1
|
Zhao W, Yu YM, Wang XY, Xia SH, Ma Y, Tang H, Tao M, Li H, Xu Z, Yang JX, Wu P, Zhang H, Ding HL, Cao JL. CRF regulates pain sensation by enhancement of corticoaccumbal excitatory synaptic transmission. Mol Psychiatry 2024; 29:2170-2184. [PMID: 38454083 DOI: 10.1038/s41380-024-02488-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 03/09/2024]
Abstract
Both peripheral and central corticotropin-releasing factor (CRF) systems have been implicated in regulating pain sensation. However, compared with the peripheral, the mechanisms underlying central CRF system in pain modulation have not yet been elucidated, especially at the neural circuit level. The corticoaccumbal circuit, a structure rich in CRF receptors and CRF-positive neurons, plays an important role in behavioral responses to stressors including nociceptive stimuli. The present study was designed to investigate whether and how CRF signaling in this circuit regulated pain sensation under physiological and pathological pain conditions. Our studies employed the viral tracing and circuit-, and cell-specific electrophysiological methods to label the CRF-containing circuit from the medial prefrontal cortex to the nucleus accumbens shell (mPFCCRF-NAcS) and record its neuronal propriety. Combining optogenetic and chemogenetic manipulation, neuropharmacological methods, and behavioral tests, we were able to precisely manipulate this circuit and depict its role in regulation of pain sensation. The current study found that the CRF signaling in the NAc shell (NAcS), but not NAc core, was necessary and sufficient for the regulation of pain sensation under physiological and pathological pain conditions. This process was involved in the CRF-mediated enhancement of excitatory synaptic transmission in the NAcS. Furthermore, we demonstrated that the mPFCCRF neurons monosynaptically connected with the NAcS neurons. Chronic pain increased the protein level of CRF in NAcS, and then maintained the persistent NAcS neuronal hyperactivity through enhancement of this monosynaptic excitatory connection, and thus sustained chronic pain behavior. These findings reveal a novel cell- and circuit-based mechanistic link between chronic pain and the mPFCCRF → NAcS circuit and provide a potential new therapeutic target for chronic pain.
Collapse
Affiliation(s)
- Weinan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yu-Mei Yu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xiao-Yi Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Department of Anesthesiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Sun-Hui Xia
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yu Ma
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Huimei Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Mingshu Tao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - He Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Zheng Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Peng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
2
|
Roy TK, Uniyal A, Tiwari V. Multifactorial pathways in burn injury-induced chronic pain: novel targets and their pharmacological modulation. Mol Biol Rep 2022; 49:12121-12132. [PMID: 35842856 DOI: 10.1007/s11033-022-07748-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022]
Abstract
Burn injuries are among the highly prevalent medical conditions worldwide that occur mainly in children, military veterans and victims of fire accidents. It is one of the leading causes of temporary as well as permanent disabilities in patients. Burn injuries are accompanied by pain that persists even after recovery from tissue damage which puts immense pressure on the healthcare system. The pathophysiology of burn pain is poorly understood due to its complex nature and lack of considerable preclinical and clinical shreds of evidence, that creates a substantial barrier to the development of new analgesics. Burns damage the skin layers supplied with nociceptors such as NAV1.7, TRPV1, and TRPA1. Burn injury-mediated co-localization and simultaneous activation of TRPA1 and TRPV1 in nociceptive primary afferent C-fibers which contributes to the development and maintenance of chronic pain. Burn injuries are accompanied by central sensitization, a key feature of pain pathophysiology mainly driven by a series of cascades involving aberrations in the glutamatergic system, microglial activation, release of neuropeptides, cytokines, and chemokines. Activation of p38 mitogen-activated protein kinase, altered endogenous opioid signaling, and distorted genomic expression are other pathophysiological factors responsible for the development and maintenance of burn pain. Here we discuss comprehensive literature on molecular mechanisms of burn pain and potential targets that could be translated into near future therapeutics.
Collapse
Affiliation(s)
- Tapas Kumar Roy
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, 221005, Varanasi, U.P, India
| | - Ankit Uniyal
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, 221005, Varanasi, U.P, India
| | - Vinod Tiwari
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, 221005, Varanasi, U.P, India.
| |
Collapse
|
3
|
Mousa SA, Khalefa BI, Shaqura M, Al-Madol M, Treskatsch S, Schäfer M. Superior control of inflammatory pain by corticotropin-releasing factor receptor 1 via opioid peptides in distinct pain-relevant brain areas. J Neuroinflammation 2022; 19:148. [PMID: 35705992 PMCID: PMC9199204 DOI: 10.1186/s12974-022-02498-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/26/2022] [Indexed: 08/30/2023] Open
Abstract
Background Under inflammatory conditions, the activation of corticotropin-releasing factor (CRF) receptor has been shown to inhibit pain through opioid peptide release from immune cells or neurons. CRF’s effects on human and animal pain modulation depend, however, on the distribution of its receptor subtypes 1 and 2 (CRF-R1 and CRF-R2) along the neuraxis of pain transmission. The objective of this study is to investigate the respective role of each CRF receptor subtype on centrally administered CRF-induced antinociception during inflammatory pain. Methods The present study investigated the role of intracerebroventricular (i.c.v.) CRF receptor agonists on nociception and the contribution of cerebral CRF-R1 and/or CRF-R2 subtypes in an animal model of Freund’s complete adjuvant (FCA)-induced hind paw inflammation. Methods used included behavioral experiments, immunofluorescence confocal analysis, and reverse transcriptase-polymerase chain reaction. Results Intracerebroventricular, but systemically inactive, doses of CRF elicited potent, dose-dependent antinociceptive effects in inflammatory pain which were significantly antagonized by i.c.v. CRF-R1-selective antagonist NBI 27914 (by approximately 60%) but less by CRF-R2-selective antagonist K41498 (by only 20%). In line with these findings, i.c.v. administration of CRF-R1 agonist stressin I produced superior control of inflammatory pain over CRF-R2 agonist urocortin-2. Intriguingly, i.c.v. opioid antagonist naloxone significantly reversed the CRF as well as CRF-R1 agonist-elicited pain inhibition. Consistent with existing evidence of high CRF concentrations in brain areas such as the thalamus, hypothalamus, locus coeruleus, and periaqueductal gray following its i.c.v. administration, double-immunofluorescence confocal microscopy demonstrated primarily CRF-R1-positive neurons that expressed opioid peptides in these pain-relevant brain areas. Finally, PCR analysis confirmed the predominant expression of the CRF-R1 over CRF-R2 in representative brain areas such as the hypothalamus. Conclusion Taken together, these findings suggest that CRF-R1 in opioid-peptide-containing brain areas plays an important role in the modulation of inflammatory pain and may be a useful therapeutic target for inflammatory pain control. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02498-8.
