1
|
Humes C, Sic A, Knezevic NN. Substance P's Impact on Chronic Pain and Psychiatric Conditions-A Narrative Review. Int J Mol Sci 2024; 25:5905. [PMID: 38892091 PMCID: PMC11172719 DOI: 10.3390/ijms25115905] [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: 04/22/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Substance P (SP) plays a crucial role in pain modulation, with significant implications for major depressive disorder (MDD), anxiety disorders, and post-traumatic stress disorder (PTSD). Elevated SP levels are linked to heightened pain sensitivity and various psychiatric conditions, spurring interest in potential therapeutic interventions. In chronic pain, commonly associated with MDD and anxiety disorders, SP emerges as a key mediator in pain and emotional regulation. This review examines SP's impact on pain perception and its contributions to MDD, anxiety disorders, and PTSD. The association of SP with increased pain sensitivity and chronic pain conditions underscores its importance in pain modulation. Additionally, SP influences the pathophysiology of MDD, anxiety disorders, and PTSD, highlighting its potential as a therapeutic target. Understanding SP's diverse effects provides valuable insights into the mechanisms underlying these psychiatric disorders and their treatment. Further research is essential to explore SP modulation in psychiatric disorders and develop more effective treatment strategies.
Collapse
Affiliation(s)
- Charles Humes
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (C.H.); (A.S.)
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Aleksandar Sic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (C.H.); (A.S.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (C.H.); (A.S.)
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
2
|
Mannangatti P, Ragu Varman D, Ramamoorthy S, Jayanthi LD. Neurokinin-1 Antagonism Distinguishes the Role of Norepinephrine Transporter from Dopamine Transporter in Mediating Amphetamine Behaviors. Pharmacology 2021; 106:597-605. [PMID: 34515205 DOI: 10.1159/000518033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/21/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Amphetamine (AMPH) and other psychostimulants act on the norepinephrine (NE) transporter (NET) and the dopamine (DA) transporter (DAT) and enhance NE and DA signaling. Both NET and DAT share anatomical and functional characteristics and are regulated similarly by psychostimulants and receptor-linked signaling pathways. We and others have demonstrated that NET and DAT are downregulated by AMPH and substance P/neurokinin-1 receptor (NK1R)-mediated protein kinase C pathway. OBJECTIVES Since both NET and DAT are downregulated by AMPH and NK1R activation and share high sequence homology, the objective of the study was to determine the catecholamine transporter specificity in NK1R modulation of AMPH-induced behaviors. METHODS The effect of NK1R antagonism on AMPH-induced conditioned place preference (CPP) as well as AMPH-induced NET and DAT downregulation was examined using NET and DAT knockout mice (NET-KO and DAT-KO) along with their wild-type littermates. RESULTS Aprepitant (5 mg/kg i.p.) significantly attenuated AMPH (2 mg/kg i.p.)-induced CPP in the wild-type and DAT-KO but not in the NET-KO. Locomotor activity measured during the post-conditioning test (in the absence of AMPH) showed higher locomotor activity in DAT-KO compared to wild-type or NET-KO. However, the locomotor activity of all 3 genotypes remained unchanged following aprepitant. Additionally, in the ventral striatum of wild-type, the AMPH-induced downregulation of NET function and surface expression but not that of DAT was attenuated by aprepitant. CONCLUSIONS The results from the current study demonstrate that aprepitant attenuates the expression of AMPH-induced CPP in DAT-KO mice but not in NET-KO mice suggesting a role for NK1R-mediated NET regulation in AMPH-induced behaviors.
Collapse
Affiliation(s)
- Padmanabhan Mannangatti
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Lankupalle D Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
3
|
Nyman M, Eskola O, Kajander J, Jokinen R, Penttinen J, Karjalainen T, Nummenmaa L, Hirvonen J, Burns D, Hargreaves R, Solin O, Hietala J. Brain neurokinin-1 receptor availability in never-medicated patients with major depression - A pilot study. J Affect Disord 2019; 242:188-194. [PMID: 30193189 DOI: 10.1016/j.jad.2018.08.084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/16/2018] [Accepted: 08/20/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Neurotransmitter substance P (SP) and its preferred neurokinin-1 receptor (NK1R) have been implicated in the treatment of affective and addiction disorders. Despite promising preclinical data on antidepressant action, the clinical trials of NK1R antagonists in major depression have been disappointing. There are no direct in vivo imaging studies on NK1R characteristics in patients with a major depressive disorder (MDD). METHODS In this cross-sectional case-control study, we recruited nine never-medicated patients with moderate to severe MDD and nine matched healthy controls. NK1R availability (NK1R binding potential, BPND) was measured with in vivo 3-D positron emission tomography and a specific NK1 receptor tracer [18F]SPA-RQ. Clinical symptoms were assessed with the 17-item Hamilton Rating Scale for Depression (HAM-D17). RESULTS NK1R-BPND did not differ statistically significantly between patients with MDD and healthy controls. HAM-D17 total scores (range 21-32) correlated positively with NK1R-BPND in cortical and limbic areas. HAM-D17 subscale score for anxiety symptoms correlated positively with NK1R-BPND in specific brain areas implicated in fear and anxiety. LIMITATIONS Small sample size. Low variability in the clinical HAM-D subscale ratings may affect the observed correlations. CONCLUSIONS Our preliminary results do not support a different baseline expression of NK1Rs in a representative sample of never-medicated patients with MDD during a current moderate/severe depressive episode. The modulatory effect of NK1Rs on affective symptoms is in line with early positive results on antidepressant action of NK1 antagonists. However, the effect is likely to be too weak for treatment of MDD with NK1R antagonists alone in clinical practice.
