1
|
Kawabata R, Fujita A, Oke Y, Yao I, Koga K. The elevated open platform stress suppresses excitatory synaptic transmission in the layer V anterior cingulate cortex. Neuroscience 2025; 564:243-259. [PMID: 39369946 DOI: 10.1016/j.neuroscience.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
There are various forms of stress including; physical, psychological and social stress. Exposure to physical stress can lead to physical sensations (e.g. hyperalgesia) and negative emotions including anxiety and depression in animals and humans. Recently, our studies in mice have shown that acute physical stress induced by the elevated open platform (EOP) can provoke long-lasting mechanical hypersensitivity. This effect appears to be related to activity in the anterior cingulate cortex (ACC) at the synaptic level. Indeed, EOP exposure induces synaptic plasticity in layer II/III pyramidal neurons from the ACC. However, it is still unclear whether or not EOP exposure alters intrinsic properties and synaptic transmission in layer V pyramidal neurons. This is essential because these neurons are known to be a primary output to subcortical structures which may ultimately impact the behavioral stress response. Here, we studied both intrinsic properties and excitatory/inhibitory synaptic transmission by using whole-cell patch-clamp method in brain slice preparations. The EOP exposure did not change intrinsic properties including resting membrane potentials and action potentials. In contrast, EOP exposure suppressed the frequency of miniature and spontaneous excitatory synaptic transmission with an alteration of kinetics of AMPA/GluK receptors. EOP exposure also reduced evoked synaptic transmission induced by electrical stimulation. Furthermore, we investigated projection-selective responses of the mediodorsal thalamus to the layer V ACC neurons. EOP exposure produced short-term depression in excitatory synaptic transmission on thalamo-ACC projections. These results suggest that the EOP stress provokes abnormal excitatory synaptic transmission in layer V pyramidal neurons of the ACC.
Collapse
Affiliation(s)
- Ryo Kawabata
- Biomedical Chemistry Major, Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan; Department of Neurophysiology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Ayumi Fujita
- Department of Neurophysiology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yoshihiko Oke
- Department of Physiology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Ikuko Yao
- Biomedical Chemistry Major, Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Kohei Koga
- Department of Neurophysiology, Faculty of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
2
|
Song Z, Sun Y, Liu P, Ruan H, He Y, Yin J, Xiao C, Ma J, Yu Y, Wang S, Gong Y, Lin ZW, Zhang Z, Chang C, Yang M. Terahertz Wave Alleviates Comorbidity Anxiety in Pain by Reducing the Binding Capacity of Nanostructured Glutamate Molecules to GluA2. RESEARCH (WASHINGTON, D.C.) 2024; 7:0535. [PMID: 39664293 PMCID: PMC11633831 DOI: 10.34133/research.0535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024]
Abstract
Comorbid anxiety in chronic pain is clinically common, with a comorbidity rate of over 50%. The main treatments are based on pharmacological, interventional, and implantable approaches, which have limited efficacy and carry a risk of side effects. Here, we report a terahertz (THz, 1012 Hz) wave stimulation (THS) technique, which exerts nonthermal, long-term modulatory effects on neuronal activity by reducing the binding between nano-sized glutamate molecules and GluA2, leading to the relief of pain and comorbid anxiety-like behaviors in mice. In mice with co-occurring anxiety and chronic pain induced by complete Freund's adjuvant (CFA) injection, hyperactivity was observed in glutamatergic neurons in the anterior cingulate cortex (ACCGlu). Using whole-cell recording in ACC slices, we demonstrated that THS (34 THz) effectively inhibited the excitability of ACCGlu. Moreover, molecular dynamics simulations showed that THS reduced the number of hydrogen bonds bound between glutamate molecules and GluA2. Furthermore, THS target to the ACC in CFA-treatment mice suppressed ACCGlu hyperactivity and, as a result, alleviated pain and anxiety-like behaviors. Consistently, inhibition of ACCGlu hyperactivity by chemogenetics mimics THS-induced antinociceptive and antianxiety behavior. Together, our study provides evidence for THS as an intervention technique for modulating neuronal activity and a viable clinical treatment strategy for pain and comorbid anxiety.
Collapse
Affiliation(s)
- Zihua Song
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Yuankun Sun
- School of Electronic Science and Engineering,
University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Pan Liu
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Hao Ruan
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Yuanyuan He
- School of Safety Engineering,
North China Institute of Science and Technology, Hebei 065201, China
| | - Junkai Yin
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Chun Xiao
- School of Life Sciences,
Tsinghua University, Beijing 100081, China
| | - Jing Ma
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Yun Yu
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
- School of Life Science and Technology and Frontier Institute of Science and Technology,
Xi’an Jiaotong University, Xi’an 710049, China
| | - Shaomeng Wang
- School of Electronic Science and Engineering,
University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yubin Gong
- School of Electronic Science and Engineering,
University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Z. W. Lin
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Zhi Zhang
- Division of Life Sciences and Medicine, Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale,
University of Science and Technology of China, Hefei 230026, China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
- School of Physics,
Peking University, Beijing 100081, China
| | - Maojun Yang
- School of Life Sciences,
Tsinghua University, Beijing 100081, China
| |
Collapse
|
3
|
Andrade-González RD, Montes-Ángeles CD, Perrusquia-Hernández E, González-Alva P, Hernández-Campos ME, Pérez-Martínez IO. Role of Anterior Cingulate Cortex in the exacerbated facial response to mechanical stimuli as a sign of early orofacial neuropathic pain. THE JOURNAL OF PAIN 2024; 27:104756. [PMID: 39662862 DOI: 10.1016/j.jpain.2024.104756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
The study of orofacial neuropathic pain necessitates the use of innovative assessment techniques, such as the facial expression of pain, which mirrors the internal state of the animals and could be utilized to identify the neural correlations involved. The Anterior Cingulate Cortex (ACC) is a crucial center in the processing of sensory and affective components of acute and neuropathic pain. However, its role in the facial response to pain remains a mystery. In this project, we set out to determine the changes in the facial response of early trigeminal neuropathic pain and the contribution of ACC in this process. We evaluated the facial response to mechanical stimulation using a machine-vision analysis in a head-fixed system before and after mental nerve compression in C57BL/6 mice. The role of ACC in the facial response was characterized via acute electrophysiological recording, and both glutamatergic ACC neural-ablation and optogenetic inhibition in a cre-dependent manner. Our results indicate that trigeminal nerve injury leads to an exacerbation of the intensity of the pain-like facial response to aversive stimuli in an early period. ACC neuronal activity is modulated by mechanical stimulation and during the dynamics of the facial response in different proportions before and after the lesion onset. The neuropathic pain-induced changes in the intensity of the facial response are nullified by the ablation or optogenetic inhibition of ACC glutamatergic neurons. Our study underscores the significant role of ACC in the development of signs of orofacial neuropathic pain, such as exacerbated facial response to mechanical stimuli. PERSPECTIVE: This article presents evidence on the sensory coding of mechanical stimulation in a neuropathic pain model in the Anterior Cingulate Cortex, using facial expression as a manifestation of the internal painful state. This evaluation provides a novel approach to evaluating the well-being of animals with neuropathic pain.
Collapse
Affiliation(s)
- Rey David Andrade-González
- Laboratory of Neurobiology of Orofacial Sensations and Movements. FES Iztacala, National Autonomous University of Mexico, Mexico; Postgraduate Studies and Research Section, School of Higher Education in Medicine. National Polytechnic Institute, México
| | - Claudia Daniela Montes-Ángeles
- Laboratory of Neurobiology of Orofacial Sensations and Movements. FES Iztacala, National Autonomous University of Mexico, Mexico
| | - Elías Perrusquia-Hernández
- Laboratory of Neurobiology of Orofacial Sensations and Movements. FES Iztacala, National Autonomous University of Mexico, Mexico; Postgraduate Studies and Research Section, School of Higher Education in Medicine. National Polytechnic Institute, México
| | - Patricia González-Alva
- Laboratory of Tissue Bioengineering, College of Dentistry, National Autonomous University of Mexico, Mexico
| | - María Elena Hernández-Campos
- Postgraduate Studies and Research Section, School of Higher Education in Medicine. National Polytechnic Institute, México
| | - Isaac Obed Pérez-Martínez
- Laboratory of Neurobiology of Orofacial Sensations and Movements. FES Iztacala, National Autonomous University of Mexico, Mexico.
| |
Collapse
|
4
|
Hor CC, Duan B. Lateral parabrachial nucleus: the commander-in-chief for nocifensive behavior expression in cold allodynia. Pain 2024:00006396-990000000-00777. [PMID: 39715171 DOI: 10.1097/j.pain.0000000000003469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 12/25/2024]
Affiliation(s)
- Chia Chun Hor
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.
| | | |
Collapse
|
5
|
Mindaye SA, Chen WH, Lin SC, Chen YC, Abdelaziz MA, Tzeng YS, Shih ACC, Chen SY, Yang SB, Chen CC. Separate anterior paraventricular thalamus projections differentially regulate sensory and affective aspects of pain. Cell Rep 2024; 43:114946. [PMID: 39499617 DOI: 10.1016/j.celrep.2024.114946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
The experience of pain is complex, involving both sensory and affective components, yet the underlying neural mechanisms remain elusive. Here, we show that formalin-induced pain behaviors coincide with increased responses in glutamatergic neurons within the anterior paraventricular nucleus of the thalamus (PVA). Furthermore, we describe non-overlapping subpopulations of PVAVgluT2 neurons involved in sensory and affective pain processing, whose activity varies across different pain states. Activating PVA glutamatergic neurons is sufficient to induce mechanical hypersensitivity and aversion behaviors, whereas suppression ameliorates formalin-induced pain. Furthermore, we identify the segregation of PVAVgluT2 projections to the bed nucleus of the stria terminalis (BNST) and nucleus accumbens (NAc), each influencing specific aspects of pain-like behavior. This finding provides an important insight into the mechanism of distinct components of pain, highlighting the pivotal role of PVA in mediating different aspects of pain-like behavior with distinct circuits.
Collapse
Affiliation(s)
- Selomon Assefa Mindaye
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wei-Hsin Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Shih-Che Lin
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Taiwan University and Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yong-Cyuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Mohamed Abbas Abdelaziz
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Shiuan Tzeng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - Shih-Yu Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shi-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
6
|
Motzkin JC, Basbaum AI, Crowther AJ. Neuroanatomy of the nociceptive system: From nociceptors to brain networks. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:1-39. [PMID: 39580210 DOI: 10.1016/bs.irn.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
This chapter reviews the neuroanatomy of the nociceptive system and its functional organization. We describe three main compartments of the nervous system that underlie normal nociception and the resulting pain percept: Peripheral, Spinal Cord, and Brain. We focus on how ascending nociceptive processing streams traverse these anatomical compartments, culminating in the multidimensional experience of pain. We also describe neuropathic pain conditions, in which nociceptive processing is abnormal, not only because of the primary effects of a lesion or disease affecting peripheral nerves or the central nervous system (CNS), but also due to secondary effects on ascending pathways and brain networks. We discuss how the anatomical components (circuits/networks) reorganize under various etiologies of neuropathic pain and how these changes can give rise to pathological pain states.
Collapse
Affiliation(s)
- Julian C Motzkin
- Department of Neurology and Department Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States.
| | - Allan I Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Andrew J Crowther
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
7
|
Franciosa F, Acuña MA, Nevian NE, Nevian T. A cellular mechanism contributing to pain-induced analgesia. Pain 2024; 165:2517-2529. [PMID: 38968393 PMCID: PMC11474934 DOI: 10.1097/j.pain.0000000000003315] [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: 09/29/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 07/07/2024]
Abstract
ABSTRACT The anterior cingulate cortex (ACC) plays a crucial role in the perception of pain. It is consistently activated by noxious stimuli and its hyperactivity in chronic pain indicates plasticity in the local neuronal network. However, the way persistent pain effects and modifies different neuronal cell types in the ACC and how this contributes to sensory sensitization is not completely understood. This study confirms the existence of 2 primary subtypes of pyramidal neurons in layer 5 of the rostral, agranular ACC, which we could classify as intratelencephalic (IT) and cortico-subcortical (SC) projecting neurons, similar to other cortical brain areas. Through retrograde labeling, whole-cell patch-clamp recording, and morphological analysis, we thoroughly characterized their different electrophysiological and morphological properties. When examining the effects of peripheral inflammatory pain on these neuronal subtypes, we observed time-dependent plastic changes in excitability. During the acute phase, both subtypes exhibited reduced excitability, which normalized to pre-inflammatory levels after day 7. Daily conditioning with nociceptive stimuli during this period induced an increase in excitability specifically in SC neurons, which was correlated with a decrease in mechanical sensitization. Subsequent inhibition of the activity of SC neurons projecting to the periaqueductal gray with in vivo chemogenetics, resulted in reinstatement of the hypersensitivity. Accordingly, it was sufficient to enhance the excitability of these neurons chemogenetically in the inflammatory pain condition to induce hypoalgesia. These findings suggest a cell type-specific effect on the descending control of nociception and a cellular mechanism for pain-induced analgesia. Furthermore, increased excitability in this neuronal population is hypoalgesic rather than hyperalgesic.
