1
|
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; 28:1012-1023. [PMID: 40128392 DOI: 10.1038/s41593-025-01921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
2
|
Ghitani N, von Buchholtz LJ, MacDonald DI, Falgairolle M, Nguyen MQ, Licholai JA, Ryba NJP, Chesler AT. A distributed coding logic for thermosensation and inflammatory pain. Nature 2025:10.1038/s41586-025-08875-6. [PMID: 40269164 DOI: 10.1038/s41586-025-08875-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/06/2025] [Indexed: 04/25/2025]
Abstract
Somatosensory neurons encode detailed information about touch and temperature and are the peripheral drivers of pain1,2. Here by combining functional imaging with multiplexed in situ hybridization3, we determined how heat and mechanical stimuli are encoded across neuronal classes and how inflammation transforms this representation to induce heat hypersensitivity, mechanical allodynia and continuing pain. Our data revealed that trigeminal neurons innervating the cheek exhibited complete segregation of responses to gentle touch and heat. By contrast, heat and noxious mechanical stimuli broadly activated nociceptor classes, including cell types proposed to trigger select percepts and behaviours4-6. Injection of the inflammatory mediator prostaglandin E2 caused long-lasting activity and thermal sensitization in select classes of nociceptors, providing a cellular basis for continuing inflammatory pain and heat hypersensitivity. We showed that the capsaicin receptor TRPV1 (ref. 7) has a central role in heat sensitization but not in spontaneous nociceptor activity. Unexpectedly, the responses to mechanical stimuli were minimally affected by inflammation, suggesting that tactile allodynia results from the continuing firing of nociceptors coincident with touch. Indeed, we have demonstrated that nociceptor activity is both necessary and sufficient for inflammatory tactile allodynia. Together, these findings refine models of sensory coding and discrimination at the cellular and molecular levels, demonstrate that touch and temperature are broadly but differentially encoded across transcriptomically distinct populations of sensory cells and provide insight into how cellular-level responses are reshaped by inflammation to trigger diverse aspects of pain.
Collapse
Affiliation(s)
- Nima Ghitani
- National Center for Complementary and Integrative Health, Bethesda, MD, USA
| | | | | | | | - Minh Q Nguyen
- National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - Julia A Licholai
- National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - Nicholas J P Ryba
- National Institute of Dental and Craniofacial Research, Bethesda, MD, USA.
| | - Alexander T Chesler
- National Center for Complementary and Integrative Health, Bethesda, MD, USA.
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
3
|
Upadhyay A, Gradwell MA, Vajtay TJ, Conner J, Sanyal AA, Azadegan C, Patel KR, Thackray JK, Bohic M, Imai F, Ogundare SO, Yoshida Y, Abdus-Saboor I, Azim E, Abraira VE. The dorsal column nuclei scale mechanical sensitivity in naive and neuropathic pain states. Cell Rep 2025; 44:115556. [PMID: 40202848 PMCID: PMC12093272 DOI: 10.1016/j.celrep.2025.115556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
During pathological conditions, tactile stimuli can aberrantly engage nociceptive pathways leading to the perception of touch as pain, known as mechanical allodynia. The brain stem dorsal column nuclei integrate tactile inputs, yet their role in mediating tactile sensitivity and allodynia remains understudied. We found that gracile nucleus (Gr) inhibitory interneurons and thalamus-projecting neurons are differentially innervated by primary afferents and spinal inputs. Functional manipulations of these distinct Gr neuronal populations bidirectionally shifted tactile sensitivity but did not affect noxious mechanical or thermal sensitivity. During neuropathic pain, Gr neurons exhibited increased sensory-evoked activity and asynchronous excitatory drive from primary afferents. Silencing Gr projection neurons or activating Gr inhibitory neurons in neuropathic mice reduced tactile hypersensitivity, and enhancing inhibition ameliorated paw-withdrawal signatures of neuropathic pain and induced conditioned place preference. These results suggest that Gr activity contributes to tactile sensitivity and affective, pain-associated phenotypes of mechanical allodynia.
Collapse
Affiliation(s)
- Aman Upadhyay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA; Graduate Program in Neuroscience, Rutgers Robert Wood Johnson Medical School, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA
| | - Mark A Gradwell
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| | - Thomas J Vajtay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - James Conner
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Arnab A Sanyal
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Chloe Azadegan
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Komal R Patel
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Joshua K Thackray
- Human Genetics Institute of New Jersey, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA
| | - Manon Bohic
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| | - Fumiyasu Imai
- Burke Neurological Institute, White Plains, NY, USA; Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Simon O Ogundare
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Yutaka Yoshida
- Burke Neurological Institute, White Plains, NY, USA; Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Eiman Azim
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Victoria E Abraira
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
4
|
Yarmolinsky DA, Zeng X, MacKinnon-Booth N, Greene CA, Kim C, Cheng YT, Lenfers Turnes B, Woolf CJ. Differential modification of ascending spinal outputs in acute and chronic pain states. Neuron 2025; 113:1223-1239.e5. [PMID: 40023166 PMCID: PMC12005971 DOI: 10.1016/j.neuron.2025.01.031] [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: 04/08/2024] [Revised: 11/08/2024] [Accepted: 01/31/2025] [Indexed: 03/04/2025]
Abstract
Pain hypersensitivity arises from the induction of plasticity in peripheral and spinal somatosensory neurons, which modifies nociceptive input to the brain, altering pain perception. We applied longitudinal calcium imaging of spinal dorsal projection neurons to determine whether and how the representation of somatosensory stimuli in the anterolateral tract, the principal pathway transmitting nociceptive signals to the brain, changes between distinct pain states. In healthy mice, we identified stable outputs selective for cooling or warming and a neuronal ensemble activated by noxious thermal and mechanical stimuli. Induction of acute peripheral sensitization by topical capsaicin transiently re-tuned nociceptive output neurons to encode low-intensity stimuli. In contrast, peripheral nerve injury resulted in a persistent suppression of innocuous spinal outputs coupled with persistent activation of a normally silent population of high-threshold neurons. These results demonstrate differential modulation of spinal outputs to the brain during nociceptive and neuropathic pain states.
Collapse
Affiliation(s)
- David A Yarmolinsky
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Xiangsunze Zeng
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Caitlin A Greene
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Chloe Kim
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yu-Ting Cheng
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Bruna Lenfers Turnes
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
5
|
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; 174:106145. [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)
- Patricia Tezanos
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain; PhD Program in Neuroscience, Universidad Autónoma de Madrid-Instituto Cajal, Madrid 28002, Spain
| | - José Luis Trejo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain.
| |
Collapse
|
6
|
Abd El Hay MY, Kamm GB, Tlaie Boria A, Siemens J. Diverging roles of TRPV1 and TRPM2 in warm-temperature detection. eLife 2025; 13:RP95618. [PMID: 40215103 PMCID: PMC11991700 DOI: 10.7554/elife.95618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025] Open
Abstract
The perception of innocuous temperatures is crucial for thermoregulation. The TRP ion channels TRPV1 and TRPM2 have been implicated in warmth detection, yet their precise roles remain unclear. A key challenge is the low prevalence of warmth-sensitive sensory neurons, comprising fewer than 10% of rodent dorsal root ganglion (DRG) neurons. Using calcium imaging of >20,000 cultured mouse DRG neurons, we uncovered distinct contributions of TRPV1 and TRPM2 to warmth sensitivity. TRPV1's absence - and to a lesser extent absence of TRPM2 - reduces the number of neurons responding to warmth. Additionally, TRPV1 mediates the rapid, dynamic response to a warmth challenge. Behavioural tracking in a whole-body thermal preference assay revealed that these cellular differences shape nuanced thermal behaviours. Drift diffusion modelling of decision-making in mice exposed to varying temperatures showed that TRPV1 deletion impairs evidence accumulation, reducing the precision of thermal choice, while TRPM2 deletion increases overall preference for warmer environments that wildtype mice avoid. It remains unclear whether TRPM2 in DRG sensory neurons or elsewhere mediates thermal preference. Our findings suggest that different aspects of thermal information, such as stimulation speed and temperature magnitude, are encoded by distinct TRP channel mechanisms.
Collapse
Affiliation(s)
- Muad Y Abd El Hay
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
- Ernst Strüngmann Institute for Neuroscience in Cooperation with the Max Planck SocietyFrankfurt am MainGermany
| | - Gretel B Kamm
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
| | - Alejandro Tlaie Boria
- Ernst Strüngmann Institute for Neuroscience in Cooperation with the Max Planck SocietyFrankfurt am MainGermany
| | - Jan Siemens
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| |
Collapse
|
7
|
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
|
8
|
Rivera-Arconada I, Baccei ML, López-García JA, Bardoni R. An electrophysiologist's guide to dorsal horn excitability and pain. Front Cell Neurosci 2025; 19:1548252. [PMID: 40241846 PMCID: PMC12001243 DOI: 10.3389/fncel.2025.1548252] [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: 12/19/2024] [Accepted: 03/13/2025] [Indexed: 04/18/2025] Open
Abstract
The dorsal horn of the spinal cord represents the first site in the central nervous system (CNS) where nociceptive signals are integrated. As a result, there has been a rapid growth in the number of studies investigating the ionic mechanisms regulating the excitability of dorsal horn neurons under normal and pathological conditions. We believe that it is time to look back and to critically examine what picture emerges from this wealth of studies. What are the actual types of neurons described in the literature based on electrophysiological criteria? Are these electrophysiologically-defined subpopulations strongly linked to specific morphological, functional, or molecular traits? Are these electrophysiological properties stable, or can they change during development or in response to peripheral injury? Here we provide an in-depth overview of both early and recent publications that explore the factors influencing dorsal horn neuronal excitability (including intrinsic membrane properties and synaptic transmission), how these factors vary across distinct subtypes of dorsal horn neurons, and how such factors are altered by peripheral nerve or tissue damage. The meta-research presented below leads to the conclusion that the dorsal horn is comprised of highly heterogeneous subpopulations in which the observed electrophysiological properties of a given neuron often fail to easily predict other properties such as biochemical phenotype or morphology. This highlights the need for future studies which can more fully interrogate the properties of dorsal horn neurons in a multi-modal manner.