Collapse
Affiliation(s)
- Mikko Nyman
- Turku PET Centre, Neuropsychiatric Imaging, Turku, Finland; Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Olli Eskola
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, Turku, Finland
| | - Jaana Kajander
- Turku PET Centre, Neuropsychiatric Imaging, Turku, Finland
| | - Riitta Jokinen
- Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | - Jukka Penttinen
- Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | | | | | - Jussi Hirvonen
- Turku PET Centre, Neuropsychiatric Imaging, Turku, Finland; Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Donald Burns
- Imaging Research, Merck Research Laboratories, West Point, PA, USA
| | | | - Olof Solin
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, Turku, Finland; Department of Chemistry, University of Turku, Turku, Finland
| | - Jarmo Hietala
- Turku PET Centre, Neuropsychiatric Imaging, Turku, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland.
| |
Collapse
|
4
|
The association between substance P and white matter integrity in medication-naive patients with major depressive disorder. Sci Rep 2017; 7:9707. [PMID: 28852030 PMCID: PMC5575350 DOI: 10.1038/s41598-017-10100-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 07/28/2017] [Indexed: 01/06/2023] Open
Abstract
Substance P (SP) has been implicated in major depressive disorder (MDD), with SP antagonists being studied as potential antidepressants. Although impaired neural plasticity is considered a key mechanism in MDD pathophysiology, the association between SP and brain structural changes in depression has not been investigated. We investigated the correlations between SP levels and white matter (WM) integrity in 42 medication-naive patients with MDD and 57 healthy controls (HCs). Plasma levels of SP were determined, and diffusion tensor imaging (DTI) was performed to investigate microstructural changes in WM tracts. In patients, negative correlations between SP levels and fractional anisotropy (FA) values of the forceps minor of the corpus callosum, and positive correlations between SP levels and radial diffusivity (RD) and mean diffusivity (MD) values of the right corticospinal tract (CST) were observed, with no significant correlations in HCs. Linear regression analyses showed SP levels to significantly predict FA values of the forceps minor, and RD and MD values of the right CST in patients, but not in HCs. We consider our findings to contribute to the neurobiological evidence on the association between SP and brain structural changes in depression, which may be related with the pathophysiology and treatment of MDD.
Collapse
|
5
|
Mannangatti P, Sundaramurthy S, Ramamoorthy S, Jayanthi LD. Differential effects of aprepitant, a clinically used neurokinin-1 receptor antagonist on the expression of conditioned psychostimulant versus opioid reward. Psychopharmacology (Berl) 2017; 234:695-705. [PMID: 28013351 PMCID: PMC5266628 DOI: 10.1007/s00213-016-4504-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/08/2016] [Indexed: 01/29/2023]
Abstract
RATIONALE Neurokinin-1 receptor (NK1R) signaling modulates behaviors associated with psychostimulants and opioids. Psychostimulants, such as amphetamine (AMPH) and cocaine, bind to monoamine transporters and alter their functions. Both dopamine and norepinephrine transporters are regulated by NK1R activation suggesting a role for NK1R mediated catecholamine transporter regulation in psychostimulant-mediated behaviors. OBJECTIVES The effect of in vivo administration of aprepitant (10 mg/kg) on the expression of AMPH (0.5 and 2 mg/kg) and cocaine (5 and 20 mg/kg)-induced conditioned place preference (CPP) as well as locomotor activation was examined in C57BL/6J mice. The effect of aprepitant on morphine (1 and 5 mg/kg)-induced CPP was also examined to identify the specific actions of aprepitant on psychostimulant versus opioid-induced behaviors. RESULTS Aprepitant administration significantly attenuated the CPP expression and locomotor activation produced by AMPH and cocaine. In contrast, aprepitant significantly enhanced the expression of CPP produced by morphine while significantly suppressing the locomotor activity of the mice conditioned with morphine. Aprepitant by itself did not induce significant CPP or conditioned place aversion or locomotor activation or suppression. CONCLUSIONS Attenuation of AMPH or cocaine-induced CPP and locomotor activation by aprepitant suggests a role for NK1R signaling in psychostimulant-mediated behaviors. Stimulation of morphine-induced CPP expression and suppression of locomotor activity of morphine-conditioned mice suggest differential effects of NK1R antagonism on conditioned psychostimulant versus opioid reward. Collectively, these findings indicate that clinically used NK1R antagonist, aprepitant may serve as a potential therapeutic agent in the treatment of psychostimulant abuse.
Collapse
Affiliation(s)
| | | | | | - Lankupalle D Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
6
|
Schank JR, Heilig M. Substance P and the Neurokinin-1 Receptor: The New CRF. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:151-175. [DOI: 10.1016/bs.irn.2017.06.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
7
|
Neurokinin-1 receptor antagonism attenuates neuronal activity triggered by stress-induced reinstatement of alcohol seeking. Neuropharmacology 2015; 99:106-14. [PMID: 26188146 DOI: 10.1016/j.neuropharm.2015.07.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 11/21/2022]
Abstract
Substance P (SP) and its cognate neurokinin-1 receptor (NK1R) are involved in alcohol-related behaviors. We have previously reported that NK1R antagonism attenuates stress-induced reinstatement of alcohol seeking and suppresses escalated alcohol self-administration, but does not affect primary reinforcement or cue-induced reinstatement. Here, we administered an NK1R antagonist or vehicle prior to footshock-induced reinstatement of alcohol seeking, and mapped the resulting neuronal activation using Fos immunohistochemistry. As expected, vehicle treated animals exposed to footshock showed induction of Fos immunoreactivity in several regions of the brain stress circuitry, including the amygdala (AMG), nucleus accumbens (NAC), dorsal raphe nucleus (DR), prefrontal cortex (PFC), and bed nucleus of the stria terminalis (BNST). NK1R antagonism selectively suppressed the stress-induced increase in Fos in the DR and NAC shell. In the DR, Fos-induction by stress largely overlapped with tryptophan hydroxylase (TrpH), indicating activation of serotonergic neurons. Of NAC shell neurons activated during stress-induced reinstatement of alcohol seeking, about 30% co-expressed dynorphin (DYN), while 70% co-expressed enkephalin (ENK). Few (<1%) activated NAC shell neurons coexpressed choline acetyltransferase (ChAT), which labels the cholinergic interneurons of this region. Infusion of the NK1R antagonist L822429 into the NAC shell blocked stress-induced reinstatement of alcohol seeking. In contrast, L822429 infusion into the DR had no effect, suggesting that the influence of NK1R signaling on neuronal activity in the DR is indirect. Taken together, our results outline a potential pathway through which endogenous NK1R activation mediates stress-induced alcohol seeking.
