1
|
Lu D, Uldry Lavergne CG, Choi S, Park J, Kim J, Zhao S, Desimone Q, Lendaro E, Chen B, Han BX, Wang F, Goldstein N. General anesthesia activates a central anxiolytic center in the BNST. Cell Rep 2024; 43:114909. [PMID: 39460938 DOI: 10.1016/j.celrep.2024.114909] [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: 01/24/2024] [Revised: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Low doses of general anesthetics like ketamine and dexmedetomidine have anxiolytic properties independent of their sedative effects, but the underlying mechanisms remain unclear. We discovered a population of GABAergic neurons in the oval division of the bed nucleus of the stria terminalis that are activated by multiple anesthetics and the anxiolytic drug diazepam (ovBNSTGA). The majority of ovBNSTGA neurons express neurotensin receptor 1 (Ntsr1) and form circuits with brain regions known to regulate anxiety and stress responses. Optogenetic activation of ovBNSTGA or ovBNSTNtsr1 neurons significantly attenuated anxiety-like behaviors in both naive animals and mice with inflammatory pain, while inhibition of these cells elevated anxiety. Activation of these neurons decreased heart rate and increased heart rate variability, suggesting that they reduce anxiety by modulating autonomic responses. Our study identifies ovBNSTGA/ovBNSTNtsr1 neurons as a common neural substrate mediating the anxiolytic effect of low-dose anesthetics and a potential therapeutic target for treating anxiety-related disorders.
Collapse
Affiliation(s)
- Dongye Lu
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Camille G Uldry Lavergne
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Seonmi Choi
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jaehong Park
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Jiwoo Kim
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Shengli Zhao
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Quinn Desimone
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eva Lendaro
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bin Chen
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bao-Xia Han
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Fan Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Nitsan Goldstein
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
2
|
Robertson RV, Meylakh N, Crawford LS, Tinoco Mendoza FA, Macey PM, Macefield VG, Keay KA, Henderson LA. Differential activation of lateral parabrachial nuclei and their limbic projections during head compared with body pain: A 7-Tesla functional magnetic resonance imaging study. Neuroimage 2024; 299:120832. [PMID: 39236852 DOI: 10.1016/j.neuroimage.2024.120832] [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: 06/28/2024] [Revised: 08/16/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Pain is a complex experience that involves sensory, emotional, and motivational components. It has been suggested that pain arising from the head and orofacial regions evokes stronger emotional responses than pain from the body. Indeed, recent work in rodents reports different patterns of activation in ascending pain pathways during noxious stimulation of the skin of the face when compared to noxious stimulation of the body. Such differences may dictate different activation patterns in higher brain regions, specifically in those areas processing the affective component of pain. We aimed to use ultra-high field functional magnetic resonance imaging (fMRI at 7-Tesla) to determine whether noxious thermal stimuli applied to the surface of the face and body evoke differential activation patterns within the ascending pain pathway in awake humans (n=16). Compared to the body, noxious heat stimulation to the face evoked more widespread signal changes in prefrontal cortical regions and numerous brainstem and subcortical limbic areas. Moreover, facial pain evoked significantly different signal changes in the lateral parabrachial nucleus, substantia nigra, paraventricular hypothalamus, and paraventricular thalamus, to those evoked by body pain. These results are consistent with recent preclinical findings of differential activation in the brainstem and subcortical limbic nuclei and associated cortices during cutaneous pain of the face when compared with the body. The findings suggest one potential mechanism by which facial pain could evoke a greater emotional impact than that evoked by body pain.
Collapse
Affiliation(s)
- Rebecca V Robertson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Noemi Meylakh
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Lewis S Crawford
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Fernando A Tinoco Mendoza
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Paul M Macey
- UCLA School of Nursing and Brain Research Institute, University of California, Los Angeles, California, 90095, USA
| | | | - Kevin A Keay
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Luke A Henderson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia.
| |
Collapse
|
3
|
Casas-Limón J, Quintas S, López-Bravo A, Alpuente A, Andrés-López A, Castro-Sánchez MV, Membrilla JA, Morales-Hernández C, González-García N, Irimia P. Unravelling Migraine Stigma: A Comprehensive Review of Its Impact and Strategies for Change. J Clin Med 2024; 13:5222. [PMID: 39274435 PMCID: PMC11396411 DOI: 10.3390/jcm13175222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/16/2024] Open
Abstract
Migraine-related stigma is a pervasive issue impacting nearly half of chronic migraine patients, with significant consequences for their quality of life, disability and mental health. Despite its profound effects, migraine stigma remains under-recognised in both clinical practice and research. This narrative review explores the three primary types of stigmas affecting migraine patients: public, structural and internalised. Public stigma involves negative societal attitudes and stereotypes that trivialise the condition. Structural stigma is reflected in policies that restrict access to necessary care and resources. Internalised stigma occurs when patients absorb these negative views, leading to self-blame and diminished self-worth. Addressing these different types of stigmas is crucial for improving the understanding, diagnosis and treatment of migraine. Educational efforts, advocacy and policy reform are essential strategies in this context. A deep understanding of stigma is vital for developing effective interventions that enhance clinical management and patient quality of life. Ultimately, reducing stigma can lead to better health outcomes and a more comprehensive approach to migraine care.
Collapse
Affiliation(s)
- Javier Casas-Limón
- Headache Unit, Hospital Universitario Fundación Alcorcón, 28922 Alcorcón, Spain
| | - Sonia Quintas
- Headache Unit, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), 28006 Madrid, Spain
| | | | - Alicia Alpuente
- Headache Unit, Hospital Universitario Vall d'Hebron, 08035 Barcelona, Spain
| | - Alberto Andrés-López
- Headache Unit, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | | | | | | | | | - Pablo Irimia
- Headache Unit, Clínica Universitaria de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
4
|
Liedtke W. [Additional experience with medicinal treatment of trigeminal nerve pain]. Schmerz 2024:10.1007/s00482-024-00820-2. [PMID: 39141098 DOI: 10.1007/s00482-024-00820-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2024] [Indexed: 08/15/2024]
Affiliation(s)
- Wolfgang Liedtke
- US Facial Pain Association, 7778 McGinnis Ferry Road, 30024, Suwanee, GA, USA.
- Department of Neurology, Anesthesiology and Neurobiology, Duke University, Durham, NC, USA.
- Department of Molecular Pathobiology - Dental Pain Research, New York University, College of Dentistry, New York, NY, USA.
- New York University, Pain Research Center, New York, NY, USA.
| |
Collapse
|
5
|
Chen C, Niehaus JK, Dinc F, Huang KL, Barnette AL, Tassou A, Shuster SA, Wang L, Lemire A, Menon V, Ritola K, Hantman AW, Zeng H, Schnitzer MJ, Scherrer G. Neural circuit basis of placebo pain relief. Nature 2024; 632:1092-1100. [PMID: 39048016 PMCID: PMC11358037 DOI: 10.1038/s41586-024-07816-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
Placebo effects are notable demonstrations of mind-body interactions1,2. During pain perception, in the absence of any treatment, an expectation of pain relief can reduce the experience of pain-a phenomenon known as placebo analgesia3-6. However, despite the strength of placebo effects and their impact on everyday human experience and the failure of clinical trials for new therapeutics7, the neural circuit basis of placebo effects has remained unclear. Here we show that analgesia from the expectation of pain relief is mediated by rostral anterior cingulate cortex (rACC) neurons that project to the pontine nucleus (rACC→Pn)-a precerebellar nucleus with no established function in pain. We created a behavioural assay that generates placebo-like anticipatory pain relief in mice. In vivo calcium imaging of neural activity and electrophysiological recordings in brain slices showed that expectations of pain relief boost the activity of rACC→Pn neurons and potentiate neurotransmission in this pathway. Transcriptomic studies of Pn neurons revealed an abundance of opioid receptors, further suggesting a role in pain modulation. Inhibition of the rACC→Pn pathway disrupted placebo analgesia and decreased pain thresholds, whereas activation elicited analgesia in the absence of placebo conditioning. Finally, Purkinje cells exhibited activity patterns resembling those of rACC→Pn neurons during pain-relief expectation, providing cellular-level evidence for a role of the cerebellum in cognitive pain modulation. These findings open the possibility of targeting this prefrontal cortico-ponto-cerebellar pathway with drugs or neurostimulation to treat pain.
Collapse
Affiliation(s)
- Chong Chen
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jesse K Niehaus
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Fatih Dinc
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- CNC Program, Stanford University, Stanford, CA, USA
| | - Karen L Huang
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexander L Barnette
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - S Andrew Shuster
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Lihua Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Andrew Lemire
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Vilas Menon
- Department of Neurology, Columbia University, New York, NY, USA
| | - Kimberly Ritola
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam W Hantman
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mark J Schnitzer
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- CNC Program, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
- James H. Clark Center for Biomedical Engineering & Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
6
|
Yan Y, Jiao Y, Liang E, Lei X, Zhang N, Xu S, Zhang L, Wang J, Luo T, Yuan J, Yuan C, Yang H, Dong H, Yu T, Yu W. Parabrachial nucleus Vglut2 expressing neurons projection to the extended amygdala involved in the regulation of wakefulness during sevoflurane anesthesia in mice. CNS Neurosci Ther 2024; 30:e70001. [PMID: 39154359 PMCID: PMC11330651 DOI: 10.1111/cns.70001] [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: 02/21/2024] [Revised: 07/24/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024] Open
Abstract
AIMS The parabrachial nucleus (PBN) promotes wakefulness states under general anesthesia. Recent studies have shown that glutamatergic neurons within the PBN play a crucial role in facilitating emergence from anesthesia. Our previous study indicates that vesicular glutamate transporter 2 (vglut2) expression neurons of the PBN extend into the extended amygdala (EA). However, the modulation of PBNvglut2-EA in general anesthesia remains poorly understood. This study aims to investigate the role of PBNvglut2-EA in alterations of consciousness during sevoflurane anesthesia. METHODS We first validated vglut2-expressing neuron projections from the PBN to the EA using anterograde tracing. Then, we conducted immunofluorescence staining of c-Fos to investigate the role of the EA involved in the regulation of consciousness during sevoflurane anesthesia. After, we performed calcium fiber photometry recordings to determine the changes in PBNvglut2-EA activity. Lastly, we modulated PBNvglut2-EA activity under sevoflurane anesthesia using optogenetics, and electroencephalogram (EEG) was recorded during specific optogenetic modulation. RESULTS The expression of vglut2 in PBN neurons projected to the EA, and c-Fos expression in the EA was significantly reduced during sevoflurane anesthesia. Fiber photometry revealed that activity in the PBNvglut2-EA pathway was suppressed during anesthesia induction but restored upon awakening. Optogenetic activation of the PBNvglut2-EA delayed the induction of anesthesia. Meanwhile, EEG recordings showed significantly decreased δ oscillations and increased β and γ oscillations compared to the EYFP group. Furthermore, optogenetic activation of the PBNvglut2-EA resulted in an acceleration of awakening from anesthesia, accompanied by decreased δ oscillations on EEG recordings. Optogenetic inhibition of PBNvglut2-EA accelerated anesthesia induction. Surprisingly, we found a sex-specific regulation of PBNvglut2-EA in this study. The activity of PBNvglut2-EA was lower in males during the induction of anesthesia and decreased more rapidly during sevoflurane anesthesia compared to females. Photoactivation of the PBNvglut2-EA reduced the sensitivity of males to sevoflurane, showing more pronounced wakefulness behavior and EEG changes than females. CONCLUSIONS PBNvglut2-EA is involved in the promotion of wakefulness under sevoflurane anesthesia. Furthermore, PBNvglut2-EA shows sex differences in the changes of consciousness induced by sevoflurane anesthesia.
Collapse
|
7
|
Onishi T, Hirose K, Sakaba T. Molecular tools to capture active neural circuits. Front Neural Circuits 2024; 18:1449459. [PMID: 39100199 PMCID: PMC11294111 DOI: 10.3389/fncir.2024.1449459] [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: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
To understand how neurons and neural circuits function during behaviors, it is essential to record neuronal activity in the brain in vivo. Among the various technologies developed for recording neuronal activity, molecular tools that induce gene expression in an activity-dependent manner have attracted particular attention for their ability to clarify the causal relationships between neuronal activity and behavior. In this review, we summarize recently developed activity-dependent gene expression tools and their potential contributions to the study of neural circuits.
