanical hypersensitivity. Our results identify RARα as a crucial molecular effector for neuropathic pain and a potential target for its treatment.
Collapse
Affiliation(s)
- Bing Cao
- Department of Neurosurgery, Wu Tsai Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gregory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lu Chen
- Department of Neurosurgery, Wu Tsai Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
60
|
From Low-Grade Inflammation in Osteoarthritis to Neuropsychiatric Sequelae: A Narrative Review. Int J Mol Sci 2022; 23:ijms232416031. [PMID: 36555670 PMCID: PMC9784931 DOI: 10.3390/ijms232416031] [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: 11/14/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Nowadays, osteoarthritis (OA), a common, multifactorial musculoskeletal disease, is considered to have a low-grade inflammatory pathogenetic component. Lately, neuropsychiatric sequelae of the disease have gained recognition. However, a link between the peripheral inflammatory process of OA and the development of neuropsychiatric pathology is not completely understood. In this review, we provide a narrative that explores the development of neuropsychiatric disease in the presence of chronic peripheral low-grade inflammation with a focus on its signaling to the brain. We describe the development of a pro-inflammatory environment in the OA-affected joint. We discuss inflammation-signaling pathways that link the affected joint to the central nervous system, mainly using primary sensory afferents and blood circulation via circumventricular organs and cerebral endothelium. The review describes molecular and cellular changes in the brain, recognized in the presence of chronic peripheral inflammation. In addition, changes in the volume of gray matter and alterations of connectivity important for the assessment of the efficacy of treatment in OA are discussed in the given review. Finally, the narrative considers the importance of the use of neuropsychiatric diagnostic tools for a disease with an inflammatory component in the clinical setting.
Collapse
|
61
|
Ishibashi T, Sueto D, Yoshikawa Y, Koga K, Yamaura K, Tsuda M. Identification of Spinal Inhibitory Interneurons Required for Attenuating Effect of Duloxetine on Neuropathic Allodynia-like Signs in Rats. Cells 2022; 11:cells11244051. [PMID: 36552814 PMCID: PMC9777279 DOI: 10.3390/cells11244051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Neuropathic pain is a chronic pain condition that occurs after nerve damage; allodynia, which refers to pain caused by generally innocuous stimuli, is a hallmark symptom. Although allodynia is often resistant to analgesics, the antidepressant duloxetine has been used as an effective therapeutic option. Duloxetine increases spinal noradrenaline (NA) levels by inhibiting its transporter at NAergic terminals in the spinal dorsal horn (SDH), which has been proposed to contribute to its pain-relieving effect. However, the mechanism through which duloxetine suppresses neuropathic allodynia remains unclear. Here, we identified an SDH inhibitory interneuron subset (captured by adeno-associated viral (AAV) vectors incorporating a rat neuropeptide Y promoter; AAV-NpyP+ neurons) that is mostly depolarized by NA. Furthermore, this excitatory effect was suppressed by pharmacological blockade or genetic knockdown of α1B-adrenoceptors (ARs) in AAV-NpyP+ SDH neurons. We found that duloxetine suppressed Aβ fiber-mediated allodynia-like behavioral responses after nerve injury and that this effect was not observed in AAV-NpyP+ SDH neuron-selective α1B-AR-knockdown. These results indicate that α1B-AR and AAV-NpyP+ neurons are critical targets for spinal NA and are necessary for the therapeutic effect of duloxetine on neuropathic pain, which can support the development of novel analgesics.
Collapse
Affiliation(s)
- Tadayuki Ishibashi
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Daichi Sueto
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yu Yoshikawa
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Keisuke Koga
- Department of Neurophysiology, Hyogo College of Medicine, Nishinomiya 663-8501, Japan
| | - Ken Yamaura
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Kyushu University Institute for Advanced Study, Fukuoka 819-0395, Japan
- Correspondence:
| |
Collapse
|
62
|
Lv X, Gao F, Cao X. Skeletal interoception in bone homeostasis and pain. Cell Metab 2022; 34:1914-1931. [PMID: 36257317 PMCID: PMC9742337 DOI: 10.1016/j.cmet.2022.09.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/07/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023]
Abstract
Accumulating evidence indicates that interoception maintains proper physiological status and orchestrates metabolic homeostasis by regulating feeding behaviors, glucose balance, and lipid metabolism. Continuous skeletal remodeling consumes a tremendous amount of energy to provide skeletal scaffolding, support muscle movement, store vital minerals, and maintain a niche for hematopoiesis, which are processes that also contribute to overall metabolic balance. Although skeletal innervation has been described for centuries, recent work has shown that skeletal metabolism is tightly regulated by the nervous system and that skeletal interoception regulates bone homeostasis. Here, we provide a general discussion of interoception and its effects on the skeleton and whole-body metabolism. We also discuss skeletal interoception-mediated regulation in the context of pathological conditions and skeletal pain as well as future challenges to our understanding of these process and how they can be leveraged for more effective therapy.
Collapse
Affiliation(s)
- Xiao Lv
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Feng Gao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xu Cao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
63
|
Kim BS, Inan S, Ständer S, Sciascia T, Szepietowski JC, Yosipovitch G. Role of kappa-opioid and mu-opioid receptors in pruritus: Peripheral and central itch circuits. Exp Dermatol 2022; 31:1900-1907. [PMID: 36054458 PMCID: PMC10087456 DOI: 10.1111/exd.14669] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 08/03/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022]
Abstract
Modern genetic approaches in animal models have unveiled novel itch-specific neural pathways, emboldening a paradigm in which drugs can be developed to selectively and potently target itch in a variety of chronic pruritic conditions. In recent years, kappa-opioid receptors (KORs) and mu-opioid receptors (MORs) have been implicated in both the suppression and promotion of itch, respectively, by acting on both the peripheral and central nervous systems. The precise mechanisms by which agents that modulate these pathways alleviate itch remains an active area of investigation. Notwithstanding this, a number of agents have demonstrated efficacy in clinical trials that influence both KOR and MOR signalling. Herein, we summarize a number of opioid receptor modulators in development and their promising efficacy across a number of chronic pruritic conditions, such as atopic dermatitis, uremic pruritus and beyond.
Collapse
Affiliation(s)
- Brian S Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Mark Lebwohl Center for Neuroinflammation and Sensation, Marc and Jennifer Lipschultz Precision Immunology Institute, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Sonja Ständer
- Department of Dermatology and Center for Chronic Pruritus, University Hospital Műnster, Műnster, Germany
| | | | - Jacek C Szepietowski
- Department of Dermatology, Venereology, and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Gil Yosipovitch
- Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
64
|
Bataille A, Le Gall C, Misery L, Talagas M. Merkel Cells Are Multimodal Sensory Cells: A Review of Study Methods. Cells 2022; 11:cells11233827. [PMID: 36497085 PMCID: PMC9737130 DOI: 10.3390/cells11233827] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Merkel cells (MCs) are rare multimodal epidermal sensory cells. Due to their interactions with slowly adapting type 1 (SA1) Aβ low-threshold mechanoreceptor (Aβ-LTMRs) afferents neurons to form Merkel complexes, they are considered to be part of the main tactile terminal organ involved in the light touch sensation. This function has been explored over time by ex vivo, in vivo, in vitro, and in silico approaches. Ex vivo studies have made it possible to characterize the topography, morphology, and cellular environment of these cells. The interactions of MCs with surrounding cells continue to be studied by ex vivo but also in vitro approaches. Indeed, in vitro models have improved the understanding of communication of MCs with other cells present in the skin at the cellular and molecular levels. As for in vivo methods, the sensory role of MC complexes can be demonstrated by observing physiological or pathological behavior after genetic modification in mouse models. In silico models are emerging and aim to elucidate the sensory coding mechanisms of these complexes. The different methods to study MC complexes presented in this review may allow the investigation of their involvement in other physiological and pathophysiological mechanisms, despite the difficulties in exploring these cells, in particular due to their rarity.
Collapse
Affiliation(s)
- Adeline Bataille
- LIEN—Laboratoire Interactions Epithélium Neurones, Brest University, F-29200 Brest, France
- Correspondence:
| | - Christelle Le Gall
- LIEN—Laboratoire Interactions Epithélium Neurones, Brest University, F-29200 Brest, France
- Department of Dermatology, Brest University Hospital, F-29200 Brest, France
| | - Laurent Misery
- LIEN—Laboratoire Interactions Epithélium Neurones, Brest University, F-29200 Brest, France
- Department of Dermatology, Brest University Hospital, F-29200 Brest, France
| | - Matthieu Talagas
- LIEN—Laboratoire Interactions Epithélium Neurones, Brest University, F-29200 Brest, France
- Department of Dermatology, Brest University Hospital, F-29200 Brest, France
| |
Collapse
|
65
|
Yeo M, Zhang Q, Ding L, Shen X, Chen Y, Liedtke W. Spinal cord dorsal horn sensory gate in preclinical models of chemotherapy-induced painful neuropathy and contact dermatitis chronic itch becomes less leaky with Kcc2 gene expression-enhancing treatments. Front Mol Neurosci 2022; 15:911606. [PMID: 36504679 PMCID: PMC9731339 DOI: 10.3389/fnmol.2022.911606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 10/05/2022] [Indexed: 11/25/2022] Open
Abstract
Low intraneuronal chloride in spinal cord dorsal horn (SCDH) pain relay neurons is of critical relevance for physiological transmission of primary sensory afferents because low intraneuronal chloride dictates GABA-ergic and glycin-ergic neurotransmission to be inhibitory. If neuronal chloride rises to unphysiological levels, the primary sensory gate in the spinal cord dorsal horn becomes corrupted, with resulting behavioral hallmarks of hypersensitivity and allodynia, for example in pathological pain. Low chloride in spinal cord dorsal horn neurons relies on the robust gene expression of Kcc2 and sustained transporter function of the KCC2 chloride-extruding electroneutral transporter. Based on a recent report where we characterized the GSK3-inhibitory small molecule, kenpaullone, as a Kcc2 gene expression-enhancer that potently repaired diminished Kcc2 expression and KCC2 transporter function in SCDH pain relay neurons, we extend our recent findings by reporting (i) effective pain control in a preclinical model of taxol-induced painful peripheral neuropathy that was accomplished by topical application of a TRPV4/TRPA1 dual-inhibitory compound (compound 16-8), and was associated with the repair of diminished Kcc2 gene expression in the SCDH; and (ii) potent functioning of kenpaullone as an antipruritic in a DNFB contact dermatitis preclinical model. These observations suggest that effective peripheral treatment of chemotherapy-induced painful peripheral neuropathy impacts the pain-transmitting neural circuit in the SCDH in a beneficial manner by enhancing Kcc2 gene expression, and that chronic pruritus might be relayed in the primary sensory gate of the spinal cord, following similar principles as pathological pain, specifically relating to the critical functioning of Kcc2 gene expression and the KCC2 transporter function.