Collapse
|
8
|
Abstract
Stress can trigger drug-seeking behavior, increase self-administration rates, and enhance drug reward. A number of stress-related neuropeptides have been shown to mediate these behavioral processes. The most studied peptide in this category is corticotropin-releasing hormone (CRH), which has been shown to mediate stress-induced reinstatement of drug seeking, escalated self-administration, and drug withdrawal, but it does not seem to be involved in baseline drug self-administration or cue-induced reinstatement. This pattern of effects holds for many classes of drugs, including alcohol, opiates, and psychostimulants. The neurokinin-1 receptor (NK1R) is the preferred receptor for the endogenous stress-related neuropeptide substance P (SP). The SP/NK1R system is a major mediator of stress and anxiety, and over the last several years, it has been demonstrated that the SP/NK1R system can have effects similar to those of CRH on drug taking and drug seeking. Specifically, NK1R inhibition attenuates escalated self-administration of alcohol as well as stress-induced reinstatement of alcohol and cocaine seeking; however, in contrast to other stress systems, the NK1R also appears to have a role in primary reward and reinforcement for opiates. This review outlines the role of NK1R in drug-seeking behaviors and highlights recent results from clinical studies that suggest that the NK1R may be a promising drug target going forward.
Collapse
Affiliation(s)
- Jesse R Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia
| |
Collapse
|
9
|
Iceman KE, Harris MB. A group of non-serotonergic cells is CO2-stimulated in the medullary raphé. Neuroscience 2013; 259:203-13. [PMID: 24333211 DOI: 10.1016/j.neuroscience.2013.11.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/13/2013] [Accepted: 11/30/2013] [Indexed: 01/22/2023]
Abstract
Serotonin/substance P synthesizing cells in the raphé nuclei of the brain are candidates for designation as central chemoreceptors that are stimulated by CO2/pH. We have previously demonstrated that these neurons are CO2-stimulated in situ. Evidence also suggests that CO2-inhibited raphé neurons recorded in vitro and in situ synthesize GABA. Unknown is whether there are other types of chemosensitive cells in the raphé. Here, we showed that a previously unrecognized pool of raphé neurons also exhibit chemosensitivity, and that they are not serotonergic. We used extracellular recording of individual raphé neurons in the unanesthetized juvenile rat in situ perfused decerebrate brainstem preparation to assess chemosensitivity of raphé neurons. Subsequent juxtacellular labeling of individually recorded cells, and immunohistochemistry for the serotonin synthesizing enzyme tryptophan hydroxylase and for neurokinin-1 receptor (NK1R; the receptor for substance P) indicated a group of CO2-stimulated cells that are not serotonergic, but express NK1R and are closely apposed to surrounding serotonergic cells. CO2-stimulated 5-HT and non-5-HT cells constitute distinct groups that have different firing characteristics and hypercapnic sensitivities. Non-5-HT cells fire faster and are more robustly stimulated by CO2 than are 5-HT cells. Thus, we have characterized a previously unrecognized type of CO2-stimulated medullary raphé neuron that is not serotonergic, but may receive input from neighboring serotonin/substance P synthesizing chemosensitive neurons. The potential network properties of the three types of chemosensitive raphé neurons (the present non-5-HT cells, serotonergic cells, and CO2-inhibited cells) remain to be elucidated.
Collapse
Affiliation(s)
- K E Iceman
- Institute of Arctic Biology, University of Alaska, Fairbanks, AK 99775, USA; Department of Biology and Wildlife, University of Alaska, Fairbanks, AK 99775, USA.
| | - M B Harris
- Institute of Arctic Biology, University of Alaska, Fairbanks, AK 99775, USA; Department of Biology and Wildlife, University of Alaska, Fairbanks, AK 99775, USA
| |
Collapse
|
10
|
Muto Y, Sakai A, Sakamoto A, Suzuki H. Activation of NK₁ receptors in the locus coeruleus induces analgesia through noradrenergic-mediated descending inhibition in a rat model of neuropathic pain. Br J Pharmacol 2012; 166:1047-57. [PMID: 22188400 DOI: 10.1111/j.1476-5381.2011.01820.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE The locus coeruleus (LC) is a major source of noradrenergic projections to the dorsal spinal cord, and thereby plays an important role in the modulation of nociceptive information. The LC receives inputs from substance P (SP)-containing fibres from other regions, and expresses the NK(1) tachykinin receptor, a functional receptor for SP. In the present study, we investigated the roles of SP in the LC in neuropathic pain. EXPERIMENTAL APPROACH Chronic constriction injury (CCI) of the left sciatic nerve was performed in rats to induce neuropathic pain. After development of neuropathic pain, SP was injected into the LC and the nocifensive behaviours were assessed. The involvement of noradrenergic descending inhibition in SP-induced analgesia was examined by i.t. administration of yohimbine, an α(2) -adrenoceptor antagonist. NK(1) receptor expression in the LC was examined by immunohistochemistry. KEY RESULTS In CCI rats, mechanical allodynia was alleviated by SP injection into the LC. These effects were abolished by prior injection of WIN 51708, an NK(1) receptor antagonist, into the LC or i.t. treatment with yohimbine. NK(1) receptor-like immunoreactivity was observed in noradrenergic neurons throughout the LC in intact rats, and remained unchanged after CCI. CONCLUSION AND IMPLICATIONS SP in the LC exerted analgesic effects on neuropathic pain through NK(1) receptor activation and resulted in facilitation of spinal noradrenergic transmission. Accordingly, manipulation of the SP/NK(1) receptor signalling pathway in the LC may be a promising strategy for effective treatment of neuropathic pain.