Collapse
Affiliation(s)
| | - Mario A. Acuña
- Department of Physiology, University of Bern, Bern, Switzerland
| | | | - Thomas Nevian
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Li J, Bai Y, Ge J, Zhang Y, Zhao Q, Li D, Guo B, Gao S, Zhu Y, Cai G, Wan X, Huang J, Wu S. Cell Type-Specific Modulation of Acute Itch Processing in the Anterior Cingulate Cortex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403445. [PMID: 39316379 DOI: 10.1002/advs.202403445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/24/2024] [Indexed: 09/25/2024]
Abstract
Despite remarkable progress in understanding the fundamental bases of itching, its cortical mechanisms remain poorly understood. Herein, the causal contributions of defined anterior cingulate cortex (ACC) neuronal populations to acute itch modulation in mice are established. Using cell type-specific manipulations, the opposing functions of ACC glutamatergic and GABAergic neurons in regulating acute itching are demonstrated. Photometry studies indicated that ACC glutamatergic neurons are activated during scratching induced by both histamine and chloroquine, whereas the activation pattern of GABAergic neurons is complicated by GABAergic subpopulations and acute itch modalities. By combining cell type- and projection-specific techniques, a thalamocortical circuit is further identified from the mediodorsal thalamus driving the itch-scratching cycle related to histaminergic and non-histaminergic itching, which is contingent on the activation of postsynaptic parvalbumin-expressing neurons in the ACC. These findings reveal a cellular and circuit signature of ACC neurons orchestrating behavioral responses to itching and may provide insights into therapies for itch-related diseases.
Collapse
Affiliation(s)
- Jiaqi Li
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, 110015, China
| | - Junye Ge
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yiwen Zhang
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Qiuying Zhao
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Dangchao Li
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Baolin Guo
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Shasha Gao
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuanyuan Zhu
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Guohong Cai
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiangdong Wan
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Huang
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Shengxi Wu
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
9
|
Liu L, Liang Z, Zhang L, Feng Z, Cao F, Zhang Y, Yang X, Zhang L, Wang J, Zhu Q. Corticothalamic input derived from corticospinal neurons contributes to chronic neuropathic pain after spinal cord injury. Exp Neurol 2024; 381:114923. [PMID: 39142366 DOI: 10.1016/j.expneurol.2024.114923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
Neuropathic pain is a significant and persistent issue for individuals with spinal cord injuries (SCI), severely impacting their quality of life. While changes at the peripheral and spinal levels are known to contribute to SCI-related pain, whether and how supraspinal centers contribute to post SCI chronic neuropathic pain is poorly understood. Here, we first validated delayed development of chronic neuropathic pain in mice with moderate contusion SCI. To identify supraspinal regions involved in the pathology of neuropathic pain after SCI, we next performed an activity dependent genetic screening and identified multiple cortical and subcortical regions that were activated by innocuous tactile stimuli at a late stage following contusion SCI. Notably, chemogenetic inactivation of pain trapped neurons in the lateral thalamus alleviated neuropathic pain and reduced tactile stimuli evoked cortical overactivation. Retrograde tracing showed that contusion SCI led to enhanced corticothalamic axonal sprouting and over-activation of corticospinal neurons. Mechanistically, ablation or silencing of corticospinal neurons prevented the establishment or maintenance of chronic neuropathic pain following contusion SCI. These results highlighted a corticospinal-lateral thalamic feed-forward loop whose activation is required for the development and maintenance of chronic neuropathic pain after SCI. Our data thus shed lights into the central mechanisms underlying chronic neuropathic pain associated with SCI and the development of novel therapeutic avenues to treat refractory pain caused by traumatic brain or spinal cord injuries.
Collapse
Affiliation(s)
- Ling Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihou Liang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Feng
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
| | - Fei Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunjian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijie Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qing Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
Lian YN, Cao XW, Wu C, Pei CY, Liu L, Zhang C, Li XY. Deconstruction the feedforward inhibition changes in the layer III of anterior cingulate cortex after peripheral nerve injury. Commun Biol 2024; 7:1237. [PMID: 39354145 PMCID: PMC11445484 DOI: 10.1038/s42003-024-06849-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
The anterior cingulate cortex (ACC) is one of the critical brain areas for processing noxious information. Previous studies showed that peripheral nerve injury induced broad changes in the ACC, contributing to pain hypersensitivity. The neurons in layer 3 (L3) of the ACC receive the inputs from the mediodorsal thalamus (MD) and form the feedforward inhibition (FFI) microcircuits. The effects of peripheral nerve injury on the MD-driven FFI in L3 of ACC are unknown. In our study, we record the enhanced excitatory synaptic transmissions from the MD to L3 of the ACC in mice with common peroneal nerve ligation, affecting FFI. Chemogenetically activating the MD-to-ACC projections induces pain sensitivity and place aversion in naive mice. Furthermore, chemogenetically inactivating MD-to-ACC projections decreases pain sensitivity and promotes place preference in nerve-injured mice. Our results indicate that the peripheral nerve injury changes the MD-to-ACC projections, contributing to pain hypersensitivity and aversion.
Collapse
Affiliation(s)
- Yan-Na Lian
- Department of Psychiatry, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, 322000, China
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xiao-Wen Cao
- Department of Psychiatry, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, 322000, China
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Cheng Wu
- Department of Psychiatry, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, 322000, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang, 314400, China
| | - Chen-Yu Pei
- Department of Psychiatry, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, 322000, China
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Li Liu
- Core Facilities of the School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Chen Zhang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, Jiangsu, 210000, China.
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Laboratory of Oral Health, Capital Medical University, Beijing, 100069, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| | - Xiang-Yao Li
- Department of Psychiatry, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, 322000, China.
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang, 314400, China.
| |
Collapse
|
11
|
Zhu Y, Sun H, Xiao S, Shen Z, Zhu X, Wang Y, He X, Liu B, Jiang Y, Liang Y, Fang J, Shao X. Electroacupuncture inhibited carrageenan-induced pain aversion by activating GABAergic neurons in the ACC. Mol Brain 2024; 17:69. [PMID: 39334299 PMCID: PMC11428560 DOI: 10.1186/s13041-024-01144-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: 06/08/2023] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Pain aversion is an avoidance response to painful stimuli. Previous research has indicated that the anterior cingulate cortex (ACC) is involved in pain aversion processing. However, as interneurons, the role of GABAergic neurons in the ACC (GABAACC neurons) in pain aversion is still unclear. Electroacupuncture (EA) has been shown to ameliorate pain aversion, but the mechanism is not clarified. The present study provided evidence that inhibition of GABAACC neurons contributed to pain aversion. EA alleviated pain aversion by activating GABAACC neurons in an intensity-dependent manner. Specifically, 0.3 mA EA stimulation showed better effects on pain aversion than 0.1 mA stimulation, which could be reversed by chemical genetic inhibition of GABAACC neurons. These results provide a novel mechanism by which EA alleviates pain aversion by reversing GABAACC neurons.
Collapse
Affiliation(s)
- Yichen Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Haiju Sun
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Siqi Xiao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zui Shen
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xixiao Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yifang Wang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Boyi Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yongliang Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yi Liang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Janqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xiaomei Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
12
|
Yang XL, Gao W, Dong WY, Zheng C, Wang S, Wei HR, Luo Y, Zhang Z, Chen Y, Jin Y. A neural circuit for alcohol withdrawal-induced hyperalgesia in a nondependent state. SCIENCE ADVANCES 2024; 10:eadp8636. [PMID: 39331713 PMCID: PMC11430459 DOI: 10.1126/sciadv.adp8636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/23/2024] [Indexed: 09/29/2024]
Abstract
Alcohol use disorder is highly prevalent worldwide, with characteristically severe pain sensitivity during withdrawal. Here, we established a mouse model of hyperalgesia during ethanol withdrawal (EW) before addiction to investigate the window for onset and underlying mechanisms. Viral tracing with in vivo microendoscopic and two-photon calcium imaging identified a circuit pathway from dorsal hippocampal CA1 glutamatergic neurons (dCA1Glu) to anterior cingulate cortex glutamatergic neurons (ACCGlu) activated in EW mice with hyperalgesia. Chemogenetic inhibition of this pathway can alleviate hyperalgesia in EW mice, whereas artificial activation recapitulates EW-induced hyperalgesia in naïve mice. These findings demonstrate that the dCA1Glu → ACCGlu neuronal pathway participates in driving EW-induced hyperalgesia before ethanol dependence in mice.
Collapse
Affiliation(s)
- Xin-Lu Yang
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Anesthesiology, First Affiliated Hospital of Wannan Medical College, Wuhu 241002, China
| | - Wei Gao
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Wan-Ying Dong
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Changjian Zheng
- Department of Anesthesiology, First Affiliated Hospital of Wannan Medical College, Wuhu 241002, China
| | - Sheng Wang
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Hong-Rui Wei
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yanli Luo
- Department of Psychological Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhi Zhang
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Biophysics and Neurobiology, CAS Key laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei 230026, China
| | - Yongquan Chen
- Department of Anesthesiology, First Affiliated Hospital of Wannan Medical College, Wuhu 241002, China
| | - Yan Jin
- Department of Biophysics and Neurobiology, CAS Key laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei 230026, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| |
Collapse
|
13
|
Jaeckel ER, Arias-Hervert ER, Perez-Medina AL, Schulz S, Birdsong WT. Chronic morphine treatment induces sex- and synapse-specific cellular tolerance on thalamo-cortical mu opioid receptor signaling. J Neurophysiol 2024; 132:968-978. [PMID: 39110512 PMCID: PMC11427077 DOI: 10.1152/jn.00265.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
How cellular adaptations give rise to opioid analgesic tolerance to opioids like morphine is not well understood. For one, pain is a complex phenomenon comprising both sensory and affective components, largely mediated through separate circuits. Glutamatergic projections from the medial thalamus (MThal) to the anterior cingulate cortex (ACC) are implicated in processing of affective pain, a relatively understudied component of the pain experience. The goal of this study was to determine the effects of chronic morphine exposure on mu-opioid receptor (MOR) signaling on MThal-ACC synaptic transmission within the excitatory and feedforward inhibitory pathways. Using whole cell patch-clamp electrophysiology and optogenetics to selectively target these projections, we measured morphine-mediated inhibition of optically evoked postsynaptic currents in ACC layer V pyramidal neurons in drug-naïve and chronically morphine-treated mice. We found that morphine perfusion inhibited the excitatory and feedforward inhibitory pathways similarly in females but caused greater inhibition of the inhibitory pathway in males. Chronic morphine treatment robustly attenuated morphine presynaptic inhibition within the inhibitory pathway in males, but not females, and mildly attenuated presynaptic inhibition within the excitatory pathway in both sexes. These effects were not observed in MOR phosphorylation-deficient mice. This study indicates that chronic morphine treatment induces cellular tolerance to morphine within a thalamo-cortical circuit relevant to pain and opioid analgesia. Furthermore, it suggests this tolerance may be driven by MOR phosphorylation. Overall, these findings improve our understanding of how chronic opioid exposure alters cellular signaling in ways that may contribute to opioid analgesic tolerance.NEW & NOTEWORTHY Opioid signaling within the anterior cingulate cortex (ACC) is important for opioid modulation of affective pain. Glutamatergic medial thalamus (MThal) neurons synapse in the ACC and opioids, acting through mu opioid receptors (MORs), acutely inhibit synaptic transmission from MThal synapses. However, the effect of chronic opioid exposure on MThal-ACC synaptic transmission is not known. Here, we demonstrate that chronic morphine treatment induces cellular tolerance at these synapses in a sex-specific and phosphorylation-dependent manner.