Collapse
Affiliation(s)
- Shaaban A Mousa
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| | - Baled I Khalefa
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.,Zoology Department, Faculty of Science, AL-Zintan University, Alzintan, Libya
| | - Mohammed Shaqura
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | | | - Sascha Treskatsch
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Michael Schäfer
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| |
Collapse
|
4
|
Glaeser JD, Tawackoli W, Ju DG, Yang JH, Kanim LEA, Salehi K, Yu V, Saidara E, Vit J, Khnkoyan Z, NaPier Z, Stone LS, Bae HW, Sheyn D. Optimization of a rat lumbar IVD degeneration model for low back pain. JOR Spine 2020; 3:e1092. [PMID: 32613167 PMCID: PMC7323460 DOI: 10.1002/jsp2.1092] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/16/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Intervertebral disc (IVD) degeneration is often associated with low back pain and radiating leg pain. The purpose of this study is to develop a reproducible and standardized preclinical model of painful lumbar IVD degeneration by evaluation of structural and behavioral changes in response to IVD injury with increasing needle sizes. This model can be used to develop new therapies for IVD degeneration. METHODS Forty-five female Sprague Dawley rats underwent anterior lumbar disc needle puncture at levels L4-5 and L5-6 under fluoroscopic guidance. Animals were randomly assigned to four different experimental groups: needle sizes of 18 Gauge (G), 21G, 23G, and sham control. To monitor the progression of IVD degeneration and pain, the following methods were employed: μMRI, qRT-PCR, histology, and biobehavioral analysis. RESULTS T1- and T2-weighted μMRI analysis showed a correlation between the degree of IVD degeneration and needle diameter, with the most severe degeneration in the 18G group. mRNA expression of markers for IVD degeneration markers were dysregulated in the 18G and 21G groups, while pro-nociceptive markers were increased in the 18G group only. Hematoxylin and Eosin (H&E) and Alcian Blue/Picrosirius Red staining confirmed the most pronounced IVD degeneration in the 18G group. Randall-Selitto and von Frey tests showed increased hindpaw sensitivity in the 18G group. CONCLUSION Our findings demonstrate that anterior disc injury with an 18G needle creates severe IVD degeneration and mechanical hypersensitivity, while the 21G needle results in moderate degeneration with no increased pain sensitivity. Therefore, needle sizes should be selected depending on the desired phenotype for the pre-clinical model.
Collapse
Affiliation(s)
- Juliane D. Glaeser
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of OrthopedicsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Wafa Tawackoli
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of SurgeryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Biomedical Imaging Research InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of Biomedical SciencesCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Derek G. Ju
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of OrthopedicsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Jae H. Yang
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of Orthopedic SurgeryKorea University Guro HospitalSeoulSouth Korea
| | - Linda EA Kanim
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of OrthopedicsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Khosrowdad Salehi
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Victoria Yu
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Evan Saidara
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Jean‐Phillipe Vit
- Department of Biomedical SciencesCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Zhanna Khnkoyan
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Zachary NaPier
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of OrthopedicsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Laura S. Stone
- McGill University, Faculty of DentistryAlan Edwards Centre for Research on PainMontrealCanada
| | - Hyun W. Bae
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of OrthopedicsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Dmitriy Sheyn
- Orthopaedic Stem Cell Research LaboratoryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of OrthopedicsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of SurgeryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Department of Biomedical SciencesCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| |
Collapse
|
5
|
Hestehave S, Abelson KSP, Brønnum Pedersen T, Munro G. Stress sensitivity and cutaneous sensory thresholds before and after neuropathic injury in various inbred and outbred rat strains. Behav Brain Res 2019; 375:112149. [DOI: 10.1016/j.bbr.2019.112149] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/21/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
|
6
|
Larauche M, Moussaoui N, Biraud M, Bae W, Duboc H, Million M, Taché Y. Brain corticotropin-releasing factor signaling: Involvement in acute stress-induced visceral analgesia in male rats. Neurogastroenterol Motil 2019; 31:e13489. [PMID: 30298965 PMCID: PMC6347489 DOI: 10.1111/nmo.13489] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Water avoidance stress (WAS) induces a naloxone-independent visceral analgesia in male rats under non-invasive conditions of monitoring. The objective of the study was to examine the role of brain CRF signaling in acute stress-induced visceral analgesia (SIVA). METHODS Adult male Sprague-Dawley rats were chronically implanted with an intracerebroventricular (ICV) cannula. The visceromotor response (VMR) to graded phasic colorectal distension (CRD: 10, 20, 40, 60 mm Hg, 20 seconds, 4 minutes intervals) was monitored using manometry. The VMR to a first CRD (baseline) was recorded 5 minutes after an ICV saline injection, followed 1 hour later by ICV injection of either CRF (30, 100, or 300 ng and 1, 3, or 5 μg/rat) or saline and a second CRD, 5 minutes later. Receptor antagonists against CRF1 /CRF2 (astressin-B, 30 μg/rat), CRF2 (astressin2 -B, 10 μg/rat), oxytocin (tocinoic acid, 20 μg/rat), or vehicle were injected ICV 5 minutes before CRF (300 ng/rat, ICV) or 15 minutes before WAS (1 hour). KEY RESULTS ICV CRF (100 and 300 ng) reduced the VMR to CRD at 60 mm Hg by -36.6% ± 6.8% and -48.7% ± 11.7%, respectively, vs baseline (P < 0.001), while other doses had no effect and IP CRF (10 µg/kg) induced visceral hyperalgesia. Astressin-B and tocinoic acid injected ICV induced hyperalgesia and prevented the analgesic effect of ICV CRF (300 ng/rat) and WAS, while astressin2 -B only blocked WAS-induced SIVA. CONCLUSIONS & INFERENCES These data support a role for brain CRF signaling via CRF2 in SIVA in a model of WAS and CRD likely mediated by the activation of brain oxytocin pathway.