Collapse
Affiliation(s)
- Y Muto
- Department of Anesthesiology, Nippon Medical School, Tokyo, Japan
| | | | | | | |
Collapse
|
11
|
Schank JR, Ryabinin AE, Giardino WJ, Ciccocioppo R, Heilig M. Stress-related neuropeptides and addictive behaviors: beyond the usual suspects. Neuron 2012; 76:192-208. [PMID: 23040815 PMCID: PMC3495179 DOI: 10.1016/j.neuron.2012.09.026] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Addictive disorders are chronic, relapsing conditions that cause extensive disease burden. Genetic factors partly account for susceptibility to addiction, but environmental factors such as stressful experiences and prolonged exposure of the brain to addictive drugs promote its development. Progression to addiction involves neuroadaptations within neurocircuitry that mediates stress responses and is influenced by several peptidergic neuromodulators. While corticotrophin releasing factor is the prototypic member of this class, recent work has identified several additional stress-related neuropeptides that play an important role in regulation of drug intake and relapse, including the urocortins, nociceptin, substance P, and neuropeptide S. Here, we review this emerging literature, discussing to what extent the properties of these neuromodulators are shared or distinct and considering their potential as drug targets.
Collapse
Affiliation(s)
- Jesse R. Schank
- Laboratory of Clinical and Translational Studies, National Inst. on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Andrey E. Ryabinin
- Dept. of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239-3098
| | - William J. Giardino
- Dept. of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239-3098
| | - Roberto Ciccocioppo
- Dept. of Experimental Medicine and Public Health, Camerino University, Italy
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies, National Inst. on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
12
|
Parker LM, Tallapragada VJ, Kumar NN, Goodchild AK. Distribution and localisation of Gα proteins in the rostral ventrolateral medulla of normotensive and hypertensive rats: focus on catecholaminergic neurons. Neuroscience 2012; 218:20-34. [PMID: 22626648 DOI: 10.1016/j.neuroscience.2012.05.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 05/11/2012] [Accepted: 05/12/2012] [Indexed: 02/07/2023]
Abstract
About 860 G-protein-coupled receptors (GPCRs) mediate their actions via heterotrimeric G-proteins. Their activation releases Gα from Gβλ subunits. The type of Gα subunit dictates the major signalling proteins involved: adenylyl cyclase, PLC and rhoGEF. The rostral ventrolateral medulla (RVLM), containing the rostral C1 (rC1) cell group, sets and maintains the tonic and reflex control of blood pressure and a plethora of inputs converge onto these neurons. We determined the relative abundance of 10 Gα subunit mRNAs, representing the four major families, within the RVLM, using quantitative RT-PCR. In situ hybridisation (ISH) combined with immunohistochemistry (IHC) was used to quantify and compare this expression in rC1 with that in the A1 and A5 cell groups. The relative abundance of Gα subunit mRNAs and a comparison of gene expression levels were quantitatively determined in normotensive and hypertensive rat strains. All 10 Gα mRNAs were detected in the RVLM of Sprague-Dawley (SD) rats with relative abundance such that Gαs>Gαi2>Gαo>Gαq>GαL>Gα11>Gαi3>Gαi1>Gα12>Gα13. The high abundance of Gα mRNAs signalling via adenylyl cyclase indicates the importance of associated GPCRs. Within the rC1 and A1 groups similar differential Gα mRNA expression profiles were seen with Gαs being found in all rC1 cells, Gα11 absent and Gαi3 rarely expressed. Thus functionally distinct subgroups exist within the rC1 and A1 cell groups as differing distributions of Gα subunits must reflect the array of GPCRs that influence their activity. In contrast, all A5 cells expressed all Gα mRNAs suggesting a functionally homogeneous group. When the 10 Gα mRNAs of the RVLM in spontaneously hypertensive rats (SHR) were compared quantitatively to Wistar-Kyoto (WKY), only Gαs and Gα12 were significantly elevated. However when the expression in normotensive SD and WKY was compared with SHR no significant differences were evident. These findings demonstrate a range of GPCR signalling capabilities in brainstem neurons important for homeostasis and suggest a prominent role for signalling via adenylyl cyclase.