Collapse
Affiliation(s)
- Elizabeth R Jaeckel
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, United States
| | - Erwin R Arias-Hervert
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, United States
| | | | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | - William T Birdsong
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
14
|
Suzuki Y, Kiyosawa M, Wakakura M, Ishii K. Hyperactivity of the medial thalamus in patients with photophobia-associated migraine. Headache 2024; 64:1005-1014. [PMID: 39023425 DOI: 10.1111/head.14785] [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: 10/22/2023] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVE To examine cerebral functional alterations associated with sensory processing in patients with migraine and constant photophobia. BACKGROUND Migraine is a common headache disorder that presents with photophobia in many patients during attacks. Furthermore, some patients with migraine experience constant photophobia, even during headache-free intervals, leading to a compromised quality of life. METHODS This prospective, case-control study included 40 patients with migraine (18 male and 22 female) who were recruited at an eye hospital and eye clinic. The patients were divided into two groups: migraine with photophobia group, consisting of 22 patients (10 male and 12 female) with constant photophobia, and migraine without photophobia group, consisting of 18 patients (eight male and 10 female) without constant photophobia. We used 18F-fluorodeoxyglucose and positron emission tomography to compare cerebral glucose metabolism between the two patient groups and 42 healthy participants (16 men and 26 women). RESULTS Compared with the healthy group, both the migraine with photophobia and migraine without photophobia groups showed cerebral glucose hypermetabolism in the bilateral thalamus (p < 0.05, family-wise error-corrected). Moreover, the contrast of migraine with photophobia minus migraine without photophobia patients showed glucose hypermetabolism in the bilateral medial thalamus (p < 0.05, family-wise error-corrected). CONCLUSIONS The medial thalamus may be associated with the development of continuous photophobia in patients with migraine.
Collapse
Affiliation(s)
- Yukihisa Suzuki
- Japan Community Health Care Organization, Mishima General Hospital, Mishima, Shizuoka, Japan
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
15
|
Wu H, Xie L, Chen Q, Xu F, Dai A, Ma X, Xie S, Li H, Zhu F, Jiao C, Sun L, Xu Q, Zhou Y, Shen Y, Chen X. Activation of GABAergic neurons in the dorsal raphe nucleus alleviates hyperalgesia induced by ovarian hormone withdrawal. Pain 2024:00006396-990000000-00678. [PMID: 39106454 DOI: 10.1097/j.pain.0000000000003362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 06/25/2024] [Indexed: 08/09/2024]
Abstract
ABSTRACT Menopausal and postmenopausal women, characterized by a significant reduction in ovarian hormones, have a high prevalence of chronic pain with great pain intensity. However, the underlying mechanism of hyperalgesia induced by ovarian hormone withdrawal remains poorly understood. Here, we report that decreases in the activity and excitability of GABAergic neurons in the dorsal raphe nucleus (DRN) are associated with hyperalgesia induced by ovariectomy in mice. Supplementation with 17β-estradiol, but not progesterone, is sufficient to increase the mechanical pain threshold in ovariectomized (OVX) mice and the excitability of DRN GABAergic (DRNGABA) neurons. Moreover, activation of the DRNGABA neurons projecting to the lateral parabrachial nucleus was critical for alleviating hyperalgesia in OVX mice. These findings show the essential role of DRNGABA neurons and their modulation by estrogen in regulating hyperalgesia induced by ovarian hormone withdrawal, providing therapeutic basis for the treatment of chronic pain in physiological or surgical menopausal women.
Collapse
Affiliation(s)
- Hui Wu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linghua Xie
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qing Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ange Dai
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolin Ma
- School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Shulan Xie
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hua Li
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fangfang Zhu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cuicui Jiao
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihong Sun
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yudong Zhou
- School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yi Shen
- School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Xinzhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
16
|
Viellard J, Bouali-Benazzouz R, Benazzouz A, Fossat P. Modulating Neural Circuits of Pain in Preclinical Models: Recent Insights for Future Therapeutics. Cells 2024; 13:997. [PMID: 38920628 PMCID: PMC11202162 DOI: 10.3390/cells13120997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Chronic pain is a pathological state defined as daily pain sensation over three consecutive months. It affects up to 30% of the general population. Although significant research efforts have been made in the past 30 years, only a few and relatively low effective molecules have emerged to treat chronic pain, with a considerable translational failure rate. Most preclinical models have focused on sensory neurotransmission, with particular emphasis on the dorsal horn of the spinal cord as the first relay of nociceptive information. Beyond impaired nociceptive transmission, chronic pain is also accompanied by numerous comorbidities, such as anxiety-depressive disorders, anhedonia and motor and cognitive deficits gathered under the term "pain matrix". The emergence of cutting-edge techniques assessing specific neuronal circuits allow in-depth studies of the connections between "pain matrix" circuits and behavioural outputs. Pain behaviours are assessed not only by reflex-induced responses but also by various or more complex behaviours in order to obtain the most complete picture of an animal's pain state. This review summarises the latest findings on pain modulation by brain component of the pain matrix and proposes new opportunities to unravel the mechanisms of chronic pain.
Collapse
Affiliation(s)
- Juliette Viellard
- Université de Bordeaux, UMR 5293, F-33076 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Rabia Bouali-Benazzouz
- Université de Bordeaux, UMR 5293, F-33076 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Abdelhamid Benazzouz
- Université de Bordeaux, UMR 5293, F-33076 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Pascal Fossat
- Université de Bordeaux, UMR 5293, F-33076 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| |
Collapse
|
17
|
Faig CA, Kim GHK, Do AD, Dworsky-Fried Z, Jackson J, Taylor AMW. Claustrum projections to the anterior cingulate modulate nociceptive and pain-associated behavior. Curr Biol 2024; 34:1987-1995.e4. [PMID: 38614081 DOI: 10.1016/j.cub.2024.03.044] [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: 01/11/2024] [Revised: 03/08/2024] [Accepted: 03/22/2024] [Indexed: 04/15/2024]
Abstract
The anterior cingulate cortex (ACC) is critical for the perception and unpleasantness of pain.1,2,3,4,5,6 It receives nociceptive information from regions such as the thalamus and amygdala and projects to several cortical and subcortical regions of the pain neuromatrix.7,8 ACC hyperexcitability is one of many functional changes associated with chronic pain, and experimental activation of ACC pyramidal cells produces hypersensitivity to innocuous stimuli (i.e., allodynia).9,10,11,12,13,14 A less-well-studied projection to the ACC arises from a small forebrain region, the claustrum.15,16,17,18,19,20 Stimulation of excitatory claustrum projection neurons preferentially activates GABAergic interneurons, generating feed-forward inhibition onto excitatory cortical networks.21,22,23,24 Previous work has shown that claustrocingulate projections display altered activity in prolonged pain25,26,27; however, it remains unclear whether and how the claustrum participates in nociceptive processing and high-order pain behaviors. Inhibition of ACC activity reverses mechanical allodynia in animal models of persistent and neuropathic pain,1,9,28 suggesting claustrum inputs may function to attenuate pain processing. In this study, we sought to define claustrum function in acute and chronic pain. We found enhanced claustrum activity after a painful stimulus that was attenuated in chronic inflammatory pain. Selective inhibition of claustrocingulate projection neurons enhanced acute nociception but blocked pain learning. Inversely, chemogenetic activation of claustrocingulate neurons had no effect on basal nociception but rescued inflammation-induced mechanical allodynia. Together, these results suggest that claustrocingulate neurons are a critical component of the pain neuromatrix, and dysregulation of this connection may contribute to chronic pain.
Collapse
Affiliation(s)
- Christian A Faig
- Department of Pharmacology, University of Alberta, 8613 114 Street NW, Edmonton, AB T6G 2R3, Canada; Neuroscience and Mental Health Institute, University of Alberta, 11315 87 Avenue NW, Edmonton, AB T6G 2E1, Canada
| | - Gloria H K Kim
- Neuroscience and Mental Health Institute, University of Alberta, 11315 87 Avenue NW, Edmonton, AB T6G 2E1, Canada
| | - Alison D Do
- Department of Physiology, University of Alberta, 8613 114 Street NW, Edmonton, AB T6G 2R3, Canada
| | - Zoë Dworsky-Fried
- Department of Pharmacology, University of Alberta, 8613 114 Street NW, Edmonton, AB T6G 2R3, Canada
| | - Jesse Jackson
- Neuroscience and Mental Health Institute, University of Alberta, 11315 87 Avenue NW, Edmonton, AB T6G 2E1, Canada; Department of Physiology, University of Alberta, 8613 114 Street NW, Edmonton, AB T6G 2R3, Canada.
| | - Anna M W Taylor
- Department of Pharmacology, University of Alberta, 8613 114 Street NW, Edmonton, AB T6G 2R3, Canada; Neuroscience and Mental Health Institute, University of Alberta, 11315 87 Avenue NW, Edmonton, AB T6G 2E1, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, 11315 87 Avenue NW, Edmonton, AB T6G 2E1, Canada; Department of Anesthesiology and Pain Medicine, University of Alberta, 8440 112 Street NW, Edmonton, AB T6G 2B7, Canada.
| |
Collapse
|
18
|
James JG, McCall NM, Hsu AI, Oswell CS, Salimando GJ, Mahmood M, Wooldridge LM, Wachira M, Jo A, Sandoval Ortega RA, Wojick JA, Beattie K, Farinas SA, Chehimi SN, Rodrigues A, Ejoh LSL, Kimmey BA, Lo E, Azouz G, Vasquez JJ, Banghart MR, Creasy KT, Beier KT, Ramakrishnan C, Crist RC, Reiner BC, Deisseroth K, Yttri EA, Corder G. Mimicking opioid analgesia in cortical pain circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591113. [PMID: 38746090 PMCID: PMC11092437 DOI: 10.1101/2024.04.26.591113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The anterior cingulate cortex plays a pivotal role in the cognitive and affective aspects of pain perception. Both endogenous and exogenous opioid signaling within the cingulate mitigate cortical nociception, reducing pain unpleasantness. However, the specific functional and molecular identities of cells mediating opioid analgesia in the cingulate remain elusive. Given the complexity of pain as a sensory and emotional experience, and the richness of ethological pain-related behaviors, we developed a standardized, deep-learning platform for deconstructing the behavior dynamics associated with the affective component of pain in mice-LUPE (Light aUtomated Pain Evaluator). LUPE removes human bias in behavior quantification and accelerated analysis from weeks to hours, which we leveraged to discover that morphine altered attentional and motivational pain behaviors akin to affective analgesia in humans. Through activity-dependent genetics and single-nuclei RNA sequencing, we identified specific ensembles of nociceptive cingulate neuron-types expressing mu-opioid receptors. Tuning receptor expression in these cells bidirectionally modulated morphine analgesia. Moreover, we employed a synthetic opioid receptor promoter-driven approach for cell-type specific optical and chemical genetic viral therapies to mimic morphine's pain-relieving effects in the cingulate, without reinforcement. This approach offers a novel strategy for precision pain management by targeting a key nociceptive cortical circuit with on-demand, non-addictive, and effective analgesia.
Collapse
Affiliation(s)
- Justin G. James
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nora M. McCall
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex I. Hsu
- Dept. of Biobehavioral Health Sciences, School of Nursing, and Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Corinna S. Oswell
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory J. Salimando
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Malaika Mahmood
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa M. Wooldridge
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meghan Wachira
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adrienne Jo
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jessica A. Wojick
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine Beattie
- Dept. of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sofia A. Farinas
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samar N. Chehimi
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amrith Rodrigues
- Dept. of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lind-say L. Ejoh
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Blake A. Kimmey
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Lo
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ghalia Azouz
- Dept. of Physiology and Biophysics, University of California Irvine, CA, USA
| | - Jose J. Vasquez
- Dept. of Physiology and Biophysics, University of California Irvine, CA, USA
| | - Matthew R. Banghart
- Dept. of Neurobiology, School of Biological Sciences, University of California San Diego, CA, USA
| | - Kate Townsend Creasy
- Dept. of Biobehavioral Health Sciences, School of Nursing, and Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin T. Beier
- Dept. of Physiology and Biophysics, University of California Irvine, CA, USA
| | | | - Richard C. Crist
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin C. Reiner
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karl Deisseroth
- CNC Program, Stanford University, Stanford, CA, USA
- Dept. of Bioengineering, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
- Dept. of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Eric A. Yttri
- Dept. of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Gregory Corder
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
19
|
Chang X, Zhang H, Chen S. Neural circuits regulating visceral pain. Commun Biol 2024; 7:457. [PMID: 38615103 PMCID: PMC11016080 DOI: 10.1038/s42003-024-06148-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/05/2024] [Indexed: 04/15/2024] Open
Abstract
Visceral hypersensitivity, a common clinical manifestation of irritable bowel syndrome, may contribute to the development of chronic visceral pain, which is a major challenge for both patients and health providers. Neural circuits in the brain encode, store, and transfer pain information across brain regions. In this review, we focus on the anterior cingulate cortex and paraventricular nucleus of the hypothalamus to highlight the progress in identifying the neural circuits involved in visceral pain. We also discuss several neural circuit mechanisms and emphasize the importance of cross-species, multiangle approaches and the identification of specific neurons in determining the neural circuits that control visceral pain.
