1
|
da Silva AM, de Amorim Ferreira M, Schran RG, Lückemeyer DD, Prudente AS, Ferreira J. Investigation of the participation of the TRPV1 receptor in the irritant effect of dithranol in mice. Eur J Pharmacol 2025; 994:177291. [PMID: 39870229 DOI: 10.1016/j.ejphar.2025.177291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/19/2024] [Accepted: 01/21/2025] [Indexed: 01/29/2025]
Abstract
Dithranol is one of the most effective topical medications for treating plaque psoriasis. However, its clinical use is limited by irritative adverse reactions to the skin, such as oedema, erythema, and pruritus, caused by poorly understood mechanisms. Because TRPV1 activation mediates skin irritation caused by several drugs, we conducted blind and randomised experiments in male and female C57BL/6 mice to elucidate the role of TRPV1 in dithranol-induced irritation. Dithranol (0.01%-0.5%) or vehicle was applied topically to the right ear of the animals. Oedema, erythema, and pruritus were monitored from 2 h to 6 days after application. Treatment with 0.5% dithranol caused oedema and erythema, but not pruritus, starting at 6 h, reaching its highest point at 1 day, and persisting up to 6 days after treatment, mainly in male mice. The 0.1% dose induced erythema but not oedema. Interestingly, topical application of 1% capsaicin was shown to defunctionalise TRPV1-positive fibres and did not influence early irritation caused by dithranol (2 h-2 days). However, it increased the late phase of irritation (5-6 days). Similarly, salicylate did not reduce the early irritation caused by dithranol but also increased the late phase. Antagonism by SB366791 and 4-tert-butylcyclohexanol did not alter skin irritation. Our results suggest that, contrary to our initial hypothesis, TRPV1 appears to act protectively against skin irritation caused by dithranol, particularly in the late stage.
Collapse
Affiliation(s)
- Ana Merian da Silva
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil
| | - Marcella de Amorim Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil
| | - Roberta Giusti Schran
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil
| | - Debora Denardin Lückemeyer
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil; Pain Research Center, Department of Anesthesiology, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Arthur Silveira Prudente
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil; Pain Research Center, Department of Anesthesiology, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Juliano Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88037-000, Florianópolis, SC, Brazil.
| |
Collapse
|
2
|
Hor CC, Duan B. Lateral parabrachial nucleus: the commander-in-chief for nocifensive behavior expression in cold allodynia. Pain 2025; 166:965-966. [PMID: 39715171 DOI: 10.1097/j.pain.0000000000003469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 12/25/2024]
Affiliation(s)
- Chia Chun Hor
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.
| | | |
Collapse
|
3
|
Reddy P, Narayan Prajapati J, Chaterji S, Varughese A, Chaudhary Y, Sathyamurthy A, Barik A. Converging inputs compete at the lateral parabrachial nuclei to dictate the affective-motivational responses to cold pain. Pain 2025; 166:1105-1117. [PMID: 39715193 DOI: 10.1097/j.pain.0000000000003468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/05/2024] [Indexed: 12/25/2024]
Abstract
ABSTRACT The neural mechanisms of the affective-motivational symptoms of chronic pain are poorly understood. In chronic pain, our innate coping mechanisms fail to provide relief. Hence, these behaviors are manifested at higher frequencies. In laboratory animals, such as mice and rats, licking the affected areas is a behavioral coping mechanism and it is sensitized in chronic pain. Hence, we have focused on delineating the brain circuits mediating licking in mice with chemotherapy-induced peripheral neuropathy (CIPN). Mice with CIPN develop intense cold hypersensitivity and lick their paws upon contact with cold stimuli. We studied how the lateral parabrachial nucleus (LPBN) neurons facilitate licking behavior when mice are exposed to noxious thermal stimuli. Taking advantage of transsynaptic viral, optogenetic, and chemogenetic strategies, we observed that the LPBN neurons become hypersensitive to cold in mice with CIPN and facilitate licks. Furthermore, we found that the expression of licks depends on competing excitatory and inhibitory inputs from the spinal cord and lateral hypothalamus (LHA), respectively. We anatomically traced the postsynaptic targets of the spinal cord and LHA in the LPBN and found that they synapse onto overlapping populations. Activation of this LPBN population was sufficient to promote licking due to cold allodynia. In sum, our data indicate that the nociceptive inputs from the spinal cord and information on brain states from the hypothalamus impinge on overlapping LPBN populations to modulate their activity and, in turn, regulate the elevated affective-motivational responses in CIPN.
Collapse
Affiliation(s)
- Prannay Reddy
- Center for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka, India
| | | | | | | | | | | | | |
Collapse
|
4
|
Tezanos P, Trejo JL. Why are threatening experiences remembered so well? Insights into memory strengthening from protocols of gradual aversive learning. Neurosci Biobehav Rev 2025:106145. [PMID: 40250543 DOI: 10.1016/j.neubiorev.2025.106145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Aversive experiences often result in strong and persistent memory traces, which can sometimes lead to conditions such as Post-Traumatic Stress Disorder or phobias. Aversive stimulation tests are key tools in psychology and neuroscience for studying learning and memory. These tests typically use electric shocks as the unconditioned stimulus, allowing for precise control over the aversive content of the learning event. This feature has led to extensive research applying these tests with varying shock intensities to examine differences in learning, behavior, and memory formation between low- and high-aversive experiences. This line of research is particularly valuable for understanding the neurobiology underlying memory strengthening, but, to our knowledge, no review has yet compiled and organized the findings from this specific methodology. In this comprehensive review, we focus primarily on animal studies that have employed the same aversive test (i.e. Fear Conditioning, Passive Avoidance, Active Avoidance or Operant boxes) at different intensities. We will first outline and briefly describe the main aversive learning paradigms used in this field. Next, we will examine the relationship between aversiveness and memory strength. Finally, we will explore the neurobiological insights these studies have revealed over the years. Our aim is to gain a better understanding of how the nervous system gradually strengthens memory, while also addressing the remaining gaps and challenges in this area of research.
Collapse
Affiliation(s)
- P Tezanos
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain; PhD Program in Neuroscience, Universidad Autónoma de Madrid-Instituto Cajal, Madrid 28002, Spain
| | - J L Trejo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain.
| |
Collapse
|
5
|
Ding WQ, Song W, Shi X, Feng Z, Chen X, Xie T, Liu Y, Zhou J, Chen Y, Lin JK, Wang QM, Zhou H, Liang TY, Jiang T, Ren B, Yao H, Li YQ, Evrard HC, Poo MM, Li H, Li X, Gong H, Todd AJ, Li A, Wang X, Deng J, Sun YG. Single-neuron projectome reveals organization of somatosensory ascending pathways in the mouse brain. Neuron 2025:S0896-6273(25)00179-5. [PMID: 40209714 DOI: 10.1016/j.neuron.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/08/2024] [Accepted: 03/03/2025] [Indexed: 04/12/2025]
Abstract
Relay of multimodal somatosensory information from the spinal cord to the brain is critical for sensory perception, but the underlying circuit organization remains unclear. We have reconstructed mouse cervical spinal projection neurons at single-cell resolution and identified 19 projectome-defined subtypes exhibiting diverse projection patterns. We also reconstructed the brain-wide axonal projections of central relay neurons that receive direct spinal inputs at the single-cell resolution. We discovered parallel, divergent, and convergent projection patterns for spinal projection neurons and central relay neurons. Our results revealed the diverse pathways channeling spinal information to the cortex. Furthermore, we identified parallel lateral and medial spinal-superior colliculus-brainstem pathways, which could be involved in orienting and defensive behaviors, respectively. These data allowed us to construct a wiring diagram for ascending somatosensory pathways with projectome-defined subtype resolution. Our single-cell projectome analysis provided a new framework for understanding the complex neural circuitry underlying coordinated processing of diverse somatosensory modalities.
Collapse
Affiliation(s)
- Wen-Qun Ding
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Song
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoxue Shi
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhao Feng
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Xu Chen
- Lingang Laboratory, Shanghai 200031, China
| | - Taorong Xie
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiandong Zhou
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yu Chen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun-Kai Lin
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Qiu-Miao Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Hua Zhou
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tong-Yu Liang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Jiang
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Biyu Ren
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Haishan Yao
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, the Fourth Military Medical University, Xi'an 710032, China
| | - Henry C Evrard
- International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence, Institute of Neuroscience, Chinese Academy of Sciences, Songjiang, Shanghai, China; Werner Reichardt Center for Integrative Neuroscience, Karl Eberhard University of Tübingen, Tübingen, Germany; Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Mu-Ming Poo
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, the Fourth Military Medical University, Xi'an 710032, China
| | - Xiangning Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China
| | - Hui Gong
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China; Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Andrew J Todd
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Anan Li
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China; Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China.
| | - Xiaofei Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Yan-Gang Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
6
|
Zhang MD, Kupari J, Su J, Magnusson KA, Hu Y, Calvo-Enrique L, Usoskin D, Albisetti GW, Ceder MM, Henriksson K, Leavitt AD, Zeilhofer HU, Hökfelt T, Lagerström MC, Ernfors P. Neural ensembles that encode nocifensive mechanical and heat pain in mouse spinal cord. Nat Neurosci 2025:10.1038/s41593-025-01921-6. [PMID: 40128392 DOI: 10.1038/s41593-025-01921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/07/2025] [Indexed: 03/26/2025]
Abstract
Acute pain is an unpleasant experience caused by noxious stimuli. How the spinal neural circuits attribute differences in quality of noxious information remains unknown. By means of genetic capturing, activity manipulation and single-cell RNA sequencing, we identified distinct neural ensembles in the adult mouse spinal cord encoding mechanical and heat pain. Reactivation or silencing of these ensembles potentiated or stopped, respectively, paw shaking, lifting and licking within but not across the stimuli modalities. Within ensembles, polymodal Gal+ inhibitory neurons with monosynaptic contacts to A-fiber sensory neurons gated pain transmission independent of modality. Peripheral nerve injury led to inferred microglia-driven inflammation and an ensemble transition with decreased recruitment of Gal+ inhibitory neurons and increased excitatory drive. Forced activation of Gal+ neurons reversed hypersensitivity associated with neuropathy. Our results reveal the existence of a spinal representation that forms the neural basis of the discriminative and defensive qualities of acute pain, and these neurons are under the control of a shared feed-forward inhibition.
Collapse
Affiliation(s)
- Ming-Dong Zhang
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Jussi Kupari
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Jie Su
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Kajsa A Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Yizhou Hu
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Laura Calvo-Enrique
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain
| | - Dmitry Usoskin
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Gioele W Albisetti
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Mikaela M Ceder
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Katharina Henriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Andrew D Leavitt
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Malin C Lagerström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrik Ernfors
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
7
|
Ma W, Polgár E, Dickie AC, Hajer MA, Quillet R, Gutierrez-Mecinas M, Yadav M, Hachisuka J, Todd AJ, Bell AM. Anatomical characterisation of somatostatin-expressing neurons belonging to the anterolateral system. Sci Rep 2025; 15:9549. [PMID: 40108302 PMCID: PMC11923155 DOI: 10.1038/s41598-025-93816-6] [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: 11/21/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Anterolateral system (ALS) spinal projection neurons are essential for pain perception. However, these cells are heterogeneous, and there has been extensive debate about the roles of ALS populations in the different pain dimensions. We recently performed single-nucleus RNA sequencing on a developmentally-defined subset of ALS neurons, and identified 5 transcriptomic populations. One of these, ALS4, consists of cells that express Sst, the gene coding for somatostatin, and we reported that these were located in the lateral part of lamina V. Here we use a SstCre mouse line to characterise these cells and define their axonal projections. We find that their axons ascend mainly on the ipsilateral side, giving off collaterals throughout their course in the spinal cord. They target various brainstem nuclei, including the parabrachial internal lateral nucleus, and the posterior triangular and medial dorsal thalamic nuclei. We also show that in the L4 segment Sst is expressed by ~ 75% of ALS neurons in lateral lamina V and that there are around 120 Sst-positive lateral lamina V cells on each side. Our findings indicate that this is a relatively large population, and based on projection targets we conclude that they are likely to contribute to the affective-motivational dimension of pain.