Collapse
Affiliation(s)
- L M Parker
- The Australian School of Advanced Medicine, 2 Technology Place, Macquarie University, 2109 NSW, Australia
| | | | | | | |
Collapse
|
13
|
Lin LH, Nitschke Dragon D, Talman WT. Collateral damage and compensatory changes after injection of a toxin targeting neurons with the neurokinin-1 receptor in the nucleus tractus solitarii of rat. J Chem Neuroanat 2012; 43:141-8. [PMID: 22414622 DOI: 10.1016/j.jchemneu.2012.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 02/23/2012] [Accepted: 02/24/2012] [Indexed: 02/02/2023]
Abstract
Injection into the nucleus tractus solitarii (NTS) of toxins that target substance P (SP) receptors ablates neurons that express neurokinin-1 (NK1) receptors, attenuates baroreflexes, and results in increased lability of arterial pressure. We and others have shown that the toxin leads to loss of neurons containing SP receptors and loss of GABAergic neurons in the NTS; but given that neither type neuron is thought to be integral to baroreflex transmission in NTS, mechanisms responsible for the cardiovascular changes remained unclear. Because NK1 receptors colocalize with N-methyl-d-aspartate (NMDA) receptors and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in NTS and because glutamate transmission may be integral to baroreflex transmission in the NTS we hypothesized that the toxic lesions may interrupt mechanisms for glutamate transmission. Interruption of those mechanisms could be responsible for the cardiovascular effects. We tested the hypothesis by performing fluorescent immunohistochemistry, confocal microscopy and image analysis after injecting stabilized SP-SAP (SSP-SAP) unilaterally into the NTS. We assessed changes in immunoreactivity (IR) of NMDA receptor subunit 1 (NMDAR1), AMPA receptor subunit 2 (GluR2), and 3 types of vesicular glutamate transporters (VGluT) as well as IR of gamma-aminobutyric acid receptors type b (GABAb), neuronal nitric oxide synthase (nNOS), tyrosine hydroxylase (TH), and protein gene product 9.5 (PGP 9.5), a neuronal marker, in the NTS. When compared to that of the same section of the un-injected NTS, IR decreased significantly in the injected side for NMDAR1 (p<0.01), GluR2 (p<0.01), VGluT3 (p<0.01), GABAb (p<0.001), and PGP9.5 (p<0.001). In contrast, IR for VGluT1 (p<0.001), VGluT2 (p<0.001), nNOS (p<0.001), and TH (p<0.001) increased significantly. We conclude that pathologic effects following ablation of neurons with NK1 receptors in NTS may result from interruption of neurotransmission through other neurochemical systems associated with NK1 receptors-containing neurons.
Collapse
Affiliation(s)
- Li-Hsien Lin
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
14
|
Roux JC, Panayotis N, Dura E, Villard L. Progressive noradrenergic deficits in the locus coeruleus of Mecp2 deficient mice. J Neurosci Res 2010; 88:1500-9. [PMID: 19998492 DOI: 10.1002/jnr.22312] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Methyl-CpG binding protein 2 (MeCP2) is a transcriptional regulator. Mutations in this gene cause a wide range of neurological disorders. Mecp2 deficiency has been previously associated to catecholaminergic dysfunctions leading to autonomic defects in the brainstem and the sympathoadrenergic system of the mouse. The present study was undertaken to determine if the locus coeruleus (LC), the main noradrenergic cell group of the brain, is affected. Using real type PCR, we found a reduction of the tyrosine hydroxylase (Th) mRNA level, the rate-limiting enzyme in catecholamine synthesis, in the whole pons of P15 (-36%), P30 (-47%) and P50 (-42%) Mecp2 null male as well as in adult heterozygous female (-44%) mice. Using immunoquantification we did not observe any difference of the Th staining level in P30 null male mice. However at P50, we demonstrated a significant decrease in both the Th staining level (-24%), and the number of Th-positive neurons (-23%). We subsequently characterized a reduction (-28%) of the dendritic density of the Th-positive fibers surrounding the LC in P50 null male mice. In heterozygous female mice immunoquantification did not revealed significant modifications, but only a tendency towards reduction. Finally, we did not found any apoptotic neurons in the pons indicating that LC neurons are not dying but are more likely loosing their catecholaminergic phenotype. In conclusion, our results showing a progressive catecholaminergic deficit in the LC of Mecp2 deficient null male mice could open new perspectives to better understand the autonomic and cognitive deficits due to the lack of Mecp2.
Collapse
|
15
|
Rajkumar R, Mahesh R. Assessing the neuronal serotonergic target-based antidepressant stratagem: impact of in vivo interaction studies and knockout models. Curr Neuropharmacol 2010; 6:215-34. [PMID: 19506722 PMCID: PMC2687932 DOI: 10.2174/157015908785777256] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 06/05/2008] [Accepted: 06/12/2008] [Indexed: 12/26/2022] Open
Abstract
Depression remains a challenge in the field of affective neuroscience, despite a steady research progress. Six out of nine basic antidepressant mechanisms rely on serotonin neurotransmitter system. Preclinical studies have demonstrated the significance of serotonin receptors (5-HT1-3,6,7), its signal transduction pathways and classical down stream targets (including neurotrophins, neurokinins, other peptides and their receptors) in antidepressant drug action. Serotonergic control of depression embraces the recent molecular requirements such as influence on proliferation, neurogenesis, plasticity, synaptic (re)modeling and transmission in the central nervous system. The present progress report analyses the credibility of each protein as therapeutically relevant target of depression. In vivo interaction studies and knockout models which identified these targets are foreseen to unearth new ligands and help them transform to drug candidates. The importance of the antidepressant assay selection at the preclinical level using salient animal models/assay systems is discussed. Such test batteries would definitely provide antidepressants with faster onset, efficacy in resistant (and co-morbid) types and with least adverse effects. Apart from the selective ligands, only those molecules which bring an overall harmony, by virtue of their affinities to various receptor subtypes, could qualify as effective antidepressants. Synchronised modulation of various serotonergic sub-pathways is the basis for a unique and balanced antidepressant profile, as that of fluoxetine (most exploited antidepressant) and such a profile may be considered as a template for the upcoming antidepressants. In conclusion, 5-HT based multi-targeted antidepressant drug discovery supported by in vivo interaction studies and knockout models is advocated as a strategy to provide classic molecules for clinical trials.