Collapse
Affiliation(s)
| | - Mark L. Baccei
- Department of Anesthesiology, Pain Research Center, University of Cincinnati, Cincinnati, OH, United States
| | | | - Rita Bardoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
9
|
Liu D, Rahman M, Johnson A, Amo R, Tsutsui-Kimura I, Sullivan ZA, Pena N, Talay M, Logeman BL, Finkbeiner S, Qian L, Choi S, Capo-Battaglia A, Abdus-Saboor I, Ginty DD, Uchida N, Watabe-Uchida M, Dulac C. A hypothalamic circuit underlying the dynamic control of social homeostasis. Nature 2025; 640:1000-1010. [PMID: 40011768 PMCID: PMC12018270 DOI: 10.1038/s41586-025-08617-8] [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: 05/11/2023] [Accepted: 01/09/2025] [Indexed: 02/28/2025]
Abstract
Social grouping increases survival in many species, including humans1,2. By contrast, social isolation generates an aversive state ('loneliness') that motivates social seeking and heightens social interaction upon reunion3-5. The observed rebound in social interaction triggered by isolation suggests a homeostatic process underlying the control of social need, similar to physiological drives such as hunger, thirst or sleep3,6. In this study, we assessed social responses in several mouse strains, among which FVB/NJ mice emerged as highly, and C57BL/6J mice as moderately, sensitive to social isolation. Using both strains, we uncovered two previously uncharacterized neuronal populations in the hypothalamic preoptic nucleus that are activated during either social isolation or social rebound and orchestrate the behaviour display of social need and social satiety, respectively. We identified direct connectivity between these two populations and with brain areas associated with social behaviour, emotional state, reward and physiological needs and showed that mice require touch to assess the presence of others and fulfil their social need. These data show a brain-wide neural system underlying social homeostasis and provide significant mechanistic insights into the nature and function of circuits controlling instinctive social need and for the understanding of healthy and diseased brain states associated with social context.
Collapse
Affiliation(s)
- Ding Liu
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Mostafizur Rahman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Autumn Johnson
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Iku Tsutsui-Kimura
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Zuri A Sullivan
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Nicolai Pena
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Mustafa Talay
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Brandon L Logeman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Samantha Finkbeiner
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Lechen Qian
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Seungwon Choi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Athena Capo-Battaglia
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Ishmail Abdus-Saboor
- Department of Biological Sciences, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
10
|
Zhang L, Zhou S, Tan YF, Gan QF, Koon TH, Wang Z, Zhao S, Chen Y, Sun Y, Leong PP. Collateral projections from the lateral parabrachial nucleus to the bed nucleus of the stria terminalis and the central amygdala in mice. Neurosci Lett 2025; 853:138204. [PMID: 40113070 DOI: 10.1016/j.neulet.2025.138204] [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: 12/21/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND The lateral parabrachial nucleus (LPBn) serves a critical hub for processing and transmitting pain signals. It has two main efferent pathways, the LPBn-CeA and LPBn-BNST, which are crucial for pain regulation and the management of negative emotions. METHODS In our study, we investigated the projections from the LPBn by performing stereotaxic injections of AAV2/2Retro-hSyn-EGFP (abbreviated as EGFP) into the bed nucleus of the stria terminalis (BNST) and AAV2/2Retro-hSyn-tdTomato (abbreviated as tdTomato) into the central amygdala (CeA) in mice. The animals subsequently underwent spared nerve injury (SNI) surgery on the contralateral side to the AAV injections. To examine the expression of calcitonin gene-related peptide (CGRP) and c-Fos, we conducted immunofluorescent histochemistry. RESULTS Our results indicated that approximately 26 % of the LPBn neurons retrogradely labeled with either EGFP or tdTomato were dual-labeled with both markers. Moreover, a significant majority (85.49 %) of these double-labeled neurons were CGRP positive (CGRP+). In mice subjected to SNI, nearly all of these neurons (93.25 %) were c-Fos positive (c-Fos+), indicating that they were activated. CONCLUSION These findings suggest that a subset of CGRP+ neurons in the LPBn projects to both the BNST and CeA via axon collaterals. Notably, under SNI conditions, these neurons may play a critical role in the transmission of chronic pain.
Collapse
Affiliation(s)
- Li Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China; Pre-Clinical Sciences Department, Faculty of Medicine and Health Sciences, UTAR Sg Long Campus, Selangor, Malaysia
| | - Shuai Zhou
- Department of Human Anatomy, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China
| | | | - Quan Fu Gan
- Pre-Clinical Sciences Department, Faculty of Medicine and Health Sciences, UTAR Sg Long Campus, Selangor, Malaysia.
| | - Teoh Hoon Koon
- Pre-Clinical Sciences Department, Faculty of Medicine and Health Sciences, UTAR Sg Long Campus, Selangor, Malaysia.
| | - Zhiqiang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China.
| | - Shiqing Zhao
- First School of Clinical Medical Sciences, Binzhou Medical University, Binzhou, Shandong, China.
| | - Yixuan Chen
- First School of Clinical Medical Sciences, Binzhou Medical University, Binzhou, Shandong, China.
| | - Yi Sun
- Department of Human Anatomy, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China.
| | - Pooi Pooi Leong
- Pre-Clinical Sciences Department, Faculty of Medicine and Health Sciences, UTAR Sg Long Campus, Selangor, Malaysia.
| |
Collapse
|
11
|
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] [Download PDF] [Figures] [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
|
12
|
Zhang Y, Shi S, Mao E, Chen Y, Chen J, Tian M, Huang F, Cai Z, Li Y, Kou Z. Tac1-expressing neurons in the central amygdala predominantly mediate histamine-induced itch by receiving inputs from parabrachial Tac1-expressing neurons. Brain Res 2025; 1851:149492. [PMID: 39914640 DOI: 10.1016/j.brainres.2025.149492] [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: 11/29/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/12/2025]
Abstract
Itch is a distinct and bothersome sensation closely associated with a strong urge to scratch. Both the parabrachial nucleus (PBN) and the central amygdala (CeA) are responsive to itch stimuli and contain neurons that express tachykinin 1 (Tac1), which are known for their significant involvement in itch-induced scratching at both spinal and supraspinal levels. Significantly, the PBN neurons project their axons to form close connections with the CeA neurons. However, the role of the PBNTac1-CeATac1 pathway in modulating itch remains to be determined. We utilized immunohistochemistry, fiber photometry, chemogenetic, and behavioral techniques to investigate the role of the PBNTac1-CeATac1 pathway in itch. Our results indicate that neurons in the CeA can be more activated by acute itch than chronic itch. Notably, in response to acute itch stimuli, both CeATac1 and PBNTac1 neurons were specifically activated by histamine (His)-induced itch. Furthermore, the Tac1-positive terminals from the PBNTac1 neurons formed close connections with CeATac1 neurons. We also demonstrated that activating the PBNTac1-CeA pathway using a chemogenetic approach could increase scratching behaviors in His-induced itch, other than chloroquine (CQ)-induced itch. Conversely, inhibiting the PBNTac1-CeA pathway decreased scratching behaviors in mice with His-induced itch. Taken together, these results suggest that the PBNTac1-CeATac1 pathway may play a specific role in modulating His-induced acute itch.
Collapse
Affiliation(s)
- Yingning Zhang
- Department of Human Anatomy, Baotou Medical College, Baotou 014040 China; Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China
| | - Sujuan Shi
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China
| | - E Mao
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China
| | - Yuling Chen
- Department of Human Anatomy, Baotou Medical College, Baotou 014040 China; Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China
| | - Jing Chen
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China
| | - Miao Tian
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China
| | - Fensheng Huang
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China
| | - Zhiping Cai
- Department of Human Anatomy, Baotou Medical College, Baotou 014040 China.
| | - Yunqing Li
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China; Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006 China; Department of Anatomy, College of Basic Medicine, Dali University, Dali 671000 China.
| | - Zhenzhen Kou
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032 China; Department of Anatomy, College of Basic Medicine, Dali University, Dali 671000 China.