Collapse
Affiliation(s)
- Taichi Onishi
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo City, Bunkyo, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo City, Bunkyo, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto, Japan
| |
Collapse
|
8
|
Kramer PR, Hornung RS, Umorin M, Benson MD, Kinchington PR. Neurexin 3 Regulates Synaptic Connections Between Central Amygdala Neurons and Excitable Cells of the Lateral Parabrachial Nucleus in Rats with Varicella Zoster Induced Orofacial Pain. J Pain Res 2024; 17:2311-2324. [PMID: 38974829 PMCID: PMC11227312 DOI: 10.2147/jpr.s441706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 03/14/2024] [Indexed: 07/09/2024] Open
Abstract
Introduction Herpes Zoster in humans is the result of varicella zoster virus (VZV) infection. Injecting rats with varicella zoster virus produces pain similar to herpes zoster "shingles" pain in humans. . In a previous study, orofacial pain was induced by injecting the whisker pad of male rats with VZV and the pain response increased after attenuating neurexin 3 (Nrxn3) expression in the central amygdala. Neurons descend from the central amygdala to the lateral parabrachial nucleus and orofacial pain signals ascend to the lateral parabrachial nucleus. GABAergic neurons within the central amygdala regulate pain by inhibiting activity within the lateral parabrachial nucleus. Attenuating Nrxn3 expression in the central amygdala increased GABA release in the lateral parabrachial nucleus suggesting Nrxn3 controls pain by regulating GABA release. Nrxn3 can also control synaptic connections between neurons, and we hypothesized that Nrxn3 knockdown in the central amygdala would reduce the number of GABAergic synaptic connections in the lateral parabrachial nucleus and increase VZV associated pain. Methods To test this idea, the number of synaptic connections between GABAergic cells of the central amygdala and excitatory or dynorphin positive neurons within the lateral parabrachial nucleus were quantitated after infusion of a virus expressing synaptophysin. Synaptophysin is a synaptic vesicle protein that labels neuronal synaptic connections. These connections were measured in rats with and without whisker pad injection of VZV and knockdown of Nrxn3 within the central amygdala. Orofacial pain was measured using a place escape avoidance paradigm. Results GABAergic synaptic connections were reduced in the lateral parabrachial nucleus after Nrxn3 knockdown. Rats with a reduction in the number of connections had an increase in VZV associated orofacial pain. Immunostaining with the pain marker prodynorphin indicated that the reduction in GABAergic connections was primarily associated with prodynorphin positive neurons. Discussion The results suggest Nrxn3 reduces VZV associated orofacial pain, in part, by enhancing synaptic connections between GABA cells of the central amygdala and pain neurons within the lateral parabrachial nucleus.
Collapse
Affiliation(s)
- Phillip R Kramer
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - Rebecca S Hornung
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - Mikhail Umorin
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - M Douglas Benson
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - Paul R Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Aji A, Zhang C, Liu W, Chen T, Liu Z, Zuo J, Li H, Mi W, Mao-Ying QL, Wang Y, Zhao Q, Chu YX. Foxg1 Modulation of the Prkcd Gene in the Lateral Habenula Mediates Trigeminal Neuralgia-Associated Anxiety-Like Behaviors in Mice. Mol Neurobiol 2024; 61:4335-4351. [PMID: 38085455 DOI: 10.1007/s12035-023-03856-0] [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: 04/20/2023] [Accepted: 11/28/2023] [Indexed: 07/11/2024]
Abstract
Trigeminal Neuralgia (TN) is a debilitating disorder frequently accompanied by mood complications such as depression and anxiety. The current study sought to elucidate the molecular underpinnings that contribute to the pathogenesis of TN and its associated anxiety. Employing a partial transection of the infraorbital nerve (pT-ION) in a murine model, we successfully induced sustained primary and secondary orofacial allodynia alongside anxiety-like behavioral manifestations. Transcriptome-wide gene microarray analyses revealed a marked upregulation of Foxg1 subsequent to pT-ION. Targeted knockdown of Foxg1, achieved through bilateral microinjection of adeno-associated virus harboring Foxg1-specific shRNA into the lateral habenula (LHb), resulted in a significant attenuation of both orofacial pain and anxiety-like behaviors. Subsequent RNA sequencing implicated Prkcd as a downstream effector gene modulated by Foxg1. Pharmacological inhibition of protein kinase C delta, encoded by Prkcd, within the LHb markedly ameliorated pT-ION-induced symptomatology. The dual luciferase assay revealed that Foxg1 substantially enhances the transcriptional activity of the Prkcd gene. Collectively, these findings indicate that trigeminal nerve injury leads to Foxg1 upregulation in the LHb, which in turn elevates the expression of Prkcd, culminating in the manifestation of orofacial pain and anxiety-like behaviors. This work offers promising therapeutic targets and a conceptual framework for the clinical management of TN and its psychological comorbidities.
Collapse
Affiliation(s)
- Abudula Aji
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Chen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Wenbo Liu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Teng Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Zhechen Liu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Jiaxin Zuo
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Haojun Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Wenli Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Qi-Liang Mao-Ying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qing Zhao
- Shanghai Sunshine Rehabilitation Center, Shanghai Yangzhi Rehabilitation Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Bress KS, Cascio CJ. Sensorimotor regulation of facial expression - An untouched frontier. Neurosci Biobehav Rev 2024; 162:105684. [PMID: 38710425 DOI: 10.1016/j.neubiorev.2024.105684] [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: 01/13/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/08/2024]
Abstract
Facial expression is a critical form of nonverbal social communication which promotes emotional exchange and affiliation among humans. Facial expressions are generated via precise contraction of the facial muscles, guided by sensory feedback. While the neural pathways underlying facial motor control are well characterized in humans and primates, it remains unknown how tactile and proprioceptive information reaches these pathways to guide facial muscle contraction. Thus, despite the importance of facial expressions for social functioning, little is known about how they are generated as a unique sensorimotor behavior. In this review, we highlight current knowledge about sensory feedback from the face and how it is distinct from other body regions. We describe connectivity between the facial sensory and motor brain systems, and call attention to the other brain systems which influence facial expression behavior, including vision, gustation, emotion, and interoception. Finally, we petition for more research on the sensory basis of facial expressions, asserting that incomplete understanding of sensorimotor mechanisms is a barrier to addressing atypical facial expressivity in clinical populations.
Collapse
Affiliation(s)
- Kimberly S Bress
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| | - Carissa J Cascio
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
11
|
Korkutata M, De Luca R, Fitzgerald B, Arrigoni E, Scammell TE. Afferent projections to the Calca /CGRP-expressing parabrachial neurons in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593004. [PMID: 38766214 PMCID: PMC11100666 DOI: 10.1101/2024.05.07.593004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The parabrachial nucleus (PB), located in the dorsolateral pons, contains primarily glutamatergic neurons which regulate responses to a variety of interoceptive and cutaneous sensory signals. The lateral PB subpopulation expressing the Calca gene which produces the neuropeptide calcitonin gene-related peptide (CGRP) relays signals related to threatening stimuli such as hypercarbia, pain, and nausea, yet the afferents to these neurons are only partially understood. We mapped the afferent projections to the lateral part of the PB in mice using conventional cholera toxin B subunit (CTb) retrograde tracing, and then used conditional rabies virus retrograde tracing to map monosynaptic inputs specifically targeting the PB Calca /CGRP neurons. Using vesicular GABA (vGAT) and glutamate (vGLUT2) transporter reporter mice, we found that lateral PB neurons receive GABAergic afferents from regions such as the lateral part of the central nucleus of the amygdala, lateral dorsal subnucleus of the bed nucleus of the stria terminalis, substantia innominata, and the ventrolateral periaqueductal gray. Additionally, they receive glutamatergic afferents from the infralimbic and insular cortex, paraventricular nucleus, parasubthalamic nucleus, trigeminal complex, medullary reticular nucleus, and nucleus of the solitary tract. Using anterograde tracing and confocal microscopy, we then identified close axonal appositions between these afferents and PB Calca /CGRP neurons. Finally, we used channelrhodopsin-assisted circuit mapping to test whether some of these inputs directly synapse upon the PB Calca /CGRP neurons. These findings provide a comprehensive neuroanatomical framework for understanding the afferent projections regulating the PB Calca /CGRP neurons.
Collapse
|
12
|
Condon LF, Yu Y, Park S, Cao F, Pauli JL, Nelson TS, Palmiter RD. Parabrachial Calca neurons drive nociplasticity. Cell Rep 2024; 43:114057. [PMID: 38583149 PMCID: PMC11210282 DOI: 10.1016/j.celrep.2024.114057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/16/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Pain that persists beyond the time required for tissue healing and pain that arises in the absence of tissue injury, collectively referred to as nociplastic pain, are poorly understood phenomena mediated by plasticity within the central nervous system. The parabrachial nucleus (PBN) is a hub that relays aversive sensory information and appears to play a role in nociplasticity. Here, by preventing PBN Calca neurons from releasing neurotransmitters, we demonstrate that activation of Calca neurons is necessary for the manifestation and maintenance of chronic pain. Additionally, by directly stimulating Calca neurons, we demonstrate that Calca neuron activity is sufficient to drive nociplasticity. Aversive stimuli of multiple sensory modalities, such as exposure to nitroglycerin, cisplatin, or lithium chloride, can drive nociplasticity in a Calca-neuron-dependent manner. Aversive events drive nociplasticity in Calca neurons in the form of increased activity and excitability; however, neuroplasticity also appears to occur in downstream circuitry.
Collapse
Affiliation(s)
- Logan F Condon
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA; Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Ying Yu
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Sekun Park
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Feng Cao
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jordan L Pauli
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Tyler S Nelson
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Richard D Palmiter
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
13
|
Sato N, Takahashi Y, Sugimura YK, Kato F. Presynaptic inhibition of excitatory synaptic transmission from the calcitonin gene-related peptide-containing parabrachial neurons to the central amygdala in mice - unexpected influence of systemic inflammation thereon. J Pharmacol Sci 2024; 154:264-273. [PMID: 38485344 DOI: 10.1016/j.jphs.2024.02.004] [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: 01/09/2024] [Accepted: 02/02/2024] [Indexed: 03/19/2024] Open
Abstract
The monosynaptic connection from the lateral parabrachial nucleus (LPB) to the central amygdala (CeA) serves as a fundamental pathway for transmitting nociceptive signals to the brain. The LPB receives nociceptive information from the dorsal horn and spinal trigeminal nucleus and sends it to the "nociceptive" CeA, which modulates pain-associated emotions and nociceptive sensitivity. To elucidate the role of densely expressed mu-opioid receptors (MORs) within this pathway, we investigated the effects of exogenously applied opioids on LPB-CeA synaptic transmission, employing optogenetics in mice expressing channelrhodopsin-2 in LPB neurons with calcitonin gene-related peptide (CGRP). A MOR agonist ([D-Ala2,N-Me-Phe4,Glycinol5]-enkephalin, DAMGO) significantly reduced the amplitude of light-evoked excitatory postsynaptic currents (leEPSCs), in a manner negatively correlated with an increase in the paired-pulse ratio. An antagonist of MORs significantly attenuated these effects. Notably, this antagonist significantly increased leEPSC amplitude when applied alone, an effect further amplified in mice subjected to lipopolysaccharide injection 2 h before brain isolation, yet not observed at the 24-h mark. We conclude that opioids could shut off the ascending nociceptive signal at the LPB-CeA synapse through presynaptic mechanisms. Moreover, this gating process might be modulated by endogenous opioids, and the innate immune system influences this modulation.
Collapse
Affiliation(s)
- Naoko Sato
- Department of Neuroscience, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan; Center for Neuroscience of Pain, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Yukari Takahashi
- Department of Neuroscience, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan; Center for Neuroscience of Pain, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Yae K Sugimura
- Department of Neuroscience, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan; Center for Neuroscience of Pain, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Fusao Kato
- Department of Neuroscience, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan; Center for Neuroscience of Pain, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan.
| |
Collapse
|
14
|
Chen H, Bleimeister IH, Nguyen EK, Li J, Cui AY, Stratton HJ, Smith KM, Baccei ML, Ross SE. The functional and anatomical characterization of three spinal output pathways of the anterolateral tract. Cell Rep 2024; 43:113829. [PMID: 38421871 PMCID: PMC11025583 DOI: 10.1016/j.celrep.2024.113829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/24/2023] [Accepted: 02/03/2024] [Indexed: 03/02/2024] Open
Abstract
The nature of spinal output pathways that convey nociceptive information to the brain has been the subject of controversy. Here, we provide anatomical, molecular, and functional characterizations of two distinct anterolateral pathways: one, ascending in the lateral spinal cord, triggers nociceptive behaviors, and the other one, ascending in the ventral spinal cord, when inhibited, leads to sensorimotor deficits. Moreover, the lateral pathway consists of at least two subtypes. The first is a contralateral pathway that extends to the periaqueductal gray (PAG) and thalamus; the second is a bilateral pathway that projects to the bilateral parabrachial nucleus (PBN). Finally, we present evidence showing that activation of the contralateral pathway is sufficient for defensive behaviors such as running and freezing, whereas the bilateral pathway is sufficient for attending behaviors such as licking and guarding. This work offers insight into the complex organizational logic of the anterolateral system in the mouse.