Collapse
Affiliation(s)
- R Rajkumar
- Pharmacy Group, FD-III, Vidya Vihar, Birla Institute of Technology & Science, Pilani, Rajasthan-333031, India.
| | | |
Collapse
|
16
|
de Carvalho D, Bícego KC, de Castro OW, da Silva GS, Garcia-Cairasco N, Gargaglioni LH. Role of neurokinin-1 expressing neurons in the locus coeruleus on ventilatory and cardiovascular responses to hypercapnia. Respir Physiol Neurobiol 2010; 172:24-31. [DOI: 10.1016/j.resp.2010.04.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 04/04/2010] [Accepted: 04/08/2010] [Indexed: 10/19/2022]
|
17
|
Yan TC, McQuillin A, Thapar A, Asherson P, Hunt SP, Stanford SC, Gurling H. NK1 (TACR1) receptor gene 'knockout' mouse phenotype predicts genetic association with ADHD. J Psychopharmacol 2010; 24:27-38. [PMID: 19204064 PMCID: PMC3943619 DOI: 10.1177/0269881108100255] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Mice with functional genetic ablation of the Tacr1 (substance P-preferring receptor) gene (NK1R-/-) are hyperactive. Here, we investigated whether this is mimicked by NK1R antagonism and whether dopaminergic transmission is disrupted in brain regions that govern motor performance. The locomotor activity of NK1R-/- and wild-type mice was compared after treatment with an NK1R antagonist and/or psychostimulant (d-amphetamine or methylphenidate). The inactivation of NK1R (by gene mutation or receptor antagonism) induced hyperactivity in mice, which was prevented by both psychostimulants. Using in vivo microdialysis, we then compared the regulation of extracellular dopamine in the prefrontal cortex (PFC) and striatum in the two genotypes. A lack of functional NK1R reduced (>50%) spontaneous dopamine efflux in the prefrontal cortex and abolished the striatal dopamine response to d-amphetamine. These behavioural and neurochemical abnormalities in NK1R-/- mice, together with their atypical response to psychostimulants, echo attention deficit hyperactivity disorder (ADHD) in humans. These findings prompted genetic studies on the TACR1 gene (the human equivalent of NK1R) in ADHD patients in a case-control study of 450 ADHD patients and 600 screened supernormal controls. Four single-nucleotide polymorphisms (rs3771829, rs3771833, rs3771856, and rs1701137) at the TACR1 gene, previously known to be associated with bipolar disorder or alcoholism, were strongly associated with ADHD. In conclusion, our proposal that NK1R-/- mice offer a mouse model of ADHD was borne out by our human studies, which suggest that DNA sequence changes in and around the TACR1 gene increase susceptibility to this disorder.
Collapse
Affiliation(s)
- TC Yan
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - A McQuillin
- Molecular Psychiatry Laboratory, Department of Mental Health Sciences, Royal Free & UCL School of Medicine, London, UK
| | - A Thapar
- Department of Psychological Medicine, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - P Asherson
- ADHD genetics group, MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
| | - SP Hunt
- Department of Cell and Developmental Biology, University College London, London, UK
| | - SC Stanford
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - H Gurling
- Molecular Psychiatry Laboratory, Department of Mental Health Sciences, Royal Free & UCL School of Medicine, London, UK
| |
Collapse
|
18
|
Adrenergic mechanisms of the Kölliker-Fuse/A7 area on the control of water and sodium intake. Neuroscience 2009; 164:370-9. [DOI: 10.1016/j.neuroscience.2009.08.048] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 08/21/2009] [Accepted: 08/22/2009] [Indexed: 11/20/2022]
|
19
|
Yan TC, Hunt SP, Stanford SC. Behavioural and neurochemical abnormalities in mice lacking functional tachykinin-1 (NK1) receptors: A model of attention deficit hyperactivity disorder. Neuropharmacology 2009; 57:627-35. [DOI: 10.1016/j.neuropharm.2009.08.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 08/12/2009] [Accepted: 08/13/2009] [Indexed: 01/23/2023]
|
20
|
Commons KG. Neuronal pathways linking substance P to drug addiction and stress. Brain Res 2009; 1314:175-82. [PMID: 19913520 DOI: 10.1016/j.brainres.2009.11.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 11/03/2009] [Accepted: 11/05/2009] [Indexed: 01/14/2023]
Abstract
Accumulating evidence suggests that the neuropeptide substance P (SP) and its principal receptor neurokinin 1 (NK1) play a specific role in the behavioral response to opioids and stress that may help to initiate and maintain addictive behavior. In animal models, the NK1 receptor is required for opioids to produce their rewarding and motivational effects. SP neurotransmission is also implicated in the behavioral response to stress and in the process of drug sensitization, potentially contributing to vulnerability to addiction or relapse. However, SP neurotransmission only plays a minor role in opioid-mediated antinociception and the development of opioid tolerance. Moreover, the effects of SP on addiction-related behavior are selective for opioids and evidence supporting a role in the response to cocaine or psychostimulants is less compelling. This review will summarize the effects of SP neurotransmission on opioid-dependent behaviors and correlate them with potential contributing neural pathways. Specifically, SP neurotransmission within components of the basal forebrain particularly the nucleus accumbens and ventral pallidum as well as actions within the ascending serotonin system will be emphasized. In addition, cellular- or network-level interactions between opioids and SP signaling that may underlie the specificity of their relationship will be reviewed.