Collapse
Affiliation(s)
- M. Larauche
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States
| | - N. Moussaoui
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States,Present address: Inserm U1048/I2MC Obesity Research
Laboratory, 1 avenue Jean Poulhès BP 84225 31432 Toulouse Cedex 4,
France
| | - M. Biraud
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States,Present address: 1060 William Moore drive CVM Main
Building, RM C305, Raleigh, NC 27607, USA
| | - W.K. Bae
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States,Present address: Department of Internal Medicine, Ilsan
Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - H. Duboc
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States,Present address: CRI INSERM UMR 1149, University Paris
Diderot, Sorbonne Paris Cité and DHU Unity, APHP, F-75890 Paris, France
| | - M. Million
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States
| | - Y. Taché
- Department of Medicine, UCLA, G Oppenheimer Center for
Neurobiology of Stress and Resilience and CURE: Digestive Diseases Research Center,
Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of
Medicine, Los Angeles, CA, United States,VA Greater Los Angeles Healthcare System, Los Angeles, CA,
United States
| |
Collapse
|
7
|
Hestehave S, Abelson KSP, Brønnum Pedersen T, Munro G. The analgesic efficacy of morphine varies with rat strain and experimental pain model: implications for target validation efforts in pain drug discovery. Eur J Pain 2018; 23:539-554. [PMID: 30318662 PMCID: PMC6587867 DOI: 10.1002/ejp.1327] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 09/07/2018] [Accepted: 10/10/2018] [Indexed: 11/25/2022]
Abstract
Background Translating efficacy of analgesic drugs from animal models to humans remains challenging. Reasons are multifaceted, but lack of sufficiently rigorous preclinical study design criteria and phenotypically relevant models may be partly responsible. To begin to address this fundamental issue, we assessed the analgesic efficacy of morphine in three inbred rat strains (selected based on stress reactivity and affective/pain phenotypes), and outbred Sprague Dawley (SD) rats supplied from two vendors. Methods Sensitivity to morphine (0.3–6.0 mg/kg, s.c.) was evaluated in the hot plate test of acute thermal nociception, the Complete Freund's Adjuvant (CFA) model of inflammatory‐induced mechanical hyperalgesia, and in a locomotor motility assay in male rats from the following strains; Lewis (LEW), Fischer (F344), Wistar Kyoto (WKY), and SD's from Envigo and Charles River. Results F344 and SD rats were similarly sensitive to morphine in hot plate and CFA‐induced inflammatory hyperalgesia (Minimum Effective Dose (MED) = 3.0 mg/kg). WKY rats developed a less robust mechanical hypersensitivity after CFA injection, and were less sensitive to morphine in both pain tests (MED = 6.0 mg/kg). LEW rats were completely insensitive to morphine in the hot plate test, in contrast to the reversal of CFA‐induced hyperalgesia (MED = 3.0 mg/kg). All strains exhibited a dose‐dependent reduction in locomotor activity at 3.0–6.0 mg/kg. Conclusion Sensory phenotyping in response to acute thermal and inflammatory‐induced pain, and sensitivity to morphine in various inbred and outbred rat strains indicates that different pathophysiological mechanisms are engaged after injury. This could have profound implications for translating preclinical drug discovery efforts into pain patients. Significance The choice of rat strain used in preclinical pain research can profoundly affect the outcome of experiments in relation to (a) nociceptive threshold responses, and (b) efficacy to analgesic treatment, in assays of acute and tonic inflammatory nociceptive pain.
Collapse
Affiliation(s)
- Sara Hestehave
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,H. Lundbeck A/S, Valby, Denmark
| | - Klas S P Abelson
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Gordon Munro
- Department of Neurology, Danish Headache Center, Glostrup Research Institute, Glostrup, Copenhagen, Denmark
| |
Collapse
|
8
|
Yarushkina NI, Filaretova LP. Corticotropin-Releasing Factor (CRF) and Somatic Pain Sensitivity: the Contribution of CRF Receptors of Subtypes 1 and 2. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418020137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
The peripheral corticotropin-releasing factor (CRF)-induced analgesic effect on somatic pain sensitivity in conscious rats: involving CRF, opioid and glucocorticoid receptors. Inflammopharmacology 2018; 26:305-318. [DOI: 10.1007/s10787-018-0445-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/23/2018] [Indexed: 12/30/2022]
|
10
|
Romero A, García-Carmona JA, Laorden ML, Puig MM. Role of CRF1 receptor in post-incisional plasma extravasation and nociceptive responses in mice. Toxicol Appl Pharmacol 2017; 332:121-128. [DOI: 10.1016/j.taap.2017.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/12/2017] [Accepted: 04/19/2017] [Indexed: 12/30/2022]
|
11
|
Sosanya NM, Trevino AV, Chavez RL, Christy RJ, Cheppudira BP. Sound-stress-induced altered nociceptive behaviors are associated with increased spinal CRFR2 gene expression in a rat model of burn injury. J Pain Res 2017; 10:2135-2145. [PMID: 28979159 PMCID: PMC5589110 DOI: 10.2147/jpr.s144055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Sound stress (SS) elicits behavioral changes, including pain behaviors. However, the neuronal mechanisms underlying SS-induced pain behaviors remain to be explored. The current study examined the effects of SS on nociceptive behaviors and changes in expression of the spinal corticotropin-releasing factor (CRF) system in male Sprague Dawley rats with and without thermal pain. We also studied the effects of SS on plasma corticosterone and fecal output. Rats were exposed to 3 days of SS protocol (n = 12/group). Changes in nociceptive behaviors were assessed using thermal and mechanical pain tests. Following the induction of SS, a subgroup of rats (n = 6/group) was inflicted with thermal injury and on day 14 postburn nociceptive behaviors were reassessed. Spinal CRF receptor mRNA expression was analyzed by semiquantitative reverse transcription polymerase chain reaction (RT-PCR). In addition, plasma corticosterone and spinal CRF concentrations were quantified using enzyme-linked immunosorbent assay (ELISA). Increased defecation was observed in SS rats. SS produced transient mechanical allodynia in naive rats, whereas it exacerbated thermal pain in thermally injured rats. Spinal CRFR2 mRNA expression was unaffected by stress or thermal injury alone, but their combined effect significantly increased its expression. SS had no effect on plasma corticosterone and spinal CRF protein in postburn rats. To conclude, SS is capable of exacerbating postburn thermal pain, which is linked to increased CRFR2 gene expression in the spinal cord. Future studies have to delineate whether attenuation of CRFR2 signaling at the spinal level prevents stress-induced exacerbation of burn pain.
Collapse
Affiliation(s)
- Natasha M Sosanya
- United States Army Institute of Surgical Research, San Antonio Military Medical Center, Fort Sam Houston, San Antonio, TX, USA
| | - Alex V Trevino
- United States Army Institute of Surgical Research, San Antonio Military Medical Center, Fort Sam Houston, San Antonio, TX, USA
| | - Roger L Chavez
- United States Army Institute of Surgical Research, San Antonio Military Medical Center, Fort Sam Houston, San Antonio, TX, USA
| | - Robert J Christy
- United States Army Institute of Surgical Research, San Antonio Military Medical Center, Fort Sam Houston, San Antonio, TX, USA
| | - Bopaiah P Cheppudira
- United States Army Institute of Surgical Research, San Antonio Military Medical Center, Fort Sam Houston, San Antonio, TX, USA
| |
Collapse
|
12
|
Victoria NC, Murphy AZ. The long-term impact of early life pain on adult responses to anxiety and stress: Historical perspectives and empirical evidence. Exp Neurol 2015. [PMID: 26210872 DOI: 10.1016/j.expneurol.2015.07.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Approximately 1 in 6 infants are born prematurely each year. Typically, these infants spend 25 days in the Neonatal Intensive Care Unit (NICU) where they experience 10-18 painful and inflammatory procedures each day. Remarkably, pre-emptive analgesics and/or anesthesia are administered less than 25% of the time. Unalleviated pain during the perinatal period is associated with permanent decreases in pain sensitivity, blunted cortisol responses and high rates of neuropsychiatric disorders. To date, the mechanism(s) by which these long-term changes in stress and pain behavior occur, and whether such alterations can be prevented by appropriate analgesia at the time of insult, remains unclear. Work in our lab using a rodent model of early life pain suggests that inflammatory pain experienced on the day of birth blunts adult responses to stress- and pain-provoking stimuli, and dysregulates the hypothalamic pituitary adrenal (HPA) axis in part through a permanent upregulation in central endogenous opioid tone. This review focuses on the long-term impact of neonatal inflammatory pain on adult anxiety- and stress-related responses, and underlying neuroanatomical changes in the context of endogenous pain control and the HPA axis. These two systems are in a state of exaggerated developmental plasticity early in postnatal life, and work in concert to respond to noxious or aversive stimuli. We present empirical evidence from animal and clinical studies, and discuss historical perspectives underlying the lack of analgesia/anesthetic use for early life pain in the modern NICU.
