1
|
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 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] [Track Full Text] [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
|
2
|
Zang H, Ji X, Yao W, Wan L, Zhang C, Zhu C, Liu T. Role of efferocytosis in chronic pain -- From molecular perspective. Neurobiol Dis 2025; 207:106857. [PMID: 40015655 DOI: 10.1016/j.nbd.2025.106857] [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: 12/27/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025] Open
Abstract
The complex nature of pain pathophysiology complicates the establishment of objective diagnostic criteria and targeted treatments. The heterogeneous manifestations of pain stemming from various primary diseases contribute to the complexity and diversity of underlying mechanisms, leading to challenges in treatment efficacy and undesirable side effects. Recent evidence suggests the presence of apoptotic cells at injury sites, the distal dorsal root ganglia (DRG), spinal cord, and certain brain regions, indicating a potential link between the ineffective clearance of dead cells and debris and pain persistence. This review highlights recent research findings indicating that efferocytosis plays a significant yet often overlooked role in lesion expansion while also representing a potentially reversible impairment that could be targeted therapeutically to mitigate chronic pain progression. We examine recent advances into how efferocytosis, a process by which phagocytes clear apoptotic cells without triggering inflammation, influences pain initiation and intensity in both human diseases and animal models. This review summarizes that efferocytosis contributes to pain progression from the perspective of defective and inefficient efferocytosis and its subsequent secondary necrocytosis, cascade inflammatory response, and the shift of phenotypic plasticity and metabolism. Additionally, we investigate the roles of newly discovered genetic alterations or modifications in biological signaling pathways in pain development and chronicity, providing insights into innovative treatment strategies that modulate efferocytosis, which are promising candidates and potential avenues for further research in pain management and prevention.
Collapse
Affiliation(s)
- Hu Zang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoyu Ji
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wenlong Yao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Li Wan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chuanhan Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chang Zhu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Tongtong Liu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
3
|
Zhang MD, Kupari J, Su J, Magnusson KA, Hu Y, Calvo-Enrique L, Usoskin D, Albisetti GW, Ceder MM, Henriksson K, Leavitt AD, Zeilhofer HU, Hökfelt T, Lagerström MC, Ernfors P. Neural ensembles that encode nocifensive mechanical and heat pain in mouse spinal cord. Nat Neurosci 2025:10.1038/s41593-025-01921-6. [PMID: 40128392 DOI: 10.1038/s41593-025-01921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/07/2025] [Indexed: 03/26/2025]
Abstract
Acute pain is an unpleasant experience caused by noxious stimuli. How the spinal neural circuits attribute differences in quality of noxious information remains unknown. By means of genetic capturing, activity manipulation and single-cell RNA sequencing, we identified distinct neural ensembles in the adult mouse spinal cord encoding mechanical and heat pain. Reactivation or silencing of these ensembles potentiated or stopped, respectively, paw shaking, lifting and licking within but not across the stimuli modalities. Within ensembles, polymodal Gal+ inhibitory neurons with monosynaptic contacts to A-fiber sensory neurons gated pain transmission independent of modality. Peripheral nerve injury led to inferred microglia-driven inflammation and an ensemble transition with decreased recruitment of Gal+ inhibitory neurons and increased excitatory drive. Forced activation of Gal+ neurons reversed hypersensitivity associated with neuropathy. Our results reveal the existence of a spinal representation that forms the neural basis of the discriminative and defensive qualities of acute pain, and these neurons are under the control of a shared feed-forward inhibition.
Collapse
Affiliation(s)
- Ming-Dong Zhang
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Jussi Kupari
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Jie Su
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Kajsa A Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Yizhou Hu
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Laura Calvo-Enrique
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain
| | - Dmitry Usoskin
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Gioele W Albisetti
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Mikaela M Ceder
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Katharina Henriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Andrew D Leavitt
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Malin C Lagerström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrik Ernfors
- Department of Medical Biochemistry and Biophysics, Division of Molecular Neurobiology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Roome RB, Yadav A, Flores L, Puarr A, Nardini D, Richardson A, Waclaw RR, Arkell RM, Menon V, Johnson JE, Levine AJ. Ontogeny of the spinal cord dorsal horn. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643370. [PMID: 40161840 PMCID: PMC11952496 DOI: 10.1101/2025.03.14.643370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The dorsal horn of the mammalian spinal cord is an exquisite example of form serving function. It is comprised of diverse neuronal populations stacked into laminae, each of which receives different circuit connections and plays specialized roles in behavior. An outstanding question is how this organization emerges during development from an apparently homogeneous pool of neural progenitors. Here, we found that dorsal neurons are diversified by time, with families of related cell types born as temporal cohorts, and by a spatial-molecular gradient that specifies the full array of individual cell types. Excitatory dorsal neurons then settle in a chronotopic arrangement that transforms their progressive birthdates into anatomical order. This establishes the dorsal horn laminae, as these neurons are also required for spatial organization of inhibitory neurons and sensory axons. This work reveals essential ontogenetic principles that shape dorsal progenitors into the diverse cell types and architecture that subserve sensorimotor behavior.
Collapse
Affiliation(s)
- Robert Brian Roome
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Archana Yadav
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Lydia Flores
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Amrit Puarr
- Maternal-foetal Precision Health Laboratory, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Diana Nardini
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander Richardson
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ronald R. Waclaw
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ruth M. Arkell
- Maternal-foetal Precision Health Laboratory, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jane E. Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ariel J. Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Lead contact
| |
Collapse
|
5
|
Tang Y, Wang Z, Cao J, Tu Y. Bone-brain crosstalk in osteoarthritis: pathophysiology and interventions. Trends Mol Med 2025; 31:281-295. [PMID: 39438197 DOI: 10.1016/j.molmed.2024.09.006] [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: 07/28/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Osteoarthritis (OA) is a prevalent articular disorder characterized by joint degeneration and persistent pain; it imposes a significant burden on both individuals and society. While OA has traditionally been viewed as a localized peripheral disorder, recent preclinical and clinical studies have revealed the crucial interconnections between the bone and the brain, highlighting the systemic nature of OA. The neuronal pathway, molecular signaling, circadian rhythms, and genetic underpinnings within the bone-brain axis play vital roles in the complex interplay that contributes to OA initiation and progression. This review explores emerging evidence of the crosstalk between the bone and brain in OA progression, and discusses the potential contributions of the bone-brain axis to the development of effective interventions for managing OA.
Collapse
Affiliation(s)
- Yilan Tang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyan Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Cao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Yiheng Tu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Sławińska U, Hammar I, Jankowska E. Modulation of Sensory Input to the Spinal Cord by Peripheral Afferent Fibres via GABAergic Astrocytes. Eur J Neurosci 2025; 61:e70057. [PMID: 40123195 PMCID: PMC11931268 DOI: 10.1111/ejn.70057] [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: 12/03/2024] [Revised: 01/25/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
A long-lasting GABA-dependent increase in the excitability of afferent fibres, and thus modulation of the sensory input to the spinal cord, may be evoked by epidural polarization. However, the direct effects of fibre polarization are short-lasting and the sustained increase in their excitability appears to be secondary to the release of GABA from nearby astrocytes. We have now investigated whether the modulation of spinal sensory input by stimulation of a peripheral nerve, previously attributed to synaptically evoked intraspinal field potentials, is evoked in a similar way. However, as neither its dependence on GABA nor its relays have been investigated, we addressed the question of whether the increase in the excitability of epidurally stimulated afferent fibres following a peripheral nerve stimulation does or does not depend on GABA and whether it might be mediated by astrocytes. The effects of conditioning stimulation of the tibial nerve were evaluated from changes in the excitability of both Group I and II muscle afferents, estimated from action potentials recorded in peripheral nerves and in field potentials recorded in the dorsal horn respectively in acute experiments on deeply anaesthetized rats. The excitability of the afferents was increased by stimulation of Group II and/or cutaneous but not Group I muscle afferents. The effects were significantly weakened by blocking GABA channels by gabazine and by astrocyte toxin L-alpha-aminoadipic acid (L-AAA), indicating that the excitability of both Group I and II afferent fibres may be modulated by GABAergic astrocytes, the new role played by astrocytes.
Collapse
Affiliation(s)
- Urszula Sławińska
- Institute of Physiology and Neuroscience, Sahlgrenska AcademyGöteborg UniversityGöteborgSweden
- Nencki Institute of Experimental Biology PASWarsawPoland
| | - Ingela Hammar
- Institute of Physiology and Neuroscience, Sahlgrenska AcademyGöteborg UniversityGöteborgSweden
| | - Elzbieta Jankowska
- Institute of Physiology and Neuroscience, Sahlgrenska AcademyGöteborg UniversityGöteborgSweden
| |
Collapse
|
7
|
Kwokdinata C, Chew SY. Additive manufacturing in spatial patterning for spinal cord injury treatment. Adv Drug Deliv Rev 2025; 218:115523. [PMID: 39880332 DOI: 10.1016/j.addr.2025.115523] [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: 11/11/2024] [Revised: 01/03/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Combinatorial treatments integrating cells and biomolecules within scaffolds have been investigated to address the multifactorial nature of spinal cord injury (SCI). Current regenerative treatments have been ineffective as they do not consider the spatial positions of various cell types to effectively form functional neural pathways. Emulating the complex heterogeneity of cells in the native spinal cord requires translating the existing biological understanding of spatial patterning in neural development, as well as the influence of biomolecule and mechanical patterning on regional specification and axonal regeneration, to engineer a scaffold for spinal cord regeneration. This review explores the potential of 3D bioprinting to precisely control material, cell and drug patterns in scaffolds, achieving spatial phenotype specification and providing axonal guidance to form appropriate connections. We also discuss the application of extrusion-based and digital light processing bioprinting in integrating mechanical, chemical and biological cues within a scaffold to advance spatially patterned 3D bioprinted scaffold, as well as current challenges and future perspectives in these bioengineering strategies.
Collapse
Affiliation(s)
- Christy Kwokdinata
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University 637459 Singapore
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University 637459 Singapore; Critical Analytics for Manufacturing Personalized-Medicine Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology, Campus for Research Excellence and Technological Enterprise 138602 Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University 308232 Singapore; School of Materials Science and Engineering 639798 Singapore; National Neuroscience Institute, 11 Jalan Tan Tock Seng 308433 Singapore.
| |
Collapse
|
8
|
Paracha M, Brezinski AN, Singh R, Sinson E, Satkunendrarajah K. Targeting Spinal Interneurons for Respiratory Recovery After Spinal Cord Injury. Cells 2025; 14:288. [PMID: 39996760 PMCID: PMC11854602 DOI: 10.3390/cells14040288] [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: 01/14/2025] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
Spinal interneurons (SpINs) are pivotal to the function of neural circuits, orchestrating motor, sensory, and autonomic functions in the healthy, intact central nervous system. These interneurons (INs) are heterogeneous, with diverse types contributing to various neural systems, including those that control respiratory function. Research in the last few decades has highlighted the complex involvement of SpINs in modulating motor control. SpINs also partake in motor plasticity by aiding in adapting and rewiring neural circuits in response to injury or disease. This plasticity is crucial in the context of spinal cord injury (SCI), where damage often leads to severe and long-term breathing deficits. Such deficits are a leading cause of morbidity and mortality in individuals with SCI, emphasizing the need for effective interventions. This review will focus on SpIN circuits involved in the modulation of breathing and explore current and emerging approaches that leverage SpINs as therapeutic targets to promote respiratory recovery following SCI.