Collapse
Affiliation(s)
- Michele Yeo
- Departments of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - Qiaojuan Zhang
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States
| | - LeAnne Ding
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States
| | - Xiangjun Shen
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States
| | - Yong Chen
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States
| | - Wolfgang Liedtke
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
66
|
Katayama Y, Miura A, Sakamoto T, Takanami K, Sakamoto H. Footedness for scratching itchy eyes in rodents. Proc Biol Sci 2022; 289:20221126. [PMID: 36259204 PMCID: PMC9579771 DOI: 10.1098/rspb.2022.1126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/21/2022] [Indexed: 11/12/2022] Open
Abstract
The neural bases of itchy eye transmission remain unclear compared with those involved in body itch. Here, we show in rodents that the gastrin-releasing peptide receptor (GRPR) of the trigeminal sensory system is involved in the transmission of itchy eyes. Interestingly, we further demonstrate a difference in scratching behaviour between the left and right hindfeet in rodents; histamine instillation into the conjunctival sac of both eyes revealed right-foot biased laterality in the scratching movements. Unilateral histamine instillation specifically induced neural activation in the ipsilateral sensory pathway, with no significant difference between the activations following left- and right-eye instillations. Thus, the behavioural laterality is presumably due to right-foot preference in rodents. Genetically modified rats with specific depletion of Grpr-expressing neurons in the trigeminal sensory nucleus caudalis of the medulla oblongata exhibited fewer and shorter histamine-induced scratching movements than controls and eliminated the footedness. These results taken together indicate that the Grpr-expressing neurons are required for the transmission of itch sensation from the eyes, but that foot preference is generated centrally. These findings could open up a new field of research on the mechanisms of the laterality in vertebrates and also offer new potential therapeutic approaches to refractory pruritic eye disorders.
Collapse
Affiliation(s)
- Yukitoshi Katayama
- Ushimado Marine Institute (UMI), Graduate School of Natural Science and Technology, Okayama University, Ushimado, Setouchi, Okayama 701-4303, Japan
| | - Ayane Miura
- Ushimado Marine Institute (UMI), Graduate School of Natural Science and Technology, Okayama University, Ushimado, Setouchi, Okayama 701-4303, Japan
- Department of Biology, Faculty of Science, Okayama University, 3-1-1 Kita-ku, Tsushimanaka, Okayama 700-8530, Japan
| | - Tatsuya Sakamoto
- Ushimado Marine Institute (UMI), Graduate School of Natural Science and Technology, Okayama University, Ushimado, Setouchi, Okayama 701-4303, Japan
| | - Keiko Takanami
- Mouse Genomics Resources Laboratory, National Institute of Genetics, Yata, Mishima, Shizuoka 411-8540, Japan
- Department of Environmental Health, Faculty of Human Life and Environmental Sciences, Nara Women's University, Kitauoya Nishimachi, Nara 630-8506, Japan
| | - Hirotaka Sakamoto
- Ushimado Marine Institute (UMI), Graduate School of Natural Science and Technology, Okayama University, Ushimado, Setouchi, Okayama 701-4303, Japan
| |
Collapse
|
67
|
Wu W, Jia J, Li F, Peng W, Liu H, Chen H, Shi Y. Editorial: Pain-related neural networks and regulation mechanisms. Front Mol Neurosci 2022; 15:1051335. [PMID: 36277490 PMCID: PMC9585350 DOI: 10.3389/fnmol.2022.1051335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022] Open
Affiliation(s)
- Wen Wu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Wen Wu
| | - Jie Jia
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Fengxian Li
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Peng
- School of Psychology, Shenzhen University, Shenzhen, China
| | - Howe Liu
- Department of Physical Therapy, Allen College, Waterloo, IA, United States
| | - Hui Chen
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Shi
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
68
|
Liu X, Xu Z, Fu X, Liu Y, Jia H, Yang Z, Zhang J, Wei S, Duan X. Stable, long-term single-neuronal recording from the rat spinal cord with flexible carbon nanotube fiber electrodes. J Neural Eng 2022; 19. [DOI: 10.1088/1741-2552/ac9258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 09/15/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Objective. Flexible implantable electrodes enable months-long stable recording of single-unit signals from rat brains. Despite extensive efforts in the development of flexible probes for brain recording, thus far there are no conclusions on their application in long-term single neuronal recording from the spinal cord which is more mechanically active. To this end, we realized the chronic recording of single-unit signals from the spinal cord of freely-moving rats using flexible carbon nanotube fiber (CNTF) electrodes. Approach. We developed flexible CNTF electrodes for intraspinal recording. Continuous in vivo impedance monitoring and histology studies were conducted to explore the critical factors determining the longevity of the recording, as well as to illustrate the evolution of the electrode-tissue interface. Gait analysis were performed to evaluate the biosafety of the chronic intraspinal implantation of the CNTF electrodes. Main results. By increasing the insulation thickness of the CNTF electrodes, single-unit signals were continuously recorded from the spinal cord of freely-moving rats without electrode repositioning for 3-4 months. Single neuronal and local field potential activities in response to somatic mechanical stimulation were successfully recorded from the spinal dorsal horns. Histological data demonstrated the ability of the CNTF microelectrodes to form an improved intraspinal interfaces with greatly reduced gliosis compared to their stiff metal counterparts. Continuous impedance monitoring suggested that the longevity of the intraspinal recording with CNTF electrodes was determined by the insulation durability. Gait analysis showed that the chronic presence of the CNTF electrodes caused no noticeable locomotor deficits in rats. Significance. It was found that the chronic recording from the spinal cord faces more stringent requirements on the electrode structural durability than recording from the brain. The stable, long-term intraspinal recording provides unique capabilities for studying the physiological functions of the spinal cord relating to motor, sensation, and autonomic control in both health and disease.
Collapse
|
69
|
Fernández-Carvajal A, Fernández-Ballester G, Ferrer-Montiel A. TRPV1 in chronic pruritus and pain: Soft modulation as a therapeutic strategy. Front Mol Neurosci 2022; 15:930964. [PMID: 36117910 PMCID: PMC9478410 DOI: 10.3389/fnmol.2022.930964] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic pain and pruritus are highly disabling pathologies that still lack appropriate therapeutic intervention. At cellular level the transduction and transmission of pain and pruritogenic signals are closely intertwined, negatively modulating each other. The molecular and cellular pathways involved are multifactorial and complex, including peripheral and central components. Peripherally, pain and itch are produced by subpopulations of specialized nociceptors that recognize and transduce algesic and pruritogenic signals. Although still under intense investigation, cumulative evidence is pointing to the thermosensory channel TRPV1 as a hub for a large number of pro-algesic and itchy agents. TRPV1 appears metabolically coupled to most neural receptors that recognize algesic and pruritic molecules. Thus, targeting TRPV1 function appears as a valuable and reasonable therapeutic strategy. In support of this tenet, capsaicin, a desensitizing TRPV1 agonist, has been shown to exhibit clinically relevant analgesic, anti-inflammatory, and anti-pruritic activities. However, potent TRPV1 antagonists have been questioned due to an hyperthermic secondary effect that prevented their clinical development. Thus, softer strategies directed to modulate peripheral TRPV1 function appear warranted to alleviate chronic pain and itch. In this regard, soft, deactivatable TRPV1 antagonists for topical or local application appear as an innovative approach for improving the distressing painful and itchy symptoms of patients suffering chronic pain or pruritus. Here, we review the data on these compounds and propose that this strategy could be used to target other peripheral therapeutic targets.