Collapse
Affiliation(s)
- Haichao Chen
- Tsinghua Medicine, Tsinghua University, Beijing 100084, China; Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Isabel H Bleimeister
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eileen K Nguyen
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jie Li
- Department of Anesthesiology, Pain Research Center, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Abby Yilin Cui
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Harrison J Stratton
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kelly M Smith
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mark L Baccei
- Department of Anesthesiology, Pain Research Center, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Sarah E Ross
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
15
|
Sakurai K. Rethinking c-Fos for understanding drug action in the brain. J Biochem 2024; 175:377-381. [PMID: 38153290 DOI: 10.1093/jb/mvad110] [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: 09/04/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 12/29/2023] Open
Abstract
Understanding the mechanisms of drug action in the brain, from the genetic to the neural circuit level, is crucial for the development of new agents that act upon the central nervous system. Determining the brain regions and neurons affected by a drug is essential for revealing its mechanism of action in the brain. c-Fos, a marker of neuronal activation, has been widely used to detect neurons activated by stimuli with high spatial resolution. In this review, the use of c-Fos for the visualization and manipulation of activated neurons is introduced. I also explain that a higher temporal resolution can be achieved by changing the staining method for visualization of c-Fos. Moreover, a new method that allows labeling and manipulating commonly activated neurons using two different stimuli is proposed.
Collapse
Affiliation(s)
- Katsuyasu Sakurai
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
16
|
Katagiri A, Kishimoto S, Okamoto Y, Yamada M, Niwa H, Bereiter DA, Kato T. Effect of chronic intermittent hypoxia on ocular and intraoral mechanical allodynia mediated via the calcitonin gene-related peptide in a rat. Sleep 2024; 47:zsad332. [PMID: 38166171 PMCID: PMC10925949 DOI: 10.1093/sleep/zsad332] [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: 08/09/2023] [Revised: 12/15/2023] [Indexed: 01/04/2024] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea, a significant hypoxic condition, may exacerbate several orofacial pain conditions. The study aims to define the involvement of calcitonin gene-related peptide (CGRP) in peripheral and central sensitization and in evoking orofacial mechanical allodynia under chronic intermittent hypoxia (CIH). METHODS Male rats were exposed to CIH. Orofacial mechanical allodynia was assessed using the eyeblink test and the two-bottle preference drinking test. The CGRP-immunoreactive neurons in the trigeminal ganglion (TG), CGRP-positive primary afferents projecting to laminae I-II of the trigeminal spinal subnucleus caudalis (Vc), and neural responses in the second-order neurons of the Vc were determined by immunohistochemistry. CGRP receptor antagonist was administrated in the TG. RESULTS CIH-induced ocular and intraoral mechanical allodynia. CGRP-immunoreactive neurons and activated satellite glial cells (SGCs) were significantly increased in the TG and the number of cFos-immunoreactive cells in laminae I-II of the Vc were significantly higher in CIH rats compared to normoxic rats. Local administration of the CGRP receptor antagonist in the TG of CIH rats attenuated orofacial mechanical allodynia; the number of CGRP-immunoreactive neurons and activated SGCs in the TG, and the density of CGRP-positive primary afferent terminals and the number of cFos-immunoreactive cells in laminae I-II of the Vc were significantly lower compared to vehicle-administrated CIH rats. CONCLUSIONS An increase in CGRP in the TG induced by CIH, as well as orofacial mechanical allodynia and central sensitization of second-order neurons in the Vc, supported the notion that CGRP plays a critical role in CIH-induced orofacial mechanical allodynia.
Collapse
Affiliation(s)
- Ayano Katagiri
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Saki Kishimoto
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yoshie Okamoto
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masaharu Yamada
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Hitoshi Niwa
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - David A Bereiter
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, MN, USA
| | - Takafumi Kato
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
17
|
Torruella-Suárez ML, Neugebauer B, Flores-Felix K, Keller A, Carrasquillo Y, Cramer N. Divergent changes in PBN excitability in a mouse model of neuropathic pain. eNeuro 2024; 11:ENEURO.0416-23.2024. [PMID: 38331576 PMCID: PMC10921257 DOI: 10.1523/eneuro.0416-23.2024] [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: 10/16/2023] [Revised: 12/19/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
The transition from acute to chronic pain involves maladaptive plasticity in central nociceptive pathways. Growing evidence suggests that changes within the parabrachial nucleus (PBN), an important component of the spino-parabrachio-amygdaloid pain pathway, are key contributors to the development and maintenance of chronic pain. In animal models of chronic pain, PBN neurons become sensitive to normally innocuous stimuli and responses to noxious stimuli become amplified and more often produce after-discharges that outlast the stimulus. Using ex vivo slice electrophysiology and two mouse models of neuropathic pain, sciatic cuff and chronic constriction of the infraorbital nerve (CCI-ION), we find that changes in the firing properties of PBN neurons and a shift in inhibitory synaptic transmission may underlie this phenomenon. Compared to PBN neurons from shams, a larger proportion of PBN neurons from mice with a sciatic cuff were spontaneously active at rest, and these same neurons showed increased excitability relative to shams. In contrast, quiescent PBN neurons from cuff mice were less excitable than those from shams. Despite an increase in excitability in a subset of PBN neurons, the presence of after-discharges frequently observed in vivo were largely absent ex vivo in both injury models. However, GABAB-mediated presynaptic inhibition of GABAergic terminals is enhanced in PBN neurons after CCI-ION. These data suggest that the amplified activity of PBN neurons observed in rodent models of chronic pain arise through a combination of changes in firing properties and network excitability.Significance Statement Hyperactivity of neurons in the parabrachial nucleus (PBN) is causally linked to exaggerated pain behaviors in rodent models of chronic pain but the underlying mechanisms remain unknown. Using two mouse models of neuropathic pain, we show the intrinsic properties of PBN neurons are largely unaltered following injury. However, subsets of PBN neurons become more excitable and GABAB receptor mediated suppression of inhibitory terminals is enhanced after injury. Thus, shifts in network excitability may be a contributing factor in injury induced potentiation of PBN activity.
Collapse
Affiliation(s)
- María L Torruella-Suárez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Benjamin Neugebauer
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Krystal Flores-Felix
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| | - Asaf Keller
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yarimar Carrasquillo
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| | - Nathan Cramer
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
18
|
Torres-Rodriguez JM, Wilson TD, Singh S, Torruella-Suárez ML, Chaudhry S, Adke AP, Becker JJ, Neugebauer B, Lin JL, Martinez Gonzalez S, Soler-Cedeño O, Carrasquillo Y. The parabrachial to central amygdala pathway is critical to injury-induced pain sensitization in mice. Neuropsychopharmacology 2024; 49:508-520. [PMID: 37542159 PMCID: PMC10789863 DOI: 10.1038/s41386-023-01673-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/27/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
The spino-ponto-amygdaloid pathway is a major ascending circuit relaying nociceptive information from the spinal cord to the brain. Potentiation of excitatory synaptic transmission in the parabrachial nucleus (PBN) to central amygdala (CeA) pathway has been reported in rodent models of persistent pain. However, the functional significance of this pathway in the modulation of the somatosensory component of pain was recently challenged by studies showing that spinal nociceptive neurons do not target CeA-projecting PBN cells and that manipulations of this pathway have no effect on reflexive-defensive somatosensory responses to peripheral noxious stimulation. Here, we showed that activation of CeA-projecting PBN neurons is critical to increase both stimulus-evoked and spontaneous nociceptive responses following an injury in male and female mice. Using optogenetic-assisted circuit mapping, we confirmed a functional excitatory projection from PBN→CeA that is independent of the genetic or firing identity of CeA cells. We then showed that peripheral noxious stimulation increased the expression of the neuronal activity marker Fos in CeA-projecting PBN neurons and that chemogenetic inactivation of these cells decreased behavioral hypersensitivity in models of neuropathic and inflammatory pain without affecting baseline nociception. Lastly, we showed that chemogenetic activation of CeA-projecting PBN neurons is sufficient to induced bilateral hypersensitivity without injury. Together, our results indicate that the PBN→CeA pathway is a key modulator of pain-related behaviors that can increase reflexive-defensive and affective-motivational responses to somatosensory stimulation in injured states without affecting nociception under normal physiological conditions.
Collapse
Affiliation(s)
- Jeitzel M Torres-Rodriguez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Torri D Wilson
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Sudhuman Singh
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Maria L Torruella-Suárez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Sarah Chaudhry
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Anisha P Adke
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Jordan J Becker
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Neugebauer
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Jenny L Lin
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Santiago Martinez Gonzalez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Omar Soler-Cedeño
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | - Yarimar Carrasquillo
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA.
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
Jeon H, Lee S, Kim SA, Lee U, Lee S. Effect of Korean Medicine Treatment on Patients with Postherpetic Neuralgia: A Retrospective Chart Review. Healthcare (Basel) 2024; 12:256. [PMID: 38275536 PMCID: PMC10815056 DOI: 10.3390/healthcare12020256] [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: 12/10/2023] [Revised: 01/05/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Evidence regarding Korean medicine treatment (KMT) for neuropathic pain is lacking. We aimed to identify the effects of integrative KMT in patients with postherpetic neuralgia (PHN). We retrospectively analyzed the electronic medical records of patients with PHN who received KMT at Kyung Hee University Korean Medicine Hospital between August 2021 and July 2022. We evaluated the effects of KMT-comprising acupuncture, pharmacopuncture, herbal medicine, cupping, and moxibustion-on pain intensity using the numerical rating scale (NRS), Short-Form McGill Pain Questionnaire (SF-MPQ), Hospital Anxiety and Depression Scale-Anxiety (HADS-A), Hospital Anxiety and Depression Scale-Depression (HADS-D), Daily Sleep Interference Scale (DSIS), Fatigue Severity Scale (FSS), and EuroQol-5D. Among 53 patients with PHN, 13 were included. The NRS score for worst pain over 1 week decreased from 6.54 ± 0.64 at baseline to 3.85 ± 0.63 at 8 weeks (41% reduction, p < 0.01), while that for average pain over 1 week decreased from 4.93 ± 0.67 at baseline to 3.08 ± 0.46 at 8 weeks (37% reduction, p < 0.01). From baseline to 8 weeks, there were significant reductions in the SF-MPQ, HADS-A, FSS, and EuroQol-5D scores. No adverse events were reported after KMT. Therefore, KMT may be an effective treatment option for patients with PHN.
Collapse
Affiliation(s)
- Hyoseung Jeon
- Department of Acupuncture and Moxibustion Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea (S.L.)
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Suji Lee
- Department of Acupuncture and Moxibustion Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea (S.L.)
- Department of Acupuncture and Moxibustion, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Sung-A Kim
- Department of Acupuncture and Moxibustion Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea (S.L.)
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Unhyung Lee
- Department of Acupuncture and Moxibustion Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea (S.L.)
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Seunghoon Lee
- Department of Acupuncture and Moxibustion Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea (S.L.)
- Department of Acupuncture and Moxibustion, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| |
Collapse
|
20
|
Lu D, Choi S, Park J, Kim J, Zhao S, Uldry Lavergne CG, Desimone Q, Chen B, Han BX, Wang F, Goldstein N. General Anesthesia Activates a Central Anxiolytic Center in the BNST. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572586. [PMID: 38187782 PMCID: PMC10769264 DOI: 10.1101/2023.12.20.572586] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Low doses of general anesthetics like ketamine and dexmedetomidine have anxiolytic properties independent of their sedative effects. How these different drugs exert these anxiolytic effects is not well understood. We discovered a population of GABAergic neurons in the oval division of the bed nucleus of the stria terminalis that is activated by multiple anesthetics and the anxiolytic drug diazepam (ovBNST GA ). A majority of ovBNST GA neurons express neurotensin receptor 1 (Ntsr1) and innervate brain regions known to regulate anxiety and stress responses. Optogenetic activation ovBNST GA or ovBNST Ntsr1 neurons significantly attenuated anxiety-like behaviors in both naïve animals and mice with inflammatory pain, while inhibition of these cells increased anxiety. Notably, activation of these neurons decreased heart rate and increased heart rate variability, suggesting that they reduce anxiety through modulation of the autonomic nervous system. Our study identifies ovBNST GA /ovBNST Ntsr1 neurons as one of the brain's endogenous anxiolytic centers and a potential therapeutic target for treating anxiety-related disorders. HIGHLIGHTS General anesthetics and anxiolytics activate a population of neurons in the ovBNSTAnesthesia-activated ovBNST neurons bidirectionally modulate anxiety-like behaviorMost anesthesia-activated ovBNST neurons express neurotensin receptor 1 ovBNST Ntsr1 neuron activation shifts autonomic responses to an anxiolytic state.
Collapse
|
21
|
Sullere S, Kunczt A, McGehee DS. A cholinergic circuit that relieves pain despite opioid tolerance. Neuron 2023; 111:3414-3434.e15. [PMID: 37734381 PMCID: PMC10843525 DOI: 10.1016/j.neuron.2023.08.017] [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: 01/18/2023] [Revised: 04/19/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023]
Abstract
Chronic pain is a tremendous burden for afflicted individuals and society. Although opioids effectively relieve pain, significant adverse outcomes limit their utility and efficacy. To investigate alternate pain control mechanisms, we explored cholinergic signaling in the ventrolateral periaqueductal gray (vlPAG), a critical nexus for descending pain modulation. Biosensor assays revealed that pain states decreased acetylcholine release in vlPAG. Activation of cholinergic projections from the pedunculopontine tegmentum to vlPAG relieved pain, even in opioid-tolerant conditions, through ⍺7 nicotinic acetylcholine receptors (nAChRs). Activating ⍺7 nAChRs with agonists or stimulating endogenous acetylcholine inhibited vlPAG neuronal activity through Ca2+ and peroxisome proliferator-activated receptor α (PPAR⍺)-dependent signaling. In vivo 2-photon imaging revealed that chronic pain induces aberrant excitability of vlPAG neuronal ensembles and that ⍺7 nAChR-mediated inhibition of these cells relieves pain, even after opioid tolerance. Finally, pain relief through these cholinergic mechanisms was not associated with tolerance, reward, or withdrawal symptoms, highlighting its potential clinical relevance.