Collapse
Affiliation(s)
- Maha Paracha
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, USA
| | - Allison N. Brezinski
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
- Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rhea Singh
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
| | - Elizabeth Sinson
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
| | - Kajana Satkunendrarajah
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.P.); (A.N.B.); (R.S.); (E.S.)
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
9
|
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
|
10
|
Bryson M, Kloefkorn H, Idlett-Ali S, Carrasco DI, Noble DJ, Martin K, Sawchuk MA, Yong NA, Garraway SM, Hochman S. Emergent epileptiform activity in spinal sensory circuits drives ectopic bursting in afferent axons and sensory dysfunction after cord injury. Pain 2025; 166:e27-e35. [PMID: 39106457 PMCID: PMC11723814 DOI: 10.1097/j.pain.0000000000003364] [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: 09/29/2023] [Accepted: 06/25/2024] [Indexed: 08/09/2024]
Abstract
ABSTRACT Spinal cord injury leads to hyperexcitability and dysfunction in spinal sensory processing. As hyperexcitable circuits can become epileptiform, we explored whether such activity emerges in a thoracic spinal cord injury (SCI) contusion model of neuropathic pain. Recordings from spinal sensory axons in multiple below-lesion segmental dorsal roots demonstrated that SCI facilitated the emergence of spontaneous ectopic burst spiking in afferent axons, which were correlated across multiple adjacent dorsal roots. Burst frequency correlated with behavioral mechanosensitivity. The same bursting events were recruited by afferent stimulation, and timing interactions with ongoing spontaneous bursts revealed that recruitment was limited by a prolonged post-burst refractory period. Ectopic bursting in afferent axons was driven by GABA A receptor activation, presumably by conversion of subthreshold GABAergic interneuronal presynaptic axoaxonic inhibitory actions to suprathreshold spiking. Collectively, the emergence of stereotyped bursting circuitry with hypersynchrony, sensory input activation, post-burst refractory period, and reorganization of connectivity represent defining features of an epileptiform network. Indeed, these same features were reproduced in naive animals with the convulsant 4-aminopyridine (fampridine). We conclude that spinal cord injury promotes the emergence of epileptiform activity in spinal sensory networks that promote profound corruption of sensory signaling. This includes hyperexcitability and bursting by ectopic spiking in afferent axons that propagate bidirectionally by reentrant central and peripheral projections as well as sensory circuit hypoexcitability during the burst refractory period. More broadly, the work links circuit hyperexcitability to epileptiform circuit emergence, further strengthening it as a conceptual basis to understand features of sensory dysfunction and neuropathic pain.
Collapse
Affiliation(s)
- Matthew Bryson
- Emory University School of Medicine Department of Cell Biology (30322)
| | - Heidi Kloefkorn
- Oregon State University Department of Chemical, Biological, and. Environmental Engineering (97331)
| | | | - Dario I. Carrasco
- Emory University School of Medicine Department of Neurosurgery (30322)
| | | | - Karmarcha Martin
- Emory University School of Medicine Department of Cell Biology (30322)
| | | | - Nicholas Au Yong
- Emory University School of Medicine Department of Neurosurgery (30322)
| | | | - Shawn Hochman
- Emory University School of Medicine Department of Cell Biology (30322)
| |
Collapse
|
11
|
He Z, Zhang J, Xu J, Wang Y, Zheng X, Wang W. Differential Neuronal Activation of Nociceptive Pathways in Neuropathic Pain After Spinal Cord Injury. Cell Mol Neurobiol 2025; 45:18. [PMID: 39883258 PMCID: PMC11782389 DOI: 10.1007/s10571-025-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/12/2025] [Indexed: 01/31/2025]
Abstract
Neuropathic pain, a prevalent complication following spinal cord injury (SCI), severely impairs the life quality of patients. No ideal treatment exists due to incomplete knowledge on underlying neural processes. To explore the SCI-induced effect on nociceptive circuits, the protein expression of c-Fos was analyzed as an indicator of neuronal activation in a rat contusion model exhibiting below-level pain. Additional stimuli were delivered to mimic the different peripheral sensory inputs in daily life. Following noxious rather than innocuous or no stimulation, a greater number of spinal dorsal horn (DH) neurons were activated after SCI, mainly in the deep DH. SCI facilitated the activation of excitatory but not inhibitory DH neurons. Moreover, excitatory interneurons expressing protein kinase C gamma (PKCγ) in laminae II-III, which are known to play a role in mechanical allodynia after peripheral nerve injury, responded in larger amounts to both innocuous and noxious stimulation following SCI. Accordingly, more spinal projection neurons in lamina I were activated. Within supraspinal nuclei processing pain, differentially enhanced activation in response to noxious stimulation was detected after SCI, with a significant increase in the locus coeruleus and medial thalamus, a slight increase in the periaqueductal gray and dorsal raphe, and no change in the lateral parabrachial nucleus or primary sensory cortex. These findings indicated differential hyperexcitability along the sensory neuroaxis following SCI, with a particular emphasis on the involvement of specific neuron subtypes, such as spinal PKCγ interneurons and locus coeruleus noradrenergic neurons, which may serve as crucial targets for potential therapies.
Collapse
Affiliation(s)
- Ziyu He
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Zhang
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Neurosurgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia Xu
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Rehabilitation, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
- Stem Cell Research Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Wang
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Neurosurgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaolong Zheng
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wei Wang
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases of Chinese Ministry of Education, the School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
12
|
Stenner MP, Nossa CM, Zaehle T, Azañón E, Heinze HJ, Deliano M, Büntjen L. Prior knowledge changes initial sensory processing in the human spinal cord. SCIENCE ADVANCES 2025; 11:eadl5602. [PMID: 39813342 PMCID: PMC11734707 DOI: 10.1126/sciadv.adl5602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
Prior knowledge changes how the brain processes sensory input. Whether knowledge influences initial sensory processing upstream of the brain, in the spinal cord, is unknown. Studying electric potentials recorded invasively and noninvasively from the human spinal cord at millisecond resolution, we find that the cord generates electric potentials at 600 hertz that are modulated by prior knowledge about the time of sensory input, as early as 13 to 16 milliseconds after stimulation. Our results reveal that already in the spinal cord, sensory processing is under top-down, cognitive control, and that 600-hertz signals, which have been identified as a macroscopic marker of population spiking in other regions of the nervous system, play a role in early, context-dependent sensory processing. The possibility to examine these signals noninvasively in humans opens up avenues for research into the physiology of the spinal cord and its interaction with the brain.
Collapse
Affiliation(s)
- Max-Philipp Stenner
- Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences Magdeburg, Magdeburg, Germany
- Center for Intervention and Research on adaptive and Maladaptive Brain Circuits Underlying Mental Health, Jena-Magdeburg-Halle, Magdeburg, Germany
| | - Cindy Márquez Nossa
- Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Tino Zaehle
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences Magdeburg, Magdeburg, Germany
- Center for Intervention and Research on adaptive and Maladaptive Brain Circuits Underlying Mental Health, Jena-Magdeburg-Halle, Magdeburg, Germany
| | - Elena Azañón
- Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences Magdeburg, Magdeburg, Germany
- Center for Intervention and Research on adaptive and Maladaptive Brain Circuits Underlying Mental Health, Jena-Magdeburg-Halle, Magdeburg, Germany
| | - Hans-Jochen Heinze
- Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences Magdeburg, Magdeburg, Germany
| | - Matthias Deliano
- Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
| | - Lars Büntjen
- Department of Neurosurgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Department of Stereotactic Neurosurgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
13
|
Dong FL, Yu L, Feng PD, Ren JX, Bai XH, Lin JQ, Cao DL, Deng YT, Zhang Y, Shen HH, Gong H, Sun WX, Chi DQ, Mei Y, Ma L, Yin MZ, Li MN, Zhang PF, Hu N, Zhou BL, Liu Y, Zheng XJ, Chen YF, Zhong D, Tao YX, Yan M, Jiang BC. An atlas of neuropathic pain-associated molecular pathological characteristics in the mouse spinal cord. Commun Biol 2025; 8:70. [PMID: 39820760 PMCID: PMC11739467 DOI: 10.1038/s42003-025-07506-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
Peripheral nerve injury (PNI)-induced neuropathic pain (NP) is a severe disease with high prevalence in clinics. Gene reprogramming and tissue remodeling in the dorsal root ganglia (DRG) and spinal cord (SC) drive the development and maintenance of neuropathic pain (NP). However, our understanding of the NP-associated spatial molecular processing landscape of SC and the non-synaptic interactions between DRG neurons and SC cells remains limited. We here integrate spatial transcriptomics (ST) with single-nucleus RNA-sequencing (snRNA-seq) and bulk RNA-sequencing (bulk RNA-seq) to characterize regional pathological heterogeneity of the SC under NP conditions. First, the SC of NP mice manifests unique spatial atlases of genes, cell populations, cell-cell cross-talks, signaling pathways, and transcriptional regulatory networks compared to sham mice. We further report that injured DRG sensory neurons and the corresponding ventral horn of the SC show similar expression patterns after PNI. In addition, for the first time, we systematically exhibit "cross-talk omics" between the DRG neurons and SC dorsal horn neurons and glial cells, indicating an altered communication profile under NP conditions. Together, our findings decode the spatial and cellular heterogeneity of molecular pathological mechanisms underlying NP, providing a foundation for designing therapeutic targets for this disorder.
Collapse
Affiliation(s)
- Fu-Lu Dong
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Pathology, Medical School, Nantong University, Nantong, China
| | - Lina Yu
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Pain Perception and Neuromodulation, Hangzhou, China
| | - Pei-Da Feng
- Department of Pathology, Medical School, Nantong University, Nantong, China
| | - Jin-Xuan Ren
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue-Hui Bai
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Qi Lin
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - De-Li Cao
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Yu-Tao Deng
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Zhang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui-Hui Shen
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Gong
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Wen-Xing Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Dong-Qiu Chi
- Medical Service Center, Nantong University, Nantong, China
| | - Yixiao Mei
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Longfei Ma
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Pain Perception and Neuromodulation, Hangzhou, China
| | - Ming-Zhe Yin
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng-Na Li
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Peng-Fei Zhang
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Nan Hu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Bing-Lin Zhou
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Liu
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan-Jie Zheng
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi-Fan Chen
- Department of Pathology, Medical School, Nantong University, Nantong, China
| | - Da Zhong
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Min Yan
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory of Pain Perception and Neuromodulation, Hangzhou, China.
- Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China.
| | - Bao-Chun Jiang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory of Pain Perception and Neuromodulation, Hangzhou, China.
| |
Collapse
|
14
|
Rupprecht P, Fan W, Sullivan SJ, Helmchen F, Sdrulla AD. Spike rate inference from mouse spinal cord calcium imaging data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.17.603957. [PMID: 39829770 PMCID: PMC11741245 DOI: 10.1101/2024.07.17.603957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Calcium imaging is a key method to record the spiking activity of identified and genetically targeted neurons. However, the observed calcium signals are only an indirect readout of the underlying electrophysiological events (single spikes or bursts of spikes) and require dedicated algorithms to recover the spike rate. These algorithms for spike inference can be optimized using ground truth data from combined electrical and optical recordings, but it is not clear how such optimized algorithms perform on cell types and brain regions for which ground truth does not exist. Here, we use a state-of-the-art algorithm based on supervised deep learning (CASCADE) and a non-supervised algorithm based on non-negative deconvolution (OASIS) to test spike rate inference in spinal cord neurons. To enable these tests, we recorded specific ground truth from glutamatergic and GABAergic somatosensory neurons in the superficial dorsal horn of spinal cord in mice of both sexes. We find that CASCADE and OASIS algorithms that were designed for cortical excitatory neurons generalize well to both spinal cord cell types. However, CASCADE models re-trained on our ground truth further improved the performance, resulting in a more accurate inference of spiking activity from spinal cord neurons. We openly provide re-trained models that can be applied to spinal cord data of variable noise levels and frame rates. Together, our ground-truth recordings and analyses provide a solid foundation for the interpretation of calcium imaging data from spinal cord dorsal horn and showcase how spike rate inference can generalize between different regions of the nervous system.
Collapse
Affiliation(s)
- Peter Rupprecht
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Switzerland
| | - Wei Fan
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Steve J. Sullivan
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Fritjof Helmchen
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Switzerland
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning, University of Zurich, Switzerland
| | - Andrei D. Sdrulla
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
15
|
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
|
16
|
Bansal P, Roitman MF, Jung EE. d-Amphetamine and Feeding States Cohesively Affect Locomotion and Motor Neuron Response in Zebrafish Larvae. Brain Behav 2024; 14:e70173. [PMID: 39643450 PMCID: PMC11624004 DOI: 10.1002/brb3.70173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/05/2024] [Accepted: 11/08/2024] [Indexed: 12/09/2024] Open
Abstract
PURPOSE Amphetamine (AMPH) increases locomotor activities in animals, and the locomotor response to AMPH is further modulated by caloric deficits such as food deprivation and restriction. The increment in locomotor activity regulated by AMPH-caloric deficit concomitance can be further modulated by varying feeding schedules (e.g., acute and chronic food deprivation and acute feeding after chronic food deprivation). However, the effects of different feeding schedules on AMPH-induced locomotor activity are yet to be explicated. Here, we have explored the stimulatory responses of acutely administered D-amphetamine in locomotion under systematically varying feeding states (fed/sated and food deprivation) and schedules (chronic and acute) in zebrafish larvae. METHOD We exposed wild-type and transgenic [Tg(mnx1:GCaMP5)] zebrafish larvae to 0.7 µM concentration of AMPH and measured swimming activity and spinal motor neuron activity in vivo in real time. The analysis involved time-elapsed and cumulative manner pre- and post-AMPH treatment in four different caloric states including acute and chronic schedules of feeding and hunger. Both locomotor and motor neuron activities were compared in all four states in both fish lines. FINDINGS Our results show that locomotion and motor neuron activity increased in both chronic and acute food deprivation post-AMPH treatment cumulatively. A steady increase in locomotion was observed in acute food deprivation compared to an immediate abrupt increase in chronic food-deprivation state. The ad libitum-fed larvae exhibited a moderate increase both in locomotion and motor neuron activity. Conversely to all other caloric states, food-sated (acute feeding after chronic food deprivation) larvae moved moderately less and exhibited a mild decrease in motor neuron activity after AMPH treatment. CONCLUSION These results reveal the importance of cohesive effects of feeding schedule and AMPH treatment by revealing the changes in stimulatory characteristics of AMPH on locomotion and motor neuron activity in acute and chronic feeding states.
Collapse
Affiliation(s)
- Pushkar Bansal
- Department of Mechanical and Industrial EngineeringThe University of Illinois at ChicagoChicagoIllinoisUSA
| | - Mitchell F. Roitman
- Department of PsychologyThe University of Illinois at ChicagoChicagoIllinoisUSA
| | - Erica E. Jung
- Department of Mechanical and Industrial EngineeringThe University of Illinois at ChicagoChicagoIllinoisUSA
- Department of BioengineeringThe University of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
17
|
Arokiaraj CM, Leone MJ, Kleyman M, Chamessian A, Noh MC, Phan BN, Lopes BC, Corrigan KA, Cherupally VK, Yeramosu D, Franusich ME, Podder R, Lele S, Shiers S, Kang B, Kennedy MM, Chen V, Chen Z, Mathys H, Dum RP, Lewis DA, Qadri Y, Price TJ, Pfenning AR, Seal RP. Spatial, transcriptomic, and epigenomic analyses link dorsal horn neurons to chronic pain genetic predisposition. Cell Rep 2024; 43:114876. [PMID: 39453813 PMCID: PMC11801220 DOI: 10.1016/j.celrep.2024.114876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/07/2024] [Accepted: 09/30/2024] [Indexed: 10/27/2024] Open
Abstract
Key mechanisms underlying chronic pain occur within the dorsal horn. Genome-wide association studies (GWASs) have identified genetic variants predisposed to chronic pain. However, most of these variants lie within regulatory non-coding regions that have not been linked to spinal cord biology. Here, we take a multi-species approach to determine whether chronic pain variants impact the regulatory genomics of dorsal horn neurons. First, we generate a large rhesus macaque single-nucleus RNA sequencing (snRNA-seq) atlas and integrate it with available human and mouse datasets to produce a single unified, species-conserved atlas of neuron subtypes. Cellular-resolution spatial transcriptomics in mouse shows the precise laminar location of these neuron subtypes, consistent with our analysis of neuron-subtype-selective markers in macaque. Using this cross-species framework, we generate a mouse single-nucleus open chromatin atlas of regulatory elements that shows strong and selective relationships between the neuron-subtype-specific chromatin regions and variants from major chronic pain GWASs.
Collapse
Affiliation(s)
- Cynthia M Arokiaraj
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael J Leone
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael Kleyman
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Alexander Chamessian
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27708, USA; Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Myung-Chul Noh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - BaDoi N Phan
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Bettega C Lopes
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kelly A Corrigan
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Vijay Kiran Cherupally
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Deepika Yeramosu
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Michael E Franusich
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Riya Podder
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Sumitra Lele
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Byungsoo Kang
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Meaghan M Kennedy
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Viola Chen
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ziheng Chen
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Hansruedi Mathys
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Richard P Dum
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Yawar Qadri
- Department of Anesthesiology, Emory University, Atlanta, GA 30038, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Andreas R Pfenning
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| | - Rebecca P Seal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
18
|
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
|
19
|
Ricci A, Rubino E, Serra GP, Wallén-Mackenzie Å. Concerning neuromodulation as treatment of neurological and neuropsychiatric disorder: Insights gained from selective targeting of the subthalamic nucleus, para-subthalamic nucleus and zona incerta in rodents. Neuropharmacology 2024; 256:110003. [PMID: 38789078 DOI: 10.1016/j.neuropharm.2024.110003] [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: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
Neuromodulation such as deep brain stimulation (DBS) is advancing as a clinical intervention in several neurological and neuropsychiatric disorders, including Parkinson's disease, dystonia, tremor, and obsessive-compulsive disorder (OCD) for which DBS is already applied to alleviate severely afflicted individuals of symptoms. Tourette syndrome and drug addiction are two additional disorders for which DBS is in trial or proposed as treatment. However, some major remaining obstacles prevent this intervention from reaching its full therapeutic potential. Side-effects have been reported, and not all DBS-treated individuals are relieved of their symptoms. One major target area for DBS electrodes is the subthalamic nucleus (STN) which plays important roles in motor, affective and associative functions, with impact on for example movement, motivation, impulsivity, compulsivity, as well as both reward and aversion. The multifunctionality of the STN is complex. Decoding the anatomical-functional organization of the STN could enhance strategic targeting in human patients. The STN is located in close proximity to zona incerta (ZI) and the para-subthalamic nucleus (pSTN). Together, the STN, pSTN and ZI form a highly heterogeneous and clinically important brain area. Rodent-based experimental studies, including opto- and chemogenetics as well as viral-genetic tract tracings, provide unique insight into complex neuronal circuitries and their impact on behavior with high spatial and temporal precision. This research field has advanced tremendously over the past few years. Here, we provide an inclusive review of current literature in the pre-clinical research fields centered around STN, pSTN and ZI in laboratory mice and rats; the three highly heterogeneous and enigmatic structures brought together in the context of relevance for treatment strategies. Specific emphasis is placed on methods of manipulation and behavioral impact.
Collapse
Affiliation(s)
- Alessia Ricci
- Uppsala University, Department of Organism Biology, 756 32 Uppsala, Sweden; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Eleonora Rubino
- Uppsala University, Department of Organism Biology, 756 32 Uppsala, Sweden; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Gian Pietro Serra
- Uppsala University, Department of Organism Biology, 756 32 Uppsala, Sweden; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Åsa Wallén-Mackenzie
- Uppsala University, Department of Organism Biology, 756 32 Uppsala, Sweden; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
20
|
Xu JF, Liu L, Liu Y, Lu KX, Zhang J, Zhu YJ, Fang F, Dou YN. Spinal Nmur2-positive Neurons Play a Crucial Role in Mechanical Itch. THE JOURNAL OF PAIN 2024; 25:104504. [PMID: 38442838 DOI: 10.1016/j.jpain.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
The dorsal spinal cord is crucial for the transmission and modulation of multiple somatosensory modalities, such as itch, pain, and touch. Despite being essential for the well-being and survival of an individual, itch and pain, in their chronic forms, have increasingly been recognized as clinical problems. Although considerable progress has been made in our understanding of the neurochemical processing of nociceptive and chemical itch sensations, the neural substrate that is crucial for mechanical itch processing is still unclear. Here, using genetic and functional manipulation, we identified a population of spinal neurons expressing neuromedin U receptor 2 (Nmur2+) as critical elements for mechanical itch. We found that spinal Nmur2+ neurons are predominantly excitatory neurons, and are enriched in the superficial laminae of the dorsal horn. Pharmacogenetic activation of cervical spinal Nmur2+ neurons evoked scratching behavior. Conversely, the ablation of these neurons using a caspase-3-based method decreased von Frey filament-induced scratching behavior without affecting responses to other somatosensory modalities. Similarly, suppressing the excitability of cervical spinal Nmur2+ neurons via the overexpression of functional Kir2.1 potassium channels reduced scratching in response to innocuous mechanical stimuli, but not to pruritogen application. At the lumbar level, pharmacogenetic activation of these neurons evoked licking and lifting behaviors. However, ablating these neurons did not affect the behavior associated with acute pain. Thus, these results revealed the crucial role of spinal Nmur2+ neurons in mechanical itch. Our study provides important insights into the neural basis of mechanical itch, paving the way for developing novel therapies for chronic itch. PERSPECTIVE: Excitatory Nmur2+ neurons in the superficial dorsal spinal cord are essential for mechanical but not chemical itch information processing. These spinal Nmur2+ neurons represent a potential cellular target for future therapeutic interventions against chronic itch. Spinal and supraspinal Nmur2+ neurons may play different roles in pain signal processing.