Collapse
Affiliation(s)
- Wenhui Ma
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Erika Polgár
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Allen C Dickie
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Mai Abu Hajer
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Raphaëlle Quillet
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Maria Gutierrez-Mecinas
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Mansi Yadav
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Junichi Hachisuka
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Andrew J Todd
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK.
| | - Andrew M Bell
- School of Psychology and Neuroscience, Sir James Black Building, University of Glasgow, Glasgow, G12 8QQ, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
8
|
MacDonald DI, Jayabalan M, Seaman JT, Balaji R, Nickolls AR, Chesler AT. Pain persists in mice lacking both Substance P and CGRPα signaling. eLife 2025; 13:RP93754. [PMID: 40100256 PMCID: PMC11919252 DOI: 10.7554/elife.93754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
The neuropeptides Substance P and CGRPα have long been thought important for pain sensation. Both peptides and their receptors are expressed at high levels in pain-responsive neurons from the periphery to the brain making them attractive therapeutic targets. However, drugs targeting these pathways individually did not relieve pain in clinical trials. Since Substance P and CGRPα are extensively co-expressed, we hypothesized that their simultaneous inhibition would be required for effective analgesia. We therefore generated Tac1 and Calca double knockout (DKO) mice and assessed their behavior using a wide range of pain-relevant assays. As expected, Substance P and CGRPα peptides were undetectable throughout the nervous system of DKO mice. To our surprise, these animals displayed largely intact responses to mechanical, thermal, chemical, and visceral pain stimuli, as well as itch. Moreover, chronic inflammatory pain and neurogenic inflammation were unaffected by loss of the two peptides. Finally, neuropathic pain evoked by nerve injury or chemotherapy treatment was also preserved in peptide-deficient mice. Thus, our results demonstrate that even in combination, Substance P and CGRPα are not required for the transmission of acute and chronic pain.
Collapse
Affiliation(s)
- Donald Iain MacDonald
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Monessha Jayabalan
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Jonathan T Seaman
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Rakshita Balaji
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Alec R Nickolls
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Alexander Theodore Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| |
Collapse
|
9
|
Rigney N, Hong W. Prosocial Helping Behavior: Conceptual Issues and Neural Mechanisms. Biol Psychiatry 2025:S0006-3223(25)01049-2. [PMID: 40090565 DOI: 10.1016/j.biopsych.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
Prosocial helping behavior, characterized by voluntary actions taken to benefit others, plays a vital role in promoting cooperation and maintaining social bonds across human and animal social groups. In this review, we examine key conceptual issues surrounding prosocial behavior, focusing specifically on targeted helping and comforting actions. We outline the behavioral paradigms used to study these two types of prosocial behaviors and summarize recent insights into their underlying neural mechanisms. Drawing on findings across species and with an emphasis on rodent models, we discuss how these behaviors are regulated by molecularly and anatomically defined neural systems and how distinct neuronal populations and circuits may differentially regulate targeted helping and comforting behaviors. Lastly, we discuss the clinical relevance of this research by addressing the implications of prosocial deficits in psychiatric disorders.
Collapse
Affiliation(s)
- Nicole Rigney
- Department of Neurobiology and Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Weizhe Hong
- Department of Neurobiology and Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California.
| |
Collapse
|
10
|
Gu H, Zhao F, Liu Z, Cao P. Defense or death? A review of the neural mechanisms underlying sensory modality-triggered innate defensive behaviors. Curr Opin Neurobiol 2025; 92:102977. [PMID: 40015135 DOI: 10.1016/j.conb.2025.102977] [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: 10/30/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 03/01/2025]
Abstract
Defense or death presents a canonical dilemma for animals when encountering predators. Threatening sensory cues provide essential information that signals predator presence, driving the evolution of a spectrum of defensive behaviors. In rodents, these behaviors, as described by the classic "predatory imminence continuum" model, range from risk assessment and freezing to rapid escape responses. During the pre-encounter phase, risk assessment and avoidance responses are crucial for monitoring the environment with vigilance and cautiousness. Once detected during the post-encounter phase or physically attacked during the circa-strike phase, multiple sensory systems are rapidly activated, triggering escape responses to increase the distance from the threat. Although there are species-specific variations, the brain regions underpinning these defensive strategies, including the thalamus, hypothalamus, and midbrain, are evolutionarily conserved. This review aims to provide a comprehensive overview of the universal innate defensive circuit framework to enrich our understanding of how animals respond to life-threatening situations.
Collapse
Affiliation(s)
- Huating Gu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Feiran Zhao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Zhihui Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Cao
- National Institute of Biological Sciences, Beijing 102206, China.
| |
Collapse
|
11
|
Malewicz-Oeck NM, Skorupka N, Bartholmes F, Dombrowski A, Ebel M, Zahn PK, Meyer-Frießem CH. Pupillary dilation to monitor nociception in awake volunteers: A stimuli-randomised placebo-controlled study. Eur J Anaesthesiol 2025:00003643-990000000-00275. [PMID: 39995234 DOI: 10.1097/eja.0000000000002143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/26/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Pupillary reflex dilation (PRD) quantifies nociception in anaesthetised patients, enabling tailored opioid administration, which in turn reduces catecholamine levels and postoperative pain intensity. However, its utility in objectively assessing pain in awake individuals remains challenging. OBJECTIVE To investigate whether PRD can differentiate between painful and nonpainful stimuli in awake volunteers. DESIGN This was a randomised, placebo-controlled, stimuli-randomised study conducted after ethical approval and registration (DRKS00024791). SETTING This single-centre study was performed at BG University Hospital Bergmannsheil Bochum, Germany, between November 2021 and January 2022. VOLUNTEERS Thirty healthy volunteers (25 ± 2 years, 50% male) were included in the study. INTERVENTIONS After a rest, the following were stimuli applied to one ventral forearm: an unannounced electric pain stimulus (UPS) and a randomised sequence of either an announced painful stimulus (APS), a placebo or a nonpainful stimulus (NPS). MAIN OUTCOME MEASURES Pupil dilatation was measured as PRD (%) for 60 s during the experimental condition "rest", and during and after each stimulus application using an AlgiScan device. The participants rated stimulus pain intensity via a numeric rating scale (NRS: 0 = no pain, 10 = most intense pain imaginable). Statistics: Paired t-test, rmANOVA, Spearman's correlation and receiver operating characteristics (ROC), P < 0.05. RESULTS The subjective pain intensity ratings were higher after APS (6.0 ± 1.9) than after UPS (5.5 ± 1.7, P = 0.007), placebo (0.0 ± 0.0, P = 0.027) and NPS (0.0 ± 0.0, P = 0.001). Similarly, objective pupillary reaction to the stimuli measured using PRD was higher for APS: 13 (97.6% CI, 10.0 to 19.0)% vs. NPS 13 (97.6% CI, 7.0 to 20.0), P = 0.024). UPS elicited the highest PRD of 25 (95.7% CI, 18.0 to 30.0)% vs. rest, P < 0.001; significantly greater than placebo at 13.5 (96.4% CI, 10.0 to 22.0)%, P < 0.001); and NPS at 13 (97.6% CI, 7.0 to 20.0)%, P < 0.0001). However, no significant differences in PRD were observed between APS and UPS despite their electrical similarity. PRD correlated with pain intensity (r = 0.35, P < 0.0001). CONCLUSIONS In awake volunteers, PRD differentiates between painful and nonpainful stimuli and correlates with pain intensity. Noninvasive PRD measurement may be suitable for nociception monitoring in awake individuals. TRIAL REGISTRATION Prospectively DRKS00024791, March 2021.
Collapse
Affiliation(s)
- Nathalie M Malewicz-Oeck
- From the Department of Anaesthesiology, Intensive Care Medicine and Pain Medicine, BG University Hospital Bergmannsheil gGmbH, Bürkle-de-la-Camp-Platz 1, Bochum (NMMO, NS, FB, AD, ME, PKZ, CHMF), the Department of Anaesthesiology, Intensive Care and Pain Medicine, KLW St. Paulus GmbH, St. Marien Hospital Lünen, Lünen, Germany (CHMF)
| | | | | | | | | | | | | |
Collapse
|
12
|
Huang JY, Jin YX, Dong WY, Zhao W, Cheng PK, Miao JH, Liu A, Wang D, Li J, Zhang Z, Tao W, Zhu X. Intra-somatosensory cortical circuits mediating pain-induced analgesia. Nat Commun 2025; 16:1859. [PMID: 39984470 PMCID: PMC11845469 DOI: 10.1038/s41467-025-57050-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 02/05/2025] [Indexed: 02/23/2025] Open
Abstract
Pain in one part of the body profoundly diminishes the sensation of pain in other parts of the body in humans. Here, we found that pain-related behaviors in hindpaw are inhibited by noxious stimuli from diverse body regions in mice. Using activity-dependent cell labeling in male FosTRAP2 mice, we captured a neuronal ensemble in the layers 2-4 of secondary somatosensory cortex (S2) that was activated during pain at diverse body regions induced analgesia. Single-cell projection analysis showed that these S2 neurons receive projections from the contralateral S2 and specifically innervate the layer 4 of primary somatosensory cortex (S1). Microendoscopic calcium imaging and chemogenetic manipulation in freely moving mice showed that this S2 → S1 feedforward inhibitory circuit mediates ipsilateral pain-induced analgesia, whereas contralateral S2 innervation of the S2 → S1 circuit mediates contralateral pain-induced analgesia. Our study defines the intra-somatosensory cortical circuits underlying "pain inhibiting pain", expanding the scope of known circuit mechanisms involved in pain relief.
Collapse
Affiliation(s)
- Ji-Ye Huang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Yu-Xin Jin
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Wan-Ying Dong
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Wan Zhao
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of University of Science and Technique of China, Hefei, PR China
| | - Ping-Kai Cheng
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Jun-Hao Miao
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - An Liu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, PR China
| | - Di Wang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Juan Li
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Zhi Zhang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China.
- Department of Biophysics and Neurobiology, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, PR China.
| | - Wenjuan Tao
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, PR China.
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, PR China.
| | - Xia Zhu
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China.
| |
Collapse
|
13
|
Li Y, Ha NT, Li J, Yan Y, Chen Q, Cai L, Li W, Liu S, Li B, Cheng T, Sun Y, Wang Y, Deng J. Tachykinin signaling in the right parabrachial nucleus mediates early-phase neuropathic pain development. Neuron 2025; 113:605-619.e6. [PMID: 39719702 DOI: 10.1016/j.neuron.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/18/2024] [Accepted: 11/27/2024] [Indexed: 12/26/2024]
Abstract
The lateral parabrachial nucleus (PBN) is critically involved in neuropathic pain modulation. However, the cellular and molecular mechanisms underlying this process remain largely unknown. Here, we report that in mice, the right-sided, but not the left-sided, PBN plays an essential role in the development of hyperalgesia following nerve injury, irrespective of the injury side. Spino-parabrachial pathways targeting the right-sided PBN display short-term facilitation, and right-sided PBN neurons exhibit an increase in the excitability and activity after nerve injury. Inhibiting Tacr1-positive neurons, blocking Tacr1-encoding tachykinin 1 receptor (NK1R), or knocking down the Tacr1 gene in the right-sided, rather than left-sided, PBN alleviates neuropathic pain-induced sensory hypersensitivity. Additionally, the right-sided PBN plays a critical role in the development of hyperalgesia during the early phase of neuropathic pain. These results highlight the essential role of NK1R in the lateralized modulation of neuropathic pain by the PBN, providing new insights into the mechanisms underlying neuropathic pain.