Collapse
Affiliation(s)
- Shivang Sullere
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Alissa Kunczt
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Daniel S McGehee
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA; Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
22
|
Li J, Tian C, Yuan S, Yin Z, Wei L, Chen F, Dong X, Liu A, Wang Z, Wu T, Tian C, Niu L, Wang L, Wang P, Xie W, Cao F, Shen H. Neuropathic pain following spinal cord hemisection induced by the reorganization in primary somatosensory cortex and regulated by neuronal activity of lateral parabrachial nucleus. CNS Neurosci Ther 2023; 29:3269-3289. [PMID: 37170721 PMCID: PMC10580357 DOI: 10.1111/cns.14258] [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: 12/26/2022] [Revised: 04/04/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023] Open
Abstract
AIMS Neuropathic pain after spinal cord injury (SCI) remains a common and thorny problem, influencing the life quality severely. This study aimed to elucidate the reorganization of the primary sensory cortex (S1) and the regulatory mechanism of the lateral parabrachial nucleus (lPBN) in the presence of allodynia or hyperalgesia after left spinal cord hemisection injury (LHS). METHODS Through behavioral tests, we first identified mechanical allodynia and thermal hyperalgesia following LHS. We then applied two-photon microscopy to observe calcium activity in S1 during mechanical or thermal stimulation and long-term spontaneous calcium activity after LHS. By slice patch clamp recording, the electrophysiological characteristics of neurons in lPBN were explored. Finally, exploiting chemogenetic activation or inhibition of the neurons in lPBN, allodynia or hyperalgesia was regulated. RESULTS The calcium activity in left S1 was increased during mechanical stimulation of right hind limb and thermal stimulation of tail, whereas in right S1 it was increased only with thermal stimulation of tail. The spontaneous calcium activity in right S1 changed more dramatically than that in left S1 after LHS. The lPBN was also activated after LHS, and exploiting chemogenetic activation or inhibition of the neurons in lPBN could induce or alleviate allodynia and hyperalgesia in central neuropathic pain. CONCLUSION The neuronal activity changes in S1 are closely related to limb pain, which has accurate anatomical correspondence. After LHS, the spontaneously increased functional connectivity of calcium transient in left S1 is likely causing the mechanical allodynia in right hind limb and increased neuronal activity in bilateral S1 may induce thermal hyperalgesia in tail. This state of allodynia and hyperalgesia can be regulated by lPBN.
Collapse
Affiliation(s)
- Jing Li
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Chao Tian
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Shiyang Yuan
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Zhenyu Yin
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Liangpeng Wei
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Feng Chen
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Xi Dong
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Aili Liu
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
| | - Zhenhuan Wang
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Tongrui Wu
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Chunxiao Tian
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Lin Niu
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
| | - Lei Wang
- Department of PhysiologyZhuhai Campus of Zunyi Medical UniversityZhuhaiChina
| | - Pu Wang
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Wanyu Xie
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Fujiang Cao
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Hui Shen
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
- Innovation Research Institute of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
23
|
Condon LF, Yu Y, Park S, Cao F, Pauli JL, Nelson TS, Palmiter RD. Parabrachial Calca neurons drive nociplasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564223. [PMID: 37961621 PMCID: PMC10634894 DOI: 10.1101/2023.10.26.564223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Pain that persists beyond the time required for tissue healing and pain that arises in the absence of tissue injury are poorly understood phenomena mediated by plasticity within the central nervous system. The parabrachial nucleus (PBN) is a hub that relays aversive sensory information and appears to play a role in nociplasticity. Here, by preventing PBN Calca neurons from releasing neurotransmitter or directly stimulating them we demonstrate that activation of Calca neurons is both necessary for the manifestation of chronic pain after nerve ligation and is sufficient to drive nociplasticity in wild-type mice. Aversive stimuli such as exposure to nitroglycerin, cisplatin, or LiCl can drive nociplasticity in a Calca-neuron-dependent manner. Calcium fluorescence imaging reveals that nitroglycerin activates PBN Calca neurons and potentiates their responses to mechanical stimulation. The activity and excitability of Calca neurons increased for several days after aversive events, but prolonged nociplasticity likely occurs in downstream circuitry.
Collapse
Affiliation(s)
- Logan F Condon
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Ying Yu
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Sekun Park
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Feng Cao
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jordan L Pauli
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Tyler S Nelson
- Department of Molecular Pathobiology, College of Dentistry, New York University, NY 10010, USA
| | - Richard D Palmiter
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
- Lead Contact
| |
Collapse
|
24
|
De Preter CC, Heinricher MM. Direct and Indirect Nociceptive Input from the Trigeminal Dorsal Horn to Pain-Modulating Neurons in the Rostral Ventromedial Medulla. J Neurosci 2023; 43:5779-5791. [PMID: 37487738 PMCID: PMC10423049 DOI: 10.1523/jneurosci.0680-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023] Open
Abstract
The brain is able to amplify or suppress nociceptive signals by means of descending projections to the spinal and trigeminal dorsal horns from the rostral ventromedial medulla (RVM). Two physiologically defined cell classes within RVM, "ON-cells" and "OFF-cells," respectively facilitate and inhibit nociceptive transmission. However, sensory pathways through which nociceptive input drives changes in RVM cell activity are only now being defined. We recently showed that indirect inputs from the dorsal horn via the parabrachial complex (PB) convey nociceptive information to RVM. The purpose of the present study was to determine whether there are also direct dorsal horn inputs to RVM pain-modulating neurons. We focused on the trigeminal dorsal horn, which conveys sensory input from the face and head, and used a combination of single-cell recording with optogenetic activation and inhibition of projections to RVM and PB from the trigeminal interpolaris-caudalis transition zone (Vi/Vc) in male and female rats. We determined that a direct projection from ventral Vi/Vc to RVM carries nociceptive information to RVM pain-modulating neurons. This projection included a GABAergic component, which could contribute to nociceptive inhibition of OFF-cells. This approach also revealed a parallel, indirect, relay of trigeminal information to RVM via PB. Activation of the indirect pathway through PB produced a more sustained response in RVM compared with activation of the direct projection from Vi/Vc. These data demonstrate that a direct trigeminal output conveys nociceptive information to RVM pain-modulating neurons with a parallel indirect pathway through the parabrachial complex.SIGNIFICANCE STATEMENT Rostral ventromedial medulla (RVM) pain-modulating neurons respond to noxious stimulation, which implies that they receive input from pain-transmission circuits. However, the traditional view has been that there is no direct input to RVM pain-modulating neurons from the dorsal horn, and that nociceptive information is carried by indirect pathways. Indeed, we recently showed that noxious information can reach RVM pain-modulating neurons via the parabrachial complex (PB). Using in vivo electrophysiology and optogenetics, the present study identified a direct relay of nociceptive information from the trigeminal dorsal horn to physiologically identified pain-modulating neurons in RVM. Combined tracing and electrophysiology data revealed that the direct projection includes GABAergic neurons. Direct and indirect pathways may play distinct functional roles in recruiting pain-modulating neurons.
Collapse
Affiliation(s)
- Caitlynn C De Preter
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon 97239
| | - Mary M Heinricher
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
25
|
Berta T, Strong JA, Zhang JM, Ji RR. Targeting dorsal root ganglia and primary sensory neurons for the treatment of chronic pain: an update. Expert Opin Ther Targets 2023; 27:665-678. [PMID: 37574713 PMCID: PMC10530032 DOI: 10.1080/14728222.2023.2247563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
INTRODUCTION Current treatments for chronic pain are inadequate. Here, we provide an update on the new therapeutic strategies that target dorsal root ganglia (DRGs) in the peripheral nervous system for a better and safer treatment of chronic pain. AREAS COVERED Despite the complex nature of chronic pain and its underlying mechanisms, we do know that changes in the plasticity and modality of neurons in DRGs play a pivotal role. DRG neurons are heterogenous and offer potential pain targets for different therapeutic interventions. We discuss the last advancements of these interventions, which include the use of systemic and local administrations, selective nerve drug delivery, and gene therapy. In particular, we provide updates and further details on the molecular characterization of primary sensory neurons, new analgesics entering the market, and future gene therapy approaches. EXPERT OPINION DRGs and primary sensory neurons are promising targets for chronic pain treatment due to their key role in pain signaling, unique anatomical location, and the potential for different targeted therapeutic interventions.
Collapse
Affiliation(s)
- Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Judith A. Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
26
|
Xu J, Wu S, Huo L, Zhang Q, Liu L, Ye Z, Cao J, Ma H, Shang C, Ma C. Trigeminal nerve stimulation restores hippocampal dopamine deficiency to promote cognitive recovery in traumatic brain injury. Prog Neurobiol 2023:102477. [PMID: 37270025 DOI: 10.1016/j.pneurobio.2023.102477] [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: 01/31/2023] [Revised: 04/20/2023] [Accepted: 05/30/2023] [Indexed: 06/05/2023]
Abstract
Cognitive impairment (CI) is a common neurological disease resulting from traumatic brain injury (TBI). Trigeminal nerve stimulation (TNS) is an emerging, non-invasive, and effective neuromodulation therapy especially for patients suffering from brain function disorders. However, the treatment and recovery mechanisms of TNS remain poorly understood. By using combined advanced technologies, we revealed here that the neuroprotective potential of TNS to improve CI caused by TBI. The study results found that 40Hz TNS treatment has the ability to improve CI in TBI mice and communicates with central nervous system via the trigeminal ganglion (TG). Transsynaptic virus experiments revealed that TG is connected to the hippocampus (HPC) through the corticotropin-releasing hormone (CRH) neurons of paraventricular hypothalamic nucleus (PVN) and the dopamine transporter (DAT) neurons of substantia nigra pars compacta/ventral tegmental area (SNc/VTA). Mechanistically, the data showed that TNS can increase the release of dopamine in the HPC by activating the following neural circuit: TG→CRH+ PVN→DAT+ SNc/VTA → HPC. Bulk RNA sequencing confirmed changes in the expression of dopamine-related genes in the HPC. This work preliminarily explains the efficacy and mechanism of TNS and adds to the increasing evidence demonstrating that nerve stimulation is an effective method to treat neurological diseases. DATA AVAILABILITY: The data that support the findings of this study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Jing Xu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Shaoling Wu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Lifang Huo
- Guangzhou Laboratory, Guangzhou, 510005, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Qian Zhang
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Lijiaqi Liu
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Zhimin Ye
- Guangzhou Laboratory, Guangzhou, 510005, China
| | - Jie Cao
- Guangzhou Laboratory, Guangzhou, 510005, China
| | - Haiyun Ma
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China
| | - Congping Shang
- Guangzhou Laboratory, Guangzhou, 510005, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China; School of Basic Medical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510005, China.
| | - Chao Ma
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510030, China.
| |
Collapse
|
27
|
Wang L, Su X, Yan J, Wu Q, Xu X, Wang X, Liu X, Song X, Zhang Z, Hu W, Liu X, Zhang Y. Involvement of Mrgprd-expressing nociceptors-recruited spinal mechanisms in nerve injury-induced mechanical allodynia. iScience 2023; 26:106764. [PMID: 37250305 PMCID: PMC10214713 DOI: 10.1016/j.isci.2023.106764] [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: 08/23/2022] [Revised: 03/17/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Mechanical allodynia and hyperalgesia are intractable symptoms lacking effective clinical treatments in patients with neuropathic pain. However, whether and how mechanically responsive non-peptidergic nociceptors are involved remains elusive. Here, we showed that von Frey-evoked static allodynia and aversion, along with mechanical hyperalgesia after spared nerve injury (SNI) were reduced by ablation of MrgprdCreERT2-marked neurons. Electrophysiological recordings revealed that SNI-opened Aβ-fiber inputs to laminae I-IIo and vIIi, as well as C-fiber inputs to vIIi, were all attenuated in Mrgprd-ablated mice. In addition, priming chemogenetic or optogenetic activation of Mrgprd+ neurons drove mechanical allodynia and aversion to low-threshold mechanical stimuli, along with mechanical hyperalgesia. Mechanistically, gated Aβ and C inputs to vIIi were opened, potentially via central sensitization by dampening potassium currents. Altogether, we uncovered the involvement of Mrgprd+ nociceptors in nerve injury-induced mechanical pain and dissected the underlying spinal mechanisms, thus providing insights into potential therapeutic targets for pain management.