Collapse
Affiliation(s)
- Jun-Feng Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lian Liu
- Department of Endocrinology and Metabolic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Lingang Laboratory, Shanghai, China
| | - Ke-Xing Lu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jun Zhang
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan-Jing Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Fang Fang
- Department of Endocrinology and Metabolic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Nong Dou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
21
|
Wang D, Lee KY, Kagan ZB, Bradley K, Lee D. Frequency-Dependent Neural Modulation of Dorsal Horn Neurons by Kilohertz Spinal Cord Stimulation in Rats. Biomedicines 2024; 12:1346. [PMID: 38927553 PMCID: PMC11201430 DOI: 10.3390/biomedicines12061346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Kilohertz high-frequency spinal cord stimulation (kHF-SCS) is a rapidly advancing neuromodulatory technique in the clinical management of chronic pain. However, the precise cellular mechanisms underlying kHF-SCS-induced paresthesia-free pain relief, as well as the neural responses within spinal pain circuits, remain largely unexplored. In this study, using a novel preparation, we investigated the impact of varying kilohertz frequency SCS on dorsal horn neuron activation. Employing calcium imaging on isolated spinal cord slices, we found that extracellular electric fields at kilohertz frequencies (1, 3, 5, 8, and 10 kHz) induce distinct patterns of activation in dorsal horn neurons. Notably, as the frequency of extracellular electric fields increased, there was a clear and significant monotonic escalation in neuronal activity. This phenomenon was observed not only in superficial dorsal horn neurons, but also in those located deeper within the dorsal horn. Our study demonstrates the unique patterns of dorsal horn neuron activation in response to varying kilohertz frequencies of extracellular electric fields, and we contribute to a deeper understanding of how kHF-SCS induces paresthesia-free pain relief. Furthermore, our study highlights the potential for kHF-SCS to modulate sensory information processing within spinal pain circuits. These insights pave the way for future research aimed at optimizing kHF-SCS parameters and refining its therapeutic applications in the clinical management of chronic pain.
Collapse
|
22
|
Ferland S, Wang F, De Koninck Y, Ferrini F. An improved conflict avoidance assay reveals modality-specific differences in pain hypersensitivity across sexes. Pain 2024; 165:1304-1316. [PMID: 38277178 PMCID: PMC11090034 DOI: 10.1097/j.pain.0000000000003132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 01/27/2024]
Abstract
ABSTRACT Abnormal encoding of somatosensory modalities (ie, mechanical, cold, and heat) are a critical part of pathological pain states. Detailed phenotyping of patients' responses to these modalities have raised hopes that analgesic treatments could one day be tailored to a patient's phenotype. Such precise treatment would require a profound understanding of the underlying mechanisms of specific pain phenotypes at molecular, cellular, and circuitry levels. Although preclinical pain models have helped in that regard, the lack of a unified assay quantifying detailed mechanical, cold, and heat pain responses on the same scale precludes comparing how analgesic compounds act on different sensory phenotypes. The conflict avoidance assay is promising in that regard, but testing conditions require validation for its use with multiple modalities. In this study, we improve upon the conflict avoidance assay to provide a validated and detailed assessment of all 3 modalities within the same animal, in mice. We first optimized testing conditions to minimize the necessary amount of training and to reduce sex differences in performances. We then tested what range of stimuli produce dynamic stimulus-response relationships for different outcome measures in naive mice. We finally used this assay to show that nerve injury produces modality-specific sex differences in pain behavior. Our improved assay opens new avenues to study the basis of modality-specific abnormalities in pain behavior.
Collapse
Affiliation(s)
| | - Feng Wang
- CERVO Brain Research Centre, Québec, QC, Canada
- Faculty of Dentistry, Université Laval, Québec, QC, Canada
| | - Yves De Koninck
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
| | - Francesco Ferrini
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| |
Collapse
|
23
|
Guo Y, Jones EJ, Škarabot J, Inns TB, Phillips BE, Atherton PJ, Piasecki M. Common synaptic inputs and persistent inward currents of vastus lateralis motor units are reduced in older male adults. GeroScience 2024; 46:3249-3261. [PMID: 38238546 PMCID: PMC11009172 DOI: 10.1007/s11357-024-01063-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 01/02/2024] [Indexed: 04/13/2024] Open
Abstract
Although muscle atrophy may partially account for age-related strength decline, it is further influenced by alterations of neural input to muscle. Persistent inward currents (PIC) and the level of common synaptic inputs to motoneurons influence neuromuscular function. However, these have not yet been described in the aged human quadriceps. High-density surface electromyography (HDsEMG) signals were collected from the vastus lateralis of 15 young (mean ± SD, 23 ± 5 y) and 15 older (67 ± 9 y) men during submaximal sustained and 20-s ramped contractions. HDsEMG signals were decomposed to identify individual motor unit discharges, from which PIC amplitude and intramuscular coherence were estimated. Older participants produced significantly lower knee extensor torque (p < 0.001) and poorer force tracking ability (p < 0.001) than young. Older participants also had lower PIC amplitude (p = 0.001) and coherence estimates in the alpha frequency band (p < 0.001) during ramp contractions when compared to young. Persistent inward currents and common synaptic inputs are lower in the vastus lateralis of older males when compared to young. These data highlight altered neural input to the clinically and functionally important quadriceps, further underpinning age-related loss of function which may occur independently of the loss of muscle mass.
Collapse
Affiliation(s)
- Yuxiao Guo
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research &, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Royal Derby Hospital Centre (Room 3011), Derby, DE22 3DT, UK
| | - Eleanor J Jones
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research &, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Royal Derby Hospital Centre (Room 3011), Derby, DE22 3DT, UK
| | - Jakob Škarabot
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Thomas B Inns
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research &, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Royal Derby Hospital Centre (Room 3011), Derby, DE22 3DT, UK
| | - Bethan E Phillips
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research &, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Royal Derby Hospital Centre (Room 3011), Derby, DE22 3DT, UK
| | - Philip J Atherton
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research &, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Royal Derby Hospital Centre (Room 3011), Derby, DE22 3DT, UK
| | - Mathew Piasecki
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research &, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Royal Derby Hospital Centre (Room 3011), Derby, DE22 3DT, UK.
| |
Collapse
|
24
|
Wu Y, Temple BA, Sevilla N, Zhang J, Zhu H, Zolotavin P, Jin Y, Duarte D, Sanders E, Azim E, Nimmerjahn A, Pfaff SL, Luan L, Xie C. Ultraflexible electrodes for recording neural activity in the mouse spinal cord during motor behavior. Cell Rep 2024; 43:114199. [PMID: 38728138 PMCID: PMC11233142 DOI: 10.1016/j.celrep.2024.114199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/10/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
Implantable electrode arrays are powerful tools for directly interrogating neural circuitry in the brain, but implementing this technology in the spinal cord in behaving animals has been challenging due to the spinal cord's significant motion with respect to the vertebral column during behavior. Consequently, the individual and ensemble activity of spinal neurons processing motor commands remains poorly understood. Here, we demonstrate that custom ultraflexible 1-μm-thick polyimide nanoelectronic threads can conduct laminar recordings of many neuronal units within the lumbar spinal cord of unrestrained, freely moving mice. The extracellular action potentials have high signal-to-noise ratio, exhibit well-isolated feature clusters, and reveal diverse patterns of activity during locomotion. Furthermore, chronic recordings demonstrate the stable tracking of single units and their functional tuning over multiple days. This technology provides a path for elucidating how spinal circuits compute motor actions.
Collapse
Affiliation(s)
- Yu Wu
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA; Rice Neuroengineering Initiative, Rice University, Houston, TX 77030, USA
| | - Benjamin A Temple
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92037, USA
| | - Nicole Sevilla
- Rice Neuroengineering Initiative, Rice University, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Jiaao Zhang
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA; Rice Neuroengineering Initiative, Rice University, Houston, TX 77030, USA
| | - Hanlin Zhu
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA; Rice Neuroengineering Initiative, Rice University, Houston, TX 77030, USA
| | - Pavlo Zolotavin
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA; Rice Neuroengineering Initiative, Rice University, Houston, TX 77030, USA
| | - Yifu Jin
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA; Rice Neuroengineering Initiative, Rice University, Houston, TX 77030, USA
| | - Daniela Duarte
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Elischa Sanders
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Eiman Azim
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Samuel L Pfaff
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Lan Luan
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA; Rice Neuroengineering Initiative, Rice University, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| | - Chong Xie
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA; Rice Neuroengineering Initiative, Rice University, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Gupta S, Heinrichs E, Novitch BG, Butler SJ. Investigating the basis of lineage decisions and developmental trajectories in the dorsal spinal cord through pseudotime analyses. Development 2024; 151:dev202209. [PMID: 38804879 PMCID: PMC11166460 DOI: 10.1242/dev.202209] [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: 07/24/2023] [Accepted: 04/18/2024] [Indexed: 05/29/2024]
Abstract
Dorsal interneurons (dIs) in the spinal cord encode the perception of touch, pain, heat, itchiness and proprioception. Previous studies using genetic strategies in animal models have revealed important insights into dI development, but the molecular details of how dIs arise as distinct populations of neurons remain incomplete. We have developed a resource to investigate dI fate specification by combining a single-cell RNA-Seq atlas of mouse embryonic stem cell-derived dIs with pseudotime analyses. To validate this in silico resource as a useful tool, we used it to first identify genes that are candidates for directing the transition states that lead to distinct dI lineage trajectories, and then validated them using in situ hybridization analyses in the developing mouse spinal cord in vivo. We have also identified an endpoint of the dI5 lineage trajectory and found that dIs become more transcriptionally homogeneous during terminal differentiation. This study introduces a valuable tool for further discovery about the timing of gene expression during dI differentiation and demonstrates its utility in clarifying dI lineage relationships.
Collapse
Affiliation(s)
- Sandeep Gupta
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric Heinrichs
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Genetics and Genomics Graduate Program, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Bennett G. Novitch
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samantha J. Butler
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
26
|
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
|
27
|
Gupta S, Heinrichs E, Novitch BG, Butler SJ. Investigating the basis of lineage decisions and developmental trajectories in the dorsal spinal cord through pseudotime analyses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.24.550380. [PMID: 37546781 PMCID: PMC10402035 DOI: 10.1101/2023.07.24.550380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Dorsal interneurons (dIs) in the spinal cord encode the perception of touch, pain, heat, itch, and proprioception. While previous studies using genetic strategies in animal models have revealed important insights into dI development, the molecular details by which dIs arise as distinct populations of neurons remain incomplete. We have developed a resource to investigate dI fate specification by combining a single-cell RNA-Seq atlas of mouse ESC-derived dIs with pseudotime analyses. To validate this in silico resource as a useful tool, we used it to first identify novel genes that are candidates for directing the transition states that lead to distinct dI lineage trajectories, and then validated them using in situ hybridization analyses in the developing mouse spinal cord in vivo . We have also identified a novel endpoint of the dI5 lineage trajectory and found that dIs become more transcriptionally homogenous during terminal differentiation. Together, this study introduces a valuable tool for further discovery about the timing of gene expression during dI differentiation and demonstrates its utility clarifying dI lineage relationships. Summary statement Pseudotime analyses of embryonic stem cell-derived dorsal spinal interneurons reveals both novel regulators and lineage relationships between different interneuron populations.