Collapse
Affiliation(s)
- Yinxia Li
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Ngoc T Ha
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Juan Li
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yaxin Yan
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Qian Chen
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Liping Cai
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weike Li
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Shoupei Liu
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Bo Li
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Tianlin Cheng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China; Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai 200032, China
| | - Yangang Sun
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
14
|
Wong C, Rodriguez-Hernandez LD, Lister KC, Gu N, Cai W, Hooshmandi M, Fan J, Brown N, Nguyen V, Ribeiro-da-Silva A, Bonin RP, Khoutorsky A. Targeting spinal mechanistic target of rapamycin complex 2 alleviates inflammatory and neuropathic pain. Brain 2025; 148:675-686. [PMID: 39167538 PMCID: PMC11788203 DOI: 10.1093/brain/awae275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/06/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
The development and maintenance of chronic pain involve the reorganization of spinal nocioceptive circuits. The mechanistic target of rapamycin complex 2 (mTORC2), a central signalling hub that modulates both actin-dependent structural changes and mechanistic target of rapamycin complex 1 (mTORC1)-dependent mRNA translation, plays key roles in hippocampal synaptic plasticity and memory formation. However, its function in spinal plasticity and chronic pain is poorly understood. Here, we show that pharmacological activation of spinal mTORC2 induces pain hypersensitivity, whereas its inhibition, using downregulation of the mTORC2-defining component Rictor, alleviates both inflammatory and neuropathic pain. Cell type-specific deletion of Rictor showed that the selective inhibition of mTORC2 in a subset of excitatory neurons impairs spinal synaptic potentiation and alleviates inflammation-induced mechanical and thermal hypersensitivity and nerve injury-induced heat hyperalgesia. The ablation of mTORC2 in inhibitory interneurons strongly alleviated nerve injury-induced mechanical hypersensitivity. Our findings reveal the role of mTORC2 in chronic pain and highlight its cell type-specific functions in mediating pain hypersensitivity in response to peripheral inflammation and nerve injury.
Collapse
Affiliation(s)
- Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Luis David Rodriguez-Hernandez
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Kevin C Lister
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Ning Gu
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Weihua Cai
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Mehdi Hooshmandi
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Jonathan Fan
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Nicole Brown
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Vivienne Nguyen
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, H3A 0C7, Canada
- Alan Edwards Center for the Research on Pain, McGill University, Montreal, QC, H3A 2B4 Canada
| | - Robert P Bonin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
- University of Toronto Centre for the Study of Pain, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC, H3G 1Y6, Canada
- Alan Edwards Center for the Research on Pain, McGill University, Montreal, QC, H3A 2B4 Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, H3G 1Y6, Canada
| |
Collapse
|
15
|
Kim SH, Lee J, Jang M, Roh SE, Kim S, Lee JH, Seo J, Baek J, Hwang JY, Baek IS, Lee YS, Shigetomi E, Lee CJ, Koizumi S, Kim SK, Kim SJ. Cerebellar Bergmann glia integrate noxious information and modulate nocifensive behaviors. Nat Neurosci 2025; 28:336-345. [PMID: 39748107 DOI: 10.1038/s41593-024-01807-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/27/2024] [Indexed: 01/04/2025]
Abstract
The cerebellum is activated by noxious stimuli and pathological pain but its role in noxious information processing remains unknown. Here, we show that in mice, cutaneous noxious electrical stimuli induced noradrenaline (NA) release from locus coeruleus (LC) terminals in the cerebellar cortex. Bergmann glia (BG) accumulated these LC-NA signals by increasing intracellular calcium in an integrative manner ('flares'). BG flares were also elicited in response to an intraplantar capsaicin injection. Chemogenetic inactivation of LC terminals or BG in the cerebellar cortex or BG-specific knockdown of α1-adrenergic receptors suppressed BG flares, reduced nocifensive licking and had analgesic effects in nerve injury-induced chronic neuropathic pain. Moreover, chemogenetic activation of BG or an intraplantar capsaicin injection reduced Purkinje cell firing, which may disinhibit the output activity of the deep cerebellar nuclei. These results suggest a role for BG in computing noxious information from the LC and in modulating pain-related behaviors by regulating cerebellar output.
Collapse
Affiliation(s)
- Seung Ha Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jaegeon Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Mirae Jang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Eon Roh
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Soobin Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Hwan Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Jewoo Seo
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jinhee Baek
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Yoon Hwang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - In Seon Baek
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Memory Network Medical Research Center, Neuroscience Research Institute, Wide River Institute of Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Korea
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Korea.
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea.
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
- Memory Network Medical Research Center, Neuroscience Research Institute, Wide River Institute of Immunology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
16
|
Asahina K, Zelikowsky M. Comparative Perspectives on Neuropeptide Function and Social Isolation. Biol Psychiatry 2025:S0006-3223(25)00061-7. [PMID: 39892690 DOI: 10.1016/j.biopsych.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/07/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
Chronic social isolation alters behavior across animal species. Genetic model organisms such as mice and flies provide crucial insight into the molecular and physiological effects of social isolation on brain cells and circuits. Here, we comparatively review recent findings regarding the function of conserved neuropeptides in social isolation in mice and flies. Analogous functions of 3 classes of neuropeptides-tachykinins, cholecystokinins, and neuropeptide Y/F-in the two model organisms suggest that these molecules may be involved in modulating behavioral changes induced by social isolation across a wider range of species, including humans. Comparative approaches armed with tools to dissect neuropeptidergic function can lead to an integrated understanding of the impacts of social isolation on brain circuits and behavior.
Collapse
Affiliation(s)
- Kenta Asahina
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California.
| | - Moriel Zelikowsky
- Department of Neurobiology, School of Medicine, The University of Utah, Salt Lake City, Utah
| |
Collapse
|
17
|
Hu Y, Feng Y, Luo H, Zhu XN, Chen S, Yang K, Deng Z, Luo M, Du W, Wang Q, Wang S, Wei K, Hu J, Wang Y. Dissociation-related behaviors in mice emerge from the inhibition of retrosplenial cortex parvalbumin interneurons. Cell Rep 2025; 44:115086. [PMID: 39708317 DOI: 10.1016/j.celrep.2024.115086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/11/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
Dissociation, characterized by altered consciousness and perception, underlies multiple mental disorders, but the specific neuronal subtypes involved remain elusive. In mice, we find that dissociation-inducing doses of ketamine significantly inhibit retrosplenial cortex (RSC) parvalbumin interneurons (PV-INs), enhancing delta oscillations (1-3 Hz) and delta-gamma phase-amplitude coupling (δ-γ PAC) and inducing dissociation-like behaviors. Optogenetic inhibition of RSC PV-INs triggers delta oscillations, δ-γ PAC, and some dissociation-like behaviors without ketamine. Furthermore, activation of RSC PV-INs or knockdown of the N-methyl-D-aspartate receptor subunit NR1 and the hyperpolarization-activated cyclic nucleotide-gated channel 1 (HCN1) in RSC PV-INs attenuates ketamine-induced delta oscillations, δ-γ PAC, and certain dissociation-like behaviors. These findings reveal that PV-INs regulate delta oscillations and δ-γ PAC and identify NR1 and HCN1 as ketamine targets in PV-INs that may cooperatively affect dissociation, possibly providing potential therapeutic targets for dissociative symptoms.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yifan Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Siyu Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kexin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziqing Deng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qi Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shubai Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kai Wei
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
18
|
Dong FL, Yu L, Feng PD, Ren JX, Bai XH, Lin JQ, Cao DL, Deng YT, Zhang Y, Shen HH, Gong H, Sun WX, Chi DQ, Mei Y, Ma L, Yin MZ, Li MN, Zhang PF, Hu N, Zhou BL, Liu Y, Zheng XJ, Chen YF, Zhong D, Tao YX, Yan M, Jiang BC. An atlas of neuropathic pain-associated molecular pathological characteristics in the mouse spinal cord. Commun Biol 2025; 8:70. [PMID: 39820760 PMCID: PMC11739467 DOI: 10.1038/s42003-025-07506-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
Peripheral nerve injury (PNI)-induced neuropathic pain (NP) is a severe disease with high prevalence in clinics. Gene reprogramming and tissue remodeling in the dorsal root ganglia (DRG) and spinal cord (SC) drive the development and maintenance of neuropathic pain (NP). However, our understanding of the NP-associated spatial molecular processing landscape of SC and the non-synaptic interactions between DRG neurons and SC cells remains limited. We here integrate spatial transcriptomics (ST) with single-nucleus RNA-sequencing (snRNA-seq) and bulk RNA-sequencing (bulk RNA-seq) to characterize regional pathological heterogeneity of the SC under NP conditions. First, the SC of NP mice manifests unique spatial atlases of genes, cell populations, cell-cell cross-talks, signaling pathways, and transcriptional regulatory networks compared to sham mice. We further report that injured DRG sensory neurons and the corresponding ventral horn of the SC show similar expression patterns after PNI. In addition, for the first time, we systematically exhibit "cross-talk omics" between the DRG neurons and SC dorsal horn neurons and glial cells, indicating an altered communication profile under NP conditions. Together, our findings decode the spatial and cellular heterogeneity of molecular pathological mechanisms underlying NP, providing a foundation for designing therapeutic targets for this disorder.
Collapse
Affiliation(s)
- Fu-Lu Dong
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Pathology, Medical School, Nantong University, Nantong, China
| | - Lina Yu
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Pain Perception and Neuromodulation, Hangzhou, China
| | - Pei-Da Feng
- Department of Pathology, Medical School, Nantong University, Nantong, China
| | - Jin-Xuan Ren
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue-Hui Bai
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Qi Lin
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - De-Li Cao
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Yu-Tao Deng
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Zhang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui-Hui Shen
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Gong
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Wen-Xing Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Dong-Qiu Chi
- Medical Service Center, Nantong University, Nantong, China
| | - Yixiao Mei
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Longfei Ma
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Pain Perception and Neuromodulation, Hangzhou, China
| | - Ming-Zhe Yin
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng-Na Li
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Peng-Fei Zhang
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Nan Hu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Bing-Lin Zhou
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Liu
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan-Jie Zheng
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi-Fan Chen
- Department of Pathology, Medical School, Nantong University, Nantong, China
| | - Da Zhong
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Min Yan
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory of Pain Perception and Neuromodulation, Hangzhou, China.
- Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China.
| | - Bao-Chun Jiang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory of Pain Perception and Neuromodulation, Hangzhou, China.
| |
Collapse
|
19
|
Korkutata M, De Luca R, Fitzgerald B, Khanday MA, Arrigoni E, Scammell TE. Afferent Projections to the Calca/CGRP-Expressing Parabrachial Neurons in Mice. J Comp Neurol 2025; 533:e70018. [PMID: 39801453 PMCID: PMC11777123 DOI: 10.1002/cne.70018] [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: 06/12/2024] [Revised: 12/05/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
The parabrachial nucleus (PB), located in the dorsolateral pons, contains primarily glutamatergic neurons that regulate responses to a variety of interoceptive and cutaneous sensory signals. One lateral PB subpopulation expresses the Calca gene, which codes for the neuropeptide calcitonin gene-related peptide (CGRP). These PBCalca /CGRP neurons relay signals related to threatening stimuli such as hypercarbia, pain, and nausea, yet their inputs and their neurochemical identity are only partially understood. We mapped the afferent projections to the lateral part of the PB in mice using conventional cholera toxin B subunit (CTb) retrograde tracing and then used conditional rabies virus retrograde tracing to map monosynaptic inputs specifically targeting the PBCalca /CGRP neurons. Using vesicular GABA (vGAT) and glutamate (vGLUT2) transporter reporter mice, we found that lateral PB neurons receive GABAergic afferents from regions such as the lateral part of the central nucleus of the amygdala, lateral dorsal subnucleus of the bed nucleus of the stria terminalis, substantia innominata, and ventrolateral periaqueductal gray. Additionally, they receive glutamatergic afferents from the infralimbic and insular cortex, paraventricular nucleus, parasubthalamic nucleus, trigeminal complex, medullary reticular nucleus, and nucleus of the solitary tract. Using anterograde tracing and confocal microscopy, we then identified close axonal appositions between these afferents and PBCalca /CGRP neurons. Finally, we used channelrhodopsin-assisted circuit mapping and found that GABAergic neurons of the central nucleus of the amygdala directly inhibit the PBCalca /CGRP neurons. These findings provide a comprehensive neuroanatomical framework for understanding the afferent projections regulating the PBCalca /CGRP neurons.