Collapse
|
70
|
Imado E, Sun S, Abawa AR, Tahara T, Kochi T, Huynh TNB, Asano S, Hasebe S, Nakamura Y, Hisaoka-Nakashima K, Kotake Y, Irifune M, Tsuga K, Takuma K, Morioka N, Kiguchi N, Ago Y. Prenatal exposure to valproic acid causes allodynia associated with spinal microglial activation. Neurochem Int 2022; 160:105415. [PMID: 36027995 DOI: 10.1016/j.neuint.2022.105415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/14/2022] [Accepted: 08/19/2022] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and social interaction and the presence of restricted, repetitive behaviors. Additionally, difficulties in sensory processing commonly occur in ASD. Sensory abnormalities include heightened or reduced sensitivity to pain, but the mechanism underlying sensory phenotypes in ASD remain unknown. Emerging evidence suggests that microglia play an important role in forming and refining neuronal circuitry, and thus contribute to neuronal plasticity and nociceptive signaling. In the present study, we investigated the age-dependent tactile sensitivity in an animal model of ASD induced by prenatal exposure to valproic acid (VPA) and subsequently assessed the involvement of microglia in the spinal cord in pain processing. Pregnant ICR (CD1) mice were intraperitoneally injected with either saline or VPA (500 mg/kg) on embryonic day 12.5. Male offspring of VPA-treated mothers showed mechanical allodynia at both 4 and 8 weeks of age. In the spinal cord dorsal horn in prenatally VPA-treated mice, the numbers and staining intensities of ionized calcium-binding adapter molecule 1-positive cells were increased and the cell bodies became enlarged, indicating microglial activation. Treatment with PLX3397, a colony-stimulating factor 1 receptor inhibitor, for 10 days resulted in a decreased number of spinal microglia and attenuated mechanical allodynia in adult mice prenatally exposed to VPA. Additionally, intrathecal injection of Mac-1-saporin, a saporin-conjugated anti-CD11b antibody to deplete microglia, abolished mechanical allodynia. These findings suggest that prenatal VPA treatment causes allodynia and that spinal microglia contribute to the increased nociceptive responses.
Collapse
Affiliation(s)
- Eiji Imado
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Samnang Sun
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Faculty of Odonto-Stomatology, University of Health Sciences, #73, Monivong Blvd., Sangkat Sras Chak, Khan Daun Penh, Phnom Penh, 12201, Cambodia
| | - Abrar Rizal Abawa
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Faculty of Dental Medicine, Universitas Airlangga, Jl. Mayjen Prof. Dr. Moestopo No. 47, Surabaya, East Java, 60132, Indonesia
| | - Takeru Tahara
- Department of Neurochemistry and Environmental Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Takahiro Kochi
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Tran Ngoc Bao Huynh
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Faculty of Odonto-Stomatology, Hong Bang International University, 215 Dien Bien Phu Street, Ward 15, Binh Thanh District, Ho Chi Minh City, Viet Nam
| | - Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Shigeru Hasebe
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Yaichiro Kotake
- Department of Neurochemistry and Environmental Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Masahiro Irifune
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kazuhiro Tsuga
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kazuhiro Takuma
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, 565-0871, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Wakayama, 640-8156, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
71
|
Thalhammer A, Fontanini M, Shi J, Scaini D, Recupero L, Evtushenko A, Fu Y, Pavagada S, Bistrovic-Popov A, Fruk L, Tian B, Ballerini L. Distributed interfacing by nanoscale photodiodes enables single-neuron light activation and sensory enhancement in 3D spinal explants. SCIENCE ADVANCES 2022; 8:eabp9257. [PMID: 35960795 PMCID: PMC9374338 DOI: 10.1126/sciadv.abp9257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/29/2022] [Indexed: 05/29/2023]
Abstract
Among emerging technologies developed to interface neuronal signaling, engineering electrodes at the nanoscale would yield more precise biodevices opening to progress in neural circuit investigations and to new therapeutic potential. Despite remarkable progress in miniature electronics for less invasive neurostimulation, most nano-enabled, optically triggered interfaces are demonstrated in cultured cells, which precludes the studies of natural neural circuits. We exploit here free-standing silicon-based nanoscale photodiodes to optically modulate single, identified neurons in mammalian spinal cord explants. With near-infrared light stimulation, we show that activating single excitatory or inhibitory neurons differently affects sensory circuits processing in the dorsal horn. We successfully functionalize nano-photodiodes to target single molecules, such as glutamate AMPA receptor subunits, thus enabling light activation of specific synaptic pathways. We conclude that nano-enabled neural interfaces can modulate selected sensory networks with low invasiveness. The use of nanoscale photodiodes can thus provide original perspective in linking neural activity to specific behavioral outcome.
Collapse
Affiliation(s)
- Agnes Thalhammer
- International School for Advanced Studies (SISSA), via Bonomea 265, 34136 Trieste, Italy
| | - Mario Fontanini
- International School for Advanced Studies (SISSA), via Bonomea 265, 34136 Trieste, Italy
| | - Jiuyun Shi
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Denis Scaini
- International School for Advanced Studies (SISSA), via Bonomea 265, 34136 Trieste, Italy
- Elettra Sincrotrone Trieste S.C.p.A., Area Science Park, I-34149 Trieste, Italy
- Basque Foundation for Science, Ikerbasque, Bilbao 48013, Spain
- Universidad del País Vasco (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Luca Recupero
- International School for Advanced Studies (SISSA), via Bonomea 265, 34136 Trieste, Italy
| | - Alexander Evtushenko
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Ying Fu
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Suraj Pavagada
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Andrea Bistrovic-Popov
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Ljiljana Fruk
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Bozhi Tian
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Laura Ballerini
- International School for Advanced Studies (SISSA), via Bonomea 265, 34136 Trieste, Italy
| |
Collapse
|
72
|
Ferrini F, Goldstein PA, Labrakakis C. Editorial: CNS pain circuits in health and disease. Front Neural Circuits 2022; 16:977404. [PMID: 36034336 PMCID: PMC9414010 DOI: 10.3389/fncir.2022.977404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Turin, Italy
- Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| | - Peter A. Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Charalampos Labrakakis
- Department of Biological Applications & Technology, University of Ioannina, Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina, Ioannina, Greece
- *Correspondence: Charalampos Labrakakis
| |
Collapse
|
73
|
Gupta S, Kawaguchi R, Heinrichs E, Gallardo S, Castellanos S, Mandric I, Novitch BG, Butler SJ. In vitro atlas of dorsal spinal interneurons reveals Wnt signaling as a critical regulator of progenitor expansion. Cell Rep 2022; 40:111119. [PMID: 35858555 PMCID: PMC9414195 DOI: 10.1016/j.celrep.2022.111119] [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: 11/08/2021] [Revised: 04/12/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022] Open
Abstract
Restoring sensation after injury or disease requires a reproducible method for generating large quantities of bona fide somatosensory interneurons. Toward this goal, we assess the mechanisms by which dorsal spinal interneurons (dIs; dI1-dI6) can be derived from mouse embryonic stem cells (mESCs). Using two developmentally relevant growth factors, retinoic acid (RA) and bone morphogenetic protein (BMP) 4, we recapitulate the complete in vivo program of dI differentiation through a neuromesodermal intermediate. Transcriptional profiling reveals that mESC-derived dIs strikingly resemble endogenous dIs, with the correct molecular and functional signatures. We further demonstrate that RA specifies dI4-dI6 fates through a default multipotential state, while the addition of BMP4 induces dI1-dI3 fates and activates Wnt signaling to enhance progenitor proliferation. Constitutively activating Wnt signaling permits the dramatic expansion of neural progenitor cultures. These cultures retain the capacity to differentiate into diverse populations of dIs, thereby providing a method of increasing neuronal yield.
Collapse
Affiliation(s)
- Sandeep Gupta
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Riki Kawaguchi
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric Heinrichs
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Genetics and Genomics Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Salena Gallardo
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephanie Castellanos
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; CIRM Bridges to Research Program, California State University, Northridge, Los Angeles, CA, USA
| | - Igor Mandric
- Department of Computer Science, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual & Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samantha J Butler
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual & Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
74
|
Wei N, Yu Y, Yang Y, Wang XL, Zhong ZJ, Chen XF, Yu YQ. Inhibitions and Down-Regulation of Motor Protein Eg5 Expression in Primary Sensory Neurons Reveal a Novel Therapeutic Target for Pathological Pain. Neurotherapeutics 2022; 19:1401-1413. [PMID: 35764763 PMCID: PMC9587155 DOI: 10.1007/s13311-022-01263-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 10/17/2022] Open
Abstract
The motor protein Eg5, known as kif11 or kinesin-5, interacts with adjacent microtubules in the mitotic spindle and plays essential roles in cell division, yet the function of Eg5 in mature postmitotic neurons remains largely unknown. In this study, we investigated the contribution and molecular mechanism of Eg5 in pathological pain. Pharmacological inhibition of Eg5 and a specific shRNA-expressing viral vector reversed complete Freund's adjuvant (CFA)-induced pain and abrogated vanilloid receptor subtype 1 (VR1) expression in dorsal root ganglion (DRG) neurons. In the dorsal root, Eg5 inhibition promoted VR1 axonal transport and decreased VR1 expression. In the spinal cord, Eg5 inhibition suppressed VR1 expression in axon terminals and impaired synapse formation in superficial laminae I/II. Finally, we showed that Eg5 is necessary for PI3K/Akt signalling-mediated VR1 membrane trafficking and pathological pain. The present study provides compelling evidence of a noncanonical function of Eg5 in primary sensory neurons. These results suggest that Eg5 may be a potential therapeutic target for intractable pain.
Collapse
Affiliation(s)
- Na Wei
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yang Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Zhen-Juan Zhong
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Xue-Feng Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yao-Qing Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Baqiao, Xi'an, 710038, China.
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China.
| |
Collapse
|
75
|
Spinal ascending pathways for somatosensory information processing. Trends Neurosci 2022; 45:594-607. [PMID: 35701247 DOI: 10.1016/j.tins.2022.05.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/19/2022] [Accepted: 05/14/2022] [Indexed: 12/27/2022]
Abstract
The somatosensory system processes diverse types of information including mechanical, thermal, and chemical signals. It has an essential role in sensory perception and body movement and, thus, is crucial for organism survival. The neural network for processing somatosensory information comprises multiple key nodes. Spinal projection neurons represent the key node for transmitting somatosensory information from the periphery to the brain. Although the anatomy of spinal ascending pathways has been characterized, the mechanisms underlying somatosensory information processing by spinal ascending pathways are incompletely understood. Recent studies have begun to reveal the diversity of spinal ascending pathways and their functional roles in somatosensory information processing. Here, we review the anatomic, molecular, and functional characteristics of spinal ascending pathways.