Collapse
|
28
|
Zhang J, Wang M, Alam M, Zheng YP, Ye F, Hu X. Effects of non-invasive cervical spinal cord neuromodulation by trans-spinal electrical stimulation on cortico-muscular descending patterns in upper extremity of chronic stroke. Front Bioeng Biotechnol 2024; 12:1372158. [PMID: 38576448 PMCID: PMC10991759 DOI: 10.3389/fbioe.2024.1372158] [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: 01/17/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024] Open
Abstract
Background: Trans-spinal electrical stimulation (tsES) to the intact spinal cord poststroke may modulate the cortico-muscular control in stroke survivors with diverse lesions in the brain. This work aimed to investigate the immediate effects of tsES on the cortico-muscular descending patterns during voluntary upper extremity (UE) muscle contractions by analyzing cortico-muscular coherence (CMCoh) and electromyography (EMG) in people with chronic stroke. Methods: Twelve chronic stroke participants were recruited to perform wrist-hand extension and flexion tasks at submaximal levels of voluntary contraction for the corresponding agonist flexors and extensors. During the tasks, the tsES was delivered to the cervical spinal cord with rectangular biphasic pulses. Electroencephalography (EEG) data were collected from the sensorimotor cortex, and the EMG data were recorded from both distal and proximal UE muscles. The CMCoh, laterality index (LI) of the peak CMCoh, and EMG activation level parameters under both non-tsES and tsES conditions were compared to evaluate the immediate effects of tsES on the cortico-muscular descending pathway. Results: The CMCoh and LI of peak CMCoh in the agonist distal muscles showed significant increases (p < 0.05) during the wrist-hand extension and flexion tasks with the application of tsES. The EMG activation levels of the antagonist distal muscle during wrist-hand extension were significantly decreased (p < 0.05) with tsES. Additionally, the proximal UE muscles exhibited significant decreases (p < 0.05) in peak CMCoh and EMG activation levels by applying tsES. There was a significant increase (p < 0.05) in LI of peak CMCoh of proximal UE muscles during tsES. Conclusion: The cervical spinal cord neuromodulation via tsES enhanced the residual descending excitatory control, activated the local inhibitory circuits within the spinal cord, and reduced the cortical and proximal muscular compensatory effects. These results suggested the potential of tsES as a supplementary input for improving UE motor functions in stroke rehabilitation.
Collapse
Affiliation(s)
- Jianing Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, China
| | - Maner Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, China
| | - Monzurul Alam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, China
| | - Yong-Ping Zheng
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, China
| | - Fuqiang Ye
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, China
| | - Xiaoling Hu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, China
- Research Institute for Smart Ageing (RISA), Hong Kong SAR, China
- Research Centre of Data Science and Artificial Intelligence (RC-DSAI), Hong Kong SAR, China
- Joint Research Centre for Biosensing and Precision Theranostics, Hong Kong SAR, China
- University Research Facility in Behavioral and Systems Neuroscience (UBSN), The Hong Kong Polytechnic University, Hong Kong SAR, China
| |
Collapse
|
29
|
Tian T, Li H, Zhang S, Yang M. Characterization of sensory and motor dysfunction and morphological alterations in late stages of type 2 diabetic mice. Front Endocrinol (Lausanne) 2024; 15:1374689. [PMID: 38532899 PMCID: PMC10964478 DOI: 10.3389/fendo.2024.1374689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Diabetic neuropathy is the most common complication of diabetes and lacks effective treatments. Although sensory dysfunction during the early stages of diabetes has been extensively studied in various animal models, the functional and morphological alterations in sensory and motor systems during late stages of diabetes remain largely unexplored. In the current work, we examined the influence of diabetes on sensory and motor function as well as morphological changes in late stages of diabetes. The obese diabetic Leprdb/db mice (db/db) were used for behavioral assessments and subsequent morphological examinations. The db/db mice exhibited severe sensory and motor behavioral defects at the age of 32 weeks, including significantly higher mechanical withdrawal threshold and thermal latency of hindpaws compared with age-matched nondiabetic control animals. The impaired response to noxious stimuli was mainly associated with the remarkable loss of epidermal sensory fibers, particularly CGRP-positive nociceptive fibers. Unexpectedly, the area of CGRP-positive terminals in the spinal dorsal horn was dramatically increased in diabetic mice, which was presumably associated with microglial activation. In addition, the db/db mice showed significantly more foot slips and took longer time during the beam-walking examination compared with controls. Meanwhile, the running duration in the rotarod test was markedly reduced in db/db mice. The observed sensorimotor deficits and motor dysfunction were largely attributed to abnormal sensory feedback and muscle atrophy as well as attenuated neuromuscular transmission in aged diabetic mice. Morphological analysis of neuromuscular junctions (NMJs) demonstrated partial denervation of NMJs and obvious fragmentation of acetylcholine receptors (AChRs). Intrafusal muscle atrophy and abnormal muscle spindle innervation were also detected in db/db mice. Additionally, the number of VGLUT1-positive excitatory boutons on motor neurons was profoundly increased in aged diabetic mice as compared to controls. Nevertheless, inhibitory synaptic inputs onto motor neurons were similar between the two groups. This excitation-inhibition imbalance in synaptic transmission might be implicated in the disturbed locomotion. Collectively, these results suggest that severe sensory and motor deficits are present in late stages of diabetes. This study contributes to our understanding of mechanisms underlying neurological dysfunction during diabetes progression and helps to identify novel therapeutic interventions for patients with diabetic neuropathy.
Collapse
Affiliation(s)
- Ting Tian
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haofeng Li
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
- Cryo-EM Facility Center, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
30
|
Rankin G, Chirila AM, Emanuel AJ, Zhang Z, Woolf CJ, Drugowitsch J, Ginty DD. Nerve injury disrupts temporal processing in the spinal cord dorsal horn through alterations in PV + interneurons. Cell Rep 2024; 43:113718. [PMID: 38294904 PMCID: PMC11101906 DOI: 10.1016/j.celrep.2024.113718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 11/13/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
How mechanical allodynia following nerve injury is encoded in patterns of neural activity in the spinal cord dorsal horn (DH) remains incompletely understood. We address this in mice using the spared nerve injury model of neuropathic pain and in vivo electrophysiological recordings. Surprisingly, despite dramatic behavioral over-reactivity to mechanical stimuli following nerve injury, an overall increase in sensitivity or reactivity of DH neurons is not observed. We do, however, observe a marked decrease in correlated neural firing patterns, including the synchrony of mechanical stimulus-evoked firing, across the DH. Alterations in DH temporal firing patterns are recapitulated by silencing DH parvalbumin+ (PV+) interneurons, previously implicated in mechanical allodynia, as are allodynic pain-like behaviors. These findings reveal decorrelated DH network activity, driven by alterations in PV+ interneurons, as a prominent feature of neuropathic pain and suggest restoration of proper temporal activity as a potential therapeutic strategy to treat chronic neuropathic pain.
Collapse
Affiliation(s)
- Genelle Rankin
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Anda M Chirila
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alan J Emanuel
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Zihe Zhang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jan Drugowitsch
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Katic Secerovic N, Balaguer JM, Gorskii O, Pavlova N, Liang L, Ho J, Grigsby E, Gerszten PC, Karal-Ogly D, Bulgin D, Orlov S, Pirondini E, Musienko P, Raspopovic S, Capogrosso M. Neural population dynamics reveals disruption of spinal circuits' responses to proprioceptive input during electrical stimulation of sensory afferents. Cell Rep 2024; 43:113695. [PMID: 38245870 PMCID: PMC10962447 DOI: 10.1016/j.celrep.2024.113695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/08/2023] [Accepted: 01/06/2024] [Indexed: 01/23/2024] Open
Abstract
While neurostimulation technologies are rapidly approaching clinical applications for sensorimotor disorders, the impact of electrical stimulation on network dynamics is still unknown. Given the high degree of shared processing in neural structures, it is critical to understand if neurostimulation affects functions that are related to, but not targeted by, the intervention. Here, we approach this question by studying the effects of electrical stimulation of cutaneous afferents on unrelated processing of proprioceptive inputs. We recorded intraspinal neural activity in four monkeys while generating proprioceptive inputs from the radial nerve. We then applied continuous stimulation to the radial nerve cutaneous branch and quantified the impact of the stimulation on spinal processing of proprioceptive inputs via neural population dynamics. Proprioceptive pulses consistently produce neural trajectories that are disrupted by concurrent cutaneous stimulation. This disruption propagates to the somatosensory cortex, suggesting that electrical stimulation can perturb natural information processing across the neural axis.
Collapse
Affiliation(s)
- Natalija Katic Secerovic
- School of Electrical Engineering, University of Belgrade, 11000 Belgrade, Serbia; The Mihajlo Pupin Institute, University of Belgrade, 11060 Belgrade, Serbia; Laboratory for Neuroengineering, Institute for Robotics and Intelligent Systems, ETH Zürich, 8092 Zürich, Switzerland
| | - Josep-Maria Balaguer
- Rehab and Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Oleg Gorskii
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia; Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint-Petersburg, Russia; National University of Science and Technology "MISIS," 4 Leninskiy Pr., 119049 Moscow, Russia
| | - Natalia Pavlova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia
| | - Lucy Liang
- Rehab and Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Jonathan Ho
- Rehab and Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Erinn Grigsby
- Rehab and Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Peter C Gerszten
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Dzhina Karal-Ogly
- National Research Centre "Kurchatov Institute," 123098 Moscow, Russia
| | - Dmitry Bulgin
- National Research Centre "Kurchatov Institute," 123098 Moscow, Russia; Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Sergei Orlov
- National Research Centre "Kurchatov Institute," 123098 Moscow, Russia
| | - Elvira Pirondini
- Rehab and Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Pavel Musienko
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia; Sirius University of Science and Technology, 354340 Sochi, Russia; Life Improvement by Future Technologies Center "LIFT," 143025 Moscow, Russia
| | - Stanisa Raspopovic
- Laboratory for Neuroengineering, Institute for Robotics and Intelligent Systems, ETH Zürich, 8092 Zürich, Switzerland.
| | - Marco Capogrosso
- Rehab and Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
32
|
Xue Y, Mo S, Li Y, Cao Y, Xu X, Xie Q. Dissecting neural circuits from rostral ventromedial medulla to spinal trigeminal nucleus bidirectionally modulating craniofacial mechanical sensitivity. Prog Neurobiol 2024; 232:102561. [PMID: 38142769 DOI: 10.1016/j.pneurobio.2023.102561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/24/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Chronic craniofacial pain is intractable and its mechanisms remain unclarified. The rostral ventromedial medulla (RVM) plays a crucial role in descending pain facilitation and inhibition. It is unclear how the descending circuits from the RVM to spinal trigeminal nucleus (Sp5) are organized to bidirectionally modulate craniofacial nociception. We used viral tracing, in vivo optogenetics, calcium signaling recording, and chemogenetic manipulations to investigate the structure and function of RVM-Sp5 circuits. We found that most RVM neurons projecting to Sp5 were GABAergic or glutamatergic and facilitated or inhibited craniofacial nociception, respectively. Both GABAergic interneurons and glutamatergic projection neurons in Sp5 received RVM inputs: the former were antinociceptive, whereas the latter were pronociceptive. Furthermore, we demonstrated activation of both GABAergic and glutamatergic Sp5 neurons receiving RVM inputs in inflammation- or dysfunction-induced masseter hyperalgesia. Activating GABAergic Sp5 neurons or inhibiting glutamatergic Sp5 neurons that receive RVM projections reversed masseter hyperalgesia. Our study identifies specific cell types and projections of RVM-Sp5 circuits involved in facilitating or inhibiting craniofacial nociception respectively. Selective manipulation of RVM-Sp5 circuits can be used as potential treatment strategy to relieve chronic craniofacial muscle pain.