Collapse
Affiliation(s)
- Mustafa Korkutata
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Roberto De Luca
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Bridget Fitzgerald
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Mudasir A. Khanday
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Thomas E. Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
20
|
Chuang YC, Jiang BY, Chen CC. Effect of Advillin Knockout on Diabetic Neuropathy Induced by Multiple Low Doses of Streptozotocin. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2025; 68:11-21. [PMID: 39670415 DOI: 10.4103/ejpi.ejpi-d-24-00061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/23/2024] [Indexed: 12/14/2024]
Abstract
ABSTRACT Advillin is an actin-binding protein involved in regulating the organization of actin filaments and the dynamics of axonal growth cones. In mice, advillin is exclusively expressed in somatosensory neurons, ubiquitously expressed in all neuron subtypes during neonatal ages and particularly enriched in isolectin B4-positive (IB4 + ) non-peptidergic neurons in adulthood. We previously showed that advillin plays a key role in axon regeneration of somatosensory neurons during peripheral neuropathy. Mice lacking advillin lost the ability to recover from neuropathic pain induced by oxaliplatin, chronic compression of the sciatic nerve, and experimental autoimmune encephalitis. However, the role of advillin in painful diabetic neuropathy remains unknown. Diabetic neuropathy, a prevalent complication of types 1 and 2 diabetes mellitus, poses significant treatment challenges because of the limited efficacy and adverse side effects of current analgesics. Here we probed the effect of advillin knockout on neuropathic pain in a diabetic mouse model induced by multiple low doses of streptozotocin (STZ). STZ-induced cold allodynia was resolved in 8 weeks in wild-type ( Avil +/+ ) mice but could last more than 30 weeks in advillin-knockout ( Avil -/- ) mice. Additionally, Avi -/- but not Avil +/+ mice showed STZ-induced mechanical hypersensitivity of muscle. Consistent with the prolonged and/or worsened STZ-induced neuropathic pain, second-line coping responses to pain stimuli were greater in Avil -/- than Avil +/+ mice. On analyzing intraepidermal nerve density, STZ induced large axon degeneration in the hind paws but with distinct patterns between Avil +/+ and Avil -/- mice. We next probed whether advillin knockout could disturb capsaicin-induced axon regeneration ex vivo because capsaicin is clinically used to treat painful diabetic neuropathy by promoting axon regeneration. In a primary culture of dorsal root ganglion cells, 10-min capsaicin treatment selectively promoted neurite outgrowth of IB4 + neurons in Avil +/+ but not Avil -/- groups, which suggests that capsaicin could reprogram the intrinsic axonal regeneration by modulating the advillin-mediated actin dynamics. In conclusion, advillin knockout prolonged STZ-induced neuropathic pain in mice, which may be associated with the impaired intrinsic capacity of advillin-dependent IB4 + axon regeneration.
Collapse
Affiliation(s)
- Yu-Chia Chuang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Bo-Yang Jiang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
- Biomedical Translational Research Center, Taiwan Mouse Clinic, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
21
|
Rosa-Casillas M, Basbaum AI. Rostral ventral medulla circuits regulate both the sensory and affective dimensions of neuropathic pain: a commentary on Dogrul et al. Pain 2025; 166:7-8. [PMID: 39356207 DOI: 10.1097/j.pain.0000000000003375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 10/03/2024]
Affiliation(s)
- Mariela Rosa-Casillas
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Allan I Basbaum
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
22
|
Latypov TH, Wolfensohn A, Yakubov R, Li J, Srisaikaew P, Jörgens D, Jones A, Colak E, Mikulis D, Rudzicz F, Oh J, Hodaie M. Signatures of chronic pain in multiple sclerosis: a machine learning approach to investigate trigeminal neuralgia. Pain 2024:00006396-990000000-00789. [PMID: 39680491 DOI: 10.1097/j.pain.0000000000003497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/01/2024] [Indexed: 12/18/2024]
Abstract
ABSTRACT Chronic pain is a pervasive, disabling, and understudied feature of multiple sclerosis (MS), a progressive demyelinating and neurodegenerative disease. Current focus on motor components of MS disability combined with difficulties assessing pain symptoms present a challenge for the evaluation and management of pain in MS, highlighting the need for novel methods of assessment of neural signatures of chronic pain in MS. We investigate chronic pain in MS using MS-related trigeminal neuralgia (MS-TN) as a model condition focusing on gray matter structures as predictors of chronic pain. T1 imaging data from people with MS (n = 75) and MS-TN (n = 77) using machine learning (ML) was analyzed to derive imaging predictors at the level of cortex and subcortical gray matter. The ML classifier compared imaging metrics of patients with MS and MS-TN and distinguished between these conditions with 93.4% individual average testing accuracy. Structures within default-mode, somatomotor, salience, and visual networks (including hippocampus, primary somatosensory cortex, occipital cortex, and thalamic subnuclei) were identified as significant imaging predictors of trigeminal neuralgia pain. Our results emphasize the multifaceted nature of chronic pain and demonstrate the utility of imaging and ML in assessing and understanding MS-TN with greater objectivity.
Collapse
Affiliation(s)
- Timur H Latypov
- Division of Brain, Imaging and Behaviour, Krembil Research Institute University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Collaborative Program in Neuroscience, University of Toronto, Toronto, ON, Canada
| | - Abigail Wolfensohn
- Division of Brain, Imaging and Behaviour, Krembil Research Institute University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Faculty of Science, McGill University, Montreal, QC, Canada
| | - Rose Yakubov
- Division of Brain, Imaging and Behaviour, Krembil Research Institute University Health Network, Toronto, ON, Canada
- MD Program, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jerry Li
- Division of Brain, Imaging and Behaviour, Krembil Research Institute University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Collaborative Program in Neuroscience, University of Toronto, Toronto, ON, Canada
| | - Patcharaporn Srisaikaew
- Division of Brain, Imaging and Behaviour, Krembil Research Institute University Health Network, Toronto, ON, Canada
| | - Daniel Jörgens
- Division of Brain, Imaging and Behaviour, Krembil Research Institute University Health Network, Toronto, ON, Canada
| | - Ashley Jones
- Division of Neurology, Department of Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Errol Colak
- Department of Medical Imaging, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - David Mikulis
- Division of Brain, Imaging and Behaviour, Krembil Research Institute University Health Network, Toronto, ON, Canada
- Joint Department of Medical Imaging, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Frank Rudzicz
- Vector Institute for Artificial Intelligence, Toronto, ON, Canada
- Faculty of Computer Science, Dalhousie University, Halifax, NS, Canada
| | - Jiwon Oh
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Mojgan Hodaie
- Division of Brain, Imaging and Behaviour, Krembil Research Institute University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| |
Collapse
|
23
|
Barkai O, Zhang B, Turnes BL, Arab M, Yarmolinsky DA, Zhang Z, Barrett LB, Woolf CJ. ARBEL: A Machine Learning Tool with Light-Based Image Analysis for Automatic Classification of 3D Pain Behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.625907. [PMID: 39677681 PMCID: PMC11642810 DOI: 10.1101/2024.12.01.625907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
A detailed analysis of pain-related behaviors in rodents is essential for exploring both the mechanisms of pain and evaluating analgesic efficacy. With the advancement of pose-estimation tools, automatic single-camera video animal behavior pipelines are growing and integrating rapidly into quantitative behavioral research. However, current existing algorithms do not consider an animal's body-part contact intensity with- and distance from- the surface, a critical nuance for measuring certain pain-related responses like paw withdrawals ('flinching') with high accuracy and interpretability. Quantifying these bouts demands a high degree of attention to body part movement and currently relies on laborious and subjective human visual assessment. Here, we introduce a supervised machine learning algorithm, ARBEL: Automated Recognition of Behavior Enhanced with Light, that utilizes a combination of pose estimation together with a novel light-based analysis of body part pressure and distance from the surface, to automatically score pain-related behaviors in freely moving mice in three dimensions. We show the utility and accuracy of this algorithm for capturing a range of pain-related behavioral bouts using a bottom-up animal behavior platform, and its application for robust drug-screening. It allows for rapid objective pain behavior scoring over extended periods with high precision. This open-source algorithm is adaptable for detecting diverse behaviors across species and experimental platforms.
Collapse
Affiliation(s)
- Omer Barkai
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Biyao Zhang
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Bruna Lenfers Turnes
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Maryam Arab
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - David A Yarmolinsky
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Zihe Zhang
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Lee B Barrett
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Clifford J Woolf
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
MacDonald DI, Jayabalan M, Seaman J, Balaji R, Nickolls A, Chesler A. Pain persists in mice lacking both Substance P and CGRPα signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.15.567208. [PMID: 38076807 PMCID: PMC10705526 DOI: 10.1101/2023.11.15.567208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The neuropeptides Substance P and CGRPα have long been thought important for pain sensation. Both peptides and their receptors are expressed at high levels in pain-responsive neurons from the periphery to the brain making them attractive therapeutic targets. However, drugs targeting these pathways individually did not relieve pain in clinical trials. Since Substance P and CGRPα are extensively co-expressed we hypothesized that their simultaneous inhibition would be required for effective analgesia. We therefore generated Tac1 and Calca double knockout (DKO) mice and assessed their behavior using a wide range of pain-relevant assays. As expected, Substance P and CGRPα peptides were undetectable throughout the nervous system of DKO mice. To our surprise, these animals displayed largely intact responses to mechanical, thermal, chemical, and visceral pain stimuli, as well as itch. Moreover, chronic inflammatory pain and neurogenic inflammation were unaffected by loss of the two peptides. Finally, neuropathic pain evoked by nerve injury or chemotherapy treatment was also preserved in peptide-deficient mice. Thus, our results demonstrate that even in combination, Substance P and CGRPα are not required for the transmission of acute and chronic pain.
Collapse
Affiliation(s)
- Donald Iain MacDonald
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Monessha Jayabalan
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Jonathan Seaman
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Rakshita Balaji
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Alec Nickolls
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
| | - Alexander Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, United States
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| |
Collapse
|
25
|
Zheng Y, Lin X, Huang Y, Laureys S, Di H. Rasch Analysis of the Chinese Version of the Nociception Coma Scale-Revised in Patients with Prolonged Disorders of Consciousness. Clin Rehabil 2024; 38:1645-1657. [PMID: 39275814 DOI: 10.1177/02692155241280524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
OBJECTIVES The aim of this study was to analyze the Chinese version of the Nociception Coma Scale-Revised in patients with prolonged disorders of consciousness within the framework of Rasch modeling, including investigating the invariance of total scores across different etiologies of disorders of consciousness. DESIGN Prospective psychometric study. PARTICIPANTS Patients with prolonged disorders of consciousness from the Rehabilitation and Neurology units in hospital. INTERVENTIONS None. MAIN OUTCOME MEASURE The Nociception Coma Scale-Revised was undertaken by trained raters and the Coma Recovery Scale-Revised was used to assess patients' consciousness. The psychometric properties within the Rasch model including item-person targeting, reliability and separation, item fit, unidimensionality, and differential item functioning were assessed. RESULTS 84 patients with prolonged disorders of consciousness (mean age 53 years; mean injury 5 months; 42 with Minimally Conscious State and 42 with Unresponsive Wakefulness Syndrome) of 252 observations were enrolled in the study. Through the procedure of repeated assessment and differential item function, a lower item bias Rasch set was purified. The Rasch model assumptions were examined and met, with item reliability and validity meeting the recommended threshold. CONCLUSIONS The Chinese version of the Nociception Coma Scale-Revised demonstrated unidimensionality, good reliability and separation, and good item fit, but dissatisfied person fit and item-person targeting. The verbal subscale showed a notable discrepancy between person responses and the difficulty of the items, suggesting limited clinical significance.