| |
Collapse
|
13
|
Tran EL, Stuedemann SA, Ridlon M, Link OD, Keil Stietz KP, Crawford LK. Genetic tools that target mechanoreceptors produce reliable labeling of bladder afferents and altered mechanosensation. Am J Physiol Renal Physiol 2025; 328:F360-F374. [PMID: 39611874 DOI: 10.1152/ajprenal.00151.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/28/2024] [Accepted: 11/15/2024] [Indexed: 02/25/2025] Open
Abstract
Mechanosensitive neurons are important sensors of bladder distention, but their role in urologic disease remains unclear. Our current knowledge about how disease alters bladder sensation comes from studies that focus primarily on peptidergic nociceptors, leaving our understanding of neuropeptide-negative mechanoreceptors incomplete. In this study, we found that a substantial proportion of neurofilament heavy (NFH)-positive A-fibers innervating the bladder was calcitonin gene-related peptide (CGRP)-negative, potentially representing uncharacterized mechanoreceptors. We then identified two genetic strategies that label mechanoreceptors in mouse skin and confirmed that they likewise label bladder afferents. Cre-mediated tdTomato reporter expression driven by tropomyosin receptor kinase B (TrkB), which labels Aδ mechanoreceptors in the skin, successfully labeled bladder nerve terminals. The majority of TrkB bladder afferents were CGRP-negative and NFH-positive, with more characteristic staining patterns seen at the level of the cell body. The Ret proto-oncogene (Ret) also produced robust labeling of bladder afferents, where colocalization with CGRP and NFH was consistent with multiple afferent subtypes. Because TrkB labeling was more specific for putative mechanoreceptors, we directly tested the role of TrkB neurons in bladder mechanosensation in vivo. Using an intersectional genetic strategy, we selectively ablated TrkB afferents and measured bladder responses to mechanical distention using anesthetized cystometry. Compared with controls, mice with ablated TrkB afferents required higher distention pressure to elicit voids. Interestingly, after ablation, distention also increased the frequency of nonvoiding contractions, a poorly understood phenotype of several urologic diseases. These genetic strategies comprise critical new tools to advance the study of mechanoreceptors in bladder function and urologic disease pathophysiology.NEW & NOTEWORTHY Most mechanosensitive afferents do not express markers of peptidergic nociceptors and therefore remain largely overlooked in studies of bladder dysfunction and disease. TrkB-mediated labeling of putative Aδ mechanoreceptors emerged as a valuable tool for the study of neuropeptide-negative bladder afferents with a confirmed role in bladder mechanosensation. Targeted neuronal ablation likewise validated an intersectional genetic strategy that can now directly test the role of TrkB mechanoreceptors in bladder physiology and disease.
Collapse
Affiliation(s)
- Emily L Tran
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - Sara A Stuedemann
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - Monica Ridlon
- Department of Comparative Biosciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - Olivia D Link
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - Kimberly P Keil Stietz
- Department of Comparative Biosciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| | - LaTasha K Crawford
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin, United States
| |
Collapse
|
14
|
Maric S, Hasan M, Pounder ML, Graham BA, Browne TJ. A Viral Labelling Study of Spinal Trigeminal Nucleus Caudalis Projection Neurons Targeting the Parabrachial Nucleus. J Neurochem 2025; 169:e70028. [PMID: 40050251 PMCID: PMC11885192 DOI: 10.1111/jnc.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/09/2025]
Abstract
Projection neurons (PNs) in the Spinal Trigeminal Nucleus Caudalis (Sp5C) relay orofacial nociceptive information to higher brain regions such as the thalamus and the parabrachial nucleus (PBN). Our understanding of Sp5C PN organisation and function has advanced less than the parallel spinal cord output system despite their corresponding roles for transmission of nociceptive signals from the orofacial region and body respectively. Viral vectors are an established approach for studying circuit connectivity in the nervous system, but different serotypes are known to produce variable results across circuits. As such, we sought to validate the utility of two common viral serotypes in spinal PN research: retrograde adeno-associated virus serotype 2 (rgAAV) and adeno-associated virus serotype 9 (AAV9), for identifying and analysing Sp5C PNs that project to the PBN. Following unilateral injections of either viral serotype into the PBN, many Sp5C projection neurons were retrogradely labelled. For both serotypes, these injections labelled Sp5C PNs bilaterally with a strong bias to the ipsilateral Sp5C. Within Sp5C, similar levels of PN labelling were present in both superficial and deep regions, contrasting previous work in spinal PNs that showed greater labelling by AAV9 versus rgAAV. Comparisons of the age dependence of labelling showed greater retrograde labelling of Sp5C projection neurons when injections were made in young adult animals. Finally, we demonstrate successful Cre-dependent recombination to selectively express channelrhodopsin-2 in Sp5C projection neurons. Together, these experiments show that rgAAV and AAV9 produce strong Sp5C PN transduction and provide a basis for future study of the afferent and efferent functions of the Sp5C PN population in health and disease.
Collapse
Affiliation(s)
- Sophie Maric
- School of Biomedical Sciences & Pharmacy, Faculty of HealthUniversity of NewcastleCallaghanAustralia
- Hunter Medical Research Institute (HMRI)New Lambton HeightsNewcastleNew South WalesAustralia
| | - Mehedi Hasan
- School of Biomedical Sciences & Pharmacy, Faculty of HealthUniversity of NewcastleCallaghanAustralia
- Hunter Medical Research Institute (HMRI)New Lambton HeightsNewcastleNew South WalesAustralia
| | - Madison L. Pounder
- School of Biomedical Sciences & Pharmacy, Faculty of HealthUniversity of NewcastleCallaghanAustralia
- Hunter Medical Research Institute (HMRI)New Lambton HeightsNewcastleNew South WalesAustralia
| | - Brett A. Graham
- School of Biomedical Sciences & Pharmacy, Faculty of HealthUniversity of NewcastleCallaghanAustralia
- Hunter Medical Research Institute (HMRI)New Lambton HeightsNewcastleNew South WalesAustralia
| | - Tyler J. Browne
- School of Biomedical Sciences & Pharmacy, Faculty of HealthUniversity of NewcastleCallaghanAustralia
- Hunter Medical Research Institute (HMRI)New Lambton HeightsNewcastleNew South WalesAustralia
| |
Collapse
|
15
|
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
|
16
|
Lucas-Romero J, Bandres MF, McPherson JG. Targeted inactivation of spinal α2 adrenoceptors promotes paradoxical anti-nociception. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636935. [PMID: 39975256 PMCID: PMC11839011 DOI: 10.1101/2025.02.06.636935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Noradrenergic drive from the brainstem to the spinal cord varies in a context-dependent manner to regulate the patterns of sensory and motor transmission that govern perception and action. In sensory networks, it is traditionally assumed that activation of spinal α2 receptors is anti-nociceptive, while spinal α2 blockade is pro-nociceptive. Here, however, we demonstrate in vivo in rats that targeted blockade of spinal α2 receptors can promote anti-nociception. The anti-nociceptive effects are not contingent upon supraspinal actions, as they persist below a chronic spinal cord injury and are enhanced by direct spinal application of antagonist. They are also evident throughout sensory-dominant, sensorimotor integrative, and motor-dominant regions of the gray matter, and neither global changes in spinal neural excitability nor off-target activation of spinal α1 adrenoceptors or 5HT 1A receptors abolished the anti-nociception. Together, these findings challenge the current understanding of noradrenergic modulation of spinal nociceptive transmission.
Collapse
|
17
|
Du L, Cheng H, Cui X, Cao Q, Li X, Wang S, Wang X, Liu Y, Zhu B, Gao X, Liu K. Mrgprb4-lineage neurons indispensable in pressure induced pleasant sensation are polymodal. iScience 2025; 28:111940. [PMID: 40034120 PMCID: PMC11872644 DOI: 10.1016/j.isci.2025.111940] [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: 09/25/2024] [Revised: 11/22/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Pharmacogenetic activation of the Mas-related G-protein-coupled receptor b4 (Mrgprb4) neurons in the dorsal root ganglia is positively reinforcing, and these neurons can be activated by innocuous or noxious mechanical stimuli. However, direct evidence regarding the role of these neurons and how they encode diverse somatic inputs remains unclear. To address this, the mild pressure conditioned place preference (MP-CPP) was conducted to evaluate the indispensability of Mrgprb4-lineage neurons in the pleasantness caused by pressure. Mice without Mrgprb4-lineage neurons lost the preference for pressure. The number of Mrgprb4-lineage neurons activated by pressure was significantly higher than that of brush and pinch. The Ca2+ transients activated by pressure and brush were higher than that of pinch. Further analysis of co-activating mechano-thermosensitive neurons showed that pressure evoked higher fluorescence than that of 0°C and 43°C. In brief, Mrgprb4-lineage neurons are needed to transmit pleasant sensation and exhibit functional polymodality.
Collapse
Affiliation(s)
- Longhua Du
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongyi Cheng
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiang Cui
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qianan Cao
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Xia Li
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuya Wang
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxi Wang
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yun Liu
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bing Zhu
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyan Gao
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kun Liu
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Melvin B, Wright R, McNally A, Elmofty D. Allodynia: A Review Article. Curr Pain Headache Rep 2025; 29:49. [PMID: 39964609 DOI: 10.1007/s11916-025-01370-7] [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] [Accepted: 02/10/2025] [Indexed: 05/10/2025]
Abstract
PURPOSE OF REVIEW Allodynia is characterized by a painful response to a non-noxious stimulus. This article reviews the pathophysiology, clinical presentation, differential diagnosis, diagnostic testing, and management approaches for the causes of allodynia. RECENT FINDINGS Allodynia remains difficult to evaluate and manage. Despite ongoing research, significant progress is still needed to optimize the management of allodynia. Allodynia is a debilitating condition that can be difficult to treat. Diagnostic modalities and treatment options are limited. Advancements in diagnostic and treatment options are necessary to improve patient care.