Collapse
|
76
|
Wang H, Chen W, Dong Z, Xing G, Cui W, Yao L, Zou WJ, Robinson HL, Bian Y, Liu Z, Zhao K, Luo B, Gao N, Zhang H, Ren X, Yu Z, Meixiong J, Xiong WC, Mei L. A novel spinal neuron connection for heat sensation. Neuron 2022; 110:2315-2333.e6. [PMID: 35561677 DOI: 10.1016/j.neuron.2022.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 03/14/2022] [Accepted: 04/19/2022] [Indexed: 12/30/2022]
Abstract
Heat perception enables acute avoidance responses to prevent tissue damage and maintain body thermal homeostasis. Unlike other modalities, how heat signals are processed in the spinal cord remains unclear. By single-cell gene profiling, we identified ErbB4, a transmembrane tyrosine kinase, as a novel marker of heat-sensitive spinal neurons in mice. Ablating spinal ErbB4+ neurons attenuates heat sensation. These neurons receive monosynaptic inputs from TRPV1+ nociceptors and form excitatory synapses onto target neurons. Activation of ErbB4+ neurons enhances the heat response, while inhibition reduces the heat response. We showed that heat sensation is regulated by NRG1, an activator of ErbB4, and it involves dynamic activity of the tyrosine kinase that promotes glutamatergic transmission. Evidence indicates that the NRG1-ErbB4 signaling is also engaged in hypersensitivity of pathological pain. Together, these results identify a spinal neuron connection consisting of ErbB4+ neurons for heat sensation and reveal a regulatory mechanism by the NRG1-ErbB4 signaling.
Collapse
Affiliation(s)
- Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wenbing Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zhaoqi Dong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Guanglin Xing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wanpeng Cui
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Lingling Yao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wen-Jun Zou
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Heath L Robinson
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Yaoyao Bian
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zhipeng Liu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Kai Zhao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Bin Luo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Nannan Gao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Hongsheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Xiao Ren
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng Yu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - James Meixiong
- Solomon H. Snyder Department of Neuroscience and Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
77
|
Kanehisa K, Koga K, Maejima S, Shiraishi Y, Asai K, Shiratori-Hayashi M, Xiao MF, Sakamoto H, Worley PF, Tsuda M. Neuronal pentraxin 2 is required for facilitating excitatory synaptic inputs onto spinal neurons involved in pruriceptive transmission in a model of chronic itch. Nat Commun 2022; 13:2367. [PMID: 35501343 PMCID: PMC9061767 DOI: 10.1038/s41467-022-30089-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 04/14/2022] [Indexed: 11/08/2022] Open
Abstract
An excitatory neuron subset in the spinal dorsal horn (SDH) that expresses gastrin-releasing peptide receptors (GRPR) is critical for pruriceptive transmission. Here, we show that glutamatergic excitatory inputs onto GRPR+ neurons are facilitated in mouse models of chronic itch. In these models, neuronal pentraxin 2 (NPTX2), an activity-dependent immediate early gene product, is upregulated in the dorsal root ganglion (DRG) neurons. Electron microscopy reveals that NPTX2 is present at presynaptic terminals connected onto postsynaptic GRPR+ neurons. NPTX2-knockout prevents the facilitation of synaptic inputs to GRPR+ neurons, and repetitive scratching behavior. DRG-specific NPTX2 expression rescues the impaired behavioral phenotype in NPTX2-knockout mice. Moreover, ectopic expression of a dominant-negative form of NPTX2 in DRG neurons reduces chronic itch-like behavior in mice. Our findings indicate that the upregulation of NPTX2 expression in DRG neurons contributes to the facilitation of glutamatergic inputs onto GRPR+ neurons under chronic itch-like conditions, providing a potential therapeutic target.
Collapse
Affiliation(s)
- Kensho Kanehisa
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keisuke Koga
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Neurophysiology, Hyogo College of Medicine, Nishinomiya, Hyogo, 663-8501, Japan
| | - Sho Maejima
- Ushimado Marine Institute, Graduate School of Natural Science and Technology, Okayama University, 130-17 Kashino, Ushimado, Setouchi, 701-4303, Japan
| | - Yuto Shiraishi
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Konatsu Asai
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Miho Shiratori-Hayashi
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Mei-Fang Xiao
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, United States
| | - Hirotaka Sakamoto
- Ushimado Marine Institute, Graduate School of Natural Science and Technology, Okayama University, 130-17 Kashino, Ushimado, Setouchi, 701-4303, Japan
| | - Paul F Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, United States
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
78
|
Hu L, Jiang GY, Wang YP, Hu ZB, Zhou BY, Zhang L, Song NN, Huang Y, Chai GD, Chen JY, Lang B, Xu L, Liu JL, Li Y, Wang QX, Ding YQ. The role of PTEN in primary sensory neurons in processing itch and thermal information in mice. Cell Rep 2022; 39:110724. [PMID: 35443189 DOI: 10.1016/j.celrep.2022.110724] [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: 08/26/2021] [Revised: 03/03/2022] [Accepted: 03/30/2022] [Indexed: 12/01/2022] Open
Abstract
PTEN is known as a tumor suppressor and plays essential roles in brain development. Here, we report that PTEN in primary sensory neurons is involved in processing itch and thermal information in adult mice. Deletion of PTEN in the dorsal root ganglia (DRG) is achieved in adult Drg11-CreER: PTENflox/flox (PTEN CKO) mice with oral administration of tamoxifen, and CKO mice develop pathological itch and elevated itch responses on exposure to various pruritogens. PTEN deletion leads to ectopic expression of TRPV1 and MrgprA3 in IB4+ non-peptidergic DRG neurons, and the TRPV1 is responsive to capsaicin. Importantly, the elevated itch responses are no longer present in Drg11-CreER: PTENflox/flox: TRPV1flox/flox (PTEN: TRPV1 dCKO) mice. In addition, thermal stimulation is enhanced in PTEN CKO mice but blunted in dCKO mice. PTEN-involved regulation of itch-related gene expression in DRG neurons provides insights for understanding molecular mechanism of itch and thermal sensation at the spinal level.
Collapse
Affiliation(s)
- Ling Hu
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Guan-Yu Jiang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ying-Ping Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Zhi-Bin Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Bing-Yao Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ying Huang
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
| | - Guo-Dong Chai
- Department of Anesthesiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jia-Yin Chen
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China; Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Lin Xu
- Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Jun-Ling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Qing-Xiu Wang
- Department of Anesthesiology, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Yu-Qiang Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
79
|
Xie YK, Luo H, Zhang SX, Chen XY, Guo R, Qiu XY, Liu S, Wu H, Chen WB, Zhen XH, Ma Q, Tian JL, Li S, Chen X, Han Q, Duan S, Shen C, Yang F, Xu ZZ. GPR177 in A-fiber sensory neurons drives diabetic neuropathic pain via WNT-mediated TRPV1 activation. Sci Transl Med 2022; 14:eabh2557. [PMID: 35385340 DOI: 10.1126/scitranslmed.abh2557] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Diabetic neuropathic pain (DNP) is a common and devastating complication in patients with diabetes. The mechanisms mediating DNP are not completely elucidated, and effective treatments are lacking. A-fiber sensory neurons have been shown to mediate the development of mechanical allodynia in neuropathic pain, yet the molecular basis underlying the contribution of A-fiber neurons is still unclear. Here, we report that the orphan G protein-coupled receptor 177 (GPR177) in A-fiber neurons drives DNP via WNT5a-mediated activation of transient receptor potential vanilloid receptor-1 (TRPV1) ion channel. GPR177 is mainly expressed in large-diameter A-fiber dorsal root ganglion (DRG) neurons and required for the development of DNP in mice. Mechanistically, we found that GPR177 mediated the secretion of WNT5a from A-fiber DRG neurons into cerebrospinal fluid (CSF), which was necessary for the maintenance of DNP. Extracellular perfusion of WNT5a induced rapid currents in both TRPV1-expressing heterologous cells and nociceptive DRG neurons. Computer simulations revealed that WNT5a has the potential to bind the residues at the extracellular S5-S6 loop of TRPV1. Using a peptide able to disrupt the predicted WNT5a/TRPV1 interaction suppressed DNP- and WNT5a-induced neuropathic pain symptoms in rodents. We confirmed GPR177/WNT5A coexpression in human DRG neurons and WNT5A secretion in CSF from patients with DNP. Thus, our results reveal a role for WNT5a as an endogenous and potent TRPV1 agonist, and the GPR177-WNT5a-TRPV1 axis as a driver of DNP pathogenesis in rodents. Our findings identified a potential analgesic target that might relieve neuropathic pain in patients with diabetes.