Collapse
Affiliation(s)
- Xiaoli Chang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Haiyan Zhang
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shaozong Chen
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
20
|
Li Z, Zhang L, Zhang F, Yue L, Hu L. Deciphering Authentic Nociceptive Thalamic Responses in Rats. RESEARCH (WASHINGTON, D.C.) 2024; 7:0348. [PMID: 38617991 PMCID: PMC11014087 DOI: 10.34133/research.0348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/12/2024] [Indexed: 04/16/2024]
Abstract
The thalamus and its cortical connections play a pivotal role in pain information processing, yet the exploration of its electrophysiological responses to nociceptive stimuli has been limited. Here, in 2 experiments we recorded neural responses to nociceptive laser stimuli in the thalamic (ventral posterior lateral nucleus and medial dorsal nucleus) and cortical regions (primary somatosensory cortex [S1] and anterior cingulate cortex) within the lateral and medial pain pathways. We found remarkable similarities in laser-evoked brain responses that encoded pain intensity within thalamic and cortical regions. Contrary to the expected temporal sequence of ascending information flow, the recorded thalamic response (N1) was temporally later than its cortical counterparts, suggesting that it may not be a genuine thalamus-generated response. Importantly, we also identified a distinctive component in the thalamus, i.e., the early negativity (EN) occurring around 100 ms after the onset of nociceptive stimuli. This EN component represents an authentic nociceptive thalamic response and closely synchronizes with the directional information flow from the thalamus to the cortex. These findings underscore the importance of isolating genuine thalamic neural responses, thereby contributing to a more comprehensive understanding of the thalamic function in pain processing. Additionally, these findings hold potential clinical implications, particularly in the advancement of closed-loop neuromodulation treatments for neurological diseases targeting this vital brain region.
Collapse
Affiliation(s)
- Zhenjiang Li
- CAS Key Laboratory of Mental Health, Institute of Psychology,
Chinese Academy of Sciences, 100101 Beijing, China
- Department of Psychology,
University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Libo Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology,
Chinese Academy of Sciences, 100101 Beijing, China
- Department of Psychology,
University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Fengrui Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology,
Chinese Academy of Sciences, 100101 Beijing, China
- Department of Psychology,
University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Lupeng Yue
- CAS Key Laboratory of Mental Health, Institute of Psychology,
Chinese Academy of Sciences, 100101 Beijing, China
- Department of Psychology,
University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Li Hu
- CAS Key Laboratory of Mental Health, Institute of Psychology,
Chinese Academy of Sciences, 100101 Beijing, China
- Department of Psychology,
University of Chinese Academy of Sciences, 100049 Beijing, China
| |
Collapse
|
21
|
Koga K, Kobayashi K, Tsuda M, Pickering AE, Furue H. Anterior cingulate cross-hemispheric inhibition via the claustrum resolves painful sensory conflict. Commun Biol 2024; 7:330. [PMID: 38491200 PMCID: PMC10943010 DOI: 10.1038/s42003-024-06008-9] [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: 12/08/2022] [Accepted: 03/04/2024] [Indexed: 03/18/2024] Open
Abstract
The anterior cingulate cortex (ACC) responds to noxious and innocuous sensory inputs, and integrates them to coordinate appropriate behavioral reactions. However, the role of the projections of ACC neurons to subcortical areas and their influence on sensory processing are not fully investigated. Here, we identified that ACC neurons projecting to the contralateral claustrum (ACC→contraCLA) preferentially respond to contralateral mechanical sensory stimulation. These sensory responses were enhanced during attending behavior. Optogenetic activation of ACC→contraCLA neurons silenced pyramidal neurons in the contralateral ACC by recruiting local circuit fast-spiking interneuron activation via an excitatory relay in the CLA. This circuit activation suppressed withdrawal behavior to mechanical stimuli ipsilateral to the ACC→contraCLA neurons. Chemogenetic silencing showed that the cross-hemispheric circuit has an important role in the suppression of contralateral nociceptive behavior during sensory-driven attending behavior. Our findings identify a cross-hemispheric cortical-subcortical-cortical arc allowing the brain to give attentional priority to competing innocuous and noxious inputs.
Collapse
Affiliation(s)
- Keisuke Koga
- Department of Neurophysiology, Hyogo Medical University, Nishinomiya, Japan.
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Anthony E Pickering
- Anesthesia, Pain and Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Hidemasa Furue
- Department of Neurophysiology, Hyogo Medical University, Nishinomiya, Japan.
| |
Collapse
|
22
|
Song Q, Wei A, Xu H, Gu Y, Jiang Y, Dong N, Zheng C, Wang Q, Gao M, Sun S, Duan X, Chen Y, Wang B, Huo J, Yao J, Wu H, Li H, Wu X, Jing Z, Liu X, Yang Y, Hu S, Zhao A, Wang H, Cheng X, Qin Y, Qu Q, Chen T, Zhou Z, Chai Z, Kang X, Wei F, Wang C. An ACC-VTA-ACC positive-feedback loop mediates the persistence of neuropathic pain and emotional consequences. Nat Neurosci 2024; 27:272-285. [PMID: 38172439 DOI: 10.1038/s41593-023-01519-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/04/2023] [Indexed: 01/05/2024]
Abstract
The central mechanisms underlying pain chronicity remain elusive. Here, we identify a reciprocal neuronal circuit in mice between the anterior cingulate cortex (ACC) and the ventral tegmental area (VTA) that mediates mutual exacerbation between hyperalgesia and allodynia and their emotional consequences and, thereby, the chronicity of neuropathic pain. ACC glutamatergic neurons (ACCGlu) projecting to the VTA indirectly inhibit dopaminergic neurons (VTADA) by activating local GABAergic interneurons (VTAGABA), and this effect is reinforced after nerve injury. VTADA neurons in turn project to the ACC and synapse to the initial ACCGlu neurons to convey feedback information from emotional changes. Thus, an ACCGlu-VTAGABA-VTADA-ACCGlu positive-feedback loop mediates the progression to and maintenance of persistent pain and comorbid anxiodepressive-like behavior. Disruption of this feedback loop relieves hyperalgesia and anxiodepressive-like behavior in a mouse model of neuropathic pain, both acutely and in the long term.
Collapse
Affiliation(s)
- Qian Song
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Anqi Wei
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Huadong Xu
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yuhao Gu
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Yong Jiang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Nan Dong
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Chaowen Zheng
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Qinglong Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology; Peking-Tsinghua Center for Life Sciences; and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Min Gao
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology; Peking-Tsinghua Center for Life Sciences; and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Suhua Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology; Peking-Tsinghua Center for Life Sciences; and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Xueting Duan
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Yang Chen
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Bianbian Wang
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Jingxiao Huo
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Jingyu Yao
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Hao Wu
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Hua Li
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Xuanang Wu
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Zexin Jing
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoying Liu
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Yuxin Yang
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
- College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Shaoqin Hu
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Anran Zhao
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China
| | - Hongyan Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Xu Cheng
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yuhao Qin
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Qiumin Qu
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tao Chen
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, the Fourth Military Medical University, Xi'an, China
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology; Peking-Tsinghua Center for Life Sciences; and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Zuying Chai
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Xinjiang Kang
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, China.
- College of Life Sciences, Liaocheng University, Liaocheng, China.
| | - Feng Wei
- Department of Neural and Pain Sciences, School of Dentistry; Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, USA.
| | - Changhe Wang
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, China.
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
23
|
Gulledge AT. Cholinergic Activation of Corticofugal Circuits in the Adult Mouse Prefrontal Cortex. J Neurosci 2024; 44:e1388232023. [PMID: 38050146 PMCID: PMC10860659 DOI: 10.1523/jneurosci.1388-23.2023] [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: 07/20/2023] [Revised: 10/10/2023] [Accepted: 11/08/2023] [Indexed: 12/06/2023] Open
Abstract
Acetylcholine (ACh) promotes neocortical output to the thalamus and brainstem by preferentially enhancing the postsynaptic excitability of layer 5 pyramidal tract (PT) neurons relative to neighboring intratelencephalic (IT) neurons. Less is known about how ACh regulates the excitatory synaptic drive of IT and PT neurons. To address this question, spontaneous excitatory postsynaptic potentials (sEPSPs) were recorded in dual recordings of IT and PT neurons in slices of prelimbic cortex from adult female and male mice. ACh (20 µM) enhanced sEPSP amplitudes, frequencies, rise-times, and half-widths preferentially in PT neurons. These effects were blocked by the muscarinic receptor antagonist atropine (1 µM). When challenged with pirenzepine (1 µM), an antagonist selective for M1-type muscarinic receptors, ACh instead reduced sEPSP frequencies, suggesting that ACh may generally suppress synaptic transmission in the cortex via non-M1 receptors. Cholinergic enhancement of sEPSPs in PT neurons was not sensitive to antagonism of GABA receptors with gabazine (10 µM) and CGP52432 (2.5 µM) but was blocked by tetrodotoxin (1 µM), suggesting that ACh enhances action-potential-dependent excitatory synaptic transmission in PT neurons. ACh also preferentially promoted the occurrence of synchronous sEPSPs in dual recordings of PT neurons relative to IT-PT and IT-IT parings. Finally, selective chemogenetic silencing of hM4Di-expressing PT, but not commissural IT, neurons blocked cholinergic enhancement of sEPSP amplitudes and frequencies in PT neurons. These data suggest that, in addition to selectively enhancing the postsynaptic excitability of PT neurons, M1 receptor activation promotes corticofugal output by amplifying recurrent excitation within networks of PT neurons.
Collapse
Affiliation(s)
- Allan T Gulledge
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College, Hanover 03755, New Hampshire
| |
Collapse
|
24
|
Krimmel SR, Laumann TO, Chauvin RJ, Hershey T, Roland JL, Shimony JS, Willie JT, Norris SA, Marek S, Van AN, Monk J, Scheidter KM, Whiting F, Ramirez-Perez N, Metoki A, Wang A, Kay BP, Nahman-Averbuch H, Fair DA, Lynch CJ, Raichle ME, Gordon EM, Dosenbach NUF. The brainstem's red nucleus was evolutionarily upgraded to support goal-directed action. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573730. [PMID: 38260662 PMCID: PMC10802246 DOI: 10.1101/2023.12.30.573730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The red nucleus is a large brainstem structure that coordinates limb movement for locomotion in quadrupedal animals (Basile et al., 2021). The humans red nucleus has a different pattern of anatomical connectivity compared to quadrupeds, suggesting a unique purpose (Hatschek, 1907). Previously the function of the human red nucleus remained unclear at least partly due to methodological limitations with brainstem functional neuroimaging (Sclocco et al., 2018). Here, we used our most advanced resting-state functional connectivity (RSFC) based precision functional mapping (PFM) in highly sampled individuals (n = 5) and large group-averaged datasets (combined N ~ 45,000), to precisely examine red nucleus functional connectivity. Notably, red nucleus functional connectivity to motor-effector networks (somatomotor hand, foot, and mouth) was minimal. Instead, red nucleus functional connectivity along the central sulcus was specific to regions of the recently discovered somato-cognitive action network (SCAN; (Gordon et al., 2023)). Outside of primary motor cortex, red nucleus connectivity was strongest to the cingulo-opercular (CON) and salience networks, involved in action/cognitive control (Dosenbach et al., 2007; Newbold et al., 2021) and reward/motivated behavior (Seeley, 2019), respectively. Functional connectivity to these two networks was organized into discrete dorsal-medial and ventral-lateral zones. Red nucleus functional connectivity to the thalamus recapitulated known structural connectivity of the dento-rubral thalamic tract (DRTT) and could prove clinically useful in functionally targeting the ventral intermediate (VIM) nucleus. In total, our results indicate that far from being a 'motor' structure, the red nucleus is better understood as a brainstem nucleus for implementing goal-directed behavior, integrating behavioral valence and action plans.
Collapse
Affiliation(s)
- Samuel R Krimmel
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Timothy O Laumann
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Roselyne J Chauvin
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tamara Hershey
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychological & Brain Sciences, Washington University, St. Louis, Missouri, USA
| | - Jarod L Roland
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jon T Willie
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, New York, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri
| | - Scott A Norris
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Scott Marek
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrew N Van
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Julia Monk
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kristen M Scheidter
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Forrest Whiting
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nadeshka Ramirez-Perez
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Athanasia Metoki
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Anxu Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Division of Computation and Data Science, Washington University, St. Louis, Missouri, USA
| | - Benjamin P Kay
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hadas Nahman-Averbuch
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Damien A Fair
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, Minnesota, USA
- Institute of Child Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Charles J Lynch
- Department of Psychiatry, Weill Cornell Medicine, New York, New York, USA
| | - Marcus E Raichle
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychological & Brain Sciences, Washington University, St. Louis, Missouri, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Evan M Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nico U F Dosenbach
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Psychological & Brain Sciences, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
- Program in Occupational Therapy, Washington University, St. Louis, Missouri, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
25
|
Xue M, Chen QY, Shi W, Zhou Z, Li X, Xu F, Bi G, Yang X, Lu JS, Zhuo M. Whole-brain mapping of afferents to the anterior cingulate cortex in adult mice. Mol Pain 2024; 20:17448069241300990. [PMID: 39614717 DOI: 10.1177/17448069241300990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024] Open
Abstract
The anterior cingulate cortex (ACC) is critical for pain perception, emotion and cognition. Previous studies showed that the ACC has a complex network architecture, which can receive some projection fibers from many brain regions, including the thalamus, the cerebral cortex and other brain regions. However, there was still a lack of whole-brain mapping of the ACC in adult mice. In the present study, we utilized a rabies virus-based retrograde trans-monosynaptic tracing system to map whole-brain afferents to the unilateral ACC in adult mice. We also combined with a new high-throughput, high-speed and high-resolution VISoR imaging technique to generate a three-dimensional whole-brain reconstruction. Our results showed that several principal groups of brain structures send direct monosynaptic inputs to the ACC, including the cerebral cortex, amygdala, striatum, the thalamus, and the brainstem. We also found that cortical neurons in the ACC mainly receive ipsilateral monosynaptic projections. Some cortical areas and forebrain regions also bilaterally projected to the ACC. These findings provide a complete analysis of the afferents to the ACC in adult mice, and whole-brain mapping of ACC afferents would provide important anatomic evidence for the study of pain, memory, and cognition.