Collapse
Affiliation(s)
- Liangbiao Wang
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiaojing Su
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Jinjin Yan
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Qiaofeng Wu
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiang Xu
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xinyue Wang
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiaoqing Liu
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaoyuan Song
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhi Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Wei Hu
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xinfeng Liu
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yan Zhang
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| |
Collapse
|
28
|
Zheng Y, Xu C, Sun J, Ming W, Dai S, Shao Y, Qiu X, Li M, Shen C, Xu J, Fei F, Fang J, Jiang X, Zheng G, Hu W, Wang Y, Wang S, Ding M, Chen Z. Excitatory somatostatin interneurons in the dentate gyrus drive a widespread seizure network in cortical dysplasia. Signal Transduct Target Ther 2023; 8:186. [PMID: 37193687 DOI: 10.1038/s41392-023-01404-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 02/19/2023] [Accepted: 03/05/2023] [Indexed: 05/18/2023] Open
Abstract
Seizures due to cortical dysplasia are notorious for their poor prognosis even with medications and surgery, likely due to the widespread seizure network. Previous studies have primarily focused on the disruption of dysplastic lesions, rather than remote regions such as the hippocampus. Here, we first quantified the epileptogenicity of the hippocampus in patients with late-stage cortical dysplasia. We further investigated the cellular substrates leading to the epileptic hippocampus, using multiscale tools including calcium imaging, optogenetics, immunohistochemistry and electrophysiology. For the first time, we revealed the role of hippocampal somatostatin-positive interneurons in cortical dysplasia-related seizures. Somatostatin-positive were recruited during cortical dysplasia-related seizures. Interestingly, optogenetic studies suggested that somatostatin-positive interneurons paradoxically facilitated seizure generalization. By contrast, parvalbumin-positive interneurons retained an inhibitory role as in controls. Electrophysiological recordings and immunohistochemical studies revealed glutamate-mediated excitatory transmission from somatostatin-positive interneurons in the dentate gyrus. Taken together, our study reveals a novel role of excitatory somatostatin-positive neurons in the seizure network and brings new insights into the cellular basis of cortical dysplasia.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Cenglin Xu
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Jinyi Sun
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wenjie Ming
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Sijie Dai
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuying Shao
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Menghan Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chunhong Shen
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jinghong Xu
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Fan Fei
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiajia Fang
- Department of Neurology, Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Xuhong Jiang
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Guoqing Zheng
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China
| | - Weiwei Hu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yi Wang
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Shuang Wang
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Meiping Ding
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Zhong Chen
- Department of Neurology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310060, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Epilepsy Center, Department of Neurology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
29
|
Suttle A, Wang P, Dias FC, Zhang Q, Luo Y, Simmons L, Bortsov A, Tchivileva IE, Nackley AG, Chen Y. Sensory Neuron-TRPV4 Modulates Temporomandibular Disorder Pain Via CGRP in Mice. THE JOURNAL OF PAIN 2023; 24:782-795. [PMID: 36509176 PMCID: PMC10164682 DOI: 10.1016/j.jpain.2022.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
Temporomandibular disorder (TMD) pain that involves inflammation and injury in the temporomandibular joint (TMJ) and/or masticatory muscle is the most common form of orofacial pain. We recently found that transient receptor potential vanilloid-4 (TRPV4) in trigeminal ganglion (TG) neurons is upregulated after TMJ inflammation, and TRPV4 coexpresses with calcitonin gene-related peptide (CGRP) in TMJ-innervating TG neurons. Here, we extended these findings to determine the specific contribution of TRPV4 in TG neurons to TMD pain, and examine whether sensory neuron-TRPV4 modulates TMD pain via CGRP. In mouse models of TMJ inflammation or masseter muscle injury, sensory neuron-Trpv4 conditional knockout (cKO) mice displayed reduced pain. Coexpression of TRPV4 and CGRP in TMJ- or masseter muscle-innervating TG neurons was increased after TMJ inflammation and masseter muscle injury, respectively. Activation of TRPV4-expressing TG neurons triggered secretion of CGRP, which was associated with increased levels of CGRP in peri-TMJ tissues, masseter muscle, spinal trigeminal nucleus, and plasma in both models. Local injection of CGRP into the TMJ or masseter muscle evoked acute pain in naïve mice, while blockade of CGRP receptor attenuated pain in mouse models of TMD. These results suggest that TRPV4 in TG neurons contributes to TMD pain by potentiating CGRP secretion. PERSPECTIVE: This study demonstrates that activation of TRPV4 in TG sensory neurons drives pain by potentiating the release of pain mediator CGRP in mouse models of TMJ inflammation and masseter muscle injury. Targeting TRPV4 and CGRP may be of clinical potential in alleviating TMD pain.
Collapse
Affiliation(s)
- Abbie Suttle
- Department of Neurology, Duke University, Durham, North Carolina
| | - Peng Wang
- Department of Neurology, Duke University, Durham, North Carolina
| | - Fabiana C Dias
- Department of Neurology, Duke University, Durham, North Carolina
| | - Qiaojuan Zhang
- Department of Neurology, Duke University, Durham, North Carolina
| | - Yuhui Luo
- Department of Neurology, Duke University, Durham, North Carolina
| | - Lauren Simmons
- Department of Neurology, Duke University, Durham, North Carolina
| | - Andrey Bortsov
- Department of Endodontics, Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina
| | - Inna E Tchivileva
- Center for Pain Research and Innovation, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andrea G Nackley
- Department of Endodontics, Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina; Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Yong Chen
- Department of Neurology, Duke University, Durham, North Carolina; Department of Endodontics, Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina; Department of Pathology, Duke University, Durham, North Carolina.
| |
Collapse
|
30
|
Chen LQ, Lv XJ, Guo QH, Lv SS, Lv N, Xu WD, Yu J, Zhang YQ. Asymmetric activation of microglia in the hippocampus drives anxiodepressive consequences of trigeminal neuralgia in rodents. Br J Pharmacol 2023; 180:1090-1113. [PMID: 36443951 DOI: 10.1111/bph.15994] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/26/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Patients suffering from trigeminal neuralgia are often accompanied by anxiety and depression. Microglia-mediated neuroinflammation is involved in the development of neuropathic pain and anxiodepression pathogenesis. Whether and how microglia are involved in trigeminal neuralgia-induced anxiodepression remains unclear. EXPERIMENTAL APPROACH Unilateral constriction of the infraorbital nerve (CION) was performed to establish trigeminal neuralgia in rat and mouse models. Mechanical allodynia and anxiodepressive-like behaviours were measured. Optogenetic and pharmacological manipulations were employed to investigate the role of hippocampal microglia in anxiety and depression caused by trigeminal neuralgia. KEY RESULTS Trigeminal neuralgia activated ipsilateral but not contralateral hippocampal microglia, up-regulated ipsilateral hippocampal ATP and interleukin-1β (IL-1β) levels, impaired ipsilateral hippocampal long-term potentiation (LTP) and induced anxiodepressive-like behaviours in a time-dependent manner in rodents. Pharmacological or optogenetic inhibition of ipsilateral hippocampal microglia completely blocked trigeminal neuralgia-induced anxiodepressive-like behaviours. Activation of unilateral hippocampal microglia directly elicited an anxiodepressive state and impaired hippocampal LTP. Knockdown of ipsilateral hippocampal P2X7 receptors prevented trigeminal neuralgia-induced microglial activation and anxiodepressive-like behaviours. Furthermore, we demonstrated that microglia-derived IL-1β mediated microglial activation-induced anxiodepressive-like behaviours and LTP impairment. CONCLUSION AND IMPLICATIONS These findings suggest that priming of microglia with ATP/P2X7 receptors in the ipsilateral hippocampus drives pain-related anxiodepressive-like behaviours via IL-1β. An asymmetric role of the bilateral hippocampus in trigeminal neuralgia-induced anxiety and depression was uncovered. The approaches targeting microglia and P2X7 signalling might offer novel therapies for trigeminal neuralgia-related anxiety and depressive disorder.
Collapse
Affiliation(s)
- Li-Qiang Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xue-Jing Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qing-Huan Guo
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Su-Su Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ning Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wen-Dong Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China.,Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Khan J, Singer SR, Young A, Tanaiutchawoot N, Kalladka M, Mupparapu M. Pathogenesis and Differential Diagnosis of Temporomandibular Joint Disorders. Dent Clin North Am 2023; 67:259-280. [PMID: 36965930 DOI: 10.1016/j.cden.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Temporomandibular disorders (TMDs) are an umbrella term including disorders of the temporomandibular joint and muscles of the masticatory system. They are the most common nonodontogenic cause of pain in the orofacial region. A clear understanding of various conditions, underlying mechanisms, clinical presentation, and examination skills is required to effectively diagnose and manage these patients.
Collapse
Affiliation(s)
- Junad Khan
- Orofacial Pain and TMJD, Eastman Institute for Oral Health, 625 Elmwood Avenue, Rochester, NY 14620, USA.
| | - Steven R Singer
- Department of Diagnostic Sciences Division of Oral & Maxillofacial Radiology, Rutgers School of Dental Medicine, 110 Bergen Street | PO Box 1709, Newark, NJ 07101-1709, USA
| | - Andrew Young
- Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA, USA
| | - Naruthorn Tanaiutchawoot
- Department of Diagnostic Sciences Division of Oral & Maxillofacial Radiology, Rutgers School of Dental Medicine, 110 Bergen Street | PO Box 1709, Newark, NJ 07101-1709, USA
| | - Mythili Kalladka
- Orofacial Pain and TMJD, Eastman Institute for Oral Health, 625 Elmwood Avenue, Rochester, NY 14620, USA
| | - Mel Mupparapu
- Penn Dental Medicine, 240 S 40th Street, Philadelphia, PA 19104, USA
| |
Collapse
|
32
|
An SB, Cho YS, Park SK, Kim YS, Bae YC. Synaptic connectivity of the TRPV1-positive trigeminal afferents in the rat lateral parabrachial nucleus. Front Cell Neurosci 2023; 17:1162874. [PMID: 37066077 PMCID: PMC10098450 DOI: 10.3389/fncel.2023.1162874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Recent studies have shown a direct projection of nociceptive trigeminal afferents into the lateral parabrachial nucleus (LPBN). Information about the synaptic connectivity of these afferents may help understand how orofacial nociception is processed in the LPBN, which is known to be involved primarily in the affective aspect of pain. To address this issue, we investigated the synapses of the transient receptor potential vanilloid 1-positive (TRPV1+) trigeminal afferent terminals in the LPBN by immunostaining and serial section electron microscopy. TRPV1 + afferents arising from the ascending trigeminal tract issued axons and terminals (boutons) in the LPBN. TRPV1+ boutons formed synapses of asymmetric type with dendritic shafts and spines. Almost all (98.3%) TRPV1+ boutons formed synapses with one (82.6%) or two postsynaptic dendrites, suggesting that, at a single bouton level, the orofacial nociceptive information is predominantly transmitted to a single postsynaptic neuron with a small degree of synaptic divergence. A small fraction (14.9%) of the TRPV1+ boutons formed synapses with dendritic spines. None of the TRPV1+ boutons were involved in axoaxonic synapses. Conversely, in the trigeminal caudal nucleus (Vc), TRPV1+ boutons often formed synapses with multiple postsynaptic dendrites and were involved in axoaxonic synapses. Number of dendritic spine and total number of postsynaptic dendrites per TRPV1+ bouton were significantly fewer in the LPBN than Vc. Thus, the synaptic connectivity of the TRPV1+ boutons in the LPBN differed significantly from that in the Vc, suggesting that the TRPV1-mediated orofacial nociception is relayed to the LPBN in a distinctively different manner than in the Vc.
Collapse
|
33
|
He B, Wang W, Zhang R, Xu Y, Wei X, Yang Z, Cao Y. Fluorescence visualization of the neuropathic pain triad in trigeminal neuralgia. JOURNAL OF BIOPHOTONICS 2023; 16:e202200301. [PMID: 36369929 DOI: 10.1002/jbio.202200301] [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: 10/02/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
Trigeminal neuralgia (TN), an exemplary condition of neuropathic facial pain, seriously affects the physical and mental health of patients, becoming a major medical and social problem. So far, the mechanism of TN and its relation to neuronal activity remain unclear, largely limited by the spatial resolution of visualization methods. In the meanwhile, current therapeutic strategies targeting neurons have not achieved satisfactory outcome. Here, we investigate the neuropathic pain triad in TN by establishing an animal model of TN by chronic constriction injury of the unilateral infraorbital nerve (ION-CCI) and leveraging the single-cell resolution of confocal microscopy, including neuronal hyperexcitability, glial activation, and macrophage polarization. These results can broaden the understanding of TN pathogenesis from neurons to the neuropathic pain triad, and suggest that optical microscopy can provide new opportunities for understanding the complex pathogenesis of TN at single-cell resolution, potentially contributing to the identification of more precise therapeutic targets and the development of more effective treatment modalities.