Collapse
Affiliation(s)
- Yang Xue
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Siyi Mo
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Yuan Li
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Ye Cao
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China.
| | - Xiaoxiang Xu
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China.
| | - Qiufei Xie
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China.
| |
Collapse
|
33
|
Dai D, Zhao T, Li Z, Li W, Chen A, Tang Y, Gao XF, Xiong L. The plasticity of neuropeptide Y-Y1 receptor system on Tac2 neurons contributes to mechanical hyperknesis during chronic itch. Theranostics 2024; 14:363-378. [PMID: 38164144 PMCID: PMC10750199 DOI: 10.7150/thno.89433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/26/2023] [Indexed: 01/03/2024] Open
Abstract
Rationale: In the physiological states, the act of scratching protects the person from harmful substances, while in certain pathological conditions, the patient suffers from chronic itch, both physically and mentally. Chronic itch sufferers are more sensitive to mechanical stimuli, and mechanical hyperknesis relief is essential for chronic itch treatment. While neuropeptide Y-Y1 receptor (NPY-Y1R) system is known to play a crucial role in modulating mechanical itch in physiological conditions, it is elusive how they are altered during chronic itch. We hypothesize that the negative regulatory effect of Y1Rs on Tac2 neurons, the key neurons that transmit mechanical itch, declines during chronic itch. Methods: We combined transgenic mice, chemogenetic manipulation, immunofluorescence, rabies virus circuit tracing, and electrophysiology to investigate the plasticity of Y1Rs on Tac2 neurons during chronic itch. Results: We found that Tac2 neurons receive direct input from Npy neurons and that inhibition of Npy neurons induces activation of Tac2 neurons. Moreover, the expression of Y1Rs on Tac2 neurons is reduced, and the regulatory effect is also reduced during chronic itch. Conclusion: Our study clarifies the plasticity of Y1Rs on Tac2 neurons during chronic itch and further elucidates the mechanism by which NPY-Y1R system is responsible for modulating mechanical itch. We highlight Y1Rs as a promising therapeutic target for mechanical hyperknesis during chronic itch.
Collapse
Affiliation(s)
- Danqing Dai
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Tiantian Zhao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Zhen Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Wanrong Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Aiwen Chen
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Yali Tang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Xiao-Fei Gao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No. 1279, Sanmen Road, Shanghai 200434, China
| |
Collapse
|
34
|
Shekhtmeyster P, Duarte D, Carey EM, Ngo A, Gao G, Olmstead JA, Nelson NA, Nimmerjahn A. Trans-segmental imaging in the spinal cord of behaving mice. Nat Biotechnol 2023; 41:1729-1733. [PMID: 36879007 PMCID: PMC11229418 DOI: 10.1038/s41587-023-01700-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/01/2023] [Indexed: 03/08/2023]
Abstract
Spinal cord circuits play crucial roles in transmitting pain, but the underlying activity patterns within and across spinal segments in behaving mice have remained elusive. We developed a wearable widefield macroscope with a 7.9-mm2 field of view, ~3- to 4-μm lateral resolution, 2.7-mm working distance and <10-g overall weight and show that highly localized painful mechanical stimuli evoke widespread, coordinated astrocyte excitation across multiple spinal segments.
Collapse
Affiliation(s)
- Pavel Shekhtmeyster
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Electrical and Computer Engineering Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Daniela Duarte
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Erin M Carey
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Alexander Ngo
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Grace Gao
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jack A Olmstead
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Nicholas A Nelson
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
35
|
Kinany N, Pirondini E, Micera S, Van De Ville D. Spinal Cord fMRI: A New Window into the Central Nervous System. Neuroscientist 2023; 29:715-731. [PMID: 35822665 PMCID: PMC10623605 DOI: 10.1177/10738584221101827] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
With the brain, the spinal cord forms the central nervous system. Initially considered a passive relay between the brain and the periphery, the spinal cord is now recognized as being active and plastic. Yet, it remains largely overlooked by the human neuroscience community, in stark contrast with the wealth of research investigating the brain. In this review, we argue that fMRI, traditionally used to image cerebral function, can be extended beyond the brain to help unravel spinal mechanisms involved in human behaviors. To this end, we first outline strategies that have been proposed to tackle the challenges inherent to spinal cord fMRI. Then, we discuss how they have been utilized to provide insights into the functional organization of spinal sensorimotor circuits, highlighting their potential to address fundamental and clinical questions. By summarizing guidelines and applications of spinal cord fMRI, we hope to stimulate and support further research into this promising yet underexplored field.
Collapse
Affiliation(s)
- Nawal Kinany
- Department of Radiology and Medical Informatics, University of Geneva, Geneva, Switzerland
- Medical Image Processing Laboratory, Center for Neuroprosthetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
- Bertarelli Foundation Chair in Translational Neuroengineering, Center for Neuroprosthetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Elvira Pirondini
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
- Department of BioEngineering, University of Pittsburgh, PA, USA
- Rehabilitation Neural Engineering Laboratories, University of Pittsburgh, Pittsburgh, PA, USA
| | - Silvestro Micera
- Bertarelli Foundation Chair in Translational Neuroengineering, Center for Neuroprosthetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
- Translational Neural Engineering Area, The Biorobotics Institute, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Dimitri Van De Ville
- Department of Radiology and Medical Informatics, University of Geneva, Geneva, Switzerland
- Medical Image Processing Laboratory, Center for Neuroprosthetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| |
Collapse
|
36
|
Zheng X, Liu Z, He Z, Xu J, Wang Y, Gong C, Zhang R, Zhang SC, Chen H, Wang W. Preclinical long-term safety of intraspinal transplantation of human dorsal spinal GABA neural progenitor cells. iScience 2023; 26:108306. [PMID: 38026209 PMCID: PMC10661464 DOI: 10.1016/j.isci.2023.108306] [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: 08/10/2023] [Revised: 09/28/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Human pluripotent stem cell (hPSC)-derived neurons have shown promise in treating spinal cord injury (SCI). We previously showed that hPSC-derived dorsal spinal γ-aminobutyric acid (GABA) neurons can alleviate spasticity and promote locomotion in rats with SCI, but their long-term safety remains elusive. Here, we characterized the long-term fate and safety of human dorsal spinal GABA neural progenitor cells (NPCs) in naive rats over one year. All grafted NPCs had undergone differentiation, yielding mainly neurons and astrocytes. Fully mature human neurons grew many axons and formed numerous synapses with rat neural circuits, together with mature human astrocytes that structurally integrated into the rat spinal cord. The sensorimotor function of rats was not impaired by intraspinal transplantation, even when human neurons were activated or inhibited by designer receptors exclusively activated by designer drugs (DREADDs). These findings represent a significant step toward the clinical translation of human spinal neuron transplantation for treating SCI.
Collapse
Affiliation(s)
- Xiaolong Zheng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhixian Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ziyu He
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - YaNan Wang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - ChenZi Gong
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ruoying Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Su-Chun Zhang
- Waisman Center, Department of Neuroscience and Department of Neurology, University of Wisconsin, Madison, WI, USA
- Program in Neuroscience & Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Neurological Diseases of Chinese Ministry of Education, the School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
37
|
Mogilski S, Kubacka M, Świerczek A, Wyska E, Szczepańska K, Sapa J, Kieć-Kononowicz K, Łażewska D. Efficacy of the Multi-Target Compound E153 in Relieving Pain and Pruritus of Different Origins. Pharmaceuticals (Basel) 2023; 16:1481. [PMID: 37895952 PMCID: PMC10609854 DOI: 10.3390/ph16101481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Itch and pain are closely related but distinct sensations that share largely overlapping mediators and receptors. We hypothesized that the novel, multi-target compound E153 has the potential to attenuate pain and pruritus of different origins. After the evaluation of sigma receptor affinity and pharmacokinetic studies, we tested the compound using different procedures and models of pain and pruritus. Additionally, we used pharmacological tools, such as PRE-084, RAMH, JNJ 5207852, and S1RA, to precisely determine the role of histamine H3 and sigma 1 receptors in the analgesic and antipruritic effects of the compound. In vitro studies revealed that the test compound had potent affinity for sigma 1 and sigma 2 receptors, moderate affinity for opioid kappa receptors, and no affinity for delta or μ receptors. Pharmacokinetic studies showed that after intraperitoneal administration, the compound was present at high concentrations in both the peripheral tissues and the central nervous system. The blood-brain barrier-penetrating properties indicate its ability to act centrally at the levels of the brain and spinal cord. Furthermore, the test compound attenuated different types of pain, including acute, inflammatory, and neuropathic. It also showed a broad spectrum of antipruritic activity, attenuating histamine-dependent and histamine-independent itching. Finally, we proved that antagonism of both sigma 1 and histamine H3 receptors is involved in the analgesic activity of the compound, while the antipruritic effect to a greater extent depends on sigma 1 antagonism.
Collapse
Affiliation(s)
- Szczepan Mogilski
- Department of Pharmacodynamics, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.K.); (J.S.)
| | - Monika Kubacka
- Department of Pharmacodynamics, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.K.); (J.S.)
| | - Artur Świerczek
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (A.Ś.); (E.W.)
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (A.Ś.); (E.W.)
| | - Katarzyna Szczepańska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (K.S.); (K.K.-K.); (D.Ł.)
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Jacek Sapa
- Department of Pharmacodynamics, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.K.); (J.S.)
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (K.S.); (K.K.-K.); (D.Ł.)
| | - Dorota Łażewska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (K.S.); (K.K.-K.); (D.Ł.)
| |
Collapse
|
38
|
Zhang Z, Shao H, Liu C, Song H, Wu X, Cao D, Zhu M, Fu Y, Wang J, Gao Y. Descending dopaminergic pathway facilitates itch signal processing via activating spinal GRPR + neurons. EMBO Rep 2023; 24:e56098. [PMID: 37522391 PMCID: PMC10561366 DOI: 10.15252/embr.202256098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 08/01/2023] Open
Abstract
A11 dopaminergic neurons regulate somatosensory transduction by projecting from the diencephalon to the spinal cord, but the function of this descending projection in itch remained elusive. Here, we report that dopaminergic projection neurons from the A11 nucleus to the spinal dorsal horn (dopaminergicA11-SDH ) are activated by pruritogens. Inhibition of these neurons alleviates itch-induced scratching behaviors. Furthermore, chemogenetic inhibition of spinal dopamine receptor D1-expressing (DRD1+ ) neurons decreases acute or chronic itch-induced scratching. Mechanistically, spinal DRD1+ neurons are excitatory and mostly co-localize with gastrin-releasing peptide (GRP), an endogenous neuropeptide for itch. In addition, DRD1+ neurons form synapses with GRP receptor-expressing (GRPR+ ) neurons and activate these neurons via AMPA receptor (AMPAR). Finally, spontaneous itch and enhanced acute itch induced by activating spinal DRD1+ neurons are relieved by antagonists against AMPAR and GRPR. Thus, the descending dopaminergic pathway facilitates spinal itch transmission via activating DRD1+ neurons and releasing glutamate and GRP, which directly augments GRPR signaling. Interruption of this descending pathway may be used to treat chronic itch.