Collapse
Affiliation(s)
- Brittany Melvin
- Physical Medicine & Rehabilitation Resident, Department of Physical Medicine & Rehabilitation, Sinai Health System, Chicago, IL, USA
| | - Raven Wright
- Department of Anesthesia and Critical Care, University of Chicago Medical Center, Chicago, IL, USA
| | - Alexandra McNally
- Department of Anesthesia and Critical Care, University of Chicago Medical Center, Chicago, IL, USA
| | - Dalia Elmofty
- Department of Anesthesia and Critical Care, University of Chicago Medical Center, Chicago, IL, USA.
| |
Collapse
|
21
|
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
|
22
|
Yang M, Keller D, Dobolyi A, Valtcheva S. The lateral thalamus: a bridge between multisensory processing and naturalistic behaviors. Trends Neurosci 2025; 48:33-46. [PMID: 39672783 DOI: 10.1016/j.tins.2024.11.005] [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: 06/27/2024] [Revised: 10/14/2024] [Accepted: 11/19/2024] [Indexed: 12/15/2024]
Abstract
The lateral thalamus (LT) receives input from primary sensory nuclei and responds to multimodal stimuli. The LT is also involved in regulating innate and social behaviors through its projections to cortical and limbic networks. However, the importance of multisensory processing within the LT in modulating behavioral output has not been explicitly addressed. Here, we discuss recent findings primarily from rodent studies that extend the classical view of the LT as a passive relay, by underscoring its involvement in associating multimodal features and encoding the salience, valence, and social relevance of sensory signals. We propose that the primary function of the LT is to integrate sensory and non-sensory aspects of multisensory input to gate naturalistic behaviors.
Collapse
Affiliation(s)
- Mingyu Yang
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany
| | - Dávid Keller
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany; Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest 1094, Hungary
| | - Arpád Dobolyi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest 1094, Hungary; Department of Physiology and Neurobiology, Eotvos Lorand University, Budapest 1117, Hungary.
| | - Silvana Valtcheva
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne and University Clinic Cologne, Cologne 50931, Germany.
| |
Collapse
|
23
|
Yin G, Duan K, Dong D, Du F, Guo C, Zhang C, Liu X, Sun Y, Huang T, Cui G, Cheng L. Central control of opioid-induced mechanical hypersensitivity and tolerance in mice. Neuron 2024; 112:3897-3923.e10. [PMID: 39406237 DOI: 10.1016/j.neuron.2024.09.014] [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: 02/13/2024] [Revised: 07/18/2024] [Accepted: 09/16/2024] [Indexed: 12/07/2024]
Abstract
Repetitive use of morphine (MF) and other opioids can trigger two major pain-related side effects: opioid-induced hypersensitivity (OIH) and analgesic tolerance, which can be subclassified as mechanical and thermal. The central mechanisms underlying mechanical OIH/tolerance remain unresolved. Here, we report that a brain-to-spinal opioid pathway, starting from μ-opioid receptor (MOR)-expressing neuron in the lateral parabrachial nucleus (lPBNMOR+) via dynorphin (Dyn) neuron in the paraventricular hypothalamic nucleus (PVHDyn+) to κ-opioid receptor (KOR)-expressing GABAergic neuron in the spinal dorsal horn (SDHKOR-GABA), controls repeated systemic administration of MF-induced mechanical OIH and tolerance in mice. The above effect is likely mediated by disruption of dorsal horn gate control for MF-resistant mechanical pain via silencing of the Dyn-positive GABAergic neurons in the SDH (lPBNMOR+ → PVHDyn+ → SDHKOR-GABA → SDHDyn-GABA). Repetitive binding of MF to MORs during repeated MF administration disrupted the above circuits. Targeting the above brain-to-spinal opioid pathways rescued repetitive MF-induced mechanical OIH and tolerance.
Collapse
Affiliation(s)
- Guangjuan Yin
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kaifang Duan
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dong Dong
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Feng Du
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chao Guo
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Changyi Zhang
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xi Liu
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuanjie Sun
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tianwen Huang
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Guangfu Cui
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Longzhen Cheng
- Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
24
|
Zhang FC, Weng RX, Li D, Li YC, Dai XX, Hu S, Sun Q, Li R, Xu GY. A vagus nerve dominant tetra-synaptic ascending pathway for gastric pain processing. Nat Commun 2024; 15:9824. [PMID: 39537596 PMCID: PMC11561356 DOI: 10.1038/s41467-024-54056-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric pain has limited treatment options and the mechanisms within the central circuitry remain largely unclear. This study investigates the central circuitry in gastric pain induced by noxious gastric distension (GD) in mice. Here, we identified that the nucleus tractus solitarius (NTS) serves as the first-level center of gastric pain, primarily via the vagus nerve. The prelimbic cortex (PL) is engaged in the perception of gastric pain. The lateral parabrachial nucleus (LPB) and the paraventricular thalamic nucleus (PVT) are crucial regions for synaptic transmission from the NTS to the PL. The glutamatergic tetra-synaptic NTS-LPB-PVT-PL circuitry is necessary and sufficient for the processing of gastric pain. Overall, our finding reveals a glutamatergic tetra-synaptic NTS-LPB-PVT-PL circuitry that transmits gastric nociceptive signaling by the vagus nerve in mice. It provides an insight into the gastric pain ascending pathway and offers potential therapeutic targets for relieving visceral pain.
Collapse
Affiliation(s)
- Fu-Chao Zhang
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Rui-Xia Weng
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Di Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yong-Chang Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xiao-Xuan Dai
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Shufen Hu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Qian Sun
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Guang-Yin Xu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.
| |
Collapse
|
25
|
Zhang D, Turecek J, Choi S, Delisle M, Pamplona CL, Meltzer S, Ginty DD. C-LTMRs evoke wet dog shakes via the spinoparabrachial pathway. Science 2024; 386:686-692. [PMID: 39509513 PMCID: PMC11753759 DOI: 10.1126/science.adq8834] [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: 06/10/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024]
Abstract
Many hairy mammals perform rapid oscillations of their body, called wet dog shakes, to remove water and irritants from their back hairy skin. The somatosensory mechanisms that underlie this behavior are unclear. We report that Piezo2-dependent mechanosensation mediates wet dog shakes evoked by water or oil droplets applied to back hairy skin of mice. Unmyelinated C-fiber low-threshold mechanoreceptors (C-LTMRs) were activated by oil droplets, and their optogenetic activation elicited wet dog shakes. Ablation of C-LTMRs attenuated this behavior. Moreover, C-LTMRs synaptically couple to spinoparabrachial neurons, and optogenetically inhibiting spinoparabrachial neuron synapses and excitatory neurons in the parabrachial nucleus impaired both oil droplet- and C-LTMR-evoked wet dog shakes. Thus, a C-LTMR-spinoparabrachial pathway promotes wet dog shakes for removal of water and mechanical irritants from back hairy skin.
Collapse
Affiliation(s)
- Dawei Zhang
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Seungwon Choi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michelle Delisle
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Caroline Leal Pamplona
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Shan Meltzer
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - David D. Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
26
|
Mitchell JR, Vincelette L, Tuberman S, Sheppard V, Bergeron E, Calitri R, Clark R, Cody C, Kannan A, Keith J, Parakoyi A, Pikus M, Vance V, Ziane L, Brenhouse H, Laine MA, Shansky RM. Behavioral and neural correlates of diverse conditioned fear responses in male and female rats. Neurobiol Stress 2024; 33:100675. [PMID: 39391589 PMCID: PMC11465128 DOI: 10.1016/j.ynstr.2024.100675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Pavlovian fear conditioning is a widely used tool that models associative learning in rodents. For decades the field has used predominantly male rodents and focused on a sole conditioned fear response: freezing. However, recent work from our lab and others has identified darting as a female-biased conditioned response, characterized by an escape-like movement across a fear conditioning chamber. It is also accompanied by a behavioral phenotype: Darters reliably show decreased freezing compared to Non-darters and males and reach higher velocities in response to the foot shock ("shock response"). However, the relationship between shock response and conditioned darting is not known. This study investigated if this link is due to differences in general processing of aversive stimuli between Darters, Non-darters and males. Across a variety of modalities, including corticosterone measures, the acoustic startle test, and sensitivity to thermal pain, Darters were found not to be more reactive or sensitive to aversive stimuli, and, in some cases, they appear less reactive to Non-darters and males. Analyses of cFos activity in regions involved in pain and fear processing following fear conditioning identified discrete patterns of expression among Darters, Non-darters, and males exposed to low and high intensity foot shocks. The results from these studies further our understanding of the differences between Darters, Non-darters and males and highlight the importance of studying individual differences in fear conditioning as indicators of fear state.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rose Clark
- Northeastern University, Boston, MA, USA
| | | | | | - Jack Keith
- Northeastern University, Boston, MA, USA
| | | | | | | | | | | | - Mikaela A. Laine
- Northeastern University, Boston, MA, USA
- Smith College, Northampton, MA, USA
| | | |
Collapse
|
27
|
Browne TJ, Smith KM, Gradwell MA, Dayas CV, Callister RJ, Hughes DI, Graham BA. Lateral lamina V projection neuron axon collaterals connect sensory processing across the dorsal horn of the mouse spinal cord. Sci Rep 2024; 14:26354. [PMID: 39487174 PMCID: PMC11530558 DOI: 10.1038/s41598-024-73620-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 09/19/2024] [Indexed: 11/04/2024] Open
Abstract
Spinal projection neurons (PNs) are defined by long axons that travel from their origin in the spinal cord to the brain where they relay sensory information from the body. The existence and function of a substantial axon collateral network, also arising from PNs and remaining within the spinal cord, is less well appreciated. Here we use a retrograde viral transduction strategy to characterise a novel subpopulation of deep dorsal horn spinoparabrachial neurons. Brainbow assisted analysis confirmed that virally labelled PN cell bodies formed a discrete cell column in the lateral part of Lamina V (LVlat) and the adjoining white matter. These PNs exhibited large dendritic territories biased to regions lateral and ventral to the cell body column and extending considerable rostrocaudal distances. Optogenetic activation of LVLat PNs confirmed this population mediates widespread signalling within spinal cord circuits, including activation in the superficial dorsal horn. This signalling was also demonstrated with patch clamp recordings during LVLat PN photostimulation, with a range of direct and indirect connections identified and evidence of a postsynaptic population of inhibitory interneurons. Together, these findings confirm a substantial role for PNs in local spinal sensory processing, as well as relay of sensory signals to the brain.