Collapse
Affiliation(s)
- Man Xue
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Qi-Yu Chen
- CAS Key Laboratory of Brain Connectome and Manipulation, Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Wantong Shi
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhaoxiang Zhou
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xuhui Li
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Fang Xu
- CAS Key Laboratory of Brain Connectome and Manipulation, Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Guoqiang Bi
- CAS Key Laboratory of Brain Connectome and Manipulation, Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xixiao Yang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jing-Shan Lu
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Min Zhuo
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
26
|
Chen K, Gupta R, Martín‐Ávila A, Cui M, Xie Z, Yang G. Anesthesia-induced hippocampal-cortical hyperactivity and tau hyperphosphorylation impair remote memory retrieval in Alzheimer's disease. Alzheimers Dement 2024; 20:494-510. [PMID: 37695022 PMCID: PMC10843666 DOI: 10.1002/alz.13464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/25/2023] [Accepted: 08/16/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Anesthesia often exacerbates memory recall difficulties in individuals with Alzheimer's disease (AD), but the underlying mechanisms remain unclear. METHODS We used in vivo Ca2+ imaging, viral-based circuit tracing, and chemogenetic approaches to investigate anesthesia-induced remote memory impairment in mouse models of presymptomatic AD. RESULTS Our study identified pyramidal neuron hyperactivity in the anterior cingulate cortex (ACC) as a significant contributor to anesthesia-induced remote memory impairment. This ACC hyperactivation arises from the disinhibition of local inhibitory circuits and increased excitatory inputs from the hippocampal CA1 region. Inhibiting hyperactivity in the CA1-ACC circuit improved memory recall after anesthesia. Moreover, anesthesia led to increased tau phosphorylation in the hippocampus, and inhibiting this hyperphosphorylation prevented ACC hyperactivity and subsequent memory impairment. DISCUSSION Hippocampal-cortical hyperactivity plays a role in anesthesia-induced remote memory impairment. Targeting tau hyperphosphorylation shows promise as a therapeutic strategy to mitigate anesthesia-induced neural network dysfunction and retrograde amnesia in AD.
Collapse
Affiliation(s)
- Kai Chen
- Department of AnesthesiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Riya Gupta
- Barnard College of Columbia UniversityNew YorkNew YorkUSA
| | | | - Meng Cui
- Department of BiologyPurdue UniversityWest LafayetteIndianaUSA
| | - Zhongcong Xie
- Geriatric Anesthesia Research UnitDepartment of AnesthesiaCritical Care and Pain MedicineMassachusetts General Hospital and Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Guang Yang
- Department of AnesthesiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
27
|
Yao D, Chen Y, Chen G. The role of pain modulation pathway and related brain regions in pain. Rev Neurosci 2023; 34:899-914. [PMID: 37288945 DOI: 10.1515/revneuro-2023-0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/18/2023] [Indexed: 06/09/2023]
Abstract
Pain is a multifaceted process that encompasses unpleasant sensory and emotional experiences. The essence of the pain process is aversion, or perceived negative emotion. Central sensitization plays a significant role in initiating and perpetuating of chronic pain. Melzack proposed the concept of the "pain matrix", in which brain regions associated with pain form an interconnected network, rather than being controlled by a singular brain region. This review aims to investigate distinct brain regions involved in pain and their interconnections. In addition, it also sheds light on the reciprocal connectivity between the ascending and descending pathways that participate in pain modulation. We review the involvement of various brain areas during pain and focus on understanding the connections among them, which can contribute to a better understanding of pain mechanisms and provide opportunities for further research on therapies for improved pain management.
Collapse
Affiliation(s)
- Dandan Yao
- Department of Anesthesiology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Gang Chen
- Department of Anesthesiology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
28
|
Chen C, Tassou A, Morales V, Scherrer G. Graph theory analysis reveals an assortative pain network vulnerable to attacks. Sci Rep 2023; 13:21985. [PMID: 38082002 PMCID: PMC10713541 DOI: 10.1038/s41598-023-49458-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/08/2023] [Indexed: 12/18/2023] Open
Abstract
The neural substrate of pain experience has been described as a dense network of connected brain regions. However, the connectivity pattern of these brain regions remains elusive, precluding a deeper understanding of how pain emerges from the structural connectivity. Here, we employ graph theory to systematically characterize the architecture of a comprehensive pain network, including both cortical and subcortical brain areas. This structural brain network consists of 49 nodes denoting pain-related brain areas, linked by edges representing their relative incoming and outgoing axonal projection strengths. Within this network, 63% of brain areas share reciprocal connections, reflecting a dense network. The clustering coefficient, a measurement of the probability that adjacent nodes are connected, indicates that brain areas in the pain network tend to cluster together. Community detection, the process of discovering cohesive groups in complex networks, successfully reveals two known subnetworks that specifically mediate the sensory and affective components of pain, respectively. Assortativity analysis, which evaluates the tendency of nodes to connect with other nodes that have similar features, indicates that the pain network is assortative. Finally, robustness, the resistance of a complex network to failures and perturbations, indicates that the pain network displays a high degree of error tolerance (local failure rarely affects the global information carried by the network) but is vulnerable to attacks (selective removal of hub nodes critically changes network connectivity). Taken together, graph theory analysis unveils an assortative structural pain network in the brain that processes nociceptive information. Furthermore, the vulnerability of this network to attack presents the possibility of alleviating pain by targeting the most connected brain areas in the network.
Collapse
Affiliation(s)
- Chong Chen
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Valentina Morales
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- New York Stem Cell Foundation ‒ Robertson Investigator, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
29
|
Xu Y, Zhu X, Chen Y, Chen Y, Zhu Y, Xiao S, Wu M, Wang Y, Zhang C, Wu Z, He X, Liu B, Shen Z, Shao X, Fang J. Electroacupuncture alleviates mechanical allodynia and anxiety-like behaviors induced by chronic neuropathic pain via regulating rostral anterior cingulate cortex-dorsal raphe nucleus neural circuit. CNS Neurosci Ther 2023; 29:4043-4058. [PMID: 37401033 PMCID: PMC10651964 DOI: 10.1111/cns.14328] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/02/2023] [Accepted: 06/17/2023] [Indexed: 07/05/2023] Open
Abstract
AIMS Epidemiological studies in patients with neuropathic pain have demonstrated a strong association between neuropathic pain and psychiatric conditions such as anxiety. Preclinical and clinical work has demonstrated that electroacupuncture (EA) effectively alleviates anxiety-like behaviors induced by chronic neuropathic pain. In this study, a potential neural circuitry underlying the therapeutic action of EA was investigated. METHODS The effects of EA stimulation on mechanical allodynia and anxiety-like behaviors in animal models of spared nerve injury (SNI) were examined. EA plus chemogenetic manipulation of glutamatergic (Glu) neurons projecting from the rostral anterior cingulate cortex (rACCGlu ) to the dorsal raphe nucleus (DRN) was used to explore the changes of mechanical allodynia and anxiety-like behaviors in SNI mice. RESULTS Electroacupuncture significantly alleviated both mechanical allodynia and anxiety-like behaviors with increased activities of glutamatergic neurons in the rACC and serotoninergic neurons in the DRN. Chemogenetic activation of the rACCGlu -DRN projections attenuated both mechanical allodynia and anxiety-like behaviors in mice at day 14 after SNI. Chemogenetic inhibition of the rACCGlu -DRN pathway did not induce mechanical allodynia and anxiety-like behaviors under physiological conditions, but inhibiting this pathway produced anxiety-like behaviors in mice at day 7 after SNI; this effect was reversed by EA. EA plus activation of the rACCGlu -DRN circuit did not produce a synergistic effect on mechanical allodynia and anxiety-like behaviors. The analgesic and anxiolytic effects of EA could be blocked by inhibiting the rACCGlu -DRN pathway. CONCLUSIONS The role of rACCGlu -DRN circuit may be different during the progression of chronic neuropathic pain and these changes may be related to the serotoninergic neurons in the DRN. These findings describe a novel rACCGlu -DRN pathway through which EA exerts analgesic and anxiolytic effects in SNI mice exhibiting anxiety-like behaviors.
Collapse
Affiliation(s)
- Yingling Xu
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain‐Machine Integration, School of Brain Science and Brain MedicineZhejiang UniversityHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
| | - Xixiao Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yuerong Chen
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yeqing Chen
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yichen Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Siqi Xiao
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Mengwei Wu
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yifang Wang
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Chi Zhang
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Zenmin Wu
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Boyu Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Zui Shen
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaomei Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceThe Third Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
30
|
Jiang X, Kuang H, Lv H, Xiong J, Li J, Hong S, Yan YI, Gu L, Jiang J. Aberrant functional and causal connectivity of the amygdala in herpes zoster and post-herpetic neuralgia patients. Br J Radiol 2023; 96:20230338. [PMID: 37750852 PMCID: PMC10646639 DOI: 10.1259/bjr.20230338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/08/2023] [Accepted: 08/10/2023] [Indexed: 09/27/2023] Open
Abstract
OBJECTIVE Resting-state functional magnetic resonance imaging (rs-fMRI) and Granger causality analysis (GCA) were used to observe the characteristics of amygdala and whole-brain effect connections in patients with herpes zoster (HZ) and post-herpetic neuralgia (PHN) and to determine their relationship with clinical features. METHODS Rs-fMRI scans were performed on 50 HZ; 50 PHN; and 50 age-, sex- and education-year-matched healthy controls (HCs). Bilateral amygdala subregions were used as seeds for functional connectivity (FC). GCA was used to analyze the effective connection of brain regions that were significantly different among groups. Then, the correlation between FC, and GCA values and clinical indices was investigated. RESULTS PHN had impaired FC between the amygdala subregion with the putamen, cortex, anterior cingulate cortex (ACC) to HCs and reduced FC of medial amygdala (MeA) with the parieto-occipital lobe and motor cortex to HZ; HZ had reduced FC of the lateral amygdala (LA) with the insula to HCs. GCA values from the bilateral LA to the bilateral ACC, left MeA to the bilateral ACC and left putamen, and right ACC to the bilateral MeA were reduced in PHN patients compared to HCs. Compared with HCs, the GCA values from the left MeA to the left ACC and right putamen were reduced in HZ. The GCA values from the amygdala subregion to the ACC were positively correlated with HAMA or HAMD scores in PHN. CONCLUSION PHN showed reduced FC between the amygdala subregions and cortico-putamen and decreased effective connectivity from the amygdala subregion to the ACC and putamen. ADVANCES IN KNOWLEDGE HZ and PHN patients had significant changes in effective connectivity in brain regions, including diverse functional areas emanating from and projecting to the amygdala. The current findings will provide a new perspective for understanding the neuropathophysiological mechanism HZ and PHN.
Collapse
Affiliation(s)
| | | | | | | | | | | | - YI Yan
- Department of Pain, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Lili Gu
- Department of Pain, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | | |
Collapse
|
31
|
Boeken OJ, Cieslik EC, Langner R, Markett S. Characterizing functional modules in the human thalamus: coactivation-based parcellation and systems-level functional decoding. Brain Struct Funct 2023; 228:1811-1834. [PMID: 36547707 PMCID: PMC10516793 DOI: 10.1007/s00429-022-02603-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
The human thalamus relays sensory signals to the cortex and facilitates brain-wide communication. The thalamus is also more directly involved in sensorimotor and various cognitive functions but a full characterization of its functional repertoire, particularly in regard to its internal anatomical structure, is still outstanding. As a putative hub in the human connectome, the thalamus might reveal its functional profile only in conjunction with interconnected brain areas. We therefore developed a novel systems-level Bayesian reverse inference decoding that complements the traditional neuroinformatics approach towards a network account of thalamic function. The systems-level decoding considers the functional repertoire (i.e., the terms associated with a brain region) of all regions showing co-activations with a predefined seed region in a brain-wide fashion. Here, we used task-constrained meta-analytic connectivity-based parcellation (MACM-CBP) to identify thalamic subregions as seed regions and applied the systems-level decoding to these subregions in conjunction with functionally connected cortical regions. Our results confirm thalamic structure-function relationships known from animal and clinical studies and revealed further associations with language, memory, and locomotion that have not been detailed in the cognitive neuroscience literature before. The systems-level decoding further uncovered large systems engaged in autobiographical memory and nociception. We propose this novel decoding approach as a useful tool to detect previously unknown structure-function relationships at the brain network level, and to build viable starting points for future studies.