Collapse
Affiliation(s)
- Zhi‐Jun Zhang
- Institute of Pain Medicine and Special Environmental Medicine, Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
- Department of Human Anatomy, School of MedicineNantong UniversityJiangsuChina
| | - Han‐Yu Shao
- Department of Human Anatomy, School of MedicineNantong UniversityJiangsuChina
| | - Chuan Liu
- Department of Human Anatomy, School of MedicineNantong UniversityJiangsuChina
| | - Hao‐Lin Song
- Department of Human Anatomy, School of MedicineNantong UniversityJiangsuChina
| | - Xiao‐Bo Wu
- Institute of Pain Medicine and Special Environmental Medicine, Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
| | - De‐Li Cao
- Institute of Pain Medicine and Special Environmental Medicine, Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
| | - Meixuan Zhu
- University of North Carolina at Chapel HillChapel HillNCUSA
| | - Yuan‐Yuan Fu
- Institute of Pain Medicine and Special Environmental Medicine, Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
| | - Juan Wang
- Department of Human Anatomy, School of MedicineNantong UniversityJiangsuChina
| | - Yong‐Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
| |
Collapse
|
39
|
Roome RB, Levine AJ. The organization of spinal neurons: Insights from single cell sequencing. Curr Opin Neurobiol 2023; 82:102762. [PMID: 37657185 PMCID: PMC10727478 DOI: 10.1016/j.conb.2023.102762] [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: 04/05/2023] [Revised: 06/16/2023] [Accepted: 07/22/2023] [Indexed: 09/03/2023]
Abstract
To understand how the spinal cord enacts complex sensorimotor functions, researchers have studied, classified, and functionally probed it's many neuronal populations for over a century. Recent developments in single-cell RNA-sequencing can characterize the gene expression signatures of the entire set of spinal neuron types and can simultaneously provide an unbiased view of their relationships to each other. This approach has revealed that the location of neurons predicts transcriptomic variability, as dorsal spinal neurons become highly distinct over development as ventral spinal neurons become less so. Temporal specification is also a major source of gene expression variation, subdividing many of the canonical embryonic lineage domains. Together, birthdate and cell body location are fundamental organizing features of spinal neuron diversity.
Collapse
Affiliation(s)
- R Brian Roome
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, USA. https://twitter.com/BrianRoome
| | - Ariel J Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health; Bethesda, MD, USA.
| |
Collapse
|
40
|
Punjani N, Deska-Gauthier D, Hachem LD, Abramian M, Fehlings MG. Neuroplasticity and regeneration after spinal cord injury. NORTH AMERICAN SPINE SOCIETY JOURNAL 2023; 15:100235. [PMID: 37416090 PMCID: PMC10320621 DOI: 10.1016/j.xnsj.2023.100235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023]
Abstract
Spinal cord injury (SCI) is a debilitating condition with significant personal, societal, and economic burden. The highest proportion of traumatic injuries occur at the cervical level, which results in severe sensorimotor and autonomic deficits. Following the initial physical damage associated with traumatic injuries, secondary pro-inflammatory, excitotoxic, and ischemic cascades are initiated further contributing to neuronal and glial cell death. Additionally, emerging evidence has begun to reveal that spinal interneurons undergo subtype specific neuroplastic circuit rearrangements in the weeks to months following SCI, contributing to or hindering functional recovery. The current therapeutic guidelines and standards of care for SCI patients include early surgery, hemodynamic regulation, and rehabilitation. Additionally, preclinical work and ongoing clinical trials have begun exploring neuroregenerative strategies utilizing endogenous neural stem/progenitor cells, stem cell transplantation, combinatorial approaches, and direct cell reprogramming. This review will focus on emerging cellular and noncellular regenerative therapies with an overview of the current available strategies, the role of interneurons in plasticity, and the exciting research avenues enhancing tissue repair following SCI.
Collapse
Affiliation(s)
- Nayaab Punjani
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Dylan Deska-Gauthier
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Laureen D. Hachem
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Division of Neurosurgery and Spine Program, University of Toronto, Toronto, ON, Canada
| | - Madlene Abramian
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Division of Neurosurgery and Spine Program, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| |
Collapse
|
41
|
Du F, Yin G, Han L, Liu X, Dong D, Duan K, Huo J, Sun Y, Cheng L. Targeting Peripheral μ-opioid Receptors or μ-opioid Receptor-Expressing Neurons Does not Prevent Morphine-induced Mechanical Allodynia and Anti-allodynic Tolerance. Neurosci Bull 2023; 39:1210-1228. [PMID: 36622575 PMCID: PMC10387027 DOI: 10.1007/s12264-022-01009-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/19/2022] [Indexed: 01/10/2023] Open
Abstract
The chronic use of morphine and other opioids is associated with opioid-induced hypersensitivity (OIH) and analgesic tolerance. Among the different forms of OIH and tolerance, the opioid receptors and cell types mediating opioid-induced mechanical allodynia and anti-allodynic tolerance remain unresolved. Here we demonstrated that the loss of peripheral μ-opioid receptors (MORs) or MOR-expressing neurons attenuated thermal tolerance, but did not affect the expression and maintenance of morphine-induced mechanical allodynia and anti-allodynic tolerance. To confirm this result, we made dorsal root ganglia-dorsal roots-sagittal spinal cord slice preparations and recorded low-threshold Aβ-fiber stimulation-evoked inputs and outputs in superficial dorsal horn neurons. Consistent with the behavioral results, peripheral MOR loss did not prevent the opening of Aβ mechanical allodynia pathways in the spinal dorsal horn. Therefore, the peripheral MOR signaling pathway may not be an optimal target for preventing mechanical OIH and analgesic tolerance. Future studies should focus more on central mechanisms.
Collapse
Affiliation(s)
- Feng Du
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guangjuan Yin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lei Han
- Department of Anesthesiology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China
| | - Xi Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dong Dong
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Kaifang Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiantao Huo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yanyan Sun
- Department of Anesthesiology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China.
| | - Longzhen Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China.
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
42
|
Bohic M, Upadhyay A, Eisdorfer JT, Keating J, Simon RC, Briones BA, Azadegan C, Nacht HD, Oputa O, Martinez AM, Bethell BN, Gradwell MA, Romanienko P, Ramer MS, Stuber GD, Abraira VE. A new Hoxb8FlpO mouse line for intersectional approaches to dissect developmentally defined adult sensorimotor circuits. Front Mol Neurosci 2023; 16:1176823. [PMID: 37603775 PMCID: PMC10437123 DOI: 10.3389/fnmol.2023.1176823] [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: 02/28/2023] [Accepted: 07/04/2023] [Indexed: 08/23/2023] Open
Abstract
Improvements in the speed and cost of expression profiling of neuronal tissues offer an unprecedented opportunity to define ever finer subgroups of neurons for functional studies. In the spinal cord, single cell RNA sequencing studies support decades of work on spinal cord lineage studies, offering a unique opportunity to probe adult function based on developmental lineage. While Cre/Flp recombinase intersectional strategies remain a powerful tool to manipulate spinal neurons, the field lacks genetic tools and strategies to restrict manipulations to the adult mouse spinal cord at the speed at which new tools develop. This study establishes a new workflow for intersectional mouse-viral strategies to dissect adult spinal function based on developmental lineages in a modular fashion. To restrict manipulations to the spinal cord, we generate a brain-sparing Hoxb8FlpO mouse line restricting Flp recombinase expression to caudal tissue. Recapitulating endogenous Hoxb8 gene expression, Flp-dependent reporter expression is present in the caudal embryo starting day 9.5. This expression restricts Flp activity in the adult to the caudal brainstem and below. Hoxb8FlpO heterozygous and homozygous mice do not develop any of the sensory or locomotor phenotypes evident in Hoxb8 heterozygous or mutant animals, suggesting normal developmental function of the Hoxb8 gene and protein in Hoxb8FlpO mice. Compared to the variability of brain recombination in available caudal Cre and Flp lines, Hoxb8FlpO activity is not present in the brain above the caudal brainstem, independent of mouse genetic background. Lastly, we combine the Hoxb8FlpO mouse line with dorsal horn developmental lineage Cre mouse lines to express GFP in developmentally determined dorsal horn populations. Using GFP-dependent Cre recombinase viruses and Cre recombinase-dependent inhibitory chemogenetics, we target developmentally defined lineages in the adult. We show how developmental knock-out versus transient adult silencing of the same ROR𝛃 lineage neurons affects adult sensorimotor behavior. In summary, this new mouse line and viral approach provides a blueprint to dissect adult somatosensory circuit function using Cre/Flp genetic tools to target spinal cord interneurons based on genetic lineage.
Collapse
Affiliation(s)
- Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Aman Upadhyay
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- Neuroscience PhD Program at Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Jaclyn T. Eisdorfer
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Jessica Keating
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- School of Medicine, Oregon Health and Science University, Portland, OR, United States
- M.D./PhD Program in Neuroscience, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Rhiana C. Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Brandy A. Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Chloe Azadegan
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Hannah D. Nacht
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Olisemeka Oputa
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Alana M. Martinez
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Bridget N. Bethell
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Mark A. Gradwell
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Peter Romanienko
- Genome Editing Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Matt S. Ramer
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Garret D. Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Victoria E. Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
43
|
Ma Q, Su D, Huo J, Yin G, Dong D, Duan K, Cheng H, Xu H, Ma J, Liu D, Mou B, Peng J, Cheng L. Microglial Depletion does not Affect the Laterality of Mechanical Allodynia in Mice. Neurosci Bull 2023; 39:1229-1245. [PMID: 36637789 PMCID: PMC10387012 DOI: 10.1007/s12264-022-01017-2] [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: 07/31/2022] [Accepted: 10/12/2022] [Indexed: 01/14/2023] Open
Abstract
Mechanical allodynia (MA), including punctate and dynamic forms, is a common and debilitating symptom suffered by millions of chronic pain patients. Some peripheral injuries result in the development of bilateral MA, while most injuries usually led to unilateral MA. To date, the control of such laterality remains poorly understood. Here, to study the role of microglia in the control of MA laterality, we used genetic strategies to deplete microglia and tested both dynamic and punctate forms of MA in mice. Surprisingly, the depletion of central microglia did not prevent the induction of bilateral dynamic and punctate MA. Moreover, in dorsal root ganglion-dorsal root-sagittal spinal cord slice preparations we recorded the low-threshold Aβ-fiber stimulation-evoked inputs and outputs of superficial dorsal horn neurons. Consistent with behavioral results, microglial depletion did not prevent the opening of bilateral gates for Aβ pathways in the superficial dorsal horn. This study challenges the role of microglia in the control of MA laterality in mice. Future studies are needed to further understand whether the role of microglia in the control of MA laterality is etiology-or species-specific.