Collapse
Affiliation(s)
- Tyler J Browne
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW, Australia.
| | - Kelly M Smith
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mark A Gradwell
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Christopher V Dayas
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW, Australia
| | - Robert J Callister
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW, Australia
| | - David I Hughes
- Institute of Neuroscience Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Brett A Graham
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW, Australia
| |
Collapse
|
28
|
Koo H, Wang J, Pariyar R, Hammond RM, La JH. Modulation of mechanosensation by endogenous dopaminergic signaling in the lateral parabrachial nucleus in mice. Pain Rep 2024; 9:e1186. [PMID: 39263005 PMCID: PMC11390053 DOI: 10.1097/pr9.0000000000001186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/09/2024] [Accepted: 06/18/2024] [Indexed: 09/13/2024] Open
Abstract
Introduction The lateral parabrachial nucleus (LPBN), a crucial hub for integrating and modulating diverse sensory information, is known to express both D1 and D2 dopamine receptors and receive dopaminergic inputs. However, the role of the LPBN's dopaminergic system in somatosensory processing remains largely unexplored. In this study, we investigated whether mechanical sensory stimulation triggers dopamine release in the LPBN and how D1- and D2-like receptor signaling in the LPBN influences mechanosensitivity in mice. Methods We used a G-protein-coupled receptor-based dopamine sensor to monitor dopamine release in the LPBN and a von Frey filament assay to measure the mechanical threshold for nocifensive withdrawal in mouse hind paws after unilateral microinjection of D1- or D2-like receptor antagonist into the LPBN. Results Noxious mechanical stimulation increased the dopamine sensor signal in the LPBN. Thresholds of nocifensive withdrawal from mechanical stimulation were decreased by the D1-like receptor antagonist SCH-23390 (0.1 µg) but increased by the D2-like receptor antagonist eticlopride (1 µg). In the intraplantar capsaicin injection model that develops mechanical hypersensitivity in the injected paw, the dopamine sensor signal in the LPBN was increased, and eticlopride (1 µg) in the LPBN significantly inhibited the capsaicin-induced mechanical hypersensitivity. Conclusions These results suggest that endogenous dopaminergic signaling occurs in the LPBN upon noxious mechanical stimulation, inhibiting mechanosensitivity through D1-like receptors while enhancing it through D2-like receptors. D2-like receptor signaling in the LPBN may contribute to an injury-induced increase in mechanical nociception, indicating that inhibiting the receptor within the LPBN could offer potential as a novel analgesic strategy.
Collapse
Affiliation(s)
- Ho Koo
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jigong Wang
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ramesh Pariyar
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Regan M Hammond
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jun-Ho La
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
29
|
Dedek A, Topcu E, Dedek C, McDermott JS, Krajewski JL, Tsai EC, Hildebrand ME. Heterogeneity of synaptic NMDA receptor responses within individual lamina I pain-processing neurons across sex in rats and humans. J Physiol 2024; 602:5309-5327. [PMID: 39316518 DOI: 10.1113/jp285521] [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: 08/18/2023] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Excitatory glutamatergic NMDA receptors (NMDARs) are key regulators of spinal pain processing, and yet the biophysical properties of NMDARs in dorsal horn nociceptive neurons remain poorly understood. Despite the clinical implications, it is unknown whether the molecular and functional properties of synaptic NMDAR responses are conserved between males and females or translate from rodents to humans. To address these translational gaps, we systematically compared individual and averaged excitatory synaptic responses from lamina I pain-processing neurons of adult Sprague-Dawley rats and human organ donors, including both sexes. By combining patch-clamp recordings of outward miniature excitatory postsynaptic currents with non-biased data analyses, we uncovered a wide range of decay constants of excitatory synaptic events within individual lamina I neurons. Decay constants of synaptic responses were distributed in a continuum from 1-20 ms to greater than 1000 ms, suggesting that individual lamina I neurons contain AMPA receptor (AMPAR)-only as well as GluN2A-, GluN2B- and GluN2D-NMDAR-dominated synaptic events. This intraneuronal heterogeneity in AMPAR- and NMDAR-mediated decay kinetics was observed across sex and species. However, we discovered an increased relative contribution of GluN2A-dominated NMDAR responses at human lamina I synapses compared with rodent synapses, suggesting a species difference relevant to NMDAR subunit-targeting therapeutic approaches. The conserved heterogeneity in decay rates of excitatory synaptic events within individual lamina I pain-processing neurons may enable synapse-specific forms of plasticity and sensory integration within dorsal horn nociceptive networks. KEY POINTS: Synaptic NMDA receptors (NMDARs) in spinal dorsal horn nociceptive neurons are key regulators of pain processing, but it is unknown whether their functional properties are conserved between males and females or translate from rodents to humans. In this study, we compared individual excitatory synaptic responses from lamina I pain-processing neurons of male and female adult Sprague-Dawley rats and human organ donors. Individual lamina I neurons from male and female rats and humans contain AMPA receptor-only as well as GluN2A, GluN2B- and GluN2D-NMDAR-dominated synaptic events. This may enable synapse-specific forms of plasticity and sensory integration within dorsal horn nociceptive networks. Human lamina I synapses have an increased relative contribution of GluN2A-dominated NMDAR responses compared with rodent synapses. These results uncover a species difference relevant to NMDAR subunit-targeting therapeutic approaches.
Collapse
Affiliation(s)
- Annemarie Dedek
- Department of Neuroscience, Carleton University, Ontario, Canada
- Neuroscience Program, Ottawa Hospital Research Institute, Ontario, Canada
- School of Pharmacy, University of Waterloo, Ontario, Canada
| | - Emine Topcu
- Department of Neuroscience, Carleton University, Ontario, Canada
| | | | - Jeff S McDermott
- Lilly Research Laboratories, Indianapolis, Indiana, United States
| | | | - Eve C Tsai
- Neuroscience Program, Ottawa Hospital Research Institute, Ontario, Canada
- Brain and Mind Research Institute, University of Ottawa, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, The Ottawa Hospital, Ontario, Canada
| | - Michael E Hildebrand
- Department of Neuroscience, Carleton University, Ontario, Canada
- Neuroscience Program, Ottawa Hospital Research Institute, Ontario, Canada
| |
Collapse
|
30
|
Sheahan TD, Warwick CA, Cui AY, Baranger DAA, Perry VJ, Smith KM, Manalo AP, Nguyen EK, Koerber HR, Ross SE. Kappa opioids inhibit spinal output neurons to suppress itch. SCIENCE ADVANCES 2024; 10:eadp6038. [PMID: 39321286 PMCID: PMC11423883 DOI: 10.1126/sciadv.adp6038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/20/2024] [Indexed: 09/27/2024]
Abstract
Itch is a protective sensation that drives scratching. Although specific cell types have been proposed to underlie itch, the neural basis for itch remains unclear. Here, we used two-photon Ca2+ imaging of the dorsal horn to visualize neuronal populations that are activated by itch-inducing agents. We identify a convergent population of spinal interneurons recruited by diverse itch-causing stimuli that represents a subset of neurons that express the gastrin-releasing peptide receptor (GRPR). Moreover, we find that itch is conveyed to the brain via GRPR-expressing spinal output neurons that target the lateral parabrachial nuclei. We then show that the kappa opioid receptor agonist nalfurafine relieves itch by selectively inhibiting GRPR spinoparabrachial neurons. These experiments provide a population-level view of the spinal neurons that respond to pruritic stimuli, pinpoint the output neurons that convey itch to the brain, and identify the cellular target of kappa opioid receptor agonists for the inhibition of itch.
Collapse
Affiliation(s)
- Tayler D Sheahan
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles A Warwick
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abby Y Cui
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A A Baranger
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Vijay J Perry
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kelly M Smith
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison P Manalo
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eileen K Nguyen
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - H Richard Koerber
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah E Ross
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
31
|
Tagini S, Varallo G, Capodaglio P, Mauro A, Scarpina F. The Hedonic Experience Associated with a Gentle Touch Is Preserved in Women with Fibromyalgia. J Clin Med 2024; 13:5649. [PMID: 39337136 PMCID: PMC11433436 DOI: 10.3390/jcm13185649] [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: 07/27/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Although manual therapies can be used for pain alleviation in fibromyalgia, there is no clear evidence about the processing of gentle, affective touch in this clinical condition. In fact, persistent painful sensations and psychological factors may impact the hedonic experience of touch. Methods: This observational cross-sectional study compared the subjective experience of affective touch between 14 women with fibromyalgia (age range: 35-70; range of years of education: 5-13) and 14 pain-free women (age range: 18-30; range of years of education: 13-19). The participants rated the pleasantness of slow and fast touches delivered by a brush, the experimenter's hand, and a plastic stick. Tactile stimuli were either imagined or real to disentangle the contribution of top-down and bottom-up sensory components. Additionally, a self-report questionnaire explored the lifetime experiences of affective touch. Results: Akin to healthy counterparts, individuals with fibromyalgia rated slow touches delivered by the experimenter's hand or a brush as more pleasant than fast touches, regardless of whether they were imagined or real. However, the intensity of pain affects only the imagined pleasantness in our participants with fibromyalgia. Furthermore, despite the fibromyalgia patients reporting fewer experiences of affective touch in childhood and adolescence, this evidence was not associated with the experimental outcomes. Conclusions: The hedonic experience of affective touch seems preserved in fibromyalgia despite poor intimate bodily contact in youth. We confirmed that bottom-up and top-down factors contribute to the affective touch perception in fibromyalgia: bodily pain may impact even more the expected pleasure than the actual experience. Future investigations may introduce neurophysiological measures of the implicit autonomic responses to affective touch in fibromyalgia. To conclude, although preliminary, our evidence may be in favor of manual therapies for pain relief in fibromyalgia.