Collapse
Affiliation(s)
- Ole J Boeken
- Faculty of Life Sciences, Department of Molecular Psychology, Humboldt-Universität Zu Berlin, Rudower Chaussee 18, 12489, Berlin, Germany.
| | - Edna C Cieslik
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour), Research Centre Jülich, Jülich, Germany
| | - Robert Langner
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour), Research Centre Jülich, Jülich, Germany
| | - Sebastian Markett
- Faculty of Life Sciences, Department of Molecular Psychology, Humboldt-Universität Zu Berlin, Rudower Chaussee 18, 12489, Berlin, Germany
| |
Collapse
|
32
|
Valentinova K, Acuña MA, Ntamati NR, Nevian NE, Nevian T. An amygdala-to-cingulate cortex circuit for conflicting choices in chronic pain. Cell Rep 2023; 42:113125. [PMID: 37733589 PMCID: PMC10636611 DOI: 10.1016/j.celrep.2023.113125] [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: 12/22/2022] [Revised: 07/12/2023] [Accepted: 08/28/2023] [Indexed: 09/23/2023] Open
Abstract
Chronic pain is a complex experience with multifaceted behavioral manifestations, often leading to pain avoidance at the expense of reward approach. How pain facilitates avoidance in situations with mixed outcomes is unknown. The anterior cingulate cortex (ACC) plays a key role in pain processing and in value-based decision-making. Distinct ACC inputs inform about the sensory and emotional quality of pain. However, whether specific ACC circuits underlie pathological conflict assessment in pain remains underexplored. Here, we demonstrate that mice with chronic pain favor cold avoidance rather than reward approach in a conflicting task. This occurs along with selective strengthening of basolateral amygdala inputs onto ACC layer 2/3 pyramidal neurons. The amygdala-cingulate projection is necessary and sufficient for the conflicting cold avoidance. Further, low-frequency stimulation of this pathway restores AMPA receptor function and reduces avoidance in pain mice. Our findings provide insights into the circuits and mechanisms underlying cognitive aspects of pain and offer potential targets for treatment.
Collapse
Affiliation(s)
- Kristina Valentinova
- Department of Physiology, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland.
| | - Mario A Acuña
- Department of Physiology, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Niels R Ntamati
- Department of Physiology, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Natalie E Nevian
- Department of Physiology, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Thomas Nevian
- Department of Physiology, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland.
| |
Collapse
|
33
|
Hervert EA, Birdsong W. The opioid peptide met-enkephalin modulates thalamo-cortical excitation inhibition balance in a medial thalamus-anterior cingulate cortex circuit. Neuropharmacology 2023; 242:109785. [PMID: 39491147 DOI: 10.1016/j.neuropharm.2023.109785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Activation of opioid receptors in the anterior cingulate cortex (ACC) mediates aspects of analgesia induced by both exogenous and endogenous opioids. We have previously shown that opioid signaling disrupts both afferent excitatory and indirect inhibitory synaptic transmission from the medial thalamus (MThal) to the ACC, but the effects of non-selective opioid peptides such as [Met]5-enkephalin (ME) within this circuit remain poorly understood. The goal of the current study was to understand how ME modulates thalamic-driven excitatory and inhibitory synaptic transmission onto layer V pyramidal neurons in the ACC. We used pharmacology, brain slice electrophysiology and optogenetic stimulation to study opioid-mediated modulation of optically evoked glutamatergic and GABAergic transmission. The results revealed that bath perfused ME inhibited both AMPA-mediated excitatory and GABA-mediated inhibitory synaptic transmission in the ACC. However, inhibitory transmission was more potently inhibited than excitatory transmission by ME. This preferential reduction in GABAA-mediated synaptic transmission was primarily due to the activation of delta opioid receptors by ME and resulted in a net disinhibition of MThal-ACC excitatory pathway. These results suggest that moderate concentrations of ME can lead to a net increase in excitatory drive of ACC circuitry and that analgesia may be associated with disinhibition rather than inhibition of ACC subcircuits.
Collapse
Affiliation(s)
- Erwin Arias Hervert
- Department of Pharmacology, University of Michigan. Ann Arbor, Michigan, USA.
| | - William Birdsong
- Department of Pharmacology, University of Michigan. Ann Arbor, Michigan, USA.
| |
Collapse
|
34
|
Duan Y, Li Q, Zhou Y, Chen S, Li Y, Zang Y. Activation of the TNF-α-Necroptosis Pathway in Parvalbumin-Expressing Interneurons of the Anterior Cingulate Cortex Contributes to Neuropathic Pain. Int J Mol Sci 2023; 24:15454. [PMID: 37895135 PMCID: PMC10607712 DOI: 10.3390/ijms242015454] [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: 09/05/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
The hyperexcitability of the anterior cingulate cortex (ACC) has been implicated in the development of chronic pain. As one of the key causes of ACC hyperexcitation, disinhibition of the ACC may be closely related to the dysfunction of inhibitory parvalbumin (PV)-expressing interneurons (PV-INs). However, the molecular mechanism underlying the ACC PV-INs injury remains unclear. The present study demonstrates that spared sciatic nerve injury (SNI) induces an imbalance in the excitation and inhibition (E/I) of the ACC. To test whether tumor necrosis factor-α (TNF-α) upregulation in the ACC after SNI activates necroptosis and participates in PV-INs damage, we performed a differential analysis of transcriptome sequencing using data from neuropathic pain models and found that the expression of genes key to the TNF-α-necroptosis pathway were upregulated. TNF-α immunoreactivity (IR) signals in the ACCs of SNI rats were co-located with p-RIP3- and PV-IR, or p-MLKL- and PV-IR signals. We then systematically detected the expression and cell localization of necroptosis-related proteins, including kinase RIP1, RIP3, MLKL, and their phosphorylated states, in the ACC of SNI rats. Except for RIP1 and MLKL, the levels of these proteins were significantly elevated in the contralateral ACC and mainly expressed in PV-INs. Blocking the ACC TNF-α-necroptosis pathway by microinjecting TNF-α neutralizing antibody or using an siRNA knockdown to block expression of MLKL in the ACC alleviated SNI-induced pain hypersensitivity and inhibited the upregulation of TNF-α and p-MLKL. Targeting TNF-α-triggered necroptosis within ACC PV-INs may help to correct PV-INs injury and E/I imbalance in the ACC in neuropathic pain.
Collapse
Affiliation(s)
- Yiwen Duan
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| | - Qiaoyun Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| | - Yaohui Zhou
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| | - Shaoxia Chen
- State Key Laboratory of Oncology in South China, Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China;
| | - Yongyong Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| | - Ying Zang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Pain Research Center, Department of Physiology, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; (Y.D.); (Q.L.); (Y.Z.); (Y.L.)
| |
Collapse
|
35
|
Journée SH, Mathis VP, Fillinger C, Veinante P, Yalcin I. Janus effect of the anterior cingulate cortex: Pain and emotion. Neurosci Biobehav Rev 2023; 153:105362. [PMID: 37595650 DOI: 10.1016/j.neubiorev.2023.105362] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/20/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Over the past 20 years, clinical and preclinical studies point to the anterior cingulate cortex (ACC) as a site of interest for several neurological and psychiatric conditions. The ACC plays a critical role in emotion, autonomic regulation, pain processing, attention, memory and decision making. An increasing number of studies have demonstrated the involvement of the ACC in the emotional component of pain and its comorbidity with emotional disorders such as anxiety and depression. Thanks to the development of animal models combined with state-of-the-art technologies, we now have a better mechanistic understanding of the functions of the ACC. Hence, the primary aim of this review is to compile the most recent preclinical studies on the role of ACC in the emotional component and consequences of chronic pain. Herein, we thus thoroughly describe the pain-induced electrophysiological, molecular and anatomical alterations in the ACC and in its related circuits. Finally, we discuss the next steps that are needed to strengthen our understanding of the involvement of the ACC in emotional and pain processing.
Collapse
Affiliation(s)
- Sarah H Journée
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Victor P Mathis
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Clémentine Fillinger
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Pierre Veinante
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France; Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
36
|
Gulledge AT. Cholinergic activation of corticofugal circuits in the adult mouse prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538437. [PMID: 37163128 PMCID: PMC10168390 DOI: 10.1101/2023.04.28.538437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
In layer 5 of the neocortex, ACh promotes cortical output to the thalamus and brainstem by preferentially enhancing the postsynaptic excitability of pyramidal tract (PT) neurons relative to neighboring intratelencephalic (IT) neurons. Less is known about how ACh regulates the excitatory synaptic drive of IT and PT neurons. To address this question, spontaneous excitatory postsynaptic potentials (sEPSPs) were recorded in pairs of IT and PT neurons in slices of prelimbic cortex from adult female and male mice. ACh (20 µM) enhanced sEPSP amplitudes, frequencies, rise-times, and half-widths preferentially in PT neurons. These effects were blocked by the muscarinic acetylcholine receptor antagonist atropine (1 µM). When challenged with pirenzepine (1 µM), an antagonist selective for M1-type muscarinic receptors, ACh instead reduced sEPSP frequencies. The cholinergic increase in sEPSP amplitudes and frequencies in PT neurons was not sensitive to blockade of GABAergic receptors with gabazine (10 µM) and CGP52432 (2.5 µM), but was blocked by tetrodotoxin (1 µM), suggesting that ACh enhances action-potential-dependent excitatory synaptic transmission in PT neurons. ACh also preferentially promoted the occurrence of synchronous sEPSPs in pairs of PT neurons relative to IT-PT and IT-IT pairs. Finally, selective chemogenetic silencing of hM4Di-expressing PT, but not IT, neurons with clozapine-N-oxide (5 µM) blocked cholinergic enhancement of sEPSP amplitudes and frequencies in PT neurons. These data suggest that, in addition to enhancing the postsynaptic excitability of PT neurons, M1 receptor activation promotes corticofugal output by preferentially amplifying recurrent excitation within networks of PT neurons.
Collapse
Affiliation(s)
- Allan T Gulledge
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College 74 College Street, Vail 601, Hanover, New Hampshire 03755, USA
| |
Collapse
|
37
|
Hervert EA, Birdsong W. The Endogenous Opioid Met-Enkephalin Modulates Thalamo-Cortical Excitation Inhibition Balance in a Medial Thalamus-Anterior Cingulate Cortex Circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.13.547220. [PMID: 37503144 PMCID: PMC10369945 DOI: 10.1101/2023.07.13.547220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Activation of opioid receptors in the anterior cingulate cortex (ACC) mediates aspects of analgesia induced by both exogenous and endogenous opioids. We have previously shown that opioid signaling disrupts both afferent excitatory and indirect inhibitory synaptic transmission from the medial thalamus (MThal) to the ACC, but the effects of endogenous opioids within this circuit remain poorly understood. The goal of the current study was to understand how the endogenous opioid, [Met]5-enkephalin (ME), modulates thalamic-driven excitatory and inhibitory synaptic transmission onto layer V pyramidal neurons in the ACC. We used pharmacology, brain slice electrophysiology and optogenetic stimulation to study opioid-mediated modulation of optically evoked glutamatergic and GABAergic transmission. The results revealed that ME inhibited both AMPA-mediated excitatory and GABA-mediated inhibitory synaptic transmission in the ACC. However, inhibitory transmission was more potently inhibited than excitatory transmission by ME. This preferential reduction in GABAA-mediated synaptic transmission was primarily due to the activation of delta opioid receptors by ME and resulted in a net disinhibition of MThal-ACC excitatory pathway. These results suggest that moderate concentrations of ME can lead to net excitation of ACC circuitry and that analgesia may be associated with disinhibition rather than inhibition of ACC subcircuits.