Collapse
Affiliation(s)
- Ya-Kai Xie
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hao Luo
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shan-Xin Zhang
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiao-Ying Chen
- Department of Biophysics, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Ran Guo
- Department of Pain, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiao-Yun Qiu
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shuai Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Hui Wu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wen-Bo Chen
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xing-Hua Zhen
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qiang Ma
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jin-Lan Tian
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shun Li
- Department of Pain, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xinzhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Qingjian Han
- State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Shumin Duan
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Chengyong Shen
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Fan Yang
- Department of Biophysics, and Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zhen-Zhong Xu
- Department of Neurobiology and Department of Anesthesiology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
80
|
Bardoni R. Experimental Protocols and Analytical Procedures for Studying Synaptic Transmission in Rodent Spinal Cord Dorsal Horn. Curr Protoc 2022; 2:e409. [PMID: 35435326 DOI: 10.1002/cpz1.409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Synaptic modulation and plasticity are key mechanisms underlying pain transmission in the spinal cord and supra-spinal centers. The study and understanding of these phenomena are fundamental to investigating both acute nociception and maladaptive changes occurring in chronic pain. This article describes experimental protocols and analytical methods utilized in electrophysiological studies to investigate synaptic modulation and plasticity at the first station of somatosensory processing, the spinal cord dorsal horn. Protocols useful for characterizing the nature of synaptic inputs, the site of modulation (pre- versus postsynaptic), and the presence of short-term synaptic plasticity are presented. These methods can be employed to study the physiology of acute nociception, the pathological mechanisms of persistent inflammatory and neuropathic pain, and the pharmacology of receptors and channels involved in pain transmission. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Spinal cord dissection and acute slice preparation Basic Protocol 2: Stimulation of the dorsal root and extracellular recording (compound action potentials and field potentials) Basic Protocol 3: Patch-clamp recording from dorsal horn neurons: action potential firing patterns and evoked synaptic inputs Basic Protocol 4: Analysis of parameters responsible for changes in synaptic efficacy Basic Protocol 5: Recording and analysis of currents mediated by astrocytic glutamate.
Collapse
Affiliation(s)
- Rita Bardoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi, Modena, Italy
| |
Collapse
|
81
|
Slick Potassium Channels Control Pain and Itch in Distinct Populations of Sensory and Spinal Neurons in Mice. Anesthesiology 2022; 136:802-822. [PMID: 35303056 DOI: 10.1097/aln.0000000000004163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Slick, a sodium-activated potassium channel, has been recently identified in somatosensory pathways, but its functional role is poorly understood. The authors of this study hypothesized that Slick is involved in processing sensations of pain and itch. METHODS Immunostaining, in situ hybridization, Western blot, and real-time quantitative reverse transcription polymerase chain reaction were used to investigate the expression of Slick in dorsal root ganglia and the spinal cord. Mice lacking Slick globally (Slick-/-) or conditionally in neurons of the spinal dorsal horn (Lbx1-Slick-/-) were assessed in behavioral models. RESULTS The authors found Slick to be enriched in nociceptive Aδ-fibers and in populations of interneurons in the spinal dorsal horn. Slick-/- mice, but not Lbx1-Slick-/- mice, showed enhanced responses to noxious heat in the hot plate and tail-immersion tests. Both Slick-/- and Lbx1-Slick-/- mice demonstrated prolonged paw licking after capsaicin injection (mean ± SD, 45.6 ± 30.1 s [95% CI, 19.8 to 71.4]; and 13.1 ± 16.1 s [95% CI, 1.8 to 28.0]; P = 0.006 [Slick-/- {n = 8} and wild-type {n = 7}, respectively]), which was paralleled by increased phosphorylation of the neuronal activity marker extracellular signal-regulated kinase in the spinal cord. In the spinal dorsal horn, Slick is colocalized with somatostatin receptor 2 (SSTR2), and intrathecal preadministration of the SSTR2 antagonist CYN-154806 prevented increased capsaicin-induced licking in Slick-/- and Lbx1-Slick-/- mice. Moreover, scratching after intrathecal delivery of the somatostatin analog octreotide was considerably reduced in Slick-/- and Lbx1-Slick-/- mice (Slick-/- [n = 8]: 6.1 ± 6.7 bouts [95% CI, 0.6 to 11.7]; wild-type [n =8]: 47.4 ± 51.1 bouts [95% CI, 4.8 to 90.2]; P = 0.039). CONCLUSIONS Slick expressed in a subset of sensory neurons modulates heat-induced pain, while Slick expressed in spinal cord interneurons inhibits capsaicin-induced pain but facilitates somatostatin-induced itch. EDITOR’S PERSPECTIVE
Collapse
|
82
|
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: 51] [Impact Index Per Article: 17.0] [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
|
83
|
Li J, Bai Y, Liang Y, Zhang Y, Zhao Q, Ge J, Li D, Zhu Y, Cai G, Tao H, Wu S, Huang J. Parvalbumin Neurons in Zona Incerta Regulate Itch in Mice. Front Mol Neurosci 2022; 15:843754. [PMID: 35299695 PMCID: PMC8920991 DOI: 10.3389/fnmol.2022.843754] [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: 12/26/2021] [Accepted: 02/08/2022] [Indexed: 12/20/2022] Open
Abstract
Pain and itch are intricately entangled at both circuitry and behavioral levels. Emerging evidence indicates that parvalbumin (PV)-expressing neurons in zona incerta (ZI) are critical for promoting nocifensive behaviors. However, the role of these neurons in itch modulation remains elusive. Herein, by combining FOS immunostaining, fiber photometry, and chemogenetic manipulation, we reveal that ZI PV neurons act as an endogenous negative diencephalic modulator for itch processing. Morphological data showed that both histamine and chloroquine stimuli induced FOS expression in ZI PV neurons. The activation of these neurons was further supported by the increased calcium signal upon scratching behavior evoked by acute itch. Behavioral data further indicated that chemogenetic activation of these neurons reduced scratching behaviors related to histaminergic and non-histaminergic acute itch. Similar neural activity and modulatory role of ZI PV neurons were seen in mice with chronic itch induced by atopic dermatitis. Together, our study provides direct evidence for the role of ZI PV neurons in regulating itch, and identifies a potential target for the remedy of chronic itch.
Collapse
Affiliation(s)
- Jiaqi Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yi Liang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Yiwen Zhang
- The Cadet Team 6 of School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Qiuying Zhao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Junye Ge
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Dangchao Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Yuanyuan Zhu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Guohong Cai
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Huiren Tao
- Department of Spine Surgery, Shenzhen University General Hospital, Shenzhen, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Jing Huang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
84
|
Rabies anterograde monosynaptic tracing allows identification of postsynaptic circuits receiving distinct somatosensory input. Neuroscience 2022; 491:75-86. [DOI: 10.1016/j.neuroscience.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 01/13/2023]
|
85
|
Chen Y, Zhou Y, Li XC, Ma X, Mi WL, Chu YX, Wang YQ, Mao-Ying QL. Neuronal GRK2 regulates microglial activation and contributes to electroacupuncture analgesia on inflammatory pain in mice. Biol Res 2022; 55:5. [PMID: 35115050 PMCID: PMC8812183 DOI: 10.1186/s40659-022-00374-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 01/22/2022] [Indexed: 12/30/2022] Open
Abstract
Background G protein coupled receptor kinase 2 (GRK2) has been demonstrated to play a crucial role in the development of chronic pain. Acupuncture is an alternative therapy widely used for pain management. In this study, we investigated the role of spinal neuronal GRK2 in electroacupuncture (EA) analgesia. Methods The mice model of inflammatory pain was built by subcutaneous injection of Complete Freund’s Adjuvant (CFA) into the plantar surface of the hind paws. The mechanical allodynia of mice was examined by von Frey test. The mice were subjected to EA treatment (BL60 and ST36 acupuncture points) for 1 week. Overexpression and downregulation of spinal neuronal GRK2 were achieved by intraspinal injection of adeno associated virus (AAV) containing neuron-specific promoters, and microglial activation and neuroinflammation were evaluated by real-time PCR. Results Intraplantar injection with CFA in mice induced the decrease of GRK2 and microglial activation along with neuroinflammation in spinal cord. EA treatment increased the spinal GRK2, reduced neuroinflammation, and significantly decreased CFA-induced mechanical allodynia. The effects of EA were markedly weakened by non-cell-specific downregulation of spinal GRK2. Further, intraspinal injection of AAV containing neuron-specific promoters specifically downregulated neuronal GRK2, and weakened the regulatory effect of EA on CFA-induced mechanical allodynia and microglial activation. Meanwhile, overexpression of spinal neuronal GRK2 decreased mechanical allodynia. All these indicated that the neuronal GRK2 mediated microglial activation and neuroinflammation, and subsequently contributed to CFA-induced inflammatory pain. Conclusion The restoration of the spinal GRK2 and subsequent suppression of microglial activation and neuroinflammation might be an important mechanism for EA analgesia. Our findings further suggested that the spinal GRK2, especially neuronal GRK2, might be the potential target for EA analgesia and pain management, and we provided a new experimental basis for the EA treatment of pain. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-022-00374-6.
Collapse
Affiliation(s)
- Yu Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yang Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiao-Chen Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xue Ma
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wen-Li Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, 200433, People's Republic of China
| | - Qi-Liang Mao-Ying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, People's Republic of China. .,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
86
|
Oliva V, Hartley-Davies R, Moran R, Pickering AE, Brooks JC. Simultaneous brain, brainstem and spinal cord pharmacological-fMRI reveals involvement of an endogenous opioid network in attentional analgesia. eLife 2022; 11:71877. [PMID: 35080494 PMCID: PMC8843089 DOI: 10.7554/elife.71877] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Pain perception is decreased by shifting attentional focus away from a threatening event. This attentional analgesia engages parallel descending control pathways from anterior cingulate (ACC) to locus coeruleus, and ACC to periaqueductal grey (PAG) – rostral ventromedial medulla (RVM), indicating possible roles for noradrenergic or opioidergic neuromodulators. To determine which pathway modulates nociceptive activity in humans, we used simultaneous whole brain-spinal cord pharmacological-fMRI (N = 39) across three sessions. Noxious thermal forearm stimulation generated somatotopic-activation of dorsal horn (DH) whose activity correlated with pain report and mirrored attentional pain modulation. Activity in an adjacent cluster reported the interaction between task and noxious stimulus. Effective connectivity analysis revealed that ACC interacts with PAG and RVM to modulate spinal cord activity. Blocking endogenous opioids with Naltrexone impairs attentional analgesia and disrupts RVM-spinal and ACC-PAG connectivity. Noradrenergic augmentation with Reboxetine did not alter attentional analgesia. Cognitive pain modulation involves opioidergic ACC-PAG-RVM descending control which suppresses spinal nociceptive activity.