Collapse
Affiliation(s)
- Nicole C Victoria
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303, USA.
| | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303, USA.
| |
Collapse
|
13
|
Johnson AC, Tran L, Greenwood-Van Meerveld B. Knockdown of corticotropin-releasing factor in the central amygdala reverses persistent viscerosomatic hyperalgesia. Transl Psychiatry 2015; 5:e517. [PMID: 25734510 PMCID: PMC4354346 DOI: 10.1038/tp.2015.16] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/18/2014] [Accepted: 01/12/2015] [Indexed: 12/13/2022] Open
Abstract
Gastrointestinal nociception is exacerbated by chronic stress through an unknown mechanism. The amygdala is a key nucleus involved in the autonomic and neuroendocrine responses to stress. The goal of this study was to test the hypothesis that prolonged exposure of the central amygdala (CeA) to stress or the stress hormone cortisol (or corticosterone in rats) induces nociceptive behaviors mediated by corticotropin-releasing factor (CRF) within the CeA. We selectively knocked down CRF in the CeA via antisense oligodeoxynucleotides (ASO) in animals with targeted, stereotaxically placed corticosterone (CORT) micropellets or following repeated water avoidance stress (WAS). CRF expression in the CeA was analyzed concurrently with the assessment of visceral hypersensitivity to colonic distension and mechanical somatic withdrawal threshold. The responses were characterized at 7 or 28 days post implantation of the CORT micropellet or following 7 days of WAS. Exposure of the CeA to elevated CORT or WAS increased CRF expression and heightened visceral and somatic sensitivity. Infusion of CRF ASO into the CeA decreased CRF expression and attenuated visceral and somatic hypersensitivity in both models. Our study provides important evidence for a CRF-mediated mechanism specifically within the CeA that regulates stress-induced visceral and somatic nociception.
Collapse
Affiliation(s)
- A C Johnson
- Oklahoma Center for Neuroscience, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - L Tran
- Oklahoma Center for Neuroscience, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - B Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Science Center, Oklahoma City, OK, USA,Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA,VA Medical Center, University of Oklahoma Health Science Center, Oklahoma City, OK, USA,VA Medical Center, Research Administration Room 151G, 921 NE 13th Street, Oklahoma City, OK 73104, USA. E-mail:
| |
Collapse
|
14
|
Park SH, Choi SS, Sim YB, Lee JK, Suh HW. Role of corticotropin-releasing hormone receptor 1 in the regulation of nociception in mice. Anim Cells Syst (Seoul) 2014. [DOI: 10.1080/19768354.2014.966857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
15
|
Strain differences in the expression of endocannabinoid genes and in cannabinoid receptor binding in the brain of Lewis and Fischer 344 rats. Prog Neuropsychopharmacol Biol Psychiatry 2014; 53:15-22. [PMID: 24607771 DOI: 10.1016/j.pnpbp.2014.02.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 02/23/2014] [Accepted: 02/26/2014] [Indexed: 01/27/2023]
Abstract
The Lewis (LEW) and Fischer 344 (F344) rat strains have been proposed as a model to study certain genetic influences on drug use. These strains differ in terms of the self-administration of several drugs, and in their expression of various components of the dopaminergic, glutamatergic, GABAergic and endogenous opioid neurotransmitter systems. As the endocannabinoid system is linked to these systems, we investigated whether these two strains exhibit differences in cannabinoid receptor binding and in the expression of cannabinoid-related genes. Quantitative autoradiography of [(3)H]-CP 55,940 binding levels and real-time PCR assays were used. F344 rats displayed higher levels of cannabinoid receptor binding in the lateral globus pallidus and weaker CNR1 gene expression in the prefrontal cortex (PFc) than LEW rats. Moreover, the N-acyl phosphatidylethanolamine-specific phospholipase D/fatty acid amide hydrolase ratio was greater in the PFc and NAcc of F344 rats. Our results suggest that the endocannabinoid system may be a mediator of the individual differences that exist in the susceptibility to the rewarding effects of drugs of abuse.
Collapse
|
16
|
Page GG, Opp MR, Kozachik SL. Reduced sleep, stress responsivity, and female sex contribute to persistent inflammation-induced mechanical hypersensitivity in rats. Brain Behav Immun 2014; 40:244-51. [PMID: 24594386 DOI: 10.1016/j.bbi.2014.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 01/22/2023] Open
Abstract
Studies in humans suggest that female sex, reduced sleep opportunities and biological stress responsivity increase risk for developing persistent pain conditions. To investigate the relative contribution of these three factors to persistent pain, we employed the Sciatic Inflammatory Neuritis (SIN) model of repeated left sciatic perineurial exposures to zymosan, an inflammatory stimulus, to determine their impact upon the development of persistent mechanical hypersensitivity. Following an initial moderate insult, a very low zymosan dose was infused daily for eight days to model a sub-threshold inflammatory perturbation to which only susceptible animals would manifest or maintain mechanical hypersensitivity. Using Sprague Dawley rats, maintaining wakefulness throughout the first one-half of the 12-h light phase resulted in a bilateral reduction in paw withdrawal thresholds (PWTs); zymosan infusion reduced ipsilateral PWTs in all animals and contralateral PWTs only in females. This sex difference was validated in Fischer 344, Lewis and Sprague Dawley rats, suggesting that females are the more susceptible phenotype for both local and centrally driven responses to repeated low-level inflammatory perturbations. Hypothalamic-pituitary-adrenal (HPA) axis hyporesponsive Lewis rats exhibited the most robust development of mechanical hypersensitivity and HPA axis hyperresponsive Fischer 344 rats matched the Lewis rats' mechanical hypersensitivity throughout the latter four days of the protocol. If HPA axis phenotype does indeed influence these findings, the more balanced responsivity of Sprague Dawley rats would seem to promote resilience in this paradigm. Taken together, these findings are consistent with what is known regarding persistent pain development in humans.