Collapse
Affiliation(s)
| | - William Birdsong
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
38
|
Darvish-Ghane S, Baumbach J, Martin LJ. Influence of Inflammatory Pain and Dopamine on Synaptic Transmission in the Mouse ACC. Int J Mol Sci 2023; 24:11113. [PMID: 37446289 DOI: 10.3390/ijms241311113] [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: 06/07/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Dopamine (DA) inhibits excitatory synaptic transmission in the anterior cingulate cortex (ACC), a brain region involved in the sensory and affective processing of pain. However, the DA modulation of inhibitory synaptic transmission in the ACC and its alteration of the excitatory/inhibitory (E/I) balance remains relatively understudied. Using patch-clamp recordings, we demonstrate that neither DA applied directly to the tissue slice nor complete Freund's adjuvant (CFA) injected into the hind paw significantly impacted excitatory currents (eEPSCs) in the ACC, when recorded without pharmacological isolation. However, individual neurons exhibited varied responses to DA, with some showing inhibition, potentiation, or no response. The degree of eEPSC inhibition by DA was higher in naïve slices compared to that in the CFA condition. The baseline inhibitory currents (eIPSCs) were greater in the CFA-treated slices, and DA specifically inhibited eIPSCs in the CFA-treated, but not naïve group. DA and CFA treatment did not alter the balance between excitatory and inhibitory currents. Spontaneous synaptic activity revealed that DA reduced the frequency of the excitatory currents in CFA-treated mice and decreased the amplitude of the inhibitory currents, specifically in CFA-treated mice. However, the overall synaptic drive remained similar between the naïve and CFA-treated mice. Additionally, GABAergic currents were pharmacologically isolated and found to be robustly inhibited by DA through postsynaptic D2 receptors and G-protein activity. Overall, the study suggests that CFA-induced inflammation and DA do not significantly affect the balance between excitatory and inhibitory currents in ACC neurons, but activity-dependent changes may be observed in the DA modulation of presynaptic glutamate release in the presence of inflammation.
Collapse
Affiliation(s)
- Soroush Darvish-Ghane
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jennet Baumbach
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Loren J Martin
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
39
|
Song ZH, Song XJ, Yang CL, Cao P, Mao Y, Jin Y, Xu MY, Wang HT, Zhu X, Wang W, Zhang Z, Tao WJ. Up-regulation of microglial chemokine CXCL12 in anterior cingulate cortex mediates neuropathic pain in diabetic mice. Acta Pharmacol Sin 2023; 44:1337-1349. [PMID: 36697977 PMCID: PMC10310783 DOI: 10.1038/s41401-022-01046-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/23/2022] [Indexed: 01/26/2023] Open
Abstract
Diabetic patients frequently experience neuropathic pain, which currently lacks effective treatments. The mechanisms underlying diabetic neuropathic pain remain unclear. The anterior cingulate cortex (ACC) is well-known to participate in the processing and transformation of pain information derived from internal and external sensory stimulation. Accumulating evidence shows that dysfunction of microglia in the central nervous system contributes to many diseases, including chronic pain and neurodegenerative diseases. In this study, we investigated the role of microglial chemokine CXCL12 and its neuronal receptor CXCR4 in diabetic pain development in a mouse diabetic model established by injection of streptozotocin (STZ). Pain sensitization was assessed by the left hindpaw pain threshold in von Frey filament test. Iba1+ microglia in ACC was examined using combined immunohistochemistry and three-dimensional reconstruction. The activity of glutamatergic neurons in ACC (ACCGlu) was detected by whole-cell recording in ACC slices from STZ mice, in vivo multi-tetrode electrophysiological and fiber photometric recordings. We showed that microglia in ACC was significantly activated and microglial CXCL12 expression was up-regulated at the 7-th week post-injection, resulting in hyperactivity of ACCGlu and pain sensitization. Pharmacological inhibition of microglia or blockade of CXCR4 in ACC by infusing minocycline or AMD3100 significantly alleviated diabetic pain through preventing ACCGlu hyperactivity in STZ mice. In addition, inhibition of microglia by infusing minocycline markedly decreased STZ-induced upregulation of microglial CXCL12. Together, this study demonstrated that microglia-mediated ACCGlu hyperactivity drives the development of diabetic pain via the CXCL12/CXCR4 signaling, thus revealing viable therapeutic targets for the treatment of diabetic pain.
Collapse
Affiliation(s)
- Zi-Hua Song
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100071, China
| | - Xiang-Jie Song
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Chen-Ling Yang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230022, China
- College & Hospital of stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230022, China
| | - Peng Cao
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yu Mao
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yan Jin
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Meng-Yun Xu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Hai-Tao Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Xia Zhu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Wei Wang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Zhi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Wen-Juan Tao
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230022, China.
- College & Hospital of stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230022, China.
| |
Collapse
|
40
|
Qiu XT, Guo C, Ma LT, Li XN, Zhang QY, Huang FS, Zhang MM, Bai Y, Liang GB, Li YQ. Transcriptomic and proteomic profiling of the anterior cingulate cortex in neuropathic pain model rats. Front Mol Neurosci 2023; 16:1164426. [PMID: 37396788 PMCID: PMC10311218 DOI: 10.3389/fnmol.2023.1164426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
Background Neuropathic pain (NP) takes a heavy toll on individual life quality, yet gaps in its molecular characterization persist and effective therapy is lacking. This study aimed to provide comprehensive knowledge by combining transcriptomic and proteomic data of molecular correlates of NP in the anterior cingulate cortex (ACC), a cortical hub responsible for affective pain processing. Methods The NP model was established by spared nerve injury (SNI) in Sprague-Dawley rats. RNA sequencing and proteomic data from the ACC tissue isolated from sham and SNI rats 2 weeks after surgery were integrated to compare their gene and protein expression profiles. Bioinformatic analyses were performed to figure out the functions and signaling pathways of the differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) enriched in. Results Transcriptomic analysis identified a total of 788 DEGs (with 49 genes upregulated) after SNI surgery, while proteomic analysis found 222 DEPs (with 89 proteins upregulated). While Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses of the DEGs suggested that most of the altered genes were involved in synaptic transmission and plasticity, bioinformatics analysis of the DEPs revealed novel critical pathways associated with autophagy, mitophagy, and peroxisome. Notably, we noticed functionally important NP-related changes in the protein that occurred in the absence of corresponding changes at the level of transcription. Venn diagram analysis of the transcriptomic and proteomic data identified 10 overlapping targets, among which only three genes (XK-related protein 4, NIPA-like domain-containing 3, and homeodomain-interacting protein kinase 3) showed concordance in the directions of change and strong correlations between mRNA and protein levels. Conclusion The present study identified novel pathways in the ACC in addition to confirming previously reported mechanisms for NP etiology, and provided novel mechanistic insights for future research on NP treatment. These findings also imply that mRNA profiling alone fails to provide a complete landscape of molecular pain in the ACC. Therefore, explorations of changes at the level of protein are necessary to understand NP processes that are not transcriptionally modulated.
Collapse
Affiliation(s)
- Xin-Tong Qiu
- Department of Anatomy, Histology and Embryology, Preclinical School of Medicine, Air Force Medical University, Xi’an, China
| | - Chen Guo
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Li-Tian Ma
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Xin-Ning Li
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Qi-Yan Zhang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Fen-Sheng Huang
- Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Ming-Ming Zhang
- Department of Anatomy, Histology and Embryology, Preclinical School of Medicine, Air Force Medical University, Xi’an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Guo-Biao Liang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology, Preclinical School of Medicine, Air Force Medical University, Xi’an, China
- Department of Geriatrics, Tangdu Hospital, Air Force Medical University, Xi’an, China
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi, China
- Department of Anatomy, College of Basic Medicine, Dali University, Dali, China
| |
Collapse
|
41
|
Song XJ, Yang CL, Chen D, Yang Y, Mao Y, Cao P, Jiang A, Wang W, Zhang Z, Tao W. Up-regulation of LCN2 in the anterior cingulate cortex contributes to neural injury-induced chronic pain. Front Cell Neurosci 2023; 17:1140769. [PMID: 37362002 PMCID: PMC10285483 DOI: 10.3389/fncel.2023.1140769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/02/2023] [Indexed: 06/28/2023] Open
Abstract
Chronic pain caused by disease or injury affects more than 30% of the general population. The molecular and cellular mechanisms underpinning the development of chronic pain remain unclear, resulting in scant effective treatments. Here, we combined electrophysiological recording, in vivo two-photon (2P) calcium imaging, fiber photometry, Western blotting, and chemogenetic methods to define a role for the secreted pro-inflammatory factor, Lipocalin-2 (LCN2), in chronic pain development in mice with spared nerve injury (SNI). We found that LCN2 expression was upregulated in the anterior cingulate cortex (ACC) at 14 days after SNI, resulting in hyperactivity of ACC glutamatergic neurons (ACCGlu) and pain sensitization. By contrast, suppressing LCN2 protein levels in the ACC with viral constructs or exogenous application of neutralizing antibodies leads to significant attenuation of chronic pain by preventing ACCGlu neuronal hyperactivity in SNI 2W mice. In addition, administering purified recombinant LCN2 protein in the ACC could induce pain sensitization by inducing ACCGlu neuronal hyperactivity in naïve mice. This study provides a mechanism by which LCN2-mediated hyperactivity of ACCGlu neurons contributes to pain sensitization, and reveals a new potential target for treating chronic pain.
Collapse
Affiliation(s)
- Xiang-Jie Song
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chen-Ling Yang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Danyang Chen
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yumeng Yang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yu Mao
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Peng Cao
- Department of Neurology, Stroke Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Aijun Jiang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhi Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenjuan Tao
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
42
|
Seymour B, Crook RJ, Chen ZS. Post-injury pain and behaviour: a control theory perspective. Nat Rev Neurosci 2023; 24:378-392. [PMID: 37165018 PMCID: PMC10465160 DOI: 10.1038/s41583-023-00699-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2023] [Indexed: 05/12/2023]
Abstract
Injuries of various types occur commonly in the lives of humans and other animals and lead to a pattern of persistent pain and recuperative behaviour that allows safe and effective recovery. In this Perspective, we propose a control-theoretic framework to explain the adaptive processes in the brain that drive physiological post-injury behaviour. We set out an evolutionary and ethological view on how animals respond to injury, illustrating how the behavioural state associated with persistent pain and recuperation may be just as important as phasic pain in ensuring survival. Adopting a normative approach, we suggest that the brain implements a continuous optimal inference of the current state of injury from diverse sensory and physiological signals. This drives the various effector control mechanisms of behavioural homeostasis, which span the modulation of ongoing motivation and perception to drive rest and hyper-protective behaviours. However, an inherent problem with this is that these protective behaviours may partially obscure information about whether injury has resolved. Such information restriction may seed a tendency to aberrantly or persistently infer injury, and may thus promote the transition to pathological chronic pain states.
Collapse
Affiliation(s)
- Ben Seymour
- Institute for Biomedical Engineering, University of Oxford, Oxford, UK.
- Wellcome Centre for Integrative Neuroimaging, John Radcliffe Hospital, Headington, Oxford, UK.
| | - Robyn J Crook
- Department of Biology, San Francisco State University, San Francisco, CA, USA.
| | - Zhe Sage Chen
- Departments of Psychiatry, Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY, USA.
- Interdisciplinary Pain Research Program, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
43
|
Pan Q, Guo SS, Chen M, Su XY, Gao ZL, Wang Q, Xu TL, Liu MG, Hu J. Representation and control of pain and itch by distinct prefrontal neural ensembles. Neuron 2023:S0896-6273(23)00342-2. [PMID: 37224813 DOI: 10.1016/j.neuron.2023.04.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 02/18/2023] [Accepted: 04/27/2023] [Indexed: 05/26/2023]
Abstract
Pain and itch are two closely related but essentially distinct sensations that elicit different behavioral responses. However, it remains mysterious how pain and itch information is encoded in the brain to produce differential perceptions. Here, we report that nociceptive and pruriceptive signals are separately represented and processed by distinct neural ensembles in the prelimbic (PL) subdivision of the medial prefrontal cortex (mPFC) in mice. Pain- and itch-responsive cortical neural ensembles were found to significantly differ in electrophysiological properties, input-output connectivity profiles, and activity patterns to nociceptive or pruriceptive stimuli. Moreover, these two groups of cortical neural ensembles oppositely modulate pain- or itch-related sensory and emotional behaviors through their preferential projections to specific downstream regions such as the mediodorsal thalamus (MD) and basolateral amygdala (BLA). These findings uncover separate representations of pain and itch by distinct prefrontal neural ensembles and provide a new framework for understanding somatosensory information processing in the brain.