Collapse
Affiliation(s)
- Valeria Oliva
- Department of Anesthesiology, University of California, San Diego, La Jolla, United States
| | - Ron Hartley-Davies
- School of Psychological Science, University of Bristol, Bristol, United Kingdom
| | - Rosalyn Moran
- Department of Neuroimaging, King's College London, London, United Kingdom
| | - Anthony E Pickering
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | | |
Collapse
|
87
|
Modulation of itch and pain signals processing in ventrobasal thalamus by thalamic reticular nucleus. iScience 2022; 25:103625. [PMID: 35106466 PMCID: PMC8786640 DOI: 10.1016/j.isci.2021.103625] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/16/2021] [Accepted: 12/10/2021] [Indexed: 01/03/2023] Open
Abstract
Thalamic reticular nucleus (TRN) is known to be crucial for dynamically modulating sensory processing. Recently, the functional role of TRN in itch and pain sensation processing has drawn much attention. We found that ventrobasal thalamus (VB) neurons exhibited scratching behavior-related and nociceptive behavior-related neuronal activity changes, and most of VB neurons responsive to pruritic stimulus were also activated by nociceptive stimulus. Inhibition of VB could relieve itch-induced scratching behaviors and pathological pain without affecting basal nociceptive thresholds, and activation of VB could facilitate scratching behaviors. Tracing and electrophysiology recording results showed that VB mainly received inhibitory inputs from ventral TRN. Furthermore, optogenetic activation of TRN-VB projections suppressed scratching behaviors, and ablation of TRN enhanced scratching behaviors. In addition, activation of TRN-VB projections relieved the pathological pain without affecting basal nociceptive thresholds. Thus, our study indicates that TRN modulates itch and pain signals processing via TRN-VB inhibitory projections. VB is involved in both itch and pain signals processing Manipulation of VB or TRN-VB inhibitory projections modulates both itch and pain Enhancing the inhibitory tone might be a strategy for treating itch and pain
Collapse
|
88
|
Chen Z, Lin MT, Zhan C, Zhong NS, Mu D, Lai KF, Liu MJ. A descending pathway emanating from the periaqueductal gray mediates the development of cough-like hypersensitivity. iScience 2022; 25:103641. [PMID: 35028531 PMCID: PMC8741493 DOI: 10.1016/j.isci.2021.103641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/11/2021] [Accepted: 12/13/2021] [Indexed: 01/10/2023] Open
Abstract
Chronic cough is a common refractory symptom of various respiratory diseases. However, the neural mechanisms that modulate the cough sensitivity and mediate chronic cough remain elusive. Here, we report that GABAergic neurons in the lateral/ventrolateral periaqueductal gray (l/vlPAG) suppress cough processing via a descending pathway. We found that l/vlPAG neurons are activated by coughing-like behaviors and that tussive agent-evoked coughing-like behaviors are impaired after activation of l/vlPAG neurons. In addition, we showed that l/vlPAG neurons form inhibitory synapses with the nucleus of the solitary tract (NTS) neurons. The synaptic strength of these inhibitory projections is weaker in cough hypersensitivity model mice than in naïve mice. Important, activation of l/vlPAG GABAergic neurons projecting to the NTS decreases coughing-like behaviors. In contrast, suppressing these neurons enhances cough sensitivity. These results support the notion that l/vlPAG GABAergic neurons play important roles in cough hypersensitivity and chronic cough through disinhibition of cough processing at the medullary level. GABAergic neurons in the l/vlPAG inhibit coughing-like behaviors The l/vlPAG sends predominately inhibitory projections to the NTS l/vlPAG GABAergic neurons modulate coughing-like behaviors via descending projections l/vlPAG-NTS projections mediate cough hypersensitivity via disinhibitory mechanisms
Collapse
Affiliation(s)
- Zhe Chen
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China.,Laboratory of Cough, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu 215300, China
| | - Ming-Tong Lin
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Chen Zhan
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Di Mu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| | - Ke-Fang Lai
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Mingzhe J Liu
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| |
Collapse
|
89
|
Imambocus BN, Zhou F, Formozov A, Wittich A, Tenedini FM, Hu C, Sauter K, Macarenhas Varela E, Herédia F, Casimiro AP, Macedo A, Schlegel P, Yang CH, Miguel-Aliaga I, Wiegert JS, Pankratz MJ, Gontijo AM, Cardona A, Soba P. A neuropeptidergic circuit gates selective escape behavior of Drosophila larvae. Curr Biol 2021; 32:149-163.e8. [PMID: 34798050 DOI: 10.1016/j.cub.2021.10.069] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 10/05/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
Animals display selective escape behaviors when faced with environmental threats. Selection of the appropriate response by the underlying neuronal network is key to maximizing chances of survival, yet the underlying network mechanisms are so far not fully understood. Using synapse-level reconstruction of the Drosophila larval network paired with physiological and behavioral readouts, we uncovered a circuit that gates selective escape behavior for noxious light through acute and input-specific neuropeptide action. Sensory neurons required for avoidance of noxious light and escape in response to harsh touch, each converge on discrete domains of neuromodulatory hub neurons. We show that acute release of hub neuron-derived insulin-like peptide 7 (Ilp7) and cognate relaxin family receptor (Lgr4) signaling in downstream neurons are required for noxious light avoidance, but not harsh touch responses. Our work highlights a role for compartmentalized circuit organization and neuropeptide release from regulatory hubs, acting as central circuit elements gating escape responses.
Collapse
Affiliation(s)
- Bibi Nusreen Imambocus
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Fangmin Zhou
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Andrey Formozov
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Annika Wittich
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Federico M Tenedini
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Chun Hu
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Kathrin Sauter
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Ednilson Macarenhas Varela
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Fabiana Herédia
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Andreia P Casimiro
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - André Macedo
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Philipp Schlegel
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Chung-Hui Yang
- Department of Neurobiology, Duke University Medical School, 427E Bryan Research, Durham, NC 27710, USA
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - J Simon Wiegert
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Michael J Pankratz
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Alisson M Gontijo
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Albert Cardona
- HHMI Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Physiology, Development, and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| |
Collapse
|
90
|
Shin SM, Moehring F, Itson-Zoske B, Fan F, Stucky CL, Hogan QH, Yu H. Piezo2 mechanosensitive ion channel is located to sensory neurons and nonneuronal cells in rat peripheral sensory pathway: implications in pain. Pain 2021; 162:2750-2768. [PMID: 34285153 PMCID: PMC8526381 DOI: 10.1097/j.pain.0000000000002356] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/18/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Piezo2 mechanotransduction channel is a crucial mediator of sensory neurons for sensing and transducing touch, vibration, and proprioception. We here characterized Piezo2 expression and cell specificity in rat peripheral sensory pathway using a validated Piezo2 antibody. Immunohistochemistry using this antibody revealed Piezo2 expression in pan primary sensory neurons of dorsal root ganglia in naïve rats, which was actively transported along afferent axons to both central presynaptic terminals innervating the spinal dorsal horn (DH) and peripheral afferent terminals in the skin. Piezo2 immunoreactivity (IR) was also detected in the postsynaptic neurons of the DH and in the motor neurons of the ventral horn, but not in spinal glial fibrillary acidic protein-positive and Iba1-positive glia. Notably, Piezo2-IR was clearly identified in peripheral nonneuronal cells, including perineuronal glia, Schwann cells in the sciatic nerve and surrounding cutaneous afferent endings, as well as in skin epidermal Merkel cells and melanocytes. Immunoblots showed increased Piezo2 in dorsal root ganglia ipsilateral to plantar injection of complete Freund's adjuvant, and immunostaining revealed increased Piezo2-IR intensity in the DH ipsilateral to complete Freund's adjuvant injection. This elevation of DH Piezo2-IR was also evident in various neuropathic pain models and monosodium iodoacetate knee osteoarthritis pain model, compared with controls. We conclude that (1) the pan neuronal profile of Piezo2 expression suggests that Piezo2 may function extend beyond simply touch or proprioception mediated by large-sized low-threshold mechanosensitive primary sensory neurons; (2) Piezo2 may have functional roles involving sensory processing in the spinal cord, Schwann cells, and skin melanocytes; and (3) aberrant Piezo2 expression may contribute pain pathogenesis.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Francie Moehring
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Fan Fan
- Department of Pharmacology and Toxicology, Mississippi University Medical Center, Jackson, Mississippi 39216
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin 53295
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin 53295
| |
Collapse
|
91
|
Yeo M, Chen Y, Jiang C, Chen G, Wang K, Chandra S, Bortsov A, Lioudyno M, Zeng Q, Wang P, Wang Z, Busciglio J, Ji RR, Liedtke W. Repurposing cancer drugs identifies kenpaullone which ameliorates pathologic pain in preclinical models via normalization of inhibitory neurotransmission. Nat Commun 2021; 12:6208. [PMID: 34707084 PMCID: PMC8551327 DOI: 10.1038/s41467-021-26270-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/24/2021] [Indexed: 11/13/2022] Open
Abstract
Inhibitory GABA-ergic neurotransmission is fundamental for the adult vertebrate central nervous system and requires low chloride concentration in neurons, maintained by KCC2, a neuroprotective ion transporter that extrudes intracellular neuronal chloride. To identify Kcc2 gene expression‑enhancing compounds, we screened 1057 cell growth-regulating compounds in cultured primary cortical neurons. We identified kenpaullone (KP), which enhanced Kcc2/KCC2 expression and function in cultured rodent and human neurons by inhibiting GSK3ß. KP effectively reduced pathologic pain-like behavior in mouse models of nerve injury and bone cancer. In a nerve-injury pain model, KP restored Kcc2 expression and GABA-evoked chloride reversal potential in the spinal cord dorsal horn. Delta-catenin, a phosphorylation-target of GSK3ß in neurons, activated the Kcc2 promoter via KAISO transcription factor. Transient spinal over-expression of delta-catenin mimicked KP analgesia. Our findings of a newly repurposed compound and a novel, genetically-encoded mechanism that each enhance Kcc2 gene expression enable us to re-normalize disrupted inhibitory neurotransmission through genetic re-programming.