Collapse
Affiliation(s)
- Quan Ma
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dongmei Su
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiantao Huo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guangjuan Yin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dong Dong
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Kaifang Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Hong Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Huiling Xu
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiao Ma
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dong Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Bin Mou
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Jiyun Peng
- Institute of Life Science, Nanchang University, Nanchang, 330031, China.
| | - Longzhen Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
- Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen, 518055, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
| |
Collapse
|
44
|
Boyle KA, Polgar E, Gutierrez-Mecinas M, Dickie AC, Cooper AH, Bell AM, Jumolea E, Casas-Benito A, Watanabe M, Hughes DI, Weir GA, Riddell JS, Todd AJ. Neuropeptide Y-expressing dorsal horn inhibitory interneurons gate spinal pain and itch signalling. eLife 2023; 12:RP86633. [PMID: 37490401 PMCID: PMC10392120 DOI: 10.7554/elife.86633] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023] Open
Abstract
Somatosensory information is processed by a complex network of interneurons in the spinal dorsal horn. It has been reported that inhibitory interneurons that express neuropeptide Y (NPY), either permanently or during development, suppress mechanical itch, with no effect on pain. Here, we investigate the role of interneurons that continue to express NPY (NPY-INs) in the adult mouse spinal cord. We find that chemogenetic activation of NPY-INs reduces behaviours associated with acute pain and pruritogen-evoked itch, whereas silencing them causes exaggerated itch responses that depend on cells expressing the gastrin-releasing peptide receptor. As predicted by our previous studies, silencing of another population of inhibitory interneurons (those expressing dynorphin) also increases itch, but to a lesser extent. Importantly, NPY-IN activation also reduces behavioural signs of inflammatory and neuropathic pain. These results demonstrate that NPY-INs gate pain and itch transmission at the spinal level, and therefore represent a potential treatment target for pathological pain and itch.
Collapse
Affiliation(s)
- Kieran A Boyle
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Erika Polgar
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Maria Gutierrez-Mecinas
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Allen C Dickie
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrew H Cooper
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrew M Bell
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Evelline Jumolea
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Adrian Casas-Benito
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - David I Hughes
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Gregory A Weir
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - John S Riddell
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrew J Todd
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
45
|
Bryson M, Kloefkorn H, Idlett-Ali S, Martin K, Garraway SM, Hochman S. Emergent epileptiform activity drives spinal sensory circuits to generate ectopic bursting in intraspinal afferent axons after cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547522. [PMID: 37461440 PMCID: PMC10349934 DOI: 10.1101/2023.07.03.547522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/14/2024]
Abstract
Spinal cord injury ( SCI ) leads to hyperexcitability and dysfunction in spinal sensory processing. As hyperexcitable circuits can become epileptiform elsewhere, we explored whether such activity emerges in spinal sensory circuits in a thoracic SCI contusion model of neuropathic pain. Recordings from spinal sensory axons in multiple below-lesion segmental dorsal roots ( DRs ) demonstrated that SCI facilitated the emergence of spontaneous ectopic burst spiking in afferent axons, which synchronized across multiple adjacent DRs. Burst frequency correlated with behavioral mechanosensitivity. The same bursting events were recruited by afferent stimulation, and timing interactions with ongoing spontaneous bursts revealed that recruitment was limited by a prolonged post-burst refractory period. Ectopic bursting in afferent axons was driven by GABA A receptor activation, presumably via shifting subthreshold GABAergic interneuronal presynaptic axoaxonic inhibitory actions to suprathreshold spiking. Collectively, the emergence of stereotyped bursting circuitry with hypersynchrony, sensory input activation, post-burst refractory period, and reorganization of connectivity represent defining features of epileptiform networks. Indeed, these same features were reproduced in naïve animals with the convulsant 4-aminopyridine ( 4-AP ). We conclude that SCI promotes the emergence of epileptiform activity in spinal sensory networks that promotes profound corruption of sensory signaling. This corruption includes downstream actions driven by ectopic afferent bursts that propagate via reentrant central and peripheral projections and GABAergic presynaptic circuit hypoexcitability during the refractory period.
Collapse
|
46
|
Cerqueira ARA, Rodrigues L, Coavoy-Sánchez SA, Teixeira SA, Feitosa KB, Taniguchi EY, Lopes LR, Cassola AC, Muscará MN, Sá-Nunes A, Costa SKP. Aedes aegypti salivary gland extract alleviates acute itching by blocking TRPA1 channels. Front Physiol 2023; 14:1055706. [PMID: 37441000 PMCID: PMC10333701 DOI: 10.3389/fphys.2023.1055706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Aedes aegypti (Ae. aegypti) saliva induces a variety of anti-inflammatory and immunomodulatory activities. Interestingly, although it is known that mosquito bites cause allergic reactions in sensitised hosts, the primary exposure of humans to Ae. aegypti does not evoke significant itching. Whether active components in the saliva of Ae. aegypti can counteract the normal itch reaction to injury produced by a histaminergic or non-histaminergic pathway in vertebrate hosts is unknown. This study investigated the effects of Ae. aegypti mosquito salivary gland extract (SGE) on sensitive reactions such as itching and associated skin inflammation. Acute pruritus and plasma extravasation were induced in mice by the intradermal injection of either compound 48/80 (C48/80), the Mas-related G protein-coupled receptor (Mrgpr) agonist chloroquine (CQ), or the transient receptor potential ankyrin 1 (TRPA1) agonist allyl isothiocyanate (AITC). The i.d. co-injection of Ae. aegypti SGE inhibited itching, plasma extravasation, and neutrophil influx evoked by C48/80, but it did not significantly affect mast cell degranulation in situ or in vitro. Additionally, SGE partially reduced CQ- and AITC-induced pruritus in vivo, suggesting that SGE affects pruriceptive nerve firing independently of the histaminergic pathway. Activation of TRPA1 significantly increased intracellular Ca2+ in TRPA-1-transfected HEK293t lineage, which was attenuated by SGE addition. We showed for the first time that Ae. aegypti SGE exerts anti-pruriceptive effects, which are partially regulated by the histamine-independent itch TRPA1 pathway. Thus, SGE may possess bioactive molecules with therapeutic potential for treating nonhistaminergic itch.
Collapse
Affiliation(s)
- Anderson R. A. Cerqueira
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Leandro Rodrigues
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Simone A. Teixeira
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Karla B. Feitosa
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Erika Y. Taniguchi
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Lucia R. Lopes
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Antônio C. Cassola
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo N. Muscará
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Anderson Sá-Nunes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, Brazil
| | - Soraia K. P. Costa
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
47
|
Wang K, Cai B, Song Y, Chen Y, Zhang X. Somatosensory neuron types and their neural networks as revealed via single-cell transcriptomics. Trends Neurosci 2023:S0166-2236(23)00130-3. [PMID: 37268541 DOI: 10.1016/j.tins.2023.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 06/04/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) has allowed profiling cell types of the dorsal root ganglia (DRG) and their transcriptional states in physiology and chronic pain. However, the evaluation criteria used in previous studies to classify DRG neurons varied, which presents difficulties in determining the various types of DRG neurons. In this review, we aim to integrate findings from previous transcriptomic studies of the DRG. We first briefly introduce the history of DRG-neuron cell-type profiling, and discuss the advantages and disadvantages of different scRNA-seq methods. We then examine the classification of DRG neurons based on single-cell profiling under physiological and pathological conditions. Finally, we propose further studies on the somatosensory system at the molecular, cellular, and neural network levels.
Collapse
Affiliation(s)
- Kaikai Wang
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Bing Cai
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Yurang Song
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Yan Chen
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China; Xuhui Central Hospital, Shanghai, 200031, China
| | - Xu Zhang
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; SIMR Joint Lab of Drug Innovation, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China; Xuhui Central Hospital, Shanghai, 200031, China.
| |
Collapse
|
48
|
Wilson AC, Sweeney LB. Spinal cords: Symphonies of interneurons across species. Front Neural Circuits 2023; 17:1146449. [PMID: 37180760 PMCID: PMC10169611 DOI: 10.3389/fncir.2023.1146449] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/23/2023] [Indexed: 05/16/2023] Open
Abstract
Vertebrate movement is orchestrated by spinal inter- and motor neurons that, together with sensory and cognitive input, produce dynamic motor behaviors. These behaviors vary from the simple undulatory swimming of fish and larval aquatic species to the highly coordinated running, reaching and grasping of mice, humans and other mammals. This variation raises the fundamental question of how spinal circuits have changed in register with motor behavior. In simple, undulatory fish, exemplified by the lamprey, two broad classes of interneurons shape motor neuron output: ipsilateral-projecting excitatory neurons, and commissural-projecting inhibitory neurons. An additional class of ipsilateral inhibitory neurons is required to generate escape swim behavior in larval zebrafish and tadpoles. In limbed vertebrates, a more complex spinal neuron composition is observed. In this review, we provide evidence that movement elaboration correlates with an increase and specialization of these three basic interneuron types into molecularly, anatomically, and functionally distinct subpopulations. We summarize recent work linking neuron types to movement-pattern generation across fish, amphibians, reptiles, birds and mammals.
Collapse
Affiliation(s)
| | - Lora B. Sweeney
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Lower Austria, Austria
| |
Collapse
|
49
|
Huo J, Du F, Duan K, Yin G, Liu X, Ma Q, Dong D, Sun M, Hao M, Su D, Huang T, Ke J, Lai S, Zhang Z, Guo C, Sun Y, Cheng L. Identification of brain-to-spinal circuits controlling the laterality and duration of mechanical allodynia in mice. Cell Rep 2023; 42:112300. [PMID: 36952340 DOI: 10.1016/j.celrep.2023.112300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 12/22/2022] [Accepted: 03/07/2023] [Indexed: 03/24/2023] Open
Abstract
Mechanical allodynia (MA) represents one prevalent symptom of chronic pain. Previously we and others have identified spinal and brain circuits that transmit or modulate the initial establishment of MA. However, brain-derived descending pathways that control the laterality and duration of MA are still poorly understood. Here we report that the contralateral brain-to-spinal circuits, from Oprm1 neurons in the lateral parabrachial nucleus (lPBNOprm1), via Pdyn neurons in the dorsal medial regions of hypothalamus (dmHPdyn), to the spinal dorsal horn (SDH), act to prevent nerve injury from inducing contralateral MA and reduce the duration of bilateral MA induced by capsaicin. Ablating/silencing dmH-projecting lPBNOprm1 neurons or SDH-projecting dmHPdyn neurons, deleting Dyn peptide from dmH, or blocking spinal κ-opioid receptors all led to long-lasting bilateral MA. Conversely, activation of dmHPdyn neurons or their axonal terminals in SDH can suppress sustained bilateral MA induced by lPBN lesion.
Collapse
Affiliation(s)
- Jiantao Huo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Feng Du
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kaifang Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guangjuan Yin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xi Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Quan Ma
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dong Dong
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mengge Sun
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mei Hao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dongmei Su
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tianwen Huang
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Jin Ke
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Shishi Lai
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Zhi Zhang
- Division of Life Sciences and Medicine, CAS Key Laboratory of Brain Function and Diseases, University of Science and Technology of China, Hefei 230027, China
| | - Chao Guo
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuanjie Sun
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Longzhen Cheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Department of Biology, Brain Research Center, Southern University of Science and Technology, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
50
|
Tan S, Faull RLM, Curtis MA. The tracts, cytoarchitecture, and neurochemistry of the spinal cord. Anat Rec (Hoboken) 2023; 306:777-819. [PMID: 36099279 DOI: 10.1002/ar.25079] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/01/2022] [Accepted: 09/11/2022] [Indexed: 11/06/2022]
Abstract
The human spinal cord can be described using a range of nomenclatures with each providing insight into its structure and function. Here we have comprehensively reviewed the key literature detailing the general structure, configuration of tracts, the cytoarchitecture of Rexed's laminae, and the neurochemistry at the spinal segmental level. The purpose of this review is to detail current anatomical understanding of how the spinal cord is structured and to aid researchers in identifying gaps in the literature that need to be studied to improve our knowledge of the spinal cord which in turn will improve the potential of therapeutic intervention for disorders of the spinal cord.
Collapse
Affiliation(s)
- Sheryl Tan
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| |
Collapse
|