Collapse
Affiliation(s)
- Gayle G Page
- School of Nursing, Johns Hopkins University, 525 N. Wolfe St., Baltimore, MD 21205, United States.
| | - Mark R Opp
- Department of Anesthesiology & Pain Medicine, University of Washington, 325 9th Ave, Box #359724, Seattle, WA 98104, United States.
| | - Sharon L Kozachik
- School of Nursing, Johns Hopkins University, 525 N. Wolfe St., Baltimore, MD 21205, United States.
| |
Collapse
|
17
|
Chopra K, Arora V. An intricate relationship between pain and depression: clinical correlates, coactivation factors and therapeutic targets. Expert Opin Ther Targets 2013; 18:159-76. [PMID: 24295272 DOI: 10.1517/14728222.2014.855720] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION An apparent clinical relationship between pain and depression has long been recognized, which makes an enormous impact on the individual health care. At present, the practical implication of such overlapping symptomatology between pain and depression is not clear, but the prevalence estimates for depression are substantially inflated among patients with chronic pain and vice versa. This interaction has been labeled as the depression-pain syndrome or depression-pain dyad. AREAS COVERED This article discusses the neurobiological substrates and neuroanatomical pathways involved in pain-depression dyad along with newer therapeutic targets. EXPERT OPINION Several key themes emerged from our review of the relationship between depression and pain. First, the diagnosis of depression in pain or vice versa is particularly challenging, and the development of better diagnostic framework that involves both pain and depression is particularly required. Secondly, the entwined relationship between pain and depression supports the possibility of common coactivating factors that results in their neurophysiological overlap. A broad understanding of the role played by the central nervous system (CNS) in the processing of pain and depression may eventually lead to the introduction of triple reuptake inhibitors, agomelatine, vilazodone and ketamine with novel mechanism of action, hence appear to be of promising potential for pain with depression.
Collapse
Affiliation(s)
- Kanwaljit Chopra
- Panjab University, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Pharmacology Research Laboratory , Chandigarh-160 014 , India +91 172 2534105 ; +91 172 2541142 ;
| | | |
Collapse
|
18
|
Estrogen effects on the forced swim test differ in two outbred rat strains. Physiol Behav 2012; 106:81-6. [PMID: 22266677 DOI: 10.1016/j.physbeh.2012.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 01/02/2012] [Accepted: 01/03/2012] [Indexed: 12/31/2022]
Abstract
Changes in reproductive hormones, such as estrogen, play a role in mood regulation. The present study examined strain differences (Long-Evans vs. Wistar-Hannover) in the behavioral and biochemical effects of estrogen manipulation. Adult ovariectomized female rats were treated with estradiol, vehicle, or withdrawn from estradiol. The two strains demonstrated differential behavioral responses to short-term estradiol administration in the forced swim test; estradiol induced an antidepressant-like effect in Long-Evans rats but not in Wistar rats. Conversely, withdrawal from estradiol resulted in a depressive-like state in the Wistar rats but not in the Long-Evans rats. Western blot analyses found no differences in estrogen receptors α and β within the hippocampus or the frontal cortex, two brain areas strongly implicated in affective disorders. These data demonstrate the importance of strain as a variable when interpreting behavioral effects of estrogen.
Collapse
|
19
|
Rouwette T, Vanelderen P, Roubos E, Kozicz T, Vissers K. The amygdala, a relay station for switching on and off pain. Eur J Pain 2011; 16:782-92. [DOI: 10.1002/j.1532-2149.2011.00071.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2011] [Indexed: 12/21/2022]
Affiliation(s)
| | | | - E.W. Roubos
- Department of Cellular Animal Physiology; Donders Institute for Brain, Cognition and Behaviour; Centre for Neuroscience; Radboud University Nijmegen; Nijmegen; The Netherlands
| | - T. Kozicz
- Department of Cellular Animal Physiology; Donders Institute for Brain, Cognition and Behaviour; Centre for Neuroscience; Radboud University Nijmegen; Nijmegen; The Netherlands
| | - K. Vissers
- Department of Anesthesiology; Pain and Palliative Medicine; Radboud University Nijmegen Medical Centre; Nijmegen; The Netherlands
| |
Collapse
|
20
|
Lariviere WR, Fiorenzani P, Ceccarelli I, Massafra C, Sorda G, Di Canio C, Aloisi AM. Central CRH administration changes formalin pain responses in male and female rats. Brain Res 2011; 1383:128-34. [PMID: 21300038 DOI: 10.1016/j.brainres.2011.01.106] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 01/29/2011] [Indexed: 01/21/2023]
Abstract
Corticotropin-releasing hormone (CRH) is suggested to be involved in the regulation of pain. To better evaluate the CRH-mediated behavioral alterations in the formalin inflammatory pain test, we administered CRH or the CRH receptor antagonist α-helical CRH(9-41) (ahCRH) intracerebroventricularly to male and female rats and compared the effects with those of saline control. Nociceptive stimulation was carried out through a subcutaneous injection of dilute formalin (50μL, 10%) in the plantar surface of the hind paw. In both sexes, formalin-induced responses, recorded for 60min, were affected by CRH but not by ahCRH treatment. Paw flexing duration was decreased in both sexes during the formalin interphase period in the CRH-treated group compared to saline control groups; however, licking of the injected paw was markedly increased by the same treatment at other time periods. Treatments induced only a few changes in spontaneous non-pain behaviors, which do not account for the effects on pain response. In conclusion, these data demonstrate the ability of CRH to affect the behavioral responses to an inflammatory nociceptive stimulus, and that the effects can be in opposite directions depending on the behavioral response considered.
Collapse
Affiliation(s)
- William R Lariviere
- Department of Anesthesiology, University of Pittsburgh School of Medicine, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Peng YL, Zhang JN, Chang M, Li W, Han RW, Wang R. Effects of central neuropeptide S in the mouse formalin test. Peptides 2010; 31:1878-83. [PMID: 20603169 DOI: 10.1016/j.peptides.2010.06.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 06/24/2010] [Accepted: 06/24/2010] [Indexed: 10/19/2022]
Abstract
Neuropeptide S (NPS), a recently discovered bioactive peptide, was reported to regulate arousal, anxiety, locomotion, feeding behaviors, memory, and drug addiction. NPS receptor (NPSR) mRNA was found in several brain regions related to descending control system of pain, including the periaqueductal gray (PAG). Our previous study had shown that NPS could produce antinociception in mice. The present study was designed to evaluate whether NPS may produce antinociceptive effect observed in the mouse formalin test, a model of inflammatory pain. NPS (0.1-100 pmol) administrated intracerebroventricularly (i.c.v.) dose-dependently attenuated both first-phase and second-phase nociceptive behaviors induced by paw formalin injection. NPS (10 pmol, i.c.v.)-elicited antinociceptive effect was counteracted by co-injection with 1000 and 10,000 pmol [D-Val(5)]NPS, which alone induced neither hyperalgesia nor antinociception. The antinociception induced by NPS (10 pmol, i.c.v.) was not affected by naloxone (i.p., 10 mg/kg) and naloxone alone had no effect in the formalin test. In addition, compared to the saline (i.c.v.) treated group, NPS (10 pmol, i.c.v.) treated group increased c-Fos protein expression in nearly all subdivisions of the PAG in the formalin-injected mice. The above results revealed that NPS could produce antinociception in the formalin test through NPSR, which may be involved in the activation of PAG, suggesting that NPS-NPSR system may be a potential target for developing new analgesic drugs.