Collapse
Affiliation(s)
- Michele Yeo
- Department of Neurology, Duke University Medical Center, Durham, NC, USA.
| | - Yong Chen
- Department of Neurology, Duke University Medical Center, Durham, NC, USA.
| | - Changyu Jiang
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Gang Chen
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Kaiyuan Wang
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Sharat Chandra
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Andrey Bortsov
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Maria Lioudyno
- Department of Neurobiology & Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), Center for the Neurobiology of Learning and Memory, University of California at Irvine, Irvine, CA, USA
| | - Qian Zeng
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Peng Wang
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Zilong Wang
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Jorge Busciglio
- Department of Neurobiology & Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), Center for the Neurobiology of Learning and Memory, University of California at Irvine, Irvine, CA, USA
| | - Ru-Rong Ji
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| | - Wolfgang Liedtke
- Department of Neurology, Duke University Medical Center, Durham, NC, USA.
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
- Duke Neurology Clinics for Headache, Head-Pain and Trigeminal Sensory Disorders, Duke University Medical Center, Durham, NC, USA.
- Duke Anesthesiology Clinics for Innovative Pain Therapy, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
92
|
Non-Smad, BMP-dependent signaling protects against the effects of acute ethanol toxicity. Toxicol Lett 2021; 353:118-126. [PMID: 34687774 DOI: 10.1016/j.toxlet.2021.10.009] [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: 05/12/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 01/20/2023]
Abstract
This study explores the effect of acute Ethanol (EtOH) exposure on Bone Morphogenetic Protein (BMP)-evoked intracellular signaling, and the concomitant morphological changes induced by EtOH in C2C12 cells and DRG (Dorsal root ganglion) neurons in an in vitro model related to Fetal Alcohol Syndrome Disorder (FASD). All assays were performed within 30 min of BMP stimulation to specifically investigate the earliest events occurring in BMP-evoked intracellular signaling pathways. We show that Smad phosphorylation and nuclear translocation stimulated by BMPs was not altered following acute exposure to EtOH. In contrast, acute EtOH exposure alone caused a striking concentration-dependent decrease in Akt phosphorylation, as well as a loss of adhesion in C2C12 cells. The addition of BMPs before exposure to EtOH was associated with maintenance of Akt phosphorylation, greater cell adhesion in C2C12 cells, and preservation of growth cone complexity in DRG neurons. Thus, for both C2C12 cells and DRG neurons, BMPs, acting through non-canonical BMP signaling pathways, appear to impart some protection against the profound effects of acute EtOH exposure on cellular adhesion and structure.
Collapse
|
93
|
Mu D, Sun YG. Circuit Mechanisms of Itch in the Brain. J Invest Dermatol 2021; 142:23-30. [PMID: 34662562 DOI: 10.1016/j.jid.2021.09.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/21/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022]
Abstract
Itch is an unpleasant somatic sensation with the desire to scratch, and it consists of sensory, affective, and motivational components. Acute itch serves as a critical protective mechanism because an itch-evoked scratching response will help to remove harmful substances invading the skin. Recently, exciting progress has been made in deciphering the mechanisms of itch at both the peripheral nervous system and the CNS levels. Key neuronal subtypes and circuits have been revealed for ascending transmission and the descending modulation of itch. In this review, we mainly summarize the current understanding of the central circuit mechanisms of itch in the brain.
Collapse
Affiliation(s)
- Di Mu
- Department of Anesthesiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Gang Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China.
| |
Collapse
|
94
|
Nicola FDC, Hua I, Levine AJ. Intersectional genetic tools to study skilled reaching in mice. Exp Neurol 2021; 347:113879. [PMID: 34597682 DOI: 10.1016/j.expneurol.2021.113879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/10/2021] [Accepted: 09/24/2021] [Indexed: 11/25/2022]
Abstract
Reaching to grasp is an evolutionarily conserved behavior and a crucial part of the motor repertoire in mammals. As it is studied in the laboratory, reaching has become the prototypical example of dexterous forelimb movements, illuminating key principles of motor control throughout the spinal cord, brain, and peripheral nervous system. Here, we (1) review the motor elements or phases that comprise the reach, grasp, and retract movements of reaching behavior, (2) highlight the role of intersectional genetic tools in linking these movements to their neuronal substrates, (3) describe spinal cord cell types and their roles in skilled reaching, and (4) how descending pathways from the brain and the sensory systems contribute to skilled reaching. We emphasize that genetic perturbation experiments can pin-point the neuronal substrates of specific phases of reaching behavior.
Collapse
Affiliation(s)
- Fabricio do Couto Nicola
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Isabelle Hua
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Ariel J Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States of America.
| |
Collapse
|
95
|
Ran C, Kamalani GNA, Chen X. Modality-Specific Modulation of Temperature Representations in the Spinal Cord after Injury. J Neurosci 2021; 41:8210-8219. [PMID: 34408066 PMCID: PMC8482863 DOI: 10.1523/jneurosci.1104-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022] Open
Abstract
Different types of tissue injury, such as inflammatory and neuropathic conditions, cause modality-specific alternations on temperature perception. There are profound changes in peripheral sensory neurons after injury, but how patterned neuronal activities in the CNS encode injury-induced sensitization to temperature stimuli is largely unknown. Using in vivo calcium imaging and mouse genetics, we show that formalin- and prostaglandin E2-induced inflammation dramatically increase spinal responses to heating and decrease responses to cooling in male and female mice. The reduction of cold response is largely eliminated on ablation of TRPV1-expressing primary sensory neurons, indicating a crossover inhibition of cold response from the hyperactive heat inputs in the spinal cord. Interestingly, chemotherapy medication oxaliplatin can rapidly increase spinal responses to cooling and suppress responses to heating. Together, our results suggest a push-pull mechanism in processing cold and heat inputs and reveal a synergic mechanism to shift thermosensation after injury.SIGNIFICANCE STATEMENT In this paper, we combine our novel in vivo spinal cord two-photon calcium imaging, mouse genetics, and persistent pain models to study how tissue injury alters the sensation of temperature. We discover modality-specific changes of spinal temperature responses in different models of injury. Chemotherapy medication oxaliplatin leads to cold hypersensitivity and heat hyposensitivity. By contrast, inflammation increases heat sensitivity and decreases cold sensitivity. This decrease in cold sensitivity results from the stronger crossover inhibition from the hyperactive heat inputs. Our work reveals the bidirectional change of thermosensitivity by injury and suggests that the crossover inhibitory circuit underlies the shifted thermosensation, providing a mechanism to the biased perception toward a unique thermal modality that was observed clinically in chronic pain patients.
Collapse
Affiliation(s)
- Chen Ran
- Department of Biology, Stanford University, Stanford, California 94305
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | | | - Xiaoke Chen
- Department of Biology, Stanford University, Stanford, California 94305
| |
Collapse
|
96
|
Russ DE, Cross RBP, Li L, Koch SC, Matson KJE, Yadav A, Alkaslasi MR, Lee DI, Le Pichon CE, Menon V, Levine AJ. A harmonized atlas of mouse spinal cord cell types and their spatial organization. Nat Commun 2021; 12:5722. [PMID: 34588430 PMCID: PMC8481483 DOI: 10.1038/s41467-021-25125-1] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Single-cell RNA sequencing data can unveil the molecular diversity of cell types. Cell type atlases of the mouse spinal cord have been published in recent years but have not been integrated together. Here, we generate an atlas of spinal cell types based on single-cell transcriptomic data, unifying the available datasets into a common reference framework. We report a hierarchical structure of postnatal cell type relationships, with location providing the highest level of organization, then neurotransmitter status, family, and finally, dozens of refined populations. We validate a combinatorial marker code for each neuronal cell type and map their spatial distributions in the adult spinal cord. We also show complex lineage relationships among postnatal cell types. Additionally, we develop an open-source cell type classifier, SeqSeek, to facilitate the standardization of cell type identification. This work provides an integrated view of spinal cell types, their gene expression signatures, and their molecular organization.