Collapse
Affiliation(s)
- K G Commons
- Department of Anesthesiology, Perioperative, and Pain Medicine, Children's Hospital Boston, Department of Anaesthesia, Harvard Medical School, 300 Longwood Ave., Enders 1206, Boston, MA, USA.
| |
Collapse
|
21
|
Mukda S, Møller M, Ebadi M, Govitrapong P. The modulatory effect of substance P on rat pineal norepinephrine release and melatonin secretion. Neurosci Lett 2009; 461:258-61. [DOI: 10.1016/j.neulet.2009.06.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 06/11/2009] [Accepted: 06/13/2009] [Indexed: 11/30/2022]
|
22
|
Min MY, Shih PY, Wu YW, Lu HW, Lee ML, Yang HW. Neurokinin 1 receptor activates transient receptor potential-like currents in noradrenergic A7 neurons in rats. Mol Cell Neurosci 2009; 42:56-65. [PMID: 19463951 DOI: 10.1016/j.mcn.2009.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 05/12/2009] [Accepted: 05/14/2009] [Indexed: 10/20/2022] Open
Abstract
Noradrenergic (NAergic) A7 neurons are involved in modulating nociception by releasing noradrenaline in the dorsal spinal cord. Since NAergic A7 neurons receive dense Substance P (Sub-P) releasing terminals from ventromedial medulla, here we tested the effect of Sub-P on them. Bath application of Sub-P induced an inward current (I(Sub-P)) in NAergic neurons, which was significantly blocked by Neurokinin 1 (NK1) receptor antagonist. The I(Sub-P) was reversed at approximately -20 mV, blocked by several TRP channel blockers, enhanced by OAG and negatively regulated by PKC. Immunohistochemistry staining showed that NAergic A7 neurons express high level of TRPC6 channel proteins, which is consistent with pharmacological properties of I(Sub-P) shown above, as TRPC6 channel is shown to be augmented by OAG and inhibited by PKC. In conclusion, the above results provide mechanism underlying postsynaptic action of Sub-P on NAergic A7 neurons and a role for TRPC6 channel in NAergic pain modulation.
Collapse
Affiliation(s)
- Ming-Yuan Min
- Institute of Zoology, College of Life Science, National Taiwan University, Taipei 106, Taiwan; Department of Life Science, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | |
Collapse
|
23
|
Le Brun I, Dufour A, Crest M, Szabó G, Erdelyi F, Baude A. Differential expression of Nk1 and NK3 neurokinin receptors in neurons of the nucleus tractus solitarius and the dorsal vagal motor nucleus of the rat and mouse. Neuroscience 2008; 152:56-64. [PMID: 18222044 DOI: 10.1016/j.neuroscience.2007.12.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 12/07/2007] [Accepted: 12/11/2007] [Indexed: 12/16/2022]
Abstract
Tachykinins (substance P, neurokinin A and neurokinin B) influence autonomic functions by modulating neuron activity in nucleus tractus solitarius (NTS) and dorsal motor nucleus of the vagus (DMV) through activation of neurokinin receptors NK1 and NK3. Our purpose was to identify and define by neurochemical markers, the subpopulations of NK1 and NK3 expressing neurons in NTS and DMV of rat and mouse. Because the distribution of the NK1 and NK3 expressing neurons overlaps, co-expression for both receptors was tested. By double labeling, we show that NK1 and NK3 were not co-expressed in NTS neurons. In the DMV, most of neurons (87%) were immunoreactive for only one of the receptors and 34% of NK1 neurons, 7% of NK3 neurons and 12% of NK1-NK3 neurons were cholinergic neurons. None of the neurons immunoreactive for NK1 or NK3 were positive for tyrosine hydroxylase, suggesting that catecholaminergic cells of the NTS (A2 and C2 groups) did not express neurokinin receptors. The presence of NK1 and NK3 was examined in GABAergic interneurons of the NTS and DMV by using GAD65-EGFP transgenic mouse. Immunoreactivity for NK1 or NK3 was found in a subpopulation of GAD65-EGFP cells. A majority (60%) of NK3 cells, but only 11% of the NK1 cells, were GAD65-EGFP cells. In conclusion, tachykinins, through differential expression of neurokinin receptors, may influence the central regulation of vital functions by acting on separate neuron subpopulations in NTS and DMV. Of particular interest, tachykinins may be involved in inhibitory mechanisms by acting directly on local GABAergic interneurons. Our results support a larger contribution of NK3 compared with NK1 in mediating inhibition in NTS and DMV.
Collapse
Affiliation(s)
- I Le Brun
- Laboratoire de Neurophysiologie Cellulaire, Université de la Méditerranée, CNRS UMR 6150, IFR Jean-Roche, Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille 20, France
| | | | | | | | | | | |
Collapse
|
24
|
Nattie E, Li A. Central chemoreception is a complex system function that involves multiple brain stem sites. J Appl Physiol (1985) 2008; 106:1464-6. [PMID: 18467549 DOI: 10.1152/japplphysiol.00112.2008] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Eugene Nattie
- 706E Borwell Bldg., Dept. of Physiology, Dartmouth Medical School, Lebanon, NH 03755-0001, USA.