Collapse
Affiliation(s)
- Sofia Tagini
- “Rita Levi Montalcini” Department of Neurosciences, University of Turin, Via Cherasco, 15, 10126 Turin, Italy; (S.T.); (A.M.)
- I.R.C.C.S. Istituto Auxologico Italiano, U.O. di Neurologia e Neuroriabilitazione, Ospedale San Giuseppe, Via Cadorna, 90, 28824 Piancavallo, VCO, Italy
| | - Giorgia Varallo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Paolo Capodaglio
- I.R.C.C.S. Istituto Auxologico Italiano, U.O. di Riabilitazione Osteoarticolare, Ospedale San Giuseppe, 28824 Piancavallo, VCO, Italy;
- Department of Surgical Sciences, Physical and Rehabilitation Medicine, University of Turin, 10126 Turin, Italy
| | - Alessandro Mauro
- “Rita Levi Montalcini” Department of Neurosciences, University of Turin, Via Cherasco, 15, 10126 Turin, Italy; (S.T.); (A.M.)
- I.R.C.C.S. Istituto Auxologico Italiano, U.O. di Neurologia e Neuroriabilitazione, Ospedale San Giuseppe, Via Cadorna, 90, 28824 Piancavallo, VCO, Italy
| | - Federica Scarpina
- “Rita Levi Montalcini” Department of Neurosciences, University of Turin, Via Cherasco, 15, 10126 Turin, Italy; (S.T.); (A.M.)
- I.R.C.C.S. Istituto Auxologico Italiano, U.O. di Neurologia e Neuroriabilitazione, Ospedale San Giuseppe, Via Cadorna, 90, 28824 Piancavallo, VCO, Italy
| |
Collapse
|
32
|
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
|
33
|
Hung 洪瑋辰 WC, Chen 陳志成 CC, Yen 嚴震東 CT, Min 閔明源 MY. Presynaptic Enhancement of Transmission from Nociceptors Expressing Nav1.8 onto Lamina-I Spinothalamic Tract Neurons by Spared Nerve Injury in Mice. eNeuro 2024; 11:ENEURO.0087-24.2024. [PMID: 39256039 PMCID: PMC11391502 DOI: 10.1523/eneuro.0087-24.2024] [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: 03/01/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024] Open
Abstract
Alteration of synaptic function in the dorsal horn (DH) has been implicated as a cellular substrate for the development of neuropathic pain, but certain details remain unclear. In particular, the lack of information on the types of synapses that undergo functional changes hinders the understanding of disease pathogenesis from a synaptic plasticity perspective. Here, we addressed this issue by using optogenetic and retrograde tracing ex vivo to selectively stimulate first-order nociceptors expressing Nav1.8 (NRsNav1.8) and record the responses of spinothalamic tract neurons in spinal lamina I (L1-STTNs). We found that spared nerve injury (SNI) increased excitatory postsynaptic currents (EPSCs) in L1-STTNs evoked by photostimulation of NRsNav1.8 (referred to as Nav1.8-STTN EPSCs). This effect was accompanied by a significant change in the failure rate and paired-pulse ratio of synaptic transmission from NRsNav1.8 to L1-STTN and in the frequency (not amplitude) of spontaneous EPSCs recorded in L1-STTNs. However, no change was observed in the ratio of AMPA to NMDA receptor-mediated components of Nav1.8-STTN EPSCs or in the amplitude of unitary EPSCs constituting Nav1.8-STTN EPSCs recorded with extracellular Ca2+ replaced by Sr2+ In addition, there was a small increase (approximately 10%) in the number of L1-STTNs showing immunoreactivity for phosphorylated extracellular signal-regulated kinases in mice after SNI compared with sham. Similarly, only a small percentage of L1-STTNs showed a lower action potential threshold after SNI. In conclusion, our results show that SNI induces presynaptic modulation at NRNav1.8 (consisting of both peptidergic and nonpeptidergic nociceptors) synapses on L1-STTNs forming the lateral spinothalamic tract.
Collapse
Affiliation(s)
- Wei-Chen Hung 洪瑋辰
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Neurobiology and Cognitive Science Centre, National Taiwan University, Taipei 10617, Taiwan
| | | | - Cheng-Tung Yen 嚴震東
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Neurobiology and Cognitive Science Centre, National Taiwan University, Taipei 10617, Taiwan
| | - Ming-Yuan Min 閔明源
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Neurobiology and Cognitive Science Centre, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
34
|
Mitchell JR, Vincelette L, Tuberman S, Sheppard V, Bergeron E, Calitri R, Clark R, Cody C, Kannan A, Keith J, Parakoyi A, Pikus M, Vance V, Ziane L, Brenhouse H, Laine MA, Shansky RM. Behavioral and neural correlates of diverse conditioned fear responses in male and female rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608817. [PMID: 39229164 PMCID: PMC11370446 DOI: 10.1101/2024.08.20.608817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Pavlovian fear conditioning is a widely used tool that models associative learning in rodents. For decades the field has used predominantly male rodents and focused on a sole conditioned fear response: freezing. However, recent work from our lab and others has identified darting as a female-biased conditioned response, characterized by an escape-like movement across a fear conditioning chamber. It is also accompanied by a behavioral phenotype: Darters reliably show decreased freezing compared to Non-darters and males and reach higher velocities in response to the foot shock ("shock response"). However, the relationship between shock response and conditioned darting is not known. This study investigated if this link is due to differences in general processing of aversive stimuli between Darters, Non-darters and males. Across a variety of modalities, including corticosterone measures, the acoustic startle test, and sensitivity to thermal pain, Darters were found not to be more reactive or sensitive to aversive stimuli, and, in some cases, they appear less reactive to Non-darters and males. Analyses of cFos activity in regions involved in pain and fear processing following fear conditioning identified discrete patterns of expression among Darters, Non-darters, and males exposed to low and high intensity foot shocks. The results from these studies further our understanding of the differences between Darters, Non-darters and males and highlight the importance of studying individual differences in fear conditioning as indicators of fear state.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mikaela A. Laine
- Northeastern University, Boston, MA
- Smith College, Northampton, MA
| | | |
Collapse
|
35
|
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
|
36
|
Sun C, Fan Q, Xie R, Luo C, Hu B, Wang Q. Tetherless Optical Neuromodulation: Wavelength from Orange-red to Mid-infrared. Neurosci Bull 2024; 40:1173-1188. [PMID: 38372931 PMCID: PMC11306867 DOI: 10.1007/s12264-024-01179-1] [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: 07/06/2023] [Accepted: 11/11/2023] [Indexed: 02/20/2024] Open
Abstract
Optogenetics, a technique that employs light for neuromodulation, has revolutionized the study of neural mechanisms and the treatment of neurological disorders due to its high spatiotemporal resolution and cell-type specificity. However, visible light, particularly blue and green light, commonly used in conventional optogenetics, has limited penetration in biological tissue. This limitation necessitates the implantation of optical fibers for light delivery, especially in deep brain regions, leading to tissue damage and experimental constraints. To overcome these challenges, the use of orange-red and infrared light with greater tissue penetration has emerged as a promising approach for tetherless optical neuromodulation. In this review, we provide an overview of the development and applications of tetherless optical neuromodulation methods with long wavelengths. We first discuss the exploration of orange-red wavelength-responsive rhodopsins and their performance in tetherless optical neuromodulation. Then, we summarize two novel tetherless neuromodulation methods using near-infrared light: upconversion nanoparticle-mediated optogenetics and photothermal neuromodulation. In addition, we discuss recent advances in mid-infrared optical neuromodulation.
Collapse
Affiliation(s)
- Chao Sun
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Qi Fan
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Rougang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Bingliang Hu
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Quan Wang
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China.
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China.
| |
Collapse
|
37
|
Zhang D, Turecek J, Choi S, Delisle M, Pamplona CL, Meltzer S, Ginty DD. C-LTMRs mediate wet dog shakes via the spinoparabrachial pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.597395. [PMID: 38915692 PMCID: PMC11195135 DOI: 10.1101/2024.06.10.597395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Mammals perform rapid oscillations of their body- "wet dog shakes" -to remove water and irritants from their back hairy skin. The somatosensory mechanisms underlying this stereotypical behavior are unknown. We report that Piezo2-dependent mechanosensation mediates wet dog shakes evoked by water or oil droplets applied to hairy skin of mice. Unmyelinated low-threshold mechanoreceptors (C-LTMRs) were strongly activated by oil droplets and their optogenetic activation elicited wet dog shakes. Ablation of C-LTMRs attenuated this behavior. Moreover, C-LTMRs synaptically couple to spinoparabrachial (SPB) neurons, and optogenetically inhibiting SPB neuron synapses and excitatory neurons in the parabrachial nucleus impaired both oil droplet- and C-LTMR-evoked wet dog shakes. Thus, a C-LTMR- spinoparabrachial pathway mediates wet dog shakes for rapid and effective removal of foreign particles from back hairy skin.