Collapse
Affiliation(s)
- Bin He
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenlong Wang
- School of Physics and Optoelectronics, State Key Laboratory of Luminescent Materials and Devices, Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices, Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Runsen Zhang
- School of Physics and Optoelectronics, State Key Laboratory of Luminescent Materials and Devices, Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices, Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Yue Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Wei
- School of Physics and Optoelectronics, State Key Laboratory of Luminescent Materials and Devices, Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices, Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Zhongmin Yang
- School of Physics and Optoelectronics, State Key Laboratory of Luminescent Materials and Devices, Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices, Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
- Research Institute of Future Technology, South China Normal University, Guangzhou, China
| | - Yang Cao
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
34
|
Sirucek L, Ganley RP, Zeilhofer HU, Schweinhardt P. Diffuse noxious inhibitory controls and conditioned pain modulation: a shared neurobiology within the descending pain inhibitory system? Pain 2023; 164:463-468. [PMID: 36017879 PMCID: PMC9916052 DOI: 10.1097/j.pain.0000000000002719] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Laura Sirucek
- Department of Chiropractic Medicine, Integrative Spinal Research Group, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Robert Philip Ganley
- Institute for Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Hanns Ulrich Zeilhofer
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Institute for Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Petra Schweinhardt
- Department of Chiropractic Medicine, Integrative Spinal Research Group, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Torres-Rodriguez JM, Wilson TD, Singh S, Chaudhry S, Adke AP, Becker JJ, Lin JL, Martinez Gonzalez S, Soler-Cedeño O, Carrasquillo Y. The parabrachial to central amygdala circuit is a key mediator of injury-induced pain sensitization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527340. [PMID: 36945586 PMCID: PMC10028796 DOI: 10.1101/2023.02.08.527340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The spino-ponto-amygdaloid pathway is a major ascending circuit relaying nociceptive information from the spinal cord to the brain. Potentiation of excitatory synaptic transmission in the parabrachial nucleus (PbN) to central amygdala (CeA) pathway has been reported in rodent models of persistent pain. At the behavioral level, the PbN→CeA pathway has been proposed to serve as a general alarm system to potential threats that modulates pain-related escape behaviors, threat memory, aversion, and affective-motivational (but not somatosensory) responses to painful stimuli. Increased sensitivity to previously innocuous somatosensory stimulation is a hallmark of chronic pain. Whether the PbN→CeA circuit contributes to heightened peripheral sensitivity following an injury, however, remains unknown. Here, we demonstrate that activation of CeA-projecting PbN neurons contributes to injury-induced behavioral hypersensitivity but not baseline nociception in male and female mice. Using optogenetic assisted circuit mapping, we confirmed a functional excitatory projection from PbN→CeA that is independent of the genetic or firing identity of CeA cells. We then showed that peripheral noxious stimulation increases the expression of the neuronal activity marker c-Fos in CeA-projecting PbN neurons and chemogenetic inactivation of these cells reduces behavioral hypersensitivity in models of neuropathic and inflammatory pain without affecting baseline nociception. Lastly, we show that chemogenetic activation of CeA-projecting PbN neurons is sufficient to induce bilateral hypersensitivity without injury. Together, our results demonstrate that the PbN→CeA pathway is a key modulator of pain-related behaviors that can amplify responses to somatosensory stimulation in pathological states without affecting nociception under normal physiological conditions. Significance Statement Early studies identified the spino-ponto-amygdaloid pathway as a major ascending circuit conveying nociceptive inputs from the spinal cord to the brain. The functional significance of this circuit to injury-induced hypersensitivity, however, remains unknown. Here, we addressed this gap in knowledge using viral-mediated anatomical tracers, ex-vivo electrophysiology and chemogenetic intersectional approaches in rodent models of persistent pain. We found that activation of this pathway contributes to injury-induced hypersensitivity, directly demonstrating a critical function of the PbN→CeA circuit in pain modulation.
Collapse
Affiliation(s)
| | - Torri D. Wilson
- National Center for Complementary and Integrative Health, Bethesda, MD, United States
| | - Sudhuman Singh
- National Center for Complementary and Integrative Health, Bethesda, MD, United States
| | - Sarah Chaudhry
- National Center for Complementary and Integrative Health, Bethesda, MD, United States
| | - Anisha P. Adke
- National Center for Complementary and Integrative Health, Bethesda, MD, United States
| | - Jordan J. Becker
- National Center for Complementary and Integrative Health, Bethesda, MD, United States
| | - Jenny L. Lin
- National Center for Complementary and Integrative Health, Bethesda, MD, United States
| | | | - Omar Soler-Cedeño
- National Center for Complementary and Integrative Health, Bethesda, MD, United States
| | - Yarimar Carrasquillo
- National Center for Complementary and Integrative Health, Bethesda, MD, United States
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
36
|
Zhang WW, Chen T, Li SY, Wang XY, Liu WB, Wang YQ, Mi WL, Mao-Ying QL, Wang YQ, Chu YX. Tachykinin receptor 3 in the lateral habenula alleviates pain and anxiety comorbidity in mice. Front Immunol 2023; 14:1049739. [PMID: 36756128 PMCID: PMC9900122 DOI: 10.3389/fimmu.2023.1049739] [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: 09/21/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
The coexistence of chronic pain and anxiety is a common clinical phenomenon. Here, the role of tachykinin receptor 3 (NK3R) in the lateral habenula (LHb) in trigeminal neuralgia and in pain-associated anxiety was systematically investigated. First, electrophysiological recording showed that bilateral LHb neurons are hyperactive in a mouse model of trigeminal neuralgia made by partial transection of the infraorbital nerve (pT-ION). Chemicogenetic activation of bilateral LHb glutamatergic neurons in naive mice induced orofacial allodynia and anxiety-like behaviors, and pharmacological activation of NK3R in the LHb attenuated allodynia and anxiety-like behaviors induced by pT-ION. Electrophysiological recording showed that pharmacological activation of NK3R suppressed the abnormal excitation of LHb neurons. In parallel, pharmacological inhibition of NK3R induced orofacial allodynia and anxiety-like behavior in naive mice. The electrophysiological recording showed that pharmacological inhibition of NK3R activates LHb neurons. Neurokinin B (NKB) is an endogenous high-affinity ligand of NK3R, which binds NK3R and activates it to perform physiological functions, and further neuron projection tracing showed that the front section of the periaqueductal gray (fPAG) projects NKB-positive nerve fibers to the LHb. Optogenetics combined with electrophysiology recordings characterize the functional connections in this fPAG NKB → LHb pathway. In addition, electrophysiological recording showed that NKB-positive neurons in the fPAG were more active than NKB-negative neurons in pT-ION mice. Finally, inhibition of NKB release from the fPAG reversed the analgesic and anxiolytic effects of LHb Tacr3 overexpression in pT-ION mice, indicating that fPAG NKB → LHb regulates orofacial allodynia and pain-induced anxious behaviors. These findings for NK3R suggest the cellular mechanism behind pT-ION in the LHb and suggest that the fPAG NKB → LHb circuit is involved in pain and anxiety comorbidity. This previously unrecognized pathway might provide a potential approach for relieving the pain and anxiety associated with trigeminal neuralgia by targeting NK3R.
Collapse
Affiliation(s)
- Wen-Wen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Teng Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Shi-Yi Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Xin-Yue Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Wen-Bo Liu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yu-Quan Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Wen-Li Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Qi-Liang Mao-Ying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China,*Correspondence: Yu-Xia Chu, ; Yan-Qing Wang,
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institutes of Integrative Medicine, Fudan University, Shanghai, China,Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Institute of Acupuncture Research, Fudan University, Shanghai, China,*Correspondence: Yu-Xia Chu, ; Yan-Qing Wang,
| |
Collapse
|
37
|
Hogri R, Baltov B, Drdla-Schutting R, Mussetto V, Raphael H, Trofimova L, Sandkühler J. Probing pain aversion in rats with the "Heat Escape Threshold" paradigm. Mol Pain 2023; 19:17448069231156657. [PMID: 36717755 PMCID: PMC9996743 DOI: 10.1177/17448069231156657] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The aversive aspect of pain constitutes a major burden faced by pain patients. This has been recognized by the pain research community, leading to the development of novel methods focusing on affective-motivational behaviour in pain model animals. The most common tests used to assess pain aversion in animals require cognitive processes, such as associative learning, complicating the interpretation of results. To overcome this issue, studies in recent years have utilized unconditioned escape as a measure of aversion. However, the vast majority of these studies quantify jumping - a common escape behaviour in mice, but not in adult rats, thus limiting its use. Here, we present the "Heat Escape Threshold" (HET) paradigm for assessing heat aversion in rats. We demonstrate that this method can robustly and reproducibly detect the localized effects of an inflammatory pain model (intraplantar carrageenan) in male and female Sprague-Dawley rats. In males, a temperature that evoked unconditioned escape following carrageenan treatment also induced real-time place avoidance (RTPA). Systemic morphine more potently alleviated carrageenan-induced heat aversion (as measured by the HET and RTPA methods), as compared to reflexive responses to heat (as measured by the Hargreaves test), supporting previous findings. Next, we examined how blocking of excitatory transmission to the lateral parabrachial nucleus (LPBN), a key node in the ascending pain system, affects pain behaviour. Using the HET and Hargreaves tests, we show that intra-LPBN application of glutamate antagonists reverses the effects of carrageenan on both affective and reflexive pain behaviour, respectively. Finally, we employed the HET paradigm in a generalized opioid-withdrawal pain model. Withdrawal from a brief systemic administration of remifentanil resulted in a long-lasting and robust increase in heat aversion, but no change in reflexive responses to heat. Taken together, these data demonstrate the utility of the HET paradigm as a novel tool in preclinical pain research.
Collapse
Affiliation(s)
- Roni Hogri
- Department of Neurophysiology,
Center for
Brain Research, Medical University of
Vienna, Vienna, Austria
| | - Bozhidar Baltov
- Department of Neurophysiology,
Center for
Brain Research, Medical University of
Vienna, Vienna, Austria
| | - Ruth Drdla-Schutting
- Department of Neurophysiology,
Center for
Brain Research, Medical University of
Vienna, Vienna, Austria
| | - Valeria Mussetto
- Department of Neurophysiology,
Center for
Brain Research, Medical University of
Vienna, Vienna, Austria
| | - Holzinger Raphael
- Department of Neurophysiology,
Center for
Brain Research, Medical University of
Vienna, Vienna, Austria
| | - Lidia Trofimova
- Department of Neurophysiology,
Center for
Brain Research, Medical University of
Vienna, Vienna, Austria
| | - Jürgen Sandkühler
- Department of Neurophysiology,
Center for
Brain Research, Medical University of
Vienna, Vienna, Austria
| |
Collapse
|
38
|
Katagiri A, Tsubota K, Mikuzuki L, Nakamura S, Toyofuku A, Kato T, Bereiter DA, Iwata K. Diquafosol sodium reduces neuronal activity in trigeminal subnucleus caudalis in a rat model of chronic dry eye disease. Neurosci Lett 2023; 792:136939. [PMID: 36341926 DOI: 10.1016/j.neulet.2022.136939] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Patients with persistent and severe dry eye disease (DED) have corneal hypersensitivity, resulting in ocular pain, and diquafosol sodium, a potent P2Y2 receptor agonist, is commonly used to improve the resultant tear film stability. This study determined the effects of diquafosol instillation on the suppression of trigeminal subnucleus caudalis (Vc) neuronal activity and ocular pain by enhancing tear film stability in the model for chronic DED. The effects of diquafosol on the ocular surface were assessed by the topical application for 28 days, starting from the 14th day since unilateral exorbital gland removal (chronic DED). Loss of tear volume secretion in chronic DED rats was significantly reversed by diquafosol instillation after 28 days, compared with saline treatment. The number of eyeblinks and pERK-IR neurons in the superficial laminae of Vc following hypertonic saline administration to the ocular surface was lower in diquafosol-treated chronic DED rats than in saline-treated rats. The neuronal activity evoked by hypertonic saline and mechanical stimulation along with the spontaneous neuronal activity in the superficial laminae of the Vc were suppressed in diquafosol-treated chronic DED rats. These findings suggest that ocular surface instillation of diquafosol for 28 days attenuates the neuronal hyperactivity in the Vc and the ocular pain that often occurs in chronic DED.
Collapse
Affiliation(s)
- Ayano Katagiri
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka 565-0871, Japan.