Collapse
Affiliation(s)
- Qian Pan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Su-Shan Guo
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ming Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xin-Yu Su
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zi-Long Gao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qi Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Tian-Le Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Songjiang Hospital and Songjiang Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China.
| | - Ming-Gang Liu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai 200030, China.
| |
Collapse
|
44
|
Li JN, Wu XM, Zhao LJ, Sun HX, Hong J, Wu FL, Chen SH, Chen T, Li H, Dong YL, Li YQ. Central medial thalamic nucleus dynamically participates in acute itch sensation and chronic itch-induced anxiety-like behavior in male mice. Nat Commun 2023; 14:2539. [PMID: 37137899 PMCID: PMC10156671 DOI: 10.1038/s41467-023-38264-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/20/2023] [Indexed: 05/05/2023] Open
Abstract
Itch is an annoying sensation consisting of both sensory and emotional components. It is known to involve the parabrachial nucleus (PBN), but the following transmission nodes remain elusive. The present study identified that the PBN-central medial thalamic nucleus (CM)-medial prefrontal cortex (mPFC) pathway is essential for itch signal transmission at the supraspinal level in male mice. Chemogenetic inhibition of the CM-mPFC pathway attenuates scratching behavior or chronic itch-related affective responses. CM input to mPFC pyramidal neurons is enhanced in acute and chronic itch models. Specifically chronic itch stimuli also alter mPFC interneuron involvement, resulting in enhanced feedforward inhibition and a distorted excitatory/inhibitory balance in mPFC pyramidal neurons. The present work underscores CM as a transmit node of the itch signal in the thalamus, which is dynamically engaged in both the sensory and affective dimensions of itch with different stimulus salience.
Collapse
Affiliation(s)
- Jia-Ni Li
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xue-Mei Wu
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Human Anatomy, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Liu-Jie Zhao
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
| | - Han-Xue Sun
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Human Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Jie Hong
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Human Anatomy, Baotou Medical College Inner Mongolia University of Science and Technology, Baotou, 014040, China
| | - Feng-Ling Wu
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
| | - Si-Hai Chen
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
| | - Tao Chen
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Lin Dong
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China.
- Department of Human Anatomy, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China.
- Department of Human Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
- Department of Human Anatomy, Baotou Medical College Inner Mongolia University of Science and Technology, Baotou, 014040, China.
| |
Collapse
|
45
|
Cao P, Zhang M, Ni Z, Song XJ, Yang CL, Mao Y, Zhou W, Dong WY, Peng X, Zheng C, Zhang Z, Jin Y, Tao W. Green light induces antinociception via visual-somatosensory circuits. Cell Rep 2023; 42:112290. [PMID: 36947545 DOI: 10.1016/j.celrep.2023.112290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/19/2022] [Accepted: 03/03/2023] [Indexed: 03/23/2023] Open
Abstract
Light has been shown to relieve pain, but the underlying neural mechanisms remain unknown. Here, we show that low-intensity (200 lux) green light treatment exerts antinociceptive effects through a neural circuit from the visual cortex projecting to the anterior cingulate cortex (ACC) in mice. Specifically, viral tracing, in vivo two-photon calcium imaging, and fiber photometry recordings show that green light activated glutamatergic projections from the medial part of the secondary visual cortex (V2MGlu) to GABAergic neurons in the ACC, which drives inhibition of local glutamatergic neurons (V2MGlu→ACCGABA→Glu). Optogenetic or chemogenetic activation of the V2MGlu→ACCGABA→Glu circuit mimics green-light-induced antinociception in both neuropathic and inflammatory pain model mice. Artificial inhibition of ACC-projecting V2MGlu neurons abolishes the antinociception induced by green light. Taken together, our study shows the V2M-ACC circuit as a potential candidate mediating green-light-induced antinociceptive effects.
Collapse
Affiliation(s)
- Peng Cao
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Mingjun Zhang
- Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Ziyun Ni
- Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xiang-Jie Song
- Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Chen-Ling Yang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yu Mao
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Department of Anesthesiology and Department of Pain Management, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China
| | - Wenjie Zhou
- Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Wan-Ying Dong
- Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xiaoqi Peng
- Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Changjian Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Wannan Medical College, Wuhu 241002, China
| | - Zhi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China.
| | - Yan Jin
- Department of Biophysics and Neurobiology, Key Laboratory of Brain Function and Disease of Chinese Academy of Sciences, University of Science and Technology of China, Hefei 230027, China; Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China.
| | - Wenjuan Tao
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; College & Hospital of Stomatology, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
46
|
Weinrich JA, Liu CD, Jewell ME, Andolina CR, Bernstein MX, Benitez J, Rodriguez-Rosado S, Braz JM, Maze M, Nemenov MI, Basbaum AI. Paradoxical increases in anterior cingulate cortex activity during nitrous oxide-induced analgesia reveal a signature of pain affect. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.534475. [PMID: 37066151 PMCID: PMC10104003 DOI: 10.1101/2023.04.03.534475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
The general consensus is that increases in neuronal activity in the anterior cingulate cortex (ACC) contribute to pain's negative affect. Here, using in vivo imaging of neuronal calcium dynamics in mice, we report that nitrous oxide, a general anesthetic that reduces pain affect, paradoxically, increases ACC spontaneous activity. As expected, a noxious stimulus also increased ACC activity. However, as nitrous oxide increases baseline activity, the relative change in activity from pre-stimulus baseline was significantly less than the change in the absence of the general anesthetic. We suggest that this relative change in activity represents a neural signature of the affective pain experience. Furthermore, this signature of pain persists under general anesthesia induced by isoflurane, at concentrations in which the mouse is unresponsive. We suggest that this signature underlies the phenomenon of connected consciousness, in which use of the isolated forelimb technique revealed that pain percepts can persist in anesthetized patients.
Collapse
Affiliation(s)
- Jarret Ap Weinrich
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| | - Cindy D Liu
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94158, USA
| | - Madison E Jewell
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| | - Christopher R Andolina
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| | - Mollie X Bernstein
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jorge Benitez
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| | - Sian Rodriguez-Rosado
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| | - Joao M Braz
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
| | - Mervyn Maze
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA 94158, USA
| | - Mikhail I Nemenov
- Lasmed, Mountain View, CA 94043, USA
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Allan I Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94158, USA
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
47
|
Zhang Q, Hu S, Talay R, Xiao Z, Rosenberg D, Liu Y, Sun G, Li A, Caravan B, Singh A, Gould JD, Chen ZS, Wang J. A prototype closed-loop brain-machine interface for the study and treatment of pain. Nat Biomed Eng 2023; 7:533-545. [PMID: 34155354 PMCID: PMC9516430 DOI: 10.1038/s41551-021-00736-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/22/2021] [Indexed: 12/25/2022]
Abstract
Chronic pain is characterized by discrete pain episodes of unpredictable frequency and duration. This hinders the study of pain mechanisms and contributes to the use of pharmacological treatments associated with side effects, addiction and drug tolerance. Here, we show that a closed-loop brain-machine interface (BMI) can modulate sensory-affective experiences in real time in freely behaving rats by coupling neural codes for nociception directly with therapeutic cortical stimulation. The BMI decodes the onset of nociception via a state-space model on the basis of the analysis of online-sorted spikes recorded from the anterior cingulate cortex (which is critical for pain processing) and couples real-time pain detection with optogenetic activation of the prelimbic prefrontal cortex (which exerts top-down nociceptive regulation). In rats, the BMI effectively inhibited sensory and affective behaviours caused by acute mechanical or thermal pain, and by chronic inflammatory or neuropathic pain. The approach provides a blueprint for demand-based neuromodulation to treat sensory-affective disorders, and could be further leveraged for nociceptive control and to study pain mechanisms.
Collapse
Affiliation(s)
- Qiaosheng Zhang
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, USA
| | - Sile Hu
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Robert Talay
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, USA
| | - Zhengdong Xiao
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - David Rosenberg
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Yaling Liu
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, USA
| | - Guanghao Sun
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Anna Li
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, USA
| | - Bassir Caravan
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Amrita Singh
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, USA
| | - Jonathan D Gould
- College of Arts and Sciences, New York University, New York, NY, USA
| | - Zhe S Chen
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, New York University School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
| | - Jing Wang
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, New York University School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
48
|
Long DD, Zhang YZ, Liu A, Shen L, Wei HR, Lou QQ, Hu SS, Chen DY, Chai XQ, Wang D. Microglia sustain anterior cingulate cortex neuronal hyperactivity in nicotine-induced pain. J Neuroinflammation 2023; 20:81. [PMID: 36944965 PMCID: PMC10031886 DOI: 10.1186/s12974-023-02767-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/17/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Long-term smoking is a risk factor for chronic pain, and chronic nicotine exposure induces pain-like effects in rodents. The anterior cingulate cortex (ACC) has been demonstrated to be associated with pain and substance abuse. This study aims to investigate whether ACC microglia are altered in response to chronic nicotine exposure and their interaction with ACC neurons and subsequent nicotine-induced allodynia in mice. METHODS We utilized a mouse model that was fed nicotine water for 28 days. Brain slices of the ACC were collected for morphological analysis to evaluate the impacts of chronic nicotine on microglia. In vivo calcium imaging and whole-cell patch clamp were used to record the excitability of ACC glutamatergic neurons. RESULTS Compared to the vehicle control, the branch endpoints and the length of ACC microglial processes decreased in nicotine-treated mice, coinciding with the hyperactivity of glutamatergic neurons in the ACC. Inhibition of ACC glutamatergic neurons alleviated nicotine-induced allodynia and reduced microglial activation. On the other hand, reactive microglia sustain ACC neuronal excitability in response to chronic nicotine, and pharmacological inhibition of microglia by minocycline or liposome-clodronate reduces nicotine-induced allodynia. The neuron-microglia interaction in chronic nicotine-induced allodynia is mediated by increased expression of neuronal CX3CL1, which activates microglia by acting on CX3CR1 receptors on microglial cells. CONCLUSION Together, these findings underlie a critical role of ACC microglia in the maintenance of ACC neuronal hyperactivity and resulting nociceptive hypersensitivity in chronic nicotine-treated mice.
Collapse
Affiliation(s)
- Dan-Dan Long
- Pain Clinic, Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, China
| | - Yu-Zhuo Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
| | - An Liu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Liang Shen
- Pain Clinic, Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, China
| | - Hong-Rui Wei
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Qian-Qian Lou
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Shan-Shan Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, China
| | - Dan-Yang Chen
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Xiao-Qing Chai
- Pain Clinic, Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, China
| | - Di Wang
- Pain Clinic, Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, China.
| |
Collapse
|
49
|
Chen C, Tassou A, Morales V, Scherrer G. Graph theory analysis reveals an assortative pain network vulnerable to attacks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.08.531580. [PMID: 36945626 PMCID: PMC10028857 DOI: 10.1101/2023.03.08.531580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
The neural substrate of pain experience has been described as a dense network of connected brain regions. However, the connectivity pattern of these brain regions remains elusive, precluding a deeper understanding of how pain emerges from the structural connectivity. Here, we use graph theory to systematically characterize the architecture of a comprehensive pain network, including both cortical and subcortical brain areas. This structural brain network consists of 49 nodes denoting pain-related brain areas, linked by edges representing their relative incoming and outgoing axonal projection strengths. Sixty-three percent of brain areas in this structural pain network share reciprocal connections, reflecting a dense network. The clustering coefficient, a measurement of the probability that adjacent nodes are connected, indicates that brain areas in the pain network tend to cluster together. Community detection, the process of discovering cohesive groups in complex networks, successfully reveals two known subnetworks that specifically mediate the sensory and affective components of pain, respectively. Assortativity analysis, which evaluates the tendency of nodes to connect with other nodes with similar features, indicates that the pain network is assortative. Finally, robustness, the resistance of a complex network to failures and perturbations, indicates that the pain network displays a high degree of error tolerance (local failure rarely affects the global information carried by the network) but is vulnerable to attacks (selective removal of hub nodes critically changes network connectivity). Taken together, graph theory analysis unveils an assortative structural pain network in the brain processing nociceptive information, and the vulnerability of this network to attack opens up the possibility of alleviating pain by targeting the most connected brain areas in the network.
Collapse
|
50
|
Chen ZS. Hierarchical predictive coding in distributed pain circuits. Front Neural Circuits 2023; 17:1073537. [PMID: 36937818 PMCID: PMC10020379 DOI: 10.3389/fncir.2023.1073537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/07/2023] [Indexed: 03/06/2023] Open
Abstract
Predictive coding is a computational theory on describing how the brain perceives and acts, which has been widely adopted in sensory processing and motor control. Nociceptive and pain processing involves a large and distributed network of circuits. However, it is still unknown whether this distributed network is completely decentralized or requires networkwide coordination. Multiple lines of evidence from human and animal studies have suggested that the cingulate cortex and insula cortex (cingulate-insula network) are two major hubs in mediating information from sensory afferents and spinothalamic inputs, whereas subregions of cingulate and insula cortices have distinct projections and functional roles. In this mini-review, we propose an updated hierarchical predictive coding framework for pain perception and discuss its related computational, algorithmic, and implementation issues. We suggest active inference as a generalized predictive coding algorithm, and hierarchically organized traveling waves of independent neural oscillations as a plausible brain mechanism to integrate bottom-up and top-down information across distributed pain circuits.
Collapse
Affiliation(s)
- Zhe Sage Chen
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, United States
- Interdisciplinary Pain Research Program, NYU Langone Health, New York, NY, United States
| |
Collapse
|