Collapse
|
38
|
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
|
39
|
Maiarù M, Leese C, Silva-Hucha S, Fontana-Giusti S, Tait L, Tamagnini F, Davletov B, Hunt SP. Substance P-Botulinum Mediates Long-term Silencing of Pain Pathways that can be Re-instated with a Second Injection of the Construct in Mice. THE JOURNAL OF PAIN 2024; 25:104466. [PMID: 38218509 DOI: 10.1016/j.jpain.2024.01.331] [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: 07/11/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 01/15/2024]
Abstract
Chronic pain presents an enormous personal and economic burden and there is an urgent need for effective treatments. In a mouse model of chronic neuropathic pain, selective silencing of key neurons in spinal pain signalling networks with botulinum constructs resulted in a reduction of pain behaviours associated with the peripheral nerve. However, to establish clinical relevance it was important to know how long this silencing period lasted. Now, we show that neuronal silencing and the concomitant reduction of neuropathic mechanical and thermal hypersensitivity lasts for up to 120d following a single injection of botulinum construct. Crucially, we show that silencing and analgesia can then be reinstated with a second injection of the botulinum conjugate. Here we demonstrate that single doses of botulinum-toxin conjugates are a powerful new way of providing long-term neuronal silencing and pain relief. PERSPECTIVE: This research demonstrates that botulinum-toxin conjugates are a powerful new way of providing long-term neuronal silencing without toxicity following a single injection of the conjugate and have the potential for repeated dosing when silencing reverses.
Collapse
Affiliation(s)
- Maria Maiarù
- Department of Pharmacology, School of Pharmacy, University of Reading; Room 109, Hopkins Building, Whiteknights Campus, Reading RG6 6UB, United Kingdom
| | - Charlotte Leese
- Department of Biomedical Science, Firth Court, University of Sheffield; Sheffield S10 2TN
| | - Silvia Silva-Hucha
- Department of Cell and Developmental Biology, Medawar Building, University College London; Gower Street, London, WC1E 6BT, United Kingdom
| | - Sofia Fontana-Giusti
- Department of Pharmacology, School of Pharmacy, University of Reading; Room 109, Hopkins Building, Whiteknights Campus, Reading RG6 6UB, United Kingdom
| | - Luke Tait
- Cardiff University Brain Research Imaging Centre, Cardiff University; Cardiff, United Kingdom
| | - Francesco Tamagnini
- Department of Pharmacology, School of Pharmacy, University of Reading; Room 109, Hopkins Building, Whiteknights Campus, Reading RG6 6UB, United Kingdom
| | - Bazbek Davletov
- Department of Biomedical Science, Firth Court, University of Sheffield; Sheffield S10 2TN
| | - Stephen P Hunt
- Department of Cell and Developmental Biology, Medawar Building, University College London; Gower Street, London, WC1E 6BT, United Kingdom
| |
Collapse
|
40
|
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
|
41
|
Upadhyay A, Gradwell MA, Vajtay TJ, Conner J, Sanyal AA, Azadegan C, Patel KR, Thackray JK, Bohic M, Imai F, Ogundare SO, Yoshida Y, Abdus-Saboor I, Azim E, Abraira VE. The Dorsal Column Nuclei Scale Mechanical Sensitivity in Naive and Neuropathic Pain States. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.20.581208. [PMID: 38712022 PMCID: PMC11071288 DOI: 10.1101/2024.02.20.581208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Tactile perception relies on reliable transmission and modulation of low-threshold information as it travels from the periphery to the brain. During pathological conditions, tactile stimuli can aberrantly engage nociceptive pathways leading to the perception of touch as pain, known as mechanical allodynia. Two main drivers of peripheral tactile information, low-threshold mechanoreceptors (LTMRs) and postsynaptic dorsal column neurons (PSDCs), terminate in the brainstem dorsal column nuclei (DCN). Activity within the DRG, spinal cord, and DCN have all been implicated in mediating allodynia, yet the DCN remains understudied at the cellular, circuit, and functional levels compared to the other two. Here, we show that the gracile nucleus (Gr) of the DCN mediates tactile sensitivity for low-threshold stimuli and contributes to mechanical allodynia during neuropathic pain in mice. We found that the Gr contains local inhibitory interneurons in addition to thalamus-projecting neurons, which are differentially innervated by primary afferents and spinal inputs. Functional manipulations of these distinct Gr neuronal populations resulted in bidirectional changes to tactile sensitivity, but did not affect noxious mechanical or thermal sensitivity. During neuropathic pain, silencing Gr projection neurons or activating Gr inhibitory neurons was able to reduce tactile hypersensitivity, and enhancing inhibition was able to ameliorate paw withdrawal signatures of neuropathic pain, like shaking. Collectively, these results suggest that the Gr plays a specific role in mediating hypersensitivity to low-threshold, innocuous mechanical stimuli during neuropathic pain, and that Gr activity contributes to affective, pain-associated phenotypes of mechanical allodynia. Therefore, these brainstem circuits work in tandem with traditional spinal circuits underlying allodynia, resulting in enhanced signaling of tactile stimuli in the brain during neuropathic pain.
Collapse
Affiliation(s)
- Aman Upadhyay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
- Neuroscience PhD program at Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Mark A Gradwell
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Thomas J Vajtay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - James Conner
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Arnab A Sanyal
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Chloe Azadegan
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Komal R Patel
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Joshua K Thackray
- Human Genetics Institute of New Jersey, Rutgers University, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Manon Bohic
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Fumiyasu Imai
- Burke Neurological Institute, White Plains, New York City, New York, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York City, New York, USA
| | - Simon O Ogundare
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York City, New York, USA
| | - Yutaka Yoshida
- Burke Neurological Institute, White Plains, New York City, New York, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York City, New York, USA
| | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York City, New York, USA
| | - Eiman Azim
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Victoria E Abraira
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
- Lead contact
| |
Collapse
|
42
|
Condon LF, Yu Y, Park S, Cao F, Pauli JL, Nelson TS, Palmiter RD. Parabrachial Calca neurons drive nociplasticity. Cell Rep 2024; 43:114057. [PMID: 38583149 PMCID: PMC11210282 DOI: 10.1016/j.celrep.2024.114057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/16/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Pain that persists beyond the time required for tissue healing and pain that arises in the absence of tissue injury, collectively referred to as nociplastic pain, are poorly understood phenomena mediated by plasticity within the central nervous system. The parabrachial nucleus (PBN) is a hub that relays aversive sensory information and appears to play a role in nociplasticity. Here, by preventing PBN Calca neurons from releasing neurotransmitters, we demonstrate that activation of Calca neurons is necessary for the manifestation and maintenance of chronic pain. Additionally, by directly stimulating Calca neurons, we demonstrate that Calca neuron activity is sufficient to drive nociplasticity. Aversive stimuli of multiple sensory modalities, such as exposure to nitroglycerin, cisplatin, or lithium chloride, can drive nociplasticity in a Calca-neuron-dependent manner. Aversive events drive nociplasticity in Calca neurons in the form of increased activity and excitability; however, neuroplasticity also appears to occur in downstream circuitry.
Collapse
Affiliation(s)
- Logan F Condon
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA; Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Ying Yu
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Sekun Park
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Feng Cao
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jordan L Pauli
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Tyler S Nelson
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Richard D Palmiter
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
43
|
Gradwell MA, Ozeri-Engelhard N, Eisdorfer JT, Laflamme OD, Gonzalez M, Upadhyay A, Medlock L, Shrier T, Patel KR, Aoki A, Gandhi M, Abbas-Zadeh G, Oputa O, Thackray JK, Ricci M, George A, Yusuf N, Keating J, Imtiaz Z, Alomary SA, Bohic M, Haas M, Hernandez Y, Prescott SA, Akay T, Abraira VE. Multimodal sensory control of motor performance by glycinergic interneurons of the mouse spinal cord deep dorsal horn. Neuron 2024; 112:1302-1327.e13. [PMID: 38452762 DOI: 10.1016/j.neuron.2024.01.027] [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: 06/13/2023] [Revised: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 03/09/2024]
Abstract
Sensory feedback is integral for contextually appropriate motor output, yet the neural circuits responsible remain elusive. Here, we pinpoint the medial deep dorsal horn of the mouse spinal cord as a convergence point for proprioceptive and cutaneous input. Within this region, we identify a population of tonically active glycinergic inhibitory neurons expressing parvalbumin. Using anatomy and electrophysiology, we demonstrate that deep dorsal horn parvalbumin-expressing interneuron (dPV) activity is shaped by convergent proprioceptive, cutaneous, and descending input. Selectively targeting spinal dPVs, we reveal their widespread ipsilateral inhibition onto pre-motor and motor networks and demonstrate their role in gating sensory-evoked muscle activity using electromyography (EMG) recordings. dPV ablation altered limb kinematics and step-cycle timing during treadmill locomotion and reduced the transitions between sub-movements during spontaneous behavior. These findings reveal a circuit basis by which sensory convergence onto dorsal horn inhibitory neurons modulates motor output to facilitate smooth movement and context-appropriate transitions.