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Tsubota Laboratory, Inc., 34 Shinanomachi, Shinjuku-ku, Tokyo 160-0016 Japan
| | - Lou Mikuzuki
- Division of Geriatric Dentistry, Department of Critical Care Dentistry, Kanagawa Dental University, 82 Inaoka-cho Yokosuka-shi Kanagawa, 238-8580, Japan
| | - Shigeru Nakamura
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Akira Toyofuku
- Department of Psychosomatic Dentistry, Tokyo Medical and Dental University (TMDU) Graduate School, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Takafumi Kato
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka 565-0871, Japan
| | - David A Bereiter
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| |
Collapse
|
39
|
Jia T, Wang YD, Chen J, Zhang X, Cao JL, Xiao C, Zhou C. A nigro-subthalamo-parabrachial pathway modulates pain-like behaviors. Nat Commun 2022; 13:7756. [PMID: 36522327 PMCID: PMC9755217 DOI: 10.1038/s41467-022-35474-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The basal ganglia including the subthalamic nucleus (STN) and substantia nigra pars reticulata (SNr) are involved in pain-related responses, but how they regulate pain processing remains unknown. Here, we identify a pathway, consisting of GABAergic neurons in the SNr (SNrGABA) and glutamatergic neurons in the STN (STNGlu) and the lateral parabrachial nucleus (LPBGlu), that modulates acute and persistent pain states in both male and female mice. The activity of STN neurons was enhanced in acute and persistent pain states. This enhancement was accompanied by hypoactivity in SNrGABA neurons and strengthening of the STN-LPB glutamatergic projection. Reversing the dysfunction in the SNrGABA-STNGlu-LPBGlu pathway attenuated activity of LPBGlu neurons and mitigated pain-like behaviors. Therefore, the SNrGABA-STNGlu-LPBGlu pathway regulates pathological pain and is a potential target for pain management.
Collapse
Affiliation(s)
- Tao Jia
- grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China
| | - Ying-Di Wang
- grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China
| | - Jing Chen
- grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China
| | - Xue Zhang
- grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China
| | - Jun-Li Cao
- grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China ,grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China ,grid.417303.20000 0000 9927 0537NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China
| | - Cheng Xiao
- grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China ,grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China ,grid.417303.20000 0000 9927 0537NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China
| | - Chunyi Zhou
- grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory in Anesthesiology, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China ,grid.417303.20000 0000 9927 0537Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China ,grid.417303.20000 0000 9927 0537NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, 221004 Xuzhou, Jiangsu China
| |
Collapse
|
40
|
Pauli JL, Chen JY, Basiri ML, Park S, Carter ME, Sanz E, McKnight GS, Stuber GD, Palmiter RD. Molecular and anatomical characterization of parabrachial neurons and their axonal projections. eLife 2022; 11:e81868. [PMID: 36317965 PMCID: PMC9668336 DOI: 10.7554/elife.81868] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/31/2022] [Indexed: 11/07/2022] Open
Abstract
The parabrachial nucleus (PBN) is a major hub that receives sensory information from both internal and external environments. Specific populations of PBN neurons are involved in behaviors including food and water intake, nociceptive responses, breathing regulation, as well as learning and responding appropriately to threatening stimuli. However, it is unclear how many PBN neuron populations exist and how different behaviors may be encoded by unique signaling molecules or receptors. Here we provide a repository of data on the molecular identity, spatial location, and projection patterns of dozens of PBN neuron subclusters. Using single-cell RNA sequencing, we identified 21 subclusters of neurons in the PBN and neighboring regions. Multiplexed in situ hybridization showed many of these subclusters are enriched within specific PBN subregions with scattered cells in several other regions. We also provide detailed visualization of the axonal projections from 21 Cre-driver lines of mice. These results are all publicly available for download and provide a foundation for further interrogation of PBN functions and connections.
Collapse
Affiliation(s)
- Jordan L Pauli
- Department of Biochemistry, Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Jane Y Chen
- Department of Biochemistry, Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Marcus L Basiri
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, University of WashingtonSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Sekun Park
- Department of Biochemistry, Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Matthew E Carter
- Department of Biochemistry, Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Elisenda Sanz
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - G Stanley McKnight
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, University of WashingtonSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Richard D Palmiter
- Department of Biochemistry, Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| |
Collapse
|
41
|
Huang X, Li Z, Ma J, Huang D, Yan X, Zhou H. A novel implantable device for sensory and affective assessment of orofacial pain in rats. Front Vet Sci 2022; 9:1028147. [DOI: 10.3389/fvets.2022.1028147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background and objectiveOrofacial pain, in particular, chronic orofacial pain remains a great challenge in clinical practice. To better understand the underlying mechanism of disease, it is essential to apply a feasible and stable preclinical measurement of facial pain. Here, we introduced a novel electrical noxious stimulator in freely behavioral rodents and examined its validation in both naïve and chronic orofacial pain animals.MethodsOne subcutaneous device of electrical stimulator was implanted in the facial region for delivery of the nociceptive input. The sensory component of orofacial pain was assessed by response scoring tool, and conditioned place aversion (CPA) paradigm for pain affect respectively. To confirm its usage in chronic pain state, the chronic constriction injury of the infraorbital nerve (ION-CCI) model was then applied.ResultsWe found that responsive scores increased with stimulation intensity, and acted in a dosage-dependent manner, which can be attenuated by the administration of morphine intraperitoneally. Naïve rats displayed significant aversive reaction to the noxious electrical stimulation (25V) in the CPA testing. In addition, an obvious sensory hypersensitivity to electrical stimulation was confirmed by the increased response scores in ION-CCI rats. Furthermore, ION-CCI animal showed significant avoidance to electrical stimulation at relatively low intensity (10V), which was innoxious to naïve rats.ConclusionOur findings may provide an alternative pre-clinical measurement of orofacial pain, to quantitively assess both sensory and affective component of orofacial pain.
Collapse
|
42
|
Lee JJ, Lee S, Lee DH, Woo CW. Functional brain reconfiguration during sustained pain. eLife 2022; 11:e74463. [PMID: 36173388 PMCID: PMC9522250 DOI: 10.7554/elife.74463] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Pain is constructed through complex interactions among multiple brain systems, but it remains unclear how functional brain networks are reconfigured over time while experiencing pain. Here, we investigated the time-varying changes in the functional brain networks during 20 min capsaicin-induced sustained orofacial pain. In the early stage, the orofacial areas of the primary somatomotor cortex were separated from other areas of the somatosensory cortex and integrated with subcortical and frontoparietal regions, constituting an extended brain network of sustained pain. As pain decreased over time, the subcortical and frontoparietal regions were separated from this brain network and connected to multiple cerebellar regions. Machine-learning models based on these network features showed significant predictions of changes in pain experience across two independent datasets (n = 48 and 74). This study provides new insights into how multiple brain systems dynamically interact to construct and modulate pain experience, advancing our mechanistic understanding of sustained pain.
Collapse
Affiliation(s)
- Jae-Joong Lee
- Center for Neuroscience Imaging Research, Institute for Basic ScienceSuwonRepublic of Korea
- Department of Biomedical Engineering, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Sungwoo Lee
- Center for Neuroscience Imaging Research, Institute for Basic ScienceSuwonRepublic of Korea
- Department of Biomedical Engineering, Sungkyunkwan UniversitySuwonRepublic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Dong Hee Lee
- Center for Neuroscience Imaging Research, Institute for Basic ScienceSuwonRepublic of Korea
- Department of Biomedical Engineering, Sungkyunkwan UniversitySuwonRepublic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan UniversitySuwonRepublic of Korea
| | - Choong-Wan Woo
- Center for Neuroscience Imaging Research, Institute for Basic ScienceSuwonRepublic of Korea
- Department of Biomedical Engineering, Sungkyunkwan UniversitySuwonRepublic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan UniversitySuwonRepublic of Korea
| |
Collapse
|
43
|
Takatoh J, Prevosto V, Thompson PM, Lu J, Chung L, Harrahill A, Li S, Zhao S, He Z, Golomb D, Kleinfeld D, Wang F. The whisking oscillator circuit. Nature 2022; 609:560-568. [PMID: 36045290 PMCID: PMC10038238 DOI: 10.1038/s41586-022-05144-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/25/2022] [Indexed: 11/09/2022]
Abstract
Central oscillators are primordial neural circuits that generate and control rhythmic movements1,2. Mechanistic understanding of these circuits requires genetic identification of the oscillator neurons and their synaptic connections to enable targeted electrophysiological recording and causal manipulation during behaviours. However, such targeting remains a challenge with mammalian systems. Here we delimit the oscillator circuit that drives rhythmic whisking-a motor action that is central to foraging and active sensing in rodents3,4. We found that the whisking oscillator consists of parvalbumin-expressing inhibitory neurons located in the vibrissa intermediate reticular nucleus (vIRtPV) in the brainstem. vIRtPV neurons receive descending excitatory inputs and form recurrent inhibitory connections among themselves. Silencing vIRtPV neurons eliminated rhythmic whisking and resulted in sustained vibrissae protraction. In vivo recording of opto-tagged vIRtPV neurons in awake mice showed that these cells spike tonically when animals are at rest, and transition to rhythmic bursting at the onset of whisking, suggesting that rhythm generation is probably the result of network dynamics, as opposed to intrinsic cellular properties. Notably, ablating inhibitory synaptic inputs to vIRtPV neurons quenched their rhythmic bursting, impaired the tonic-to-bursting transition and abolished regular whisking. Thus, the whisking oscillator is an all-inhibitory network and recurrent synaptic inhibition has a key role in its rhythmogenesis.
Collapse
Affiliation(s)
- Jun Takatoh
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| | - Vincent Prevosto
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - P M Thompson
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jinghao Lu
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Leeyup Chung
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Andrew Harrahill
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shun Li
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Shengli Zhao
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - David Golomb
- Department of Physiology and Cell Biology, Ben Gurion University, Be'er Sheva, Israel
- Department of Physics, Ben Gurion University, Be'er Sheva, Israel
- Zlotowski Center for Neuroscience, Ben Gurion University, Be'er Sheva, Israel
| | - David Kleinfeld
- Department of Physics, University of California at San Diego, La Jolla, CA, USA
- Department of Neurobiology, University of California at San Diego, La Jolla, CA, USA
| | - Fan Wang
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
44
|
Bagues A, Girón R, Abalo R, Goicoechea C, Martín-Fontelles MI, Sánchez-Robles EM. SHORT-TERM STRESS SIGNIFICANTLY DECREASES MORPHINE ANALGESIA IN TRIGEMINAL BUT NOT IN SPINAL INNERVATED AREAS IN RATS. Behav Brain Res 2022; 435:114046. [PMID: 35933048 DOI: 10.1016/j.bbr.2022.114046] [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: 06/10/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/15/2022]
Abstract
Plenty information exists regarding the effects of chronic stress, although few data exist on the effects of short-lasting stressors, which would mimic daily challenges. Differences in craniofacial and spinal nociception have been observed, thus those observations obtained in spinally innervated areas cannot be directly applied to the orofacial region. Although, opioids are considered amongst the most effective analgesics, their use is sometimes hampered by the constipation they induce. Thus, our aims were to study if a short-lasting stressor, forced swim stress (FSS), modifies nociception, morphine antinociception and constipation in rats. Animals were submitted to 10-20min of FSS for three days, nociception and gastrointestinal transit were studied 24h after the last swimming session. Nociception and morphine (0.6-5mg/kg) antinociception were evaluated in the formalin and hypertonic saline tests in the orofacial area and limbs. Morphine-induced modifications in the GI transit were studied through radiographic techniques. Naloxone was administered, before each swimming session, to analyse the involvement of the endogenous opioid system on the effect of stress. Overall, stress did not alter nociception, although interestingly it reduced the effect of morphine in the orofacial tests and in the inflammatory phase of the formalin tests. Naloxone antagonized the effect of stress and normalized the effect of morphine. Stress did not modify the constipation induced by morphine. Opioid treatment may be less effective under a stressful situation, whilst adverse effects, such as constipation, are maintained. The prevention of stress may improve the level of opioid analgesia. Keywords.
Collapse
Affiliation(s)
- Ana Bagues
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada I+D+i al Instituto de Química Médica (CSIC), Alcorcón, Spain; High Performance Research Group in Experimental Pharmacology (PHARMAKOM).
| | - Rocío Girón
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada I+D+i al Instituto de Química Médica (CSIC), Alcorcón, Spain; High Performance Research Group in Experimental Pharmacology (PHARMAKOM).
| | - Raquel Abalo
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada I+D+i al Instituto de Química Médica (CSIC), Alcorcón, Spain; High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC); Working Group of Basic Sciences in Pain and Analgesia of the Sociedad Española del Dolor.
| | - Carlos Goicoechea
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada I+D+i al Instituto de Química Médica (CSIC), Alcorcón, Spain; High Performance Research Group in Experimental Pharmacology (PHARMAKOM); Working Group of Basic Sciences in Pain and Analgesia of the Sociedad Española del Dolor.
| | - Ma Isabel Martín-Fontelles
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada I+D+i al Instituto de Química Médica (CSIC), Alcorcón, Spain; High Performance Research Group in Experimental Pharmacology (PHARMAKOM); Working Group of Basic Sciences in Pain and Analgesia of the Sociedad Española del Dolor.
| | - Eva Ma Sánchez-Robles
- Área de Farmacología, Nutrición y Bromatología, Dpto. C.C. Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Unidad Asociada I+D+i al Instituto de Química Médica (CSIC), Alcorcón, Spain; High Performance Research Group in Experimental Pharmacology (PHARMAKOM).
| |
Collapse
|
45
|
Kramer PR, Umorin M, Hornung R, Benson MD, Kinchington PR. Sex Differences in the Role of Neurexin 3α in Zoster Associated Pain. Front Integr Neurosci 2022; 16:915797. [PMID: 35875508 PMCID: PMC9302461 DOI: 10.3389/fnint.2022.915797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Varicella zoster virus (VZV) induces orofacial pain and female rats show greater pain than male rats. During the proestrus phase of the estrous cycle the VZV induce pain response is attenuated in female rats. A screen of gene expression changes in diestrus and proestrus female rats indicated neurexin 3α (Nrxn3α) was elevated in the central amygdala of proestrus rats vs. diestrus rats. GABAergic neurons descend from the central amygdala to the lateral parabrachial region and Nrxn3α is important for presynaptic γ-Aminobutyric acid (GABA) release. Thus, we hypothesized that the reduced orofacial pain in male rats and proestrus female rats is the result of increased Nrxn3α within the central amygdala that increases GABA release from axon terminals within the parabrachial and inhibits ascending pain signals. To test this hypothesis Nrxn3 α expression was knocked-down by infusing shRNA constructs in the central amygdala. Then GABA release in the parabrachial was quantitated concomitant with measuring the pain response. Results revealed that knockdown of Nrxn3α expression significantly increases the pain response in both male rats and proestrus female rats vs. diestrus rats. GABA release was significantly reduced in the parabrachial of male and proestrus female rats after Nrxn3α knockdown. Neuronal activity of excitatory neurons was significantly inhibited in the parabrachial after Nrxn3α knockdown. These results are consistent with the idea that Nrxn3 within the central amygdala controls VZV associated pain by regulating GABA release in the lateral parabrachial that then modulates ascending orofacial pain signals.