Collapse
Affiliation(s)
- Daniel E Russ
- Division of Cancer Epidemiology and Genetics, Data Science Research Group, National Cancer Institute, NIH, Rockville, MD, USA
| | - Ryan B Patterson Cross
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Li Li
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Stephanie C Koch
- Department of Neuroscience, Physiology and Pharmacology, Division of Biosciences, University College London, London, UK
| | - Kaya J E Matson
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Archana Yadav
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Mor R Alkaslasi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA.,Department of Neuroscience, Brown University, Providence, RI, USA
| | - Dylan I Lee
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Claire E Le Pichon
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Vilas Menon
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Ariel J Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA.
| |
Collapse
|
97
|
Schwaner MJ, Hsieh ST, Braasch I, Bradley S, Campos CB, Collins CE, Donatelli CM, Fish FE, Fitch OE, Flammang BE, Jackson BE, Jusufi A, Mekdara PJ, Patel A, Swalla BJ, Vickaryous M, McGowan CP. Future Tail Tales: A Forward-Looking, Integrative Perspective on Tail Research. Integr Comp Biol 2021; 61:521-537. [PMID: 33999184 PMCID: PMC8680820 DOI: 10.1093/icb/icab082] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Synopsis Tails are a defining characteristic of chordates and show enormous diversity in function and shape. Although chordate tails share a common evolutionary and genetic-developmental origin, tails are extremely versatile in morphology and function. For example, tails can be short or long, thin or thick, and feathered or spiked, and they can be used for propulsion, communication, or balancing, and they mediate in predator-prey outcomes. Depending on the species of animal the tail is attached to, it can have extraordinarily multi-functional purposes. Despite its morphological diversity and broad functional roles, tails have not received similar scientific attention as, for example, the paired appendages such as legs or fins. This forward-looking review article is a first step toward interdisciplinary scientific synthesis in tail research. We discuss the importance of tail research in relation to five topics: (1) evolution and development, (2) regeneration, (3) functional morphology, (4) sensorimotor control, and (5) computational and physical models. Within each of these areas, we highlight areas of research and combinations of long-standing and new experimental approaches to move the field of tail research forward. To best advance a holistic understanding of tail evolution and function, it is imperative to embrace an interdisciplinary approach, re-integrating traditionally siloed fields around discussions on tail-related research.
Collapse
Affiliation(s)
- M J Schwaner
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, CA 92697, USA
| | - S T Hsieh
- Department of Biology, Temple University, Philadelphia, PA 19122, USA
| | - I Braasch
- Department of Integrative Biology and Program in Ecology, Evolution, and Behavior (EEB), Michigan State University, East Lansing, MI 48824, USA
| | - S Bradley
- Department of Biomedical Science, University of Guelph, Guelph N1G 2W1, Canada
| | - C B Campos
- Department of Biological Sciences, Sacramento State University, Sacramento, CA 95819, USA
| | - C E Collins
- Department of Biological Sciences, Sacramento State University, Sacramento, CA 95819, USA
| | - C M Donatelli
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - F E Fish
- Department of Biology, West Chester University, West Chester, PA 19383, USA
| | - O E Fitch
- Department of Integrative Biology and Program in Ecology, Evolution, and Behavior (EEB), Michigan State University, East Lansing, MI 48824, USA
| | - B E Flammang
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - B E Jackson
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA 23909, USA
| | - A Jusufi
- Max Planck Institute for Intelligent Systems, Stuttgart 70569, Germany
| | - P J Mekdara
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - A Patel
- Department of Electrical Engineering, University of Cape Town, Cape Town 7701, South Africa
| | - B J Swalla
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - M Vickaryous
- Department of Biomedical Science, University of Guelph, Guelph N1G 2W1, Canada
| | - C P McGowan
- Department of Integrative Anatomical Sciences, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
98
|
Miranda CO, Hegedüs K, Wildner H, Zeilhofer HU, Antal M. Morphological and neurochemical characterization of glycinergic neurons in laminae I-IV of the mouse spinal dorsal horn. J Comp Neurol 2021; 530:607-626. [PMID: 34382691 DOI: 10.1002/cne.25232] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022]
Abstract
A growing body of experimental evidence shows that glycinergic inhibition plays vital roles in spinal pain processing. In spite of this, however, our knowledge about the morphology, neurochemical characteristics, and synaptic relations of glycinergic neurons in the spinal dorsal horn is very limited. The lack of this knowledge makes our understanding about the specific contribution of glycinergic neurons to spinal pain processing quite vague. Here we investigated the morphology and neurochemical characteristics of glycinergic neurons in laminae I-IV of the spinal dorsal horn using a GlyT2::CreERT2-tdTomato transgenic mouse line. Confirming previous reports, we show that glycinergic neurons are sparsely distributed in laminae I-II, but their densities are much higher in lamina III and especially in lamina IV. First in the literature, we provide experimental evidence indicating that in addition to neurons in which glycine colocalizes with GABA, there are glycinergic neurons in laminae I-II that do not express GABA and can thus be referred to as glycine-only neurons. According to the shape and size of cell bodies and dendritic morphology, we divided the tdTomato-labeled glycinergic neurons into three and six morphological groups in laminae I-II and laminae III-IV, respectively. We also demonstrate that most of the glycinergic neurons co-express neuronal nitric oxide synthase, parvalbumin, the receptor tyrosine kinase RET, and the retinoic acid-related orphan nuclear receptor β (RORβ), but there might be others that need further neurochemical characterization. The present findings may foster our understanding about the contribution of glycinergic inhibition to spinal pain processing.
Collapse
Affiliation(s)
- Camila Oliveira Miranda
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Krisztina Hegedüs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Hendrik Wildner
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
99
|
Costa YM, Bonjardim LR, Conti PCR, Svensson P. Psychophysical evaluation of somatosensory function in oro-facial pain: achievements and challenges. J Oral Rehabil 2021; 48:1066-1076. [PMID: 34213796 DOI: 10.1111/joor.13223] [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: 04/28/2021] [Accepted: 06/03/2021] [Indexed: 11/30/2022]
Abstract
AIM This critical review describes key methodological aspects for a successful oro-facial psychophysical evaluation of the somatosensory system and highlights the diagnostic value of somatosensory assessment and management perspectives based on somatosensory profiling. METHODS This topical review was based on a non-systematic search for studies about somatosensory evaluation in oro-facial pain in PubMed and Embase. RESULTS The recent progress regarding the psychophysical evaluation of somatosensory function was largely possible due to the development and application of valid, reliable and standardised psychophysical methods. Qualitative sensory testing may be useful as a screening tool to rule out relevant somatosensory abnormalities. Nevertheless, the patient should preferably be referred to a more comprehensive assessment with the quantitative sensory testing battery if confirmation of somatosensory abnormalities is necessary. Moreover, the identification of relevant somatosensory alterations in chronic pain disorders that do not fulfil the current criteria to be regarded as neuropathic has also increased the usefulness of somatosensory evaluation as a feasible method to better characterise the patients and perhaps elucidate some underpinnings of the so-called 'nociplastic' pain disorders. Finally, an additional benefit of oro-facial pain treatment based on somatosensory profiling still needs to be demonstrated and convincing evidence of somatosensory findings as predictors of treatment efficacy in chronic oro-facial pain awaits further studies. CONCLUSION Psychophysical evaluation of somatosensory function in oro-facial pain is still in its infancy but with a clear potential to continue to improve the assessment, diagnosis and management of oro-facial pain patients.
Collapse
Affiliation(s)
- Yuri M Costa
- Department of Biosciences, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil.,Section for Orofacial Pain and Jaw Function, Department of Dentistry and Oral Health, Aarhus University, Aarhus, Denmark.,Scandinavian Center for Orofacial Neurosciences (SCON), Aarhus, Denmark.,Bauru Orofacial Pain Group, Bauru, Brazil
| | - Leonardo R Bonjardim
- Bauru Orofacial Pain Group, Bauru, Brazil.,Section of Head and Face Physiology, Department of Biological Sciences, Bauru School of Dentistry, University of Sao Paulo, Bauru, Brazil
| | - Paulo César R Conti
- Bauru Orofacial Pain Group, Bauru, Brazil.,Department of Prosthodontics, Bauru School of Dentistry, University of Sao Paulo, Bauru, Brazil
| | - Peter Svensson
- Section for Orofacial Pain and Jaw Function, Department of Dentistry and Oral Health, Aarhus University, Aarhus, Denmark.,Scandinavian Center for Orofacial Neurosciences (SCON), Aarhus, Denmark.,Faculty of Odontology, Malmo University, Malmo, Sweden
| |
Collapse
|
100
|
Wercberger R, Braz JM, Weinrich JA, Basbaum AI. Pain and itch processing by subpopulations of molecularly diverse spinal and trigeminal projection neurons. Proc Natl Acad Sci U S A 2021; 118:e2105732118. [PMID: 34234018 PMCID: PMC8285968 DOI: 10.1073/pnas.2105732118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
A remarkable molecular and functional heterogeneity of the primary sensory neurons and dorsal horn interneurons transmits pain- and or itch-relevant information, but the molecular signature of the projection neurons that convey the messages to the brain is unclear. Here, using retro-TRAP (translating ribosome affinity purification) and RNA sequencing, we reveal extensive molecular diversity of spino- and trigeminoparabrachial projection neurons. Among the many genes identified, we highlight distinct subsets of Cck+ -, Nptx2+ -, Nmb+ -, and Crh+ -expressing projection neurons. By combining in situ hybridization of retrogradely labeled neurons with Fos-based assays, we also demonstrate significant functional heterogeneity, including both convergence and segregation of pain- and itch-provoking inputs into molecularly diverse subsets of NK1R- and non-NK1R-expressing projection neurons.
Collapse
Affiliation(s)
- Racheli Wercberger
- Department of Anatomy, University of California, San Francisco, CA 94158
| | - Joao M Braz
- Department of Anatomy, University of California, San Francisco, CA 94158
| | - Jarret A Weinrich
- Department of Anatomy, University of California, San Francisco, CA 94158
| | - Allan I Basbaum
- Department of Anatomy, University of California, San Francisco, CA 94158
| |
Collapse
|