Collapse
Affiliation(s)
- Mark A Gradwell
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Nofar Ozeri-Engelhard
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Neuroscience PhD program, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Jaclyn T Eisdorfer
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Olivier D Laflamme
- Dalhousie PhD program, Dalhousie University, Halifax, NS, Canada; Department of Medical Neuroscience, Atlantic Mobility Action Project, Brain Repair Center, Dalhousie University, Halifax, NS, Canada
| | - Melissa Gonzalez
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Biomedical Engineering, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Aman Upadhyay
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Neuroscience PhD program, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Laura Medlock
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Tara Shrier
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Komal R Patel
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Adin Aoki
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Melissa Gandhi
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Gloria Abbas-Zadeh
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Olisemaka Oputa
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Joshua K Thackray
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Human Genetics Institute of New Jersey, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA; Tourette International Collaborative Genetics Study (TIC Genetics)
| | - Matthew Ricci
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Arlene George
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Nusrath Yusuf
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Neuroscience PhD program, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Jessica Keating
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Zarghona Imtiaz
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Simona A Alomary
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Michael Haas
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Yurdiana Hernandez
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Steven A Prescott
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Turgay Akay
- Department of Medical Neuroscience, Atlantic Mobility Action Project, Brain Repair Center, Dalhousie University, Halifax, NS, Canada
| | - Victoria E Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
44
|
Alexandre C, Miracca G, Holanda VD, Sharma A, Kourbanova K, Ferreira A, Bicca MA, Zeng X, Nassar VA, Lee S, Kaur S, Sarma SV, Sacré P, Scammell TE, Woolf CJ, Latremoliere A. Nociceptor spontaneous activity is responsible for fragmenting non-rapid eye movement sleep in mouse models of neuropathic pain. Sci Transl Med 2024; 16:eadg3036. [PMID: 38630850 PMCID: PMC11106840 DOI: 10.1126/scitranslmed.adg3036] [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: 12/14/2022] [Accepted: 03/27/2024] [Indexed: 04/19/2024]
Abstract
Spontaneous pain, a major complaint of patients with neuropathic pain, has eluded study because there is no reliable marker in either preclinical models or clinical studies. Here, we performed a comprehensive electroencephalogram/electromyogram analysis of sleep in several mouse models of chronic pain: neuropathic (spared nerve injury and chronic constriction injury), inflammatory (Freund's complete adjuvant and carrageenan, plantar incision) and chemical pain (capsaicin). We find that peripheral axonal injury drives fragmentation of sleep by increasing brief arousals from non-rapid eye movement sleep (NREMS) without changing total sleep amount. In contrast to neuropathic pain, inflammatory or chemical pain did not increase brief arousals. NREMS fragmentation was reduced by the analgesics gabapentin and carbamazepine, and it resolved when pain sensitivity returned to normal in a transient neuropathic pain model (sciatic nerve crush). Genetic silencing of peripheral sensory neurons or ablation of CGRP+ neurons in the parabrachial nucleus prevented sleep fragmentation, whereas pharmacological blockade of skin sensory fibers was ineffective, indicating that the neural activity driving the arousals originates ectopically in primary nociceptor neurons and is relayed through the lateral parabrachial nucleus. These findings identify NREMS fragmentation by brief arousals as an effective proxy to measure spontaneous neuropathic pain in mice.
Collapse
Affiliation(s)
- Chloe Alexandre
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Giulia Miracca
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Victor Duarte Holanda
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ashley Sharma
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kamila Kourbanova
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ashley Ferreira
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Maíra A. Bicca
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Xiangsunze Zeng
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Victoria A. Nassar
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Seungkyu Lee
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Satvinder Kaur
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Sridevi V. Sarma
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Pierre Sacré
- Department of Electrical Engineering and Computer Science, School of Engineering, University of Liège, Liège, Belgium
| | - Thomas E. Scammell
- Department of Neurology, Beth israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Clifford J. Woolf
- FM Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Latremoliere
- Department of Neurosurgery, Neurosurgery Pain Research institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
45
|
Li YD, Luo YJ, Su WK, Ge J, Crowther A, Chen ZK, Wang L, Lazarus M, Liu ZL, Qu WM, Huang ZL. Anterior cingulate cortex projections to the dorsal medial striatum underlie insomnia associated with chronic pain. Neuron 2024; 112:1328-1341.e4. [PMID: 38354737 DOI: 10.1016/j.neuron.2024.01.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/29/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Chronic pain often leads to the development of sleep disturbances. However, the precise neural circuit mechanisms responsible for sleep disorders in chronic pain have remained largely unknown. Here, we present compelling evidence that hyperactivity of pyramidal neurons (PNs) in the anterior cingulate cortex (ACC) drives insomnia in a mouse model of nerve-injury-induced chronic pain. After nerve injury, ACC PNs displayed spontaneous hyperactivity selectively in periods of insomnia. We then show that ACC PNs were both necessary for developing chronic-pain-induced insomnia and sufficient to mimic sleep loss in naive mice. Importantly, combining optogenetics and electrophysiological recordings, we found that the ACC projection to the dorsal medial striatum (DMS) underlies chronic-pain-induced insomnia through enhanced activity and plasticity of ACC-DMS dopamine D1R neuron synapses. Our findings shed light on the pivotal role of ACC PNs in developing chronic-pain-induced sleep disorders.
Collapse
Affiliation(s)
- Ya-Dong Li
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Songjiang Research Institute, Songjiang Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Emotions and Affective Disorders (LEAD), Shanghai 201699, China.
| | - Yan-Jia Luo
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wei-Kun Su
- Songjiang Research Institute, Songjiang Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Emotions and Affective Disorders (LEAD), Shanghai 201699, China
| | - Jing Ge
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Andrew Crowther
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ze-Ka Chen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lu Wang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS) and Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Zi-Long Liu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
46
|
Yarmolinsky DA, Zeng X, MacKinnon-Booth N, Greene C, Kim C, Woolf CJ. Selective modification of ascending spinal outputs in acute and neuropathic pain states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588581. [PMID: 38645252 PMCID: PMC11030409 DOI: 10.1101/2024.04.08.588581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Pain hypersensitivity arises from the plasticity of peripheral and spinal somatosensory neurons, which modifies nociceptive input to the brain and alters pain perception. We utilized chronic calcium imaging of spinal dorsal horn neurons to determine how the representation of somatosensory stimuli in the anterolateral tract, the principal pathway transmitting nociceptive signals to the brain, changes between distinct pain states. In healthy conditions, we identify stable, narrowly tuned outputs selective for cooling or warming, and a neuronal ensemble activated by intense/noxious thermal and mechanical stimuli. Induction of an acute peripheral sensitization with capsaicin selectively and transiently retunes nociceptive output neurons to encode low-intensity stimuli. In contrast, peripheral nerve injury-induced neuropathic pain results in a persistent suppression of innocuous spinal outputs coupled with activation of a normally silent population of high-threshold neurons. These results demonstrate the differential modulation of specific spinal outputs to the brain during nociceptive and neuropathic pain states.
Collapse
|
47
|
Gautam M, Yamada A, Yamada AI, Wu Q, Kridsada K, Ling J, Yu H, Dong P, Ma M, Gu J, Luo W. Distinct local and global functions of mouse Aβ low-threshold mechanoreceptors in mechanical nociception. Nat Commun 2024; 15:2911. [PMID: 38575590 PMCID: PMC10995180 DOI: 10.1038/s41467-024-47245-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024] Open
Abstract
The roles of Aβ low-threshold mechanoreceptors (LTMRs) in transmitting mechanical hyperalgesia and in alleviating chronic pain have been of great interest but remain contentious. Here we utilized intersectional genetic tools, optogenetics, and high-speed imaging to specifically examine functions of SplitCre labeled mouse Aβ-LTMRs in this regard. Genetic ablation of SplitCre-Aβ-LTMRs increased mechanical nociception but not thermosensation in both acute and chronic inflammatory pain conditions, indicating a modality-specific role in gating mechanical nociception. Local optogenetic activation of SplitCre-Aβ-LTMRs triggered nociception after tissue inflammation, whereas their broad activation at the dorsal column still alleviated mechanical hypersensitivity of chronic inflammation. Taking all data into consideration, we propose a model, in which Aβ-LTMRs play distinctive local and global roles in transmitting or alleviating mechanical hyperalgesia of chronic pain, respectively. Our model suggests a strategy of global activation plus local inhibition of Aβ-LTMRs for treating mechanical hyperalgesia.
Collapse
Affiliation(s)
- Mayank Gautam
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Akihiro Yamada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ayaka I Yamada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Qinxue Wu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kim Kridsada
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jennifer Ling
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Huasheng Yu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Peter Dong
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Minghong Ma
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jianguo Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
48
|
Safronov BV, Szucs P. Novel aspects of signal processing in lamina I. Neuropharmacology 2024; 247:109858. [PMID: 38286189 DOI: 10.1016/j.neuropharm.2024.109858] [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: 11/24/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 01/31/2024]
Abstract
The most superficial layer of the spinal dorsal horn, lamina I, is a key element of the nociceptive processing system. It contains different types of projection neurons (PNs) and local-circuit neurons (LCNs) whose functional roles in the signal processing are poorly understood. This article reviews recent progress in elucidating novel anatomical features and physiological properties of lamina I PNs and LCNs revealed by whole-cell recordings in ex vivo spinal cord. This article is part of the Special Issue on "Ukrainian Neuroscience".
Collapse
Affiliation(s)
- Boris V Safronov
- Neuronal Networks Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| | - Peter Szucs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Neuroscience Research Group, Debrecen, Hungary
| |
Collapse
|
49
|
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
|
50
|
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
|