Collapse
Affiliation(s)
- Ya-Li Peng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, State Key Laboratory of Applied Organic Chemistry, and Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tian Shui South Road, Lanzhou 730000, PR China
| | | | | | | | | | | |
Collapse
|
22
|
Kuehl LK, Michaux GP, Richter S, Schächinger H, Anton F. Increased basal mechanical pain sensitivity but decreased perceptual wind-up in a human model of relative hypocortisolism. Pain 2010; 149:539-546. [PMID: 20381248 DOI: 10.1016/j.pain.2010.03.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 03/18/2010] [Accepted: 03/19/2010] [Indexed: 12/15/2022]
Abstract
Clinical data have accumulated showing that relative hypocortisolism, which may be regarded as a neuroendocrinological correlate of chronic stress, may be a characteristic of some functional pain syndromes. However, it has not been clarified yet whether deregulations of the hypothalamus-pituitary-adrenal (HPA) axis may directly alter pain perception and thus be causally involved in the pathophysiology of these disorders. To test this hypothesis, we performed a randomized placebo-controlled crossover trial in N=20 healthy drug-free volunteers (median age 24yrs) and analyzed the effects of metyrapone-induced hypocortisolism on quantitatively assessed basal mechanical pain sensitivity (1.5-13m/s impact stimuli), perceptual wind-up (9m/s impact stimuli at 1Hz) and temporal summation of pain elicited by inter-digital web pinching (IWP; 10N pressure stimuli for 2min). Experimentally induced hypocortisolism significantly decreased pain detection thresholds and augmented temporal summation of IWP-induced pain (p<.05). The latter effect was dependent on the relative reduction in cortisol levels, and seemed to rely on a potentiated sensitization and not merely on the observed changes in basal pain sensitivity. Perceptual wind-up by contrast was reduced when cortisol synthesis was blocked (p<.05). This result is reminiscent of findings from animal studies showing a reversal of NMDA receptor activation by glucocorticoid receptor antagonists in neuropathic pain models. Our results speak in favor of a potential causal role of HPA axis alterations in pain chronicity.
Collapse
Affiliation(s)
- Linn K Kuehl
- Institute of Psychobiology, Department of Clinical Physiology, University of Trier, Johanniterufer 15, D-54290 Trier, Germany Laboratory of Psychobiology, University of Luxembourg, Campus Limpertsberg, 162A avenue de la Faïencerie, L-1511 Luxembourg, Luxembourg
| | | | | | | | | |
Collapse
|
23
|
Van Houdenhove B, Luyten P, Tiber Egle U. Stress as a Key Concept in Chronic Widespread Pain and Fatigue Disorders. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/10582450903284745] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
24
|
Chen YL, Li AH, Yeh TH, Chou AH, Wang HL. Nocistatin and nociceptin exert opposite effects on the excitability of central amygdala nucleus-periaqueductal gray projection neurons. Mol Cell Neurosci 2009; 40:76-88. [PMID: 18930828 DOI: 10.1016/j.mcn.2008.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 09/08/2008] [Accepted: 09/10/2008] [Indexed: 11/29/2022] Open
Abstract
Central amygdala nucleus (CeA)-periaqueductal gray (PAG) pathway is the component of descending antinociceptive circuitry. Nociceptin/orphanin FQ (N/OFQ) and nocistatin (NST) produce supraspinal pronociceptive and antinociceptive effects, respectively. We hypothesized that opposite effects of N/OFQ and NST on supraspinal pain modulation result from their opposing effects on the excitability of CeA-PAG projection neurons. This hypothesis was tested by investigating electrophysiological effects of N/OFQ and NST on medial CeA neurons that project to PAG (CeA(M)-PAG). N/OFQ hyperpolarized CeA(M)-PAG projection neurons by enhancing inwardly rectifying potassium conductance. In contrast, NST depolarized CeA(M)-PAG neurons by causing the opening of TRPC cation channels via G(alphaq/11)-PLC-PKC pathway. CeA(M)-PAG neurons hyperpolarized by N/OFQ express CRF or neurotensin mRNA. NST-responsive CeA(M)-PAG neurons contain CRF or substance P mRNA. Our study provides the evidence that the molecular and cellular basis for opposite effects of N/OFQ and NST on supraspinal pain regulation is their opposing effects on the excitability of peptidergic CeA(M)-PAG neurons.
Collapse
Affiliation(s)
- Ying-Ling Chen
- Department of Physiology, Chang Gung University School of Medicine, Kwei-San, Tao-Yuan, Taiwan, ROC
| | | | | | | | | |
Collapse
|
25
|
The role of hypothalamo-hypophyseal-adrenocortical system hormones in controlling pain sensitivity. ACTA ACUST UNITED AC 2008; 38:759-66. [PMID: 18802775 DOI: 10.1007/s11055-008-9044-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 07/09/2007] [Indexed: 10/21/2022]
Abstract
The present review addresses analysis of data demonstrating the role of the hypothalamo-hypophyseal-adrenocortical axis (HHACA) in controlling pain sensitivity. Experiments on rats have demonstrated the analgesic effects of exogenous hormones of all components of the HHACA - corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and glucocorticoids - in the same models, and have also shown that the opioid and non-opioid mechanisms contribute to the development of the analgesia induced by these hormones. Endogenous glucocorticoids are involved in the development of analgesia mediated by non-opioid mechanisms. Along with the non-opioid mechanisms associated with endogenous glucocorticoids, the analgesic effect of ACTH can be mediated by the opioid mechanism. Unlike the situation with ACTH, the analgesic effect of CRH is mediated exclusively by non-opioid mechanisms, one of which is associated with HHACA hormones, while the other, appearing only on systemic administration, is not associated with these hormones. The actions of glucocorticoids on pain are mediated by neurons in the central gray matter of the midbrain.