Collapse
Affiliation(s)
- Yuhang Zheng
- International Unresponsive Wakefulness Syndrome and Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
| | - Xinyou Lin
- International Unresponsive Wakefulness Syndrome and Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
| | - Yuehong Huang
- International Unresponsive Wakefulness Syndrome and Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
| | - Steven Laureys
- International Unresponsive Wakefulness Syndrome and Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
- Coma Science Group, GIGA Consciousness, University of Liège, Liège, Belgium
- CERVO Brain Research Centre, Laval University, Laval, QC, Canada
| | - Haibo Di
- International Unresponsive Wakefulness Syndrome and Consciousness Science Institute, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
26
|
Wang Q, Zhao G, Ding H, Wang Z, Wu J, Huang H, Cao L, Wang H, Gao Z, Feng J. Trpv1-lineage neuron-expressing Kcnq4 channel modulates itch sensation in mice. Pain 2024:00006396-990000000-00772. [PMID: 39560444 DOI: 10.1097/j.pain.0000000000003479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/13/2024] [Indexed: 11/20/2024]
Abstract
ABSTRACT Voltage-gated potassium channel subfamily q member 4 (Kcnq4) is predominantly expressed by hair cells and auditory neurons and regulates the neuronal excitability in the auditory pathway. Although it is further detected in myelinated large-diameter dorsal root ganglia (DRG) neurons in the periphery, the expression and function of Kcnq4 channel in nociceptors remains unknown. Here we showed that Kcnq4 is substantially expressed by unmyelinated small-diameter DRG neurons in both human and mouse. In spite of a dispensable role in acute pain and chronic skin inflammation, Kcnq4 is specifically involved in the regulation of scratching behavior through controlling action potential firing properties, evidenced by the increased neuronal excitability in small-diameter DRG neurons isolated from Kcnq4 deficient mice. Moreover, genetic ablation of Kcnq4 in Trpv1-positive neurons exacerbates both acute and chronic itch behavior in mice. Taken together, our results uncover a functional role of Trpv1-lineage neuron-expressing Kcnq4 channel in the modulation of itch-specific neuronal excitation in the periphery.
Collapse
Affiliation(s)
- Qiong Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guodun Zhao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huijuan Ding
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zihan Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianwei Wu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Han Huang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Cao
- Department of Chinese Medicine, Tangdu Hospital, Xi'an, China
| | - Hongli Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhaobing Gao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jing Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
27
|
Motzkin JC, Basbaum AI, Crowther AJ. Neuroanatomy of the nociceptive system: From nociceptors to brain networks. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:1-39. [PMID: 39580210 DOI: 10.1016/bs.irn.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
This chapter reviews the neuroanatomy of the nociceptive system and its functional organization. We describe three main compartments of the nervous system that underlie normal nociception and the resulting pain percept: Peripheral, Spinal Cord, and Brain. We focus on how ascending nociceptive processing streams traverse these anatomical compartments, culminating in the multidimensional experience of pain. We also describe neuropathic pain conditions, in which nociceptive processing is abnormal, not only because of the primary effects of a lesion or disease affecting peripheral nerves or the central nervous system (CNS), but also due to secondary effects on ascending pathways and brain networks. We discuss how the anatomical components (circuits/networks) reorganize under various etiologies of neuropathic pain and how these changes can give rise to pathological pain states.
Collapse
Affiliation(s)
- Julian C Motzkin
- Department of Neurology and Department Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States.
| | - Allan I Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Andrew J Crowther
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
28
|
Ballantyne JC, Basbaum AI. It all began in Issaquah 50 years ago. Pain 2024; 165:S3-S14. [PMID: 39560410 DOI: 10.1097/j.pain.0000000000003303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/28/2024] [Indexed: 11/20/2024]
Abstract
ABSTRACT "Somehow scientists still pursue the same questions, if now on higher levels of theoretical abstraction rooted in deeper layers of empirical evidence… To paraphrase an old philosophy joke, science is more like it is today than it has ever been. In other words, science remains as challenging as ever to human inquiry. And the need to communicate its progress… remains as essential now as then." - Tom Siegfried, Science News 2021In fact, essential questions about pain have not changed since IASP's creation in Issaquah: what causes it and how can we treat it? Are we any closer to answering these questions, or have we just widened the gap between bench and bedside? The technology used to answer questions about pain mechanisms has certainly changed, whether the focus is on sensory neurons, spinal cord circuitry, descending controls or cortical pain processing. In this paper, we will describe how transgenics, transcriptomics, optogenetics, calcium imaging, fMRI, neuroimmunology and in silico drug development have transformed the way we examine the complexity of pain processing. But does it all, as our founders hoped, help people with pain? Are voltage-gated Na channels the new holy grail for analgesic development, is there a pain biomarker, can we completely replace opioids, will proteomic analyses identify novel targets, is there a "pain matrix," and can it be targeted? Do the answers lie in our tangible discoveries, or in the seemingly intangible? Our founders could barely imagine what we know now, yet their questions remain.
Collapse
Affiliation(s)
- Jane C Ballantyne
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Allan I Basbaum
- Department of Anatomy, University of California, San Francisco, CA, United States
| |
Collapse
|
29
|
Staedtler ES, Sapio MR, King DM, Maric D, Ghetti A, Mannes AJ, Iadarola MJ. The μ-opioid receptor differentiates two distinct human nociceptive populations relevant to clinical pain. Cell Rep Med 2024; 5:101788. [PMID: 39413733 PMCID: PMC11513826 DOI: 10.1016/j.xcrm.2024.101788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/26/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
The shortfall in new analgesic agents is a major impediment to reducing reliance on opioid medications for control of severe pain. In both animals and man, attenuating nociceptive transmission from primary afferent neurons with a μ-opioid receptor agonist yields highly effective analgesia. Consequently, deeper molecular characterization of human nociceptive afferents expressing OPRM1, the μ-opioid receptor gene, is a key component for advancing analgesic drug discovery and understanding clinical pain control. A co-expression matrix for the μ-opioid receptor and a variety of nociceptive channels as well as δ- and κ-opioid receptors is established by multiplex in situ hybridization. Our results indicate an OPRM1-positive population with strong molecular resemblance to rodent peptidergic C-nociceptors associated with tissue damage pain and an OPRM1-negative population sharing molecular characteristics of murine non-peptidergic C-nociceptors. The empirical identification of two distinct human nociceptive populations that differ profoundly in their presumed responsiveness to opioids provides an actionable translational framework for human pain control.
Collapse
Affiliation(s)
- Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | | | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Palmiter RD. Parabrachial neurons promote nociplastic pain. Trends Neurosci 2024; 47:722-735. [PMID: 39147688 DOI: 10.1016/j.tins.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
The parabrachial nucleus (PBN) in the dorsal pons responds to bodily threats and transmits alarm signals to the forebrain. Parabrachial neuron activity is enhanced during chronic pain, and inactivation of PBN neurons in mice prevents the establishment of neuropathic, chronic pain symptoms. Chemogenetic or optogenetic activation of all glutamatergic neurons in the PBN, or just the subpopulation that expresses the Calca gene, is sufficient to establish pain phenotypes, including long-lasting tactile allodynia, that scale with the extent of stimulation, thereby promoting nociplastic pain, defined as diffuse pain without tissue inflammation or nerve injury. This review focuses on the role(s) of molecularly defined PBN neurons and the downstream nodes in the brain that contribute to establishing nociplastic pain.
Collapse
Affiliation(s)
- Richard D Palmiter
- Departments of Biochemistry and Genome Sciences, Investigator of the Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
31
|
Chen L, Wu MY, Chen SL, Hu R, Wang Y, Zeng W, Feng S, Ke M, Wang L, Chen S, Gu M. The Guardian of Vision: Intelligent Bacteriophage-Based Eyedrops for Clinical Multidrug-Resistant Ocular Surface Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407268. [PMID: 39091071 DOI: 10.1002/adma.202407268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Clinical multidrug-resistant Pseudomonas aeruginosa (MDR-PA) is the leading cause of refractory bacterial keratitis (BK). However, the reported BK treatment methods lack biosecurity and bioavailability, which usually causes irreversible visual impairment and even blindness. Herein, for BK caused by clinically isolated MDR-PA infection, armed phages are modularized with the type I photosensitizer (PS) ACR-DMT, and an intelligent phage eyedrop is developed for combined phagotherapy and photodynamic therapy (PDT). These eyedrops maximize the advantages of bacteriophages and ACR-DMT, enabling more robust and specific targeting killing of MDR-PA under low oxygen-dependence, penetrating and disrupting biofilms, and efficiently preventing biofilm reformation. Altering the biofilm and immune microenvironments alleviates inflammation noninvasively, promotes corneal healing without scar formation, protects ocular tissues, restores visual function, and prevents long-term discomfort and pain. This strategy exhibits strong scalability, enables at-home treatment of ocular surface infections with great patient compliance and a favorable prognosis, and has significant potential for clinical application.
Collapse
Affiliation(s)
- Luojia Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ming-Yu Wu
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Si-Ling Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Rui Hu
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yifei Wang
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Weijuan Zeng
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shun Feng
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Min Ke
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lianrong Wang
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Department of Respiratory Diseases, Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, 518026, China
| | - Shi Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Shenzhen Institute of Translational Medicine, Shenzhen University Medical School, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meijia Gu
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
32
|
Yin Y, Zhao P, Xu X, Zhou B, Chen J, Jiang X, Liu Y, Wu Y, Yue W, Xu H, Bu W. Piezoelectric Analgesia Blocks Cancer-Induced Bone Pain. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403979. [PMID: 39044708 DOI: 10.1002/adma.202403979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/23/2024] [Indexed: 07/25/2024]
Abstract
The manipulation of cell surface receptors' activity will open a new frontier for drug development and disease treatment. However, limited by the desensitization of drugs, effective physical intervention strategy remains challenging. Here, the controllable internalization of transient receptor potential vanilloid 1 (TRPV1) on neural cells by local piezoelectric field is reported. Single-cell-level local electric field is construct by synthesizing piezoelectric BiOIO3 nanosheets (BIONSs). Upon a mild ultrasound of 0.08 W cm-2, an electric field of 15.29 µV is generated on the surface of BIONSs, further inducing TRPV1 internalization in 5 min. The as-downregulated TRPV1 expression results in the reduction of Ca2+ signal in a spinal neuron and the inhibition of the activity of wide range dynamic neurons, therefore effectively preventing the transmission of cancer-induced bone pain (CIBP). This strategy not only charts a new course for CIBP alleviation, but also introduces a promising nanotechnology for regulating cell surface receptors, showing significant potential in neuropathological and receptor-related diseases.
Collapse
Affiliation(s)
- Yifei Yin
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Shanghai, 200072, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200072, China
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Peiran Zhao
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Xianyun Xu
- Department of Clinical Laboratory, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, China
| | - Bangguo Zhou
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Shanghai, 200072, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200072, China
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Jian Chen
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Xingwu Jiang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Yelin Wu
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Wenwen Yue
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Shanghai, 200072, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200072, China
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| |
Collapse
|
33
|
Chen C, Niehaus JK, Dinc F, Huang KL, Barnette AL, Tassou A, Shuster SA, Wang L, Lemire A, Menon V, Ritola K, Hantman AW, Zeng H, Schnitzer MJ, Scherrer G. Neural circuit basis of placebo pain relief. Nature 2024; 632:1092-1100. [PMID: 39048016 PMCID: PMC11358037 DOI: 10.1038/s41586-024-07816-z] [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: 12/07/2022] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
Placebo effects are notable demonstrations of mind-body interactions1,2. During pain perception, in the absence of any treatment, an expectation of pain relief can reduce the experience of pain-a phenomenon known as placebo analgesia3-6. However, despite the strength of placebo effects and their impact on everyday human experience and the failure of clinical trials for new therapeutics7, the neural circuit basis of placebo effects has remained unclear. Here we show that analgesia from the expectation of pain relief is mediated by rostral anterior cingulate cortex (rACC) neurons that project to the pontine nucleus (rACC→Pn)-a precerebellar nucleus with no established function in pain. We created a behavioural assay that generates placebo-like anticipatory pain relief in mice. In vivo calcium imaging of neural activity and electrophysiological recordings in brain slices showed that expectations of pain relief boost the activity of rACC→Pn neurons and potentiate neurotransmission in this pathway. Transcriptomic studies of Pn neurons revealed an abundance of opioid receptors, further suggesting a role in pain modulation. Inhibition of the rACC→Pn pathway disrupted placebo analgesia and decreased pain thresholds, whereas activation elicited analgesia in the absence of placebo conditioning. Finally, Purkinje cells exhibited activity patterns resembling those of rACC→Pn neurons during pain-relief expectation, providing cellular-level evidence for a role of the cerebellum in cognitive pain modulation. These findings open the possibility of targeting this prefrontal cortico-ponto-cerebellar pathway with drugs or neurostimulation to treat pain.