Collapse
Affiliation(s)
- Phillip R. Kramer
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, United States
| | - Mikhail Umorin
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, United States
| | - Rebecca Hornung
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, United States
| | - M. Douglas Benson
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, United States
| | - Paul R. Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
46
|
Hogri R, Teuchmann HL, Heinke B, Holzinger R, Trofimova L, Sandkühler J. GABAergic CaMKIIα+ Amygdala Output Attenuates Pain and Modulates Emotional-Motivational Behavior via Parabrachial Inhibition. J Neurosci 2022; 42:5373-5388. [PMID: 35667849 PMCID: PMC9270917 DOI: 10.1523/jneurosci.2067-21.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 04/14/2022] [Accepted: 04/23/2022] [Indexed: 01/09/2023] Open
Abstract
Pain and emotion are strongly regulated by neurons in the central nucleus of the amygdala (CeA), a major output of the limbic system; yet, the neuronal signaling pathways underlying this modulation are incompletely understood. Here, we characterized a subpopulation of CeA neurons that express the CaMKIIα gene (CeACAM neurons) and project to the lateral parabrachial nucleus (LPBN), a brainstem region known for its critical role in distributing nociceptive and other aversive signals throughout the brain. In male Sprague Dawley rats, we show that CeACAM-LPBN neurons are GABAergic and mostly express somatostatin. In anaesthetized rats, optogenetic stimulation of CeACAM-LPBN projections inhibited responses of LPBN neurons evoked by electrical activation of Aδ- and C-fiber primary afferents; this inhibition could be blocked by intra-LPBN application of the GABAA receptor antagonist bicuculline. CeACAM-LPBN stimulation also dampened LPBN responses to noxious mechanical, thermal, and chemical stimuli. In behaving rats, optogenetic stimulation of CeACAM-LPBN projections attenuated nocifensive responses to mechanical pressure and radiant heat, disrupted the ability of a noxious shock to drive aversive learning, reduced the defensive behaviors of thigmotaxis and freezing, induced place preference, and promoted food consumption in sated rats. Thus, we suggest that CeACAM-LPBN projections mediate a form of analgesia that is accompanied by a shift toward the positive-appetitive pole of the emotional-motivational continuum. Since the affective state of pain patients strongly influences their prognosis, we envision that recruitment of this pathway in a clinical setting could potentially promote pain resilience and recovery.SIGNIFICANCE STATEMENT Pain and emotion interact on multiple levels of the nervous system. Both positive and negative emotion may have analgesic effects. However, while the neuronal mechanisms underlying "stress-induced analgesia" have been the focus of many studies, the neuronal substrates underlying analgesia accompanied by appetitive emotional-motivational states have received far less attention. The current study focuses on a subpopulation of amygdala neurons that form inhibitory synapses within the brainstem lateral parabrachial nucleus (LPBN). We show that activation of these amygdalo-parabrachial projections inhibits pain processing, while also reducing behaviors related to negative affect and enhancing behaviors related to positive affect. We propose that recruitment of this pathway would benefit pain patients, many of whom suffer from psychological comorbidities such as anxiety and depression.
Collapse
Affiliation(s)
- Roni Hogri
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna 1190, Austria
| | - Hannah Luise Teuchmann
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna 1190, Austria
| | - Bernhard Heinke
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna 1190, Austria
| | - Raphael Holzinger
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna 1190, Austria
| | - Lidia Trofimova
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna 1190, Austria
| | - Jürgen Sandkühler
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna 1190, Austria
| |
Collapse
|
47
|
Neurexin 3α in the central amygdala has a role orofacial varicella zoster pain. Neuroscience 2022; 496:16-26. [PMID: 35679996 PMCID: PMC9329223 DOI: 10.1016/j.neuroscience.2022.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022]
Abstract
Varicella zoster virus (VZV) is responsible for chronic pain. VZV injection has similarities to herpes zoster (HZ) "shingles" pain in humans. In this study orofacial pain was induced by injecting male rats with the human VZV. The amygdala and parabrachial have been implicated to control affective/motivational orofacial pain. Recently our lab reported neurexin 3α (Nrxn3α) is expressed in the central amygdala and parabrachial. GABAergic neurons descend from the central amygdala to the lateral parabrachial region and Nrxn3α is important for presynaptic (γ-Aminobutyric acid) GABA release. Thus, we hypothesized that lateral parabrachial neuronal activity and orofacial pain are controlled by Nrxn3α within the central amygdala. To test the hypothesis Nrxn3α expression was knocked down (i.e., using short hairpin RNA or shRNA) in the central amygdala and GABA release and neuronal activity were quantitated in the parabrachial concomitant with measurement of the VZV induced pain response. Results revealed that attenuating Nrxn3 expression within the amygdala reduces GABA release in the parabrachial and increases neuronal activity within the lateral parabrachial region. Attenuating Nrxn3 expression also increases VZV associated orofacial pain. Activating GABAergic neurons within the central amygdala with opsins increase GABA release in the parabrachial and reduced the pain response after Nrxn3 shRNA treatment. These results are consistent with the idea that Nrxn3 within the central amygdala controls VZV associated pain by regulating GABA release in the lateral parabrachial that then controls the activity of ascending pain neurons.
Collapse
|
48
|
Barykina NV, Karasev MM, Verkhusha VV, Shcherbakova DM. Technologies for large-scale mapping of functional neural circuits active during a user-defined time window. Prog Neurobiol 2022; 216:102290. [PMID: 35654210 DOI: 10.1016/j.pneurobio.2022.102290] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/25/2022]
Abstract
The mapping of neural circuits activated during behavior down to individual neurons is crucial for decoding how the brain processes information. Technologies allowing activity-dependent labeling of neurons during user-defined restricted time windows are rapidly developing. Precise marking of the time window with light, in addition to chemicals, is now possible. In these technologies, genetically encoded molecules integrate molecular events resulting from neuronal activity with light/drug-dependent events. The outputs are either changes in fluorescence or activation of gene expression. Molecular reporters allow labeling of activated neurons for visualization and cell-type identification. The transcriptional readout also allows further control of activated neuronal populations using optogenetic tools as reporters. Here we review the design of these technologies and discuss their demonstrated applications to reveal previously unknown connections in the mammalian brain. We also consider the strengths and weaknesses of the current approaches and provide a perspective for the future.
Collapse
Affiliation(s)
- Natalia V Barykina
- P.K. Anokhin Institute of Normal Physiology, Moscow 125315, Russia; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Maksim M Karasev
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Vladislav V Verkhusha
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Daria M Shcherbakova
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
49
|
Wang LB, Su XJ, Wu QF, Xu X, Wang XY, Chen M, Ye JR, Maimaitiabula A, Liu XQ, Sun W, Zhang Y. Parallel Spinal Pathways for Transmitting Reflexive and Affective Dimensions of Nocifensive Behaviors Evoked by Selective Activation of the Mas-Related G Protein-Coupled Receptor D-Positive and Transient Receptor Potential Vanilloid 1-Positive Subsets of Nociceptors. Front Cell Neurosci 2022; 16:910670. [PMID: 35693883 PMCID: PMC9175034 DOI: 10.3389/fncel.2022.910670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022] Open
Abstract
The high incidence of treatment-resistant pain calls for the urgent preclinical translation of new analgesics. Understanding the behavioral readout of pain in animals is crucial for efficacy evaluation when developing novel analgesics. Mas-related G protein-coupled receptor D-positive (Mrgprd+) and transient receptor potential vanilloid 1-positive (TRPV1+) sensory neurons are two major non-overlapping subpopulations of C-fiber nociceptors. Their activation has been reported to provoke diverse nocifensive behaviors. However, what kind of behavior reliably represents subjectively conscious pain perception needs to be revisited. Here, we generated transgenic mice in which Mrgprd+ or TRPV1+ sensory neurons specifically express channelrhodopsin-2 (ChR2). Under physiological conditions, optogenetic activation of hindpaw Mrgprd+ afferents evoked reflexive behaviors (lifting, etc.), but failed to produce aversion. In contrast, TRPV1+ afferents activation evoked marked reflexive behaviors and affective responses (licking, etc.), as well as robust aversion. Under neuropathic pain conditions induced by spared nerve injury (SNI), affective behaviors and avoidance can be elicited by Mrgprd+ afferents excitation. Mechanistically, spinal cord-lateral parabrachial nucleus (lPBN) projecting neurons in superficial layers (lamina I–IIo) were activated by TRPV1+ nociceptors in naïve conditions or by Mrgprd+ nociceptors after SNI, whereas only deep spinal cord neurons were activated by Mrgprd+ nociceptors in naïve conditions. Moreover, the excitatory inputs from Mrgprd+ afferents to neurons within inner lamina II (IIi) are partially gated under normal conditions. Altogether, we conclude that optogenetic activation of the adult Mrgprd+ nociceptors drives non-pain-like reflexive behaviors via the deep spinal cord pathway under physiological conditions and drives pain-like affective behaviors via superficial spinal cord pathway under pathological conditions. The distinct spinal pathway transmitting different forms of nocifensive behaviors provides different therapeutic targets. Moreover, this study appeals to the rational evaluation of preclinical analgesic efficacy by using comprehensive and suitable behavioral assays, as well as by assessing neural activity in the two distinct pathways.
Collapse
Affiliation(s)
- Liang-Biao Wang
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiao-Jing Su
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiao-Feng Wu
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiang Xu
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xin-Yue Wang
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mo Chen
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jia-Reng Ye
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Abasi Maimaitiabula
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiao-Qing Liu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wen Sun
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Wen Sun,
| | - Yan Zhang
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Yan Zhang,
| |
Collapse
|
50
|
Liedtke W. Long March Toward Safe and Effective Analgesia by Enhancing Gene Expression of Kcc2: First Steps Taken. Front Mol Neurosci 2022; 15:865600. [PMID: 35645734 PMCID: PMC9137411 DOI: 10.3389/fnmol.2022.865600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 11/15/2022] Open
Abstract
Low intraneuronal chloride in spinal cord dorsal horn pain relay neurons is critical for physiologic transmission of primary pain afferents because low intraneuronal chloride dictates whether GABA-ergic and glycin-ergic neurotransmission is inhibitory. If the neuronal chloride elevates to pathologic levels, then spinal cord primary pain relay becomes leaky and exhibits the behavioral hallmarks of pathologic pain, namely hypersensitivity and allodynia. Low chloride in spinal cord dorsal horn neurons is maintained by proper gene expression of Kcc2 and sustained physiologic function of the KCC2 chloride extruding electroneutral transporter. Peripheral nerve injury and other forms of neural injury evoke greatly diminished Kcc2 gene expression and subsequent corruption of inhibitory neurotransmission in the spinal cord dorsal horn, thus causing derailment of the gate function for pain. Here I review key discoveries that have helped us understand these fundamentals, and focus on recent insights relating to the discovery of Kcc2 gene expression enhancing compounds via compound screens in neurons. One such study characterized the kinase inhibitor, kenpaullone, more in-depth, revealing its function as a robust and long-lasting analgesic in preclinical models of nerve injury and cancer bone pain, also elucidating its mechanism of action via GSK3β inhibition, diminishing delta-catenin phosphorylation, and facilitating its nuclear transfer and subsequent enhancement of Kcc2 gene expression by de-repressing Kaiso epigenetic transcriptional regulator. Future directions re Kcc2 gene expression enhancement are discussed, namely combination with other analgesics and analgesic methods, such as spinal cord stimulation and electroacupuncture, gene therapy, and leveraging Kcc2 gene expression-enhancing nanomaterials.
Collapse
|