Collapse
|
26
|
Fu Y, Neugebauer V. Differential mechanisms of CRF1 and CRF2 receptor functions in the amygdala in pain-related synaptic facilitation and behavior. J Neurosci 2008; 28:3861-76. [PMID: 18400885 PMCID: PMC2557030 DOI: 10.1523/jneurosci.0227-08.2008] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 02/22/2008] [Accepted: 02/22/2008] [Indexed: 02/06/2023] Open
Abstract
A major site of extrahypothalamic expression of corticotropin-releasing factor (CRF) and its G-protein-coupled CRF1 and CRF2 receptors is the amygdala, a key player in emotions and affective disorders. Pain-related plasticity in the laterocapsular division of the central nucleus of the amygdala (CeLC) generates emotional-affective responses and anxiety-like behavior. CRF1 receptor antagonists have anxiolytic effects. Although both CRF1 and CRF2 receptors couple positively to adenylyl cyclase, they can have opposite effects, but the underlying mechanism is unknown. This study addressed CRF1 and CRF2 receptor functions and mechanisms in the amygdala in a model of arthritic pain. Using whole-cell patch-clamp recordings of CeLC neurons, we found that a selective CRF1 receptor antagonist (NBI27914 [5-chloro-4-(N-(cyclopropyl)methyl-N-propylamino)-2-methyl-6-(2,4,6-trichlorophenyl)]) amino-pyridine inhibited synaptic facilitation in brain slices from arthritic rats through a postsynaptic mechanism. Inhibition of the NMDA receptor-mediated synaptic component was occluded by a protein kinase A (PKA) inhibitor, consistent with our previous demonstration of PKA-dependent increased NMDA receptor function in arthritis pain-related plasticity. NBI27914 also decreased neuronal excitability through inhibition of highly tetraethylammonium (TEA)-sensitive ion channels that contribute to action potential repolarization and firing rate. In contrast, a CRF2 receptor antagonist (astressin-2B [cyclo(31-34) [d-Phe11,His12,C alphaMeLeu13,39, Nle17, Glu31, Lys34] Ac-Sauvagine(8-40)]) facilitated synaptic transmission through presynaptic inhibition of GABAergic transmission (disinhibition). NBI27914 inhibited arthritis pain-related behaviors (audible and ultrasonic vocalizations and hindlimb withdrawal reflexes). Astressin-2B had no significant behavioral effect. The data suggest that endogenous CRF1 receptor activation in the amygdala contributes to pain-related synaptic facilitation, increased excitability, and pain behavior through a postsynaptic mechanism involving activation of PKA and highly TEA-sensitive K(+)-currents. Presynaptic CRF2 receptor-mediated inhibition does not reach behavioral significance.
Collapse
Affiliation(s)
- Yu Fu
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas 77555-1069
| | - Volker Neugebauer
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas 77555-1069
| |
Collapse
|
27
|
Ji G, Neugebauer V. Pro- and anti-nociceptive effects of corticotropin-releasing factor (CRF) in central amygdala neurons are mediated through different receptors. J Neurophysiol 2008; 99:1201-12. [PMID: 18171711 DOI: 10.1152/jn.01148.2007] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Corticotropin-releasing factor (CRF) is not only a stress hormone but also acts as a neuromodulator outside the hypothalamic-pituitary-adrenocortical axis, playing an important role in anxiety, depression, and pain modulation. The underlying mechanisms remain to be determined. A major site of extra-hypothalamic expression of CRF and its receptors is the amygdala, a key player in affect-related disorders such as anxiety. The latero-capsular division of the central nucleus of the amygdala (CeLC) is also important for pain modulation and pain affect. This study analyzed the effects of CRF on nociceptive processing in CeLC neurons and the contribution of CRF1 and CRF2 receptors and protein kinases A and C. Extracellular single-unit recordings were made from CeLC neurons in anesthetized adult rats. All neurons responded more strongly to noxious than innocuous mechanical stimulation of the knee. Evoked responses and background activity were measured before and during administration of CRF into the CeLC by microdialysis. CRF was administered alone or together with receptor antagonists or protein kinase inhibitors. CRF (0.01-1 microM; concentrations in microdialysis probe; 15 min) facilitated the evoked responses more strongly than background activity; a higher concentration (10 microM) had inhibitory effects. Facilitation by CRF (0.1 microM) was reversed by a selective CRF1 receptor antagonist (NBI27914, 10 microM) but not a CRF2 receptor antagonist (astressin-2B, 100 microM) and by a protein kinase A (PKA) inhibitor (KT5720, 100 microM) but not a protein kinase C inhibitor (GF109203X, 100 microM). Inhibitory effects of CRF (10 microM) were reversed by astressin-2B. These data suggest that CRF has dual effects on amygdala neurons: CRF1 receptor-mediated PKA-dependent facilitation and CRF2 receptor-mediated inhibition.
Collapse
Affiliation(s)
- Guangchen Ji
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-1069, USA
| | | |
Collapse
|
28
|
Mousa SA, Bopaiah CP, Richter JF, Yamdeu RS, Schäfer M. Inhibition of inflammatory pain by CRF at peripheral, spinal and supraspinal sites: involvement of areas coexpressing CRF receptors and opioid peptides. Neuropsychopharmacology 2007; 32:2530-42. [PMID: 17375137 DOI: 10.1038/sj.npp.1301393] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is conflicting evidence on the antinociceptive effects of corticotropin-releasing factor (CRF) along the neuraxis of pain transmission and the responsible anatomical sites of CRF's action at the level of the brain, spinal cord and periphery. In an animal model of tonic pain, that is, Freunds complete adjuvant (FCA) hindpaw inflammation, we systematically investigated CRF's ability to modulate inflammatory pain at those three levels of pain transmission by algesiometry following the intracerebroventricular, intrathecal, and intraplantar application of low, systemically inactive doses of CRF. At each level, CRF elicits potent antinociceptive effects, which are dose dependent and antagonized by local, but not systemic CRF receptor antagonist alpha-helical CRF indicating CRF receptor specificity. Consistently, we have identified by immunohistochemistry multiple brain areas, inhibitory interneurons within the dorsal horn of the spinal cord as well as immune cells within subcutaneous tissue--but not peripheral sensory neurons--that coexpress both CRF receptors and opioid peptides. In line with these anatomical findings, local administration of CRF together with the opioid receptor antagonist naloxone dose-dependently reversed CRF's antinociceptive effects at each of these three levels of pain transmission. Therefore, local application of low, systemically inactive doses of CRF at the level of the brain, spinal cord and periphery inhibits tonic inflammatory pain most likely through an activation of CRF receptors on cells that coexpress opioid peptides which results in opioid-mediated pain inhibition. Future studies have to delineate whether endogenous CRF at these three levels contributes to the body's response to cope with the stressful stimulus pain in an opioid-mediated manner.
Collapse
Affiliation(s)
- Shaaban A Mousa
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | | | | | | | | |
Collapse
|
29
|
Abstract
This paper is the 29th consecutive installment of the annual review of research concerning the endogenous opioid system, now spanning 30 years of research. It summarizes papers published during 2006 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurological disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY 11367, United States.
| |
Collapse
|
30
|
Weaver SA, Diorio J, Meaney MJ. Maternal separation leads to persistent reductions in pain sensitivity in female rats. THE JOURNAL OF PAIN 2007; 8:962-9. [PMID: 17686657 DOI: 10.1016/j.jpain.2007.07.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 07/03/2007] [Accepted: 07/05/2007] [Indexed: 11/24/2022]
Abstract
UNLABELLED We determined responses to noxious thermal stimuli, before and after morphine, and mu-opioid receptor binding in brain regions involved in nociception in maternally separated (MS), neonatally handled (H) and nonhandled (NH) female rats. Long-Evans dams were randomly assigned to either 180-minute (MS) or 15-minute (H) minute daily separations from their litters or left undisturbed (NH). At 120 days of age, paw lick latency (50 degrees C hot plate) was determined in offspring during diestrous. Rats were then given 1, 2, 5, or 10 mg/kg morphine and paw lick latency was measured. Rats were killed during diestrous and mu-opioid receptor binding was determined in discrete brain regions, using [(3)H]DAMGO autoradiography. MS rats had significantly longer (P < .05) paw lick latencies compared with H rats. The percent maximal possible effect of morphine was significantly (P < .05) lower in MS compared with H rats for the 5 mg/kg dose. Mu-Opioid receptor binding capacity was significantly greater (P < .05) in MS rats compared with H rats in the medial preoptic nucleus. In conclusion, MS and H treatments led to antipodal differences in pain sensitivity in female rats and differential mu-opioid receptor binding in the medial preoptic nucleus. PERSPECTIVE This article describes the persistent impact of early life adversity on pain sensitivity and the analgesic potency of morphine. Clinically, early life history may play an important role in pain symptoms and responses to opioid analgesics.