Collapse
Affiliation(s)
- Chong Chen
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jesse K Niehaus
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Fatih Dinc
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- CNC Program, Stanford University, Stanford, CA, USA
| | - Karen L Huang
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexander L Barnette
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - S Andrew Shuster
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Lihua Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Andrew Lemire
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Vilas Menon
- Department of Neurology, Columbia University, New York, NY, USA
| | - Kimberly Ritola
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam W Hantman
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mark J Schnitzer
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- CNC Program, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
- James H. Clark Center for Biomedical Engineering & Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
34
|
Tu C, Chen YB, Lai SQ, Yu YP, Huang ZW, Li HZ, Ao RF, Han D, Gao JW, Zhu GZ, Wu DZ, Huang YS, Zhao K, Meng TT, Zhong ZM. Accumulation of β-aminoisobutyric acid mediates hyperalgesia in ovariectomized mice through Mas-related G protein-coupled receptor D signaling. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167269. [PMID: 38810919 DOI: 10.1016/j.bbadis.2024.167269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/07/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
Hyperalgesia is typified by reduced pain thresholds and heightened responses to painful stimuli, with a notable prevalence in menopausal women, but the underlying mechanisms are far from understood. β-Aminoisobutyric acid (BAIBA), a product of valine and thymine catabolism, has been reported to be a novel ligand of the Mas-related G protein coupled receptor D (MrgprD), which mediates pain and hyperalgesia. Here, we established a hyperalgesia model in 8-week-old female mice through ovariectomy (OVX). A significant increase in BAIBA plasma level was observed and was associated with decline of mechanical withdrawal threshold, thermal and cold withdrawal latency in mice after 6 weeks of OVX surgery. Increased expression of MrgprD in dorsal root ganglion (DRG) was shown in OVX mice compared to Sham mice. Interestingly, chronic loading with BAIBA not only exacerbated hyperalgesia in OVX mice, but also induced hyperalgesia in gonadally intact female mice. BAIBA supplementation also upregulated the MrgprD expression in DRG of both OVX and intact female mice, and enhanced the excitability of DRG neurons in vitro. Knockout of MrgprD markedly suppressed the effects of BAIBA on hyperalgesia and excitability of DRG neurons. Collectively, our data suggest the involvement of BAIBA in the development of hyperalgesia via MrgprD-dependent pathway, and illuminate the mechanisms underlying hyperalgesia in menopausal women.
Collapse
Affiliation(s)
- Chen Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopeadics, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopedics, Guangzhou, China
| | - Yun-Biao Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Si-Qi Lai
- Department of Pathology, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopedics, Guangzhou, China
| | - Yong-Peng Yu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhi-Wei Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong-Zhou Li
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rui-Feng Ao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dong Han
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia-Wen Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guo-Zheng Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Di-Zheng Wu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu-Sheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kai Zhao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Ting-Ting Meng
- Unit of Anaesthesia and Pain Management, Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
35
|
Xu JF, Liu L, Liu Y, Lu KX, Zhang J, Zhu YJ, Fang F, Dou YN. Spinal Nmur2-positive Neurons Play a Crucial Role in Mechanical Itch. THE JOURNAL OF PAIN 2024; 25:104504. [PMID: 38442838 DOI: 10.1016/j.jpain.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
The dorsal spinal cord is crucial for the transmission and modulation of multiple somatosensory modalities, such as itch, pain, and touch. Despite being essential for the well-being and survival of an individual, itch and pain, in their chronic forms, have increasingly been recognized as clinical problems. Although considerable progress has been made in our understanding of the neurochemical processing of nociceptive and chemical itch sensations, the neural substrate that is crucial for mechanical itch processing is still unclear. Here, using genetic and functional manipulation, we identified a population of spinal neurons expressing neuromedin U receptor 2 (Nmur2+) as critical elements for mechanical itch. We found that spinal Nmur2+ neurons are predominantly excitatory neurons, and are enriched in the superficial laminae of the dorsal horn. Pharmacogenetic activation of cervical spinal Nmur2+ neurons evoked scratching behavior. Conversely, the ablation of these neurons using a caspase-3-based method decreased von Frey filament-induced scratching behavior without affecting responses to other somatosensory modalities. Similarly, suppressing the excitability of cervical spinal Nmur2+ neurons via the overexpression of functional Kir2.1 potassium channels reduced scratching in response to innocuous mechanical stimuli, but not to pruritogen application. At the lumbar level, pharmacogenetic activation of these neurons evoked licking and lifting behaviors. However, ablating these neurons did not affect the behavior associated with acute pain. Thus, these results revealed the crucial role of spinal Nmur2+ neurons in mechanical itch. Our study provides important insights into the neural basis of mechanical itch, paving the way for developing novel therapies for chronic itch. PERSPECTIVE: Excitatory Nmur2+ neurons in the superficial dorsal spinal cord are essential for mechanical but not chemical itch information processing. These spinal Nmur2+ neurons represent a potential cellular target for future therapeutic interventions against chronic itch. Spinal and supraspinal Nmur2+ neurons may play different roles in pain signal processing.
Collapse
Affiliation(s)
- Jun-Feng Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lian Liu
- Department of Endocrinology and Metabolic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Lingang Laboratory, Shanghai, China
| | - Ke-Xing Lu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jun Zhang
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan-Jing Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Fang Fang
- Department of Endocrinology and Metabolic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Nong Dou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
36
|
Lister KC, Wong C, Uttam S, Parisien M, Stecum P, Brown N, Cai W, Hooshmandi M, Gu N, Amiri M, Beaudry F, Jafarnejad SM, Tavares-Ferreira D, Inturi NN, Mazhar K, Zhao HT, Fitzsimmons B, Gkogkas CG, Sonenberg N, Price TJ, Diatchenko L, Atlasi Y, Mogil JS, Khoutorsky A. Translational control in the spinal cord regulates gene expression and pain hypersensitivity in the chronic phase of neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600539. [PMID: 38979173 PMCID: PMC11230214 DOI: 10.1101/2024.06.24.600539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Sensitization of spinal nociceptive circuits plays a crucial role in neuropathic pain. This sensitization depends on new gene expression that is primarily regulated via transcriptional and translational control mechanisms. The relative roles of these mechanisms in regulating gene expression in the clinically relevant chronic phase of neuropathic pain are not well understood. Here, we show that changes in gene expression in the spinal cord during the chronic phase of neuropathic pain are substantially regulated at the translational level. Downregulating spinal translation at the chronic phase alleviated pain hypersensitivity. Cell-type-specific profiling revealed that spinal inhibitory neurons exhibited greater changes in translation after peripheral nerve injury compared to excitatory neurons. Notably, increasing translation selectively in all inhibitory neurons or parvalbumin-positive (PV+) interneurons, but not excitatory neurons, promoted mechanical pain hypersensitivity. Furthermore, increasing translation in PV+ neurons decreased their intrinsic excitability and spiking activity, whereas reducing translation in spinal PV+ neurons prevented the nerve injury-induced decrease in excitability. Thus, translational control mechanisms in the spinal cord, particularly in inhibitory neurons, play a role in mediating neuropathic pain hypersensitivity.
Collapse
Affiliation(s)
- Kevin C. Lister
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Sonali Uttam
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Marc Parisien
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Patricia Stecum
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Nicole Brown
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Weihua Cai
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Mehdi Hooshmandi
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Ning Gu
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Mehdi Amiri
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Francis Beaudry
- Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- Centre de recherche sur le cerveau et l’apprentissage (CIRCA), Université de Montréal, Montréal, Québec, Canada
| | - Seyed Mehdi Jafarnejad
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, BT9 7AE, UK
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, 75080
| | - Nikhil Nageshwar Inturi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, 75080
| | - Khadijah Mazhar
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, 75080
| | | | | | - Christos G. Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, 75080
| | - Luda Diatchenko
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Yaser Atlasi
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, BT9 7AE, UK
| | - Jeffrey S. Mogil
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Department of Psychology, Faculty of Science, McGill University, Montreal, QC, Canada
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| |
Collapse
|
37
|
Bell AM, Utting C, Dickie AC, Kucharczyk MW, Quillet R, Gutierrez-Mecinas M, Razlan ANB, Cooper AH, Lan Y, Hachisuka J, Weir GA, Bannister K, Watanabe M, Kania A, Hoon MA, Macaulay IC, Denk F, Todd AJ. Deep sequencing of Phox2a nuclei reveals five classes of anterolateral system neurons. Proc Natl Acad Sci U S A 2024; 121:e2314213121. [PMID: 38805282 PMCID: PMC11161781 DOI: 10.1073/pnas.2314213121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 04/16/2024] [Indexed: 05/30/2024] Open
Abstract
The anterolateral system (ALS) is a major ascending pathway from the spinal cord that projects to multiple brain areas and underlies the perception of pain, itch, and skin temperature. Despite its importance, our understanding of this system has been hampered by the considerable functional and molecular diversity of its constituent cells. Here, we use fluorescence-activated cell sorting to isolate ALS neurons belonging to the Phox2a-lineage for single-nucleus RNA sequencing. We reveal five distinct clusters of ALS neurons (ALS1-5) and document their laminar distribution in the spinal cord using in situ hybridization. We identify three clusters of neurons located predominantly in laminae I-III of the dorsal horn (ALS1-3) and two clusters with cell bodies located in deeper laminae (ALS4 and ALS5). Our findings reveal the transcriptional logic that underlies ALS neuronal diversity in the adult mouse and uncover the molecular identity of two previously identified classes of projection neurons. We also show that these molecular signatures can be used to target groups of ALS neurons using retrograde viral tracing. Overall, our findings provide a valuable resource for studying somatosensory biology and targeting subclasses of ALS neurons.