| | | |
Collapse
|
25
|
Min MY, Wu YW, Shih PY, Lu HW, Lin CC, Wu Y, Li MJ, Yang HW. Physiological and morphological properties of, and effect of substance P on, neurons in the A7 catecholamine cell group in rats. Neuroscience 2008; 153:1020-33. [PMID: 18440151 DOI: 10.1016/j.neuroscience.2008.03.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 03/02/2008] [Accepted: 03/03/2008] [Indexed: 11/26/2022]
Abstract
The A7 catecholamine cell group consists of noradrenergic (NAergic) neurons that project to the dorsal horn of the spinal cord. Here, we characterized their morphology and physiology properties and tested the effect of substance P (Sub-P) on them, since the results of many morphological studies suggest that A7 neurons are densely innervated by Sub-P-releasing terminals from nuclei involved in the descending inhibitory system, such as the lateral hypothalamus and periaqueductal gray area. Whole cell recordings were made from neurons located approximately 200 microm rostral to the trigeminal motor nucleus (the presumed A7 area) in sagittal brainstem slices from rats aged 7-10 days. After recording, the neurons were injected with biocytin and immunostained with antibody against dopamine-beta-hydroxylase (DBH). DBH-immunoreactive (ir) cells were presumed to be NAergic neurons. They had a large somata diameter ( approximately 20 microm) and relatively simple dendritic branching patterns. They fired action potentials (AP) spontaneously with or without blockade of synaptic inputs, and had similar properties to those of NAergic neurons in other areas, including the existence of calcium channel-mediated APs and a voltage-dependent delay in initiation of the AP (an indicator of the existence of A-type potassium currents) and an ability to be hyperpolarized by norepinephrine. Furthermore, in all DBH-ir neurons tested, Sub-P caused depolarization of the membrane potential and an increase in neuronal firing rate by acting on neurokinin-1 receptors. Non-DBH-ir neurons with a smaller somata size were also found in the A7 area. These showed great diversity in firing patterns and about half were depolarized by Sub-P. Morphological examination suggested that the non-DBH-ir neurons form contacts with DBH-ir neurons. These results provide the first description of the intrinsic regulation of membrane properties of, and the excitatory effect of Sub-P on, A7 area neurons, which play an important role in pain regulation.
Collapse
Affiliation(s)
- M-Y Min
- Department of Life Science, College of Life Science, National Taiwan University, No. 1 Sec. 4 Roosevelt Road, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Ebner K, Singewald N. Stress-induced release of substance P in the locus coeruleus modulates cortical noradrenaline release. Naunyn Schmiedebergs Arch Pharmacol 2007; 376:73-82. [PMID: 17879086 DOI: 10.1007/s00210-007-0185-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Accepted: 08/20/2007] [Indexed: 10/22/2022]
Abstract
Several lines of evidence implicate the neuropeptide substance P (SP) in the modulation of emotional behavior. Interaction between SP and noradrenergic systems has been proposed to be important in the regulation of stress, depression, and anxiety mechanisms; however, most evidence so far is based on studies in unchallenged and/or anesthetized animals. Thus, by using a dual-probe microdialysis approach in freely moving animals, the aim of the present study was to investigate whether a relevant stressor can trigger the release of SP in the locus coeruleus (LC) and whether and how this response modulates noradrenaline (NA) transmission both in the LC and in the medial prefrontal cortex (mPFC), an important LC terminal region involved in emotional processing. While confirming previous reports that neurokinin 1 receptor (NK1R) antagonists activate cortical noradrenergic transmission under resting conditions, we present evidence that this interaction is opposite during stress challenge. Our results show that exposure to forced swimming considerably enhanced the release of SP and NA in the LC. Administration of a selective NK1R antagonist into the LC potentiated this NA response within the LC but abolished the stress-induced increase in NA release within the mPFC. These findings demonstrate stress-induced increase in endogenous extracellular SP levels within the LC exerting a facilitatory effect on the noradrenergic pathway to the mPFC. The attenuation of stress-induced hyperactivation of this pathway by NK1R antagonists, presumably via enhancing NA and autoinhibition in the LC, may contribute to the therapeutic efficacy of these drugs known to ameliorate symptoms of stress-related disorders.
Collapse
Affiliation(s)
- Karl Ebner
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | | |
Collapse
|
27
|
Fisher AS, Stewart RJ, Yan T, Hunt SP, Stanford SC. Disruption of noradrenergic transmission and the behavioural response to a novel environment in NK1R-/- mice. Eur J Neurosci 2007; 25:1195-204. [PMID: 17331215 DOI: 10.1111/j.1460-9568.2007.05369.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The behaviour of neurokinin-1-receptor gene knockout (NK1R-/-) mice, which lack functional, substance P-preferring receptors, resembles that of NK1R+/+ mice treated with an antidepressant. Because all antidepressants increase central monoamine transmission, we have investigated whether noradrenergic transmission is increased in NK1R-/- mice and, if so, whether this could influence their behaviour. In anaesthetized subjects, the concentration of extracellular noradrenaline in NK1R-/- mice was two-fourfold greater than in NK1R+/+ mice. Systemic administration of the alpha2-adrenoceptor antagonist, 2-(2,3-dihydro-2-methoxy-1,4-benzodioxan-2-yl)-4,5-dihydro-1H-imidazoline (RX 821002), in anaesthetized or freely moving animals increased extracellular noradrenaline in NK1R+/+ mice only. This suggests that the function of alpha2a-autoreceptors, which modulate noradrenergic transmission, is impaired in NK1R-/- mice. Consistent with this, [35S]GTPgammaS binding to activated alpha2a-adrenoceptors was lower (-70%) in the locus coeruleus, but not the frontal cortex, of NK1R-/- mice compared with their NK1R+/+ counterparts. RX 821002-pretreatment, followed by retrodialysis of the noradrenaline reuptake inhibitor, desipramine, into the frontal cortex of anaesthetized mice increased extracellular noradrenaline to the same extent in the two genotypes. Western blots confirmed that there was no difference in the amount of noradrenaline transporter protein in NK1R-/- and NK1R+/+ mice. Finally, the effects of RX 821002 on certain behaviours in a light/dark exploration box were blunted in NK1R-/- mice, but there was no consistent effect on anxiety-like behaviour in the two genotypes. It is concluded that the greater basal efflux of noradrenaline in NK1R-/- mice is explained by increased transmitter release, coupled with desensitization of somatodendritic alpha2a-adrenoceptors. These changes could contribute to the difference in the behavioural phenotypes.
Collapse
Affiliation(s)
- Amy S Fisher
- Department of Anatomy