Collapse
Affiliation(s)
- Shelley A Weaver
- War-Related Illness and Injury Study Center, Department of Veterans Affairs, New Jersey Health Care System, East Orange, New Jersey 07018, USA.
| | | | | |
Collapse
|
31
|
Current World Literature. Curr Opin Anaesthesiol 2007; 20:388-94. [PMID: 17620851 DOI: 10.1097/aco.0b013e3282c3a878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Ji G, Neugebauer V. Differential effects of CRF1 and CRF2 receptor antagonists on pain-related sensitization of neurons in the central nucleus of the amygdala. J Neurophysiol 2007; 97:3893-904. [PMID: 17392412 DOI: 10.1152/jn.00135.2007] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
As a hormone in the hypothalamic-pituitary-adrenocortical (HPA) axis corticotropin-releasing factor (CRF) mediates stress responses. CRF can also act as a neuromodulator of synaptic transmission outside the HPA axis. A major site of extrahypothalamic expression of CRF and its G-protein-coupled receptors is the amygdala, a key player in affect-related disorders such as anxiety. The laterocapsular division of the central nucleus of the amygdala (CeLC) is important for the modulation of pain affect. This study determined the effects of CRF1 and CRF2 receptor antagonists in CeLC neurons in an arthritis pain model. Extracellular single-unit recordings were made from CeLC neurons in anesthetized adult rats. All neurons responded more strongly to noxious than to innocuous mechanical stimulation (compression) of peripheral tissues, including the knee. Evoked responses and background activity were measured before and during the development of a kaolin/carrageenan-induced knee joint arthritis. Drugs were administered into the CeLC by microdialysis before and/or after arthritis induction. All CeLC neurons showed increased responses to mechanical stimuli ("sensitization") 5-6 h postinduction of arthritis. A selective CRF1 receptor antagonist (NBI27914; 1-100 microM, concentration in microdialysis probe; 15 min) inhibited evoked responses and background activity in arthritis (n = 9) but had no effect under normal conditions before arthritis (n = 9). In contrast, a selective CRF2 receptor antagonist (Astressin-2B; 1-100 microM, 15 min) had no effect in arthritis (n = 7) but increased the neurons' responses under normal conditions (n = 8). These data suggest that CRF1 receptors in the amygdala contribute to pain-related sensitization, whereas the normally inhibitory function of CRF2 receptors is lost in the arthritis pain model.
Collapse
Affiliation(s)
- Guangchen Ji
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-1069, USA
| | | |
Collapse
|
33
|
Einat H. Different behaviors and different strains: Potential new ways to model bipolar disorder. Neurosci Biobehav Rev 2007; 31:850-7. [PMID: 17307253 DOI: 10.1016/j.neubiorev.2006.12.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2006] [Revised: 12/19/2006] [Accepted: 12/20/2006] [Indexed: 02/04/2023]
Abstract
The state of animal models for bipolar disorder (BPD) is deficient, creating a major problem in the research related to this devastating disorder and in our ability to translate molecular findings to the clinic. An ideal model, a "bipolar animal" is most likely unattainable as long as we do not fully understand the biological basis of the disorder, and no models are currently available to reflect the cycling nature of the disease. Yet, additional, better and more practical models need to be developed to support research efforts in the field. The present paper suggests two approaches for the development of new models. The first approach, recently introduced in the literature, is based on modeling different facets of the disease with an attempt to create a test battery that will cover a number of BPD-related behaviors. Whereas each separate model may not have strong validity when used alone, additional strength may come when certain models are combined. One example for modeling a facet of the disorder is brought showing that aggressive behavior in resident mice can be ameliorated by the dissimilar mood stabilizers lithium and valproate suggesting a possible use of the model as part of the battery, representing the aggressive facet of mania. The second approach is based on identifying behavioral differences between existing strains of animals and identifying strains that may have a behavioral phenotype that resembles aspects of BPD. A similar approach has been used previously to model other psychiatric disorders and can be utilized for BPD research. An example of this possible approach is shown with the Black Swiss mice strain that appears to have more manic-like behaviors compared to other strains. Both approaches will not culminate to an ideal, all encompassing model of BPD but may provide useful and relatively uncomplicated tools for research of the disorder.
Collapse
Affiliation(s)
- Haim Einat
- University of Minnesota, College of Pharmacy, 376 Kirby Plaza, 1208 Kirby Drive, Duluth, MN 55812, USA.
| |
Collapse
|
34
|
Abstract
Inbred, histocompatible Lewis and Fischer 344 rats (LEW and FIS) have been used to identify an inverse relationship between hypothalamic-pituitary-adrenal (HPA) axis activity and susceptibility to autoimmune and chronic inflammatory disorders, with LEW showing blunted HPA axis activity and increased susceptibility toward the development of autoimmunity and chronic inflammation, and FIS showing the opposite relationship. In the present study, LEW and FIS were used to determine the relationship between HPA axis function and acute inflammatory pain (carrageenan-induced hindpaw inflammation) and neuropathic pain (partial sciatic nerve ligation; PSNL). The results showed that carrageenan-induced thermal and mechanical allodynia and hyperalgesia were greater in FIS than in LEW. Similarly, FIS showed more carrageenan-induced hindpaw swelling and higher levels of myeloperoxidase (a measure of neutrophils) in the carrageenan-inflamed hindpaw. After PSNL, LEW showed a profound mechanical allodynia and hyperalgesia, whereas mechanical sensitivity in FIS was unaltered. However, FIS, but not LEW, developed thermal allodynia and hyperalgesia after PSNL. These results provide strong evidence for a positive relationship between HPA axis activity and acute inflammatory pain. The results also support a relationship between HPA axis activity and neuropathic pain, but the relationship is complex and may depend on the pain assay.
Collapse
Affiliation(s)
- K Fecho
- Department of Anesthesiology, Division of Pain Medicine, The University of North Carolina at Chapel Hill, NC 27599-7010, USA.
| | | |
Collapse
|