Collapse
Affiliation(s)
- Andrew M. Bell
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
- Small Animal Clinical Sciences, School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| | | | - Allen C. Dickie
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| | - Mateusz W. Kucharczyk
- The Wolfson Centre for Age-Related Diseases, King’s College London, LondonWC2R 2LS, United Kingdom
- Cancer Neurophysiology Group, Lukasiewicz-PORT, Polish Center for Technology Development, Wroclaw54-066, Poland
| | - Raphaëlle Quillet
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| | - Maria Gutierrez-Mecinas
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| | - Aimi N. B. Razlan
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| | - Andrew H. Cooper
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| | - Yuxuan Lan
- Earlham Institute, NorwichNRU 7UZ, United Kingdom
| | - Junichi Hachisuka
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| | - Greg A. Weir
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| | - Kirsty Bannister
- The Wolfson Centre for Age-Related Diseases, King’s College London, LondonWC2R 2LS, United Kingdom
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo060-8638, Japan
| | - Artur Kania
- Neural Circuit Development Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Mark A. Hoon
- Molecular Genetics Section, National Institute of Dental and Craniofacial Research/NIH, Bethesda, MD 20892
| | | | - Franziska Denk
- The Wolfson Centre for Age-Related Diseases, King’s College London, LondonWC2R 2LS, United Kingdom
| | - Andrew J. Todd
- Spinal Cord Group, School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, GlasgowG12 8QQ, United Kingdom
| |
Collapse
|
38
|
Ke J, Lu WC, Jing HY, Qian S, Moon SW, Cui GF, Qian WX, Che XJ, Zhang Q, Lai SS, Zhang L, Zhu YJ, Xie JD, Huang TW. Functional dissection of parabrachial substrates in processing nociceptive information. Zool Res 2024; 45:633-647. [PMID: 38766746 PMCID: PMC11188607 DOI: 10.24272/j.issn.2095-8137.2023.412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 05/22/2024] Open
Abstract
Painful stimuli elicit first-line reflexive defensive reactions and, in many cases, also evoke second-line recuperative behaviors, the latter of which reflects the sensing of tissue damage and the alleviation of suffering. The lateral parabrachial nucleus (lPBN), composed of external- (elPBN), dorsal- (dlPBN), and central/superior-subnuclei (jointly referred to as slPBN), receives sensory inputs from spinal projection neurons and plays important roles in processing affective information from external threats and body integrity disruption. However, the organizational rules of lPBN neurons that provoke diverse behaviors in response to different painful stimuli from cutaneous and deep tissues remain unclear. In this study, we used region-specific neuronal depletion or silencing approaches combined with a battery of behavioral assays to show that slPBN neurons expressing substance P receptor ( NK1R) (lPBN NK1R) are crucial for driving pain-associated self-care behaviors evoked by sustained noxious thermal and mechanical stimuli applied to skin or bone/muscle, while elPBN neurons are dispensable for driving such reactions. Notably, lPBN NK1R neurons are specifically required for forming sustained somatic pain-induced negative teaching signals and aversive memory but are not necessary for fear-learning or escape behaviors elicited by external threats. Lastly, both lPBN NK1R and elPBN neurons contribute to chemical irritant-induced nocifensive reactions. Our results reveal the functional organization of parabrachial substrates that drive distinct behavioral outcomes in response to sustained pain versus external danger under physiological conditions.
Collapse
Affiliation(s)
- Jin Ke
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei-Cheng Lu
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Hai-Yang Jing
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Shen Qian
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Sun-Wook Moon
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Guang-Fu Cui
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Wei-Xin Qian
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiao-Jing Che
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhang
- Department of Anesthesiology, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Shi-Shi Lai
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, China
| | - Ling Zhang
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ying-Jie Zhu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China. E-mail:
| | - Jing-Dun Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China. E-mail:
| | - Tian-Wen Huang
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China. E-mail:
| |
Collapse
|
39
|
Korkutata M, De Luca R, Fitzgerald B, Arrigoni E, Scammell TE. Afferent projections to the Calca /CGRP-expressing parabrachial neurons in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593004. [PMID: 38766214 PMCID: PMC11100666 DOI: 10.1101/2024.05.07.593004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The parabrachial nucleus (PB), located in the dorsolateral pons, contains primarily glutamatergic neurons which regulate responses to a variety of interoceptive and cutaneous sensory signals. The lateral PB subpopulation expressing the Calca gene which produces the neuropeptide calcitonin gene-related peptide (CGRP) relays signals related to threatening stimuli such as hypercarbia, pain, and nausea, yet the afferents to these neurons are only partially understood. We mapped the afferent projections to the lateral part of the PB in mice using conventional cholera toxin B subunit (CTb) retrograde tracing, and then used conditional rabies virus retrograde tracing to map monosynaptic inputs specifically targeting the PB Calca /CGRP neurons. Using vesicular GABA (vGAT) and glutamate (vGLUT2) transporter reporter mice, we found that lateral PB neurons receive GABAergic afferents from regions such as the lateral part of the central nucleus of the amygdala, lateral dorsal subnucleus of the bed nucleus of the stria terminalis, substantia innominata, and the ventrolateral periaqueductal gray. Additionally, they receive glutamatergic afferents from the infralimbic and insular cortex, paraventricular nucleus, parasubthalamic nucleus, trigeminal complex, medullary reticular nucleus, and nucleus of the solitary tract. Using anterograde tracing and confocal microscopy, we then identified close axonal appositions between these afferents and PB Calca /CGRP neurons. Finally, we used channelrhodopsin-assisted circuit mapping to test whether some of these inputs directly synapse upon the PB Calca /CGRP neurons. These findings provide a comprehensive neuroanatomical framework for understanding the afferent projections regulating the PB Calca /CGRP neurons.
Collapse
|
40
|
Ye H, Lin Q, Mei Q, Liu Q, Cao S. Study on mechanism of transdermal administration of eugenol for pain treatment by network pharmacology and molecular docking technology. Heliyon 2024; 10:e29722. [PMID: 38681628 PMCID: PMC11046106 DOI: 10.1016/j.heliyon.2024.e29722] [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: 10/21/2023] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024] Open
Abstract
The objective of this study was to explore the pharmacological mechanism of transdermal administration of eugenol (EUG) for pain treatment. Firstly, network pharmacology techniques were employed to identify the potential targets responsible for the analgesic effect of EUG. Subsequently, molecular docking technology was used to validate interactions between EUG and the crystal structure of the core target protein. Finally, the impact of EUG on the expression and activation of TRPV1 receptors in HaCaT cells was evaluated through in vitro experiments, thus confirming the analysis of network pharmacology. The study suggested that the transdermal administration of EUG for pain treatment might target the TRPV1 receptor. Molecular docking revealed that EUG could spontaneously bind to the TRPV1 receptor with a high binding ability. The analysis of Western blot (WB) and intracellular Ca2+ levels demonstrated that EUG could increase the expression of TRPV1 in HaCaT cells, activating TRPV1 to induce intracellular Ca2+ influx (P < 0.05). These findings suggested that the initial application of EUG would cause a brief stimulation of TRPV1 receptors and upregulation of TRPV1 expression. Upon continued exposure, EUG would act as a TRPV1 agonist, increasing intracellular Ca2+ levels that might be associated with desensitization of pain sensations.
Collapse
Affiliation(s)
- Haoting Ye
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Qiuxiao Lin
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qinghua Mei
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Qiuqiong Liu
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Siwei Cao
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Malapert P, Robert G, Brunet E, Chemin J, Bourinet E, Moqrich A. A novel Na v1.8-FLPo driver mouse for intersectional genetics to uncover the functional significance of primary sensory neuron diversity. iScience 2024; 27:109396. [PMID: 38510134 PMCID: PMC10952036 DOI: 10.1016/j.isci.2024.109396] [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: 08/18/2023] [Revised: 11/08/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
The recent development of single-cell and single-nucleus RNA sequencing has highlighted the extraordinary diversity of dorsal root ganglia neurons. However, the few available genetic tools limit our understanding of the functional significance of this heterogeneity. We generated a new mouse line expressing the flippase recombinase from the scn10a locus. By crossing Nav1.8Ires-FLPo mice with the AdvillinCre and RC::FL-hM3Dq mouse lines in an intersectional genetics approach, we were able to obtain somatodendritic expression of hM3Dq-mCherry selectively in the Nav1.8 lineage. The bath application of clozapine N-oxide triggered strong calcium responses selectively in mCherry+ neurons. The intraplantar injection of CNO caused robust flinching, shaking, and biting responses accompanied by strong cFos activation in the ipsilateral lumbar spinal cord. The Nav1.8Ires-FLPo mouse model will be a valuable tool for extending our understanding of the in vivo functional specialization of neuronal subsets of the Nav1.8 lineage for which inducible Cre lines are available.
Collapse
Affiliation(s)
- Pascale Malapert
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, Marseille, France
| | - Guillaume Robert
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, Marseille, France
| | - Elena Brunet
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, Marseille, France
| | - Jean Chemin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Emmanuel Bourinet
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Aziz Moqrich
- Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, Marseille, France
| |
Collapse
|
42
|
Zhu H, Tao Y, Wang S, Zhu X, Lin K, Zheng N, Chen LM, Xu F, Wu R. fMRI, LFP, and anatomical evidence for hierarchical nociceptive routing pathway between somatosensory and insular cortices. Neuroimage 2024; 289:120549. [PMID: 38382864 DOI: 10.1016/j.neuroimage.2024.120549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/06/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
The directional organization of multiple nociceptive regions, particularly within obscure operculoinsular areas, underlying multidimensional pain processing remains elusive. This study aims to establish the fundamental organization between somatosensory and insular cortices in routing nociceptive information. By employing an integrated multimodal approach of high-field fMRI, intracranial electrophysiology, and transsynaptic viral tracing in rats, we observed a hierarchically organized connection of S1/S2 → posterior insula → anterior insula in routing nociceptive information. The directional nociceptive pathway determined by early fMRI responses was consistent with that examined by early evoked LFP, intrinsic effective connectivity, and anatomical projection, suggesting fMRI could provide a valuable facility to discern directional neural circuits in animals and humans non-invasively. Moreover, our knowledge of the nociceptive hierarchical organization of somatosensory and insular cortices and the interface role of the posterior insula may have implications for the development of targeted pain therapies.
Collapse
Affiliation(s)
- Hongyan Zhu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yan Tao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Siqi Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xutao Zhu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kunzhang Lin
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ning Zheng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Li Min Chen
- Vanderbilt University Institute of Imaging Science and Department of Psychology, Vanderbilt University, Nashville, TN 37232, USA.
| | - Fuqiang Xu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Ruiqi Wu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 200031, China.
| |
Collapse
|
43
|
Chen H, Bleimeister IH, Nguyen EK, Li J, Cui AY, Stratton HJ, Smith KM, Baccei ML, Ross SE. The functional and anatomical characterization of three spinal output pathways of the anterolateral tract. Cell Rep 2024; 43:113829. [PMID: 38421871 PMCID: PMC11025583 DOI: 10.1016/j.celrep.2024.113829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/24/2023] [Accepted: 02/03/2024] [Indexed: 03/02/2024] Open
Abstract
The nature of spinal output pathways that convey nociceptive information to the brain has been the subject of controversy. Here, we provide anatomical, molecular, and functional characterizations of two distinct anterolateral pathways: one, ascending in the lateral spinal cord, triggers nociceptive behaviors, and the other one, ascending in the ventral spinal cord, when inhibited, leads to sensorimotor deficits. Moreover, the lateral pathway consists of at least two subtypes. The first is a contralateral pathway that extends to the periaqueductal gray (PAG) and thalamus; the second is a bilateral pathway that projects to the bilateral parabrachial nucleus (PBN). Finally, we present evidence showing that activation of the contralateral pathway is sufficient for defensive behaviors such as running and freezing, whereas the bilateral pathway is sufficient for attending behaviors such as licking and guarding. This work offers insight into the complex organizational logic of the anterolateral system in the mouse.
Collapse
Affiliation(s)
- Haichao Chen
- Tsinghua Medicine, Tsinghua University, Beijing 100084, China; Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Isabel H Bleimeister
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eileen K Nguyen
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jie Li
- Department of Anesthesiology, Pain Research Center, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Abby Yilin Cui
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Harrison J Stratton
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kelly M Smith
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mark L Baccei
- Department of Anesthesiology, Pain Research Center, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Sarah E Ross
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
44
|
Srikanth KD, Elahi H, Chander P, Washburn HR, Hassler S, Mwirigi JM, Kume M, Loucks J, Arjarapu R, Hodge R, Shiers SI, Sankaranarayanan I, Erdjument-Bromage H, Neubert TA, Campbell ZT, Paik R, Price TJ, Dalva MB. VLK drives extracellular phosphorylation of EphB2 to govern the EphB2-NMDAR interaction and injury-induced pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585314. [PMID: 38562765 PMCID: PMC10983893 DOI: 10.1101/2024.03.18.585314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Phosphorylation of hundreds of protein extracellular domains is mediated by two kinase families, yet the significance of these kinases is underexplored. Here, we find that the presynaptic release of the tyrosine directed-ectokinase, Vertebrate Lonesome Kinase (VLK/Pkdcc), is necessary and sufficient for the direct extracellular interaction between EphB2 and GluN1 at synapses, for phosphorylation of the ectodomain of EphB2, and for injury-induced pain. Pkdcc is an essential gene in the nervous system, and VLK is found in synaptic vesicles, and is released from neurons in a SNARE-dependent fashion. VLK is expressed by nociceptive sensory neurons where presynaptic sensory neuron-specific knockout renders mice impervious to post-surgical pain, without changing proprioception. VLK defines an extracellular mechanism that regulates protein-protein interaction and non-opioid-dependent pain in response to injury.
Collapse
Affiliation(s)
- Kolluru D Srikanth
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Hajira Elahi
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Praveen Chander
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Halley R Washburn
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
- Department of Molecular Biology, Princeton University; Princeton, NJ 08544, USA
| | - Shayne Hassler
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- College of Medicine, University of Houston; Houston, TX 77004, USA
| | - Juliet M Mwirigi
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Moeno Kume
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Jessica Loucks
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
| | - Rohita Arjarapu
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
| | - Rachel Hodge
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Stephanie I Shiers
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Zachary T Campbell
- Department of Anesthesiology, University of Wisconsin-Madison; Madison, WI 53792, USA
| | - Raehum Paik
- Department of Anesthesiology, University of Wisconsin-Madison; Madison, WI 53792, USA
- Department of Genetics, University of Texas Health Science Center at San Antonio; San Antonio, TX 78229, USA
| | - Theodore J Price
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Matthew B Dalva
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
45
|
Kadakia F, Khadka A, Yazell J, Davidson S. Chemogenetic Modulation of Posterior Insula CaMKIIa Neurons Alters Pain and Thermoregulation. THE JOURNAL OF PAIN 2024; 25:766-780. [PMID: 37832899 PMCID: PMC10922377 DOI: 10.1016/j.jpain.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023]
Abstract
The posterior insular cortex (PIC) is well positioned to perform somatosensory-limbic integration; yet, the function of neuronal subsets within the PIC in processing the sensory and affective dimensions of pain remains unclear. Here, we employ bidirectional chemogenetic modulation to characterize the function of PIC CaMKIIa-expressing excitatory neurons in a comprehensive array of sensory, affective, and thermoregulatory behaviors. Excitatory pyramidal neurons in the PIC were found to be sensitized under inflammatory pain conditions. Chemogenetic activation of excitatory CaMKIIa-expressing PIC neurons in non-injured conditions produced an increase in reflexive and affective pain- and anxiety-like behaviors. Moreover, activation of PIC CaMKIIa-expressing neurons during inflammatory pain conditions exacerbated hyperalgesia and decreased pain tolerance. However, Chemogenetic activation did not alter heat nociception via hot plate latency or body temperature. Conversely, inhibiting CaMKIIa-expressing neurons did not alter either sensory or affective pain-like behaviors in non-injured or under inflammatory pain conditions, but it did decrease body temperature and decreased hot plate latency. Our findings reveal that PIC CaMKIIa-expressing neurons are a critical hub for producing both sensory and affective pain-like behaviors and important for thermoregulatory processing. PERSPECTIVE: The present study reveals that activation of the posterior insula produces hyperalgesia and negative affect, and has a role in thermal tolerance and thermoregulation. These findings highlight the insula as a key player in contributing to the multidimensionality of pain.
Collapse
Affiliation(s)
- Feni Kadakia
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Department of Anesthesiology and Pain Research Center, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Akansha Khadka
- Department of Anesthesiology and Pain Research Center, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Jake Yazell
- Department of Anesthesiology and Pain Research Center, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Steve Davidson
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Department of Anesthesiology and Pain Research Center, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
46
|
Goldstein N, Maes A, Allen HN, Nelson TS, Kruger KA, Kindel M, Smith NK, Carty JRE, Villari RE, Cho E, Marble EL, Khanna R, Taylor BK, Kennedy A, Betley JN. A parabrachial hub for the prioritization of survival behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582069. [PMID: 38464066 PMCID: PMC10925167 DOI: 10.1101/2024.02.26.582069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Long-term sustained pain in the absence of acute physical injury is a prominent feature of chronic pain conditions. While neurons responding to noxious stimuli have been identified, understanding the signals that persist without ongoing painful stimuli remains a challenge. Using an ethological approach based on the prioritization of adaptive survival behaviors, we determined that neuropeptide Y (NPY) signaling from multiple sources converges on parabrachial neurons expressing the NPY Y1 receptor to reduce sustained pain responses. Neural activity recordings and computational modeling demonstrate that activity in Y1R parabrachial neurons is elevated following injury, predicts functional coping behavior, and is inhibited by competing survival needs. Taken together, our findings suggest that parabrachial Y1 receptor-expressing neurons are a critical hub for endogenous analgesic pathways that suppress sustained pain states.
Collapse
|
47
|
Rankin G, Chirila AM, Emanuel AJ, Zhang Z, Woolf CJ, Drugowitsch J, Ginty DD. Nerve injury disrupts temporal processing in the spinal cord dorsal horn through alterations in PV + interneurons. Cell Rep 2024; 43:113718. [PMID: 38294904 PMCID: PMC11101906 DOI: 10.1016/j.celrep.2024.113718] [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: 03/15/2023] [Revised: 11/13/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
How mechanical allodynia following nerve injury is encoded in patterns of neural activity in the spinal cord dorsal horn (DH) remains incompletely understood. We address this in mice using the spared nerve injury model of neuropathic pain and in vivo electrophysiological recordings. Surprisingly, despite dramatic behavioral over-reactivity to mechanical stimuli following nerve injury, an overall increase in sensitivity or reactivity of DH neurons is not observed. We do, however, observe a marked decrease in correlated neural firing patterns, including the synchrony of mechanical stimulus-evoked firing, across the DH. Alterations in DH temporal firing patterns are recapitulated by silencing DH parvalbumin+ (PV+) interneurons, previously implicated in mechanical allodynia, as are allodynic pain-like behaviors. These findings reveal decorrelated DH network activity, driven by alterations in PV+ interneurons, as a prominent feature of neuropathic pain and suggest restoration of proper temporal activity as a potential therapeutic strategy to treat chronic neuropathic pain.
Collapse
Affiliation(s)
- Genelle Rankin
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Anda M Chirila
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alan J Emanuel
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Zihe Zhang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jan Drugowitsch
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Bai HH, Wang KL, Zeng XR, Li J, Li Y, Xu JY, Zhang Y, Jiang HF, Yang X, Suo ZW, Hu XD. GPR39 regulated spinal glycinergic inhibition and mechanical inflammatory pain. SCIENCE ADVANCES 2024; 10:eadj3808. [PMID: 38306424 PMCID: PMC10836721 DOI: 10.1126/sciadv.adj3808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024]
Abstract
G protein-coupled receptor 39 (GPR39) senses the change of extracellular divalent zinc ion and signals through multiple G proteins to a broad spectrum of downstream effectors. Here, we found that GPR39 was prevalent at inhibitory synapses of spinal cord somatostatin-positive (SOM+) interneurons, a mechanosensitive subpopulation that is critical for the conveyance of mechanical pain. GPR39 complexed specifically with inhibitory glycine receptors (GlyRs) and helped maintain glycinergic transmission in a manner independent of G protein signalings. Targeted knockdown of GPR39 in SOM+ interneurons reduced the glycinergic inhibition and facilitated the excitatory output from SOM+ interneurons to spinoparabrachial neurons that engaged superspinal neural circuits encoding both the sensory discriminative and affective motivational domains of pain experience. Our data showed that pharmacological activation of GPR39 or augmenting GPR39 interaction with GlyRs at the spinal level effectively alleviated the sensory and affective pain induced by complete Freund's adjuvant and implicated GPR39 as a promising therapeutic target for the treatment of inflammatory mechanical pain.
Collapse
Affiliation(s)
- Hu-Hu Bai
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- School of Life Science, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Kang-Li Wang
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiang-Ru Zeng
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jing Li
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yuan Li
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jia-Yu Xu
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yue Zhang
- School of Public Health, Gansu University of Chinese medicine, Lanzhou, Gansu 730000, P.R. China
| | - Hai-Feng Jiang
- School of Public Health, Gansu University of Chinese medicine, Lanzhou, Gansu 730000, P.R. China
| | - Xian Yang
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhan-Wei Suo
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiao-Dong Hu
- Department of Molecular Pharmacology, School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
49
|
Zhang M, Wu YE, Jiang M, Hong W. Cortical regulation of helping behaviour towards others in pain. Nature 2024; 626:136-144. [PMID: 38267578 PMCID: PMC10925558 DOI: 10.1038/s41586-023-06973-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
Humans and animals exhibit various forms of prosocial helping behaviour towards others in need1-3. Although previous research has investigated how individuals may perceive others' states4,5, the neural mechanisms of how they respond to others' needs and goals with helping behaviour remain largely unknown. Here we show that mice engage in a form of helping behaviour towards other individuals experiencing physical pain and injury-they exhibit allolicking (social licking) behaviour specifically towards the injury site, which aids the recipients in coping with pain. Using microendoscopic imaging, we found that single-neuron and ensemble activity in the anterior cingulate cortex (ACC) encodes others' state of pain and that this representation is different from that of general stress in others. Furthermore, functional manipulations demonstrate a causal role of the ACC in bidirectionally controlling targeted allolicking. Notably, this behaviour is represented in a population code in the ACC that differs from that of general allogrooming, a distinct type of prosocial behaviour elicited by others' emotional stress. These findings advance our understanding of the neural coding and regulation of helping behaviour.
Collapse
Affiliation(s)
- Mingmin Zhang
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ye Emily Wu
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mengping Jiang
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Weizhe Hong
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
50
|
Torres-Rodriguez JM, Wilson TD, Singh S, Torruella-Suárez ML, Chaudhry S, Adke AP, Becker JJ, Neugebauer B, Lin JL, Martinez Gonzalez S, Soler-Cedeño O, Carrasquillo Y. The parabrachial to central amygdala pathway is critical to injury-induced pain sensitization in mice. Neuropsychopharmacology 2024; 49:508-520. [PMID: 37542159 PMCID: PMC10789863 DOI: 10.1038/s41386-023-01673-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/27/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
The spino-ponto-amygdaloid pathway is a major ascending circuit relaying nociceptive information from the spinal cord to the brain. Potentiation of excitatory synaptic transmission in the parabrachial nucleus (PBN) to central amygdala (CeA) pathway has been reported in rodent models of persistent pain. However, the functional significance of this pathway in the modulation of the somatosensory component of pain was recently challenged by studies showing that spinal nociceptive neurons do not target CeA-projecting PBN cells and that manipulations of this pathway have no effect on reflexive-defensive somatosensory responses to peripheral noxious stimulation. Here, we showed that activation of CeA-projecting PBN neurons is critical to increase both stimulus-evoked and spontaneous nociceptive responses following an injury in male and female mice. Using optogenetic-assisted circuit mapping, we confirmed a functional excitatory projection from PBN→CeA that is independent of the genetic or firing identity of CeA cells. We then showed that peripheral noxious stimulation increased the expression of the neuronal activity marker Fos in CeA-projecting PBN neurons and that chemogenetic inactivation of these cells decreased behavioral hypersensitivity in models of neuropathic and inflammatory pain without affecting baseline nociception. Lastly, we showed that chemogenetic activation of CeA-projecting PBN neurons is sufficient to induced bilateral hypersensitivity without injury. Together, our results indicate that the PBN→CeA pathway is a key modulator of pain-related behaviors that can increase reflexive-defensive and affective-motivational responses to somatosensory stimulation in injured states without affecting nociception under normal physiological conditions.
Collapse
Affiliation(s)
- Jeitzel M Torres-Rodriguez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Torri D Wilson
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Sudhuman Singh
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Maria L Torruella-Suárez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Sarah Chaudhry
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Anisha P Adke
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Jordan J Becker
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Neugebauer
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Jenny L Lin
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Santiago Martinez Gonzalez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Omar Soler-Cedeño
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Yarimar Carrasquillo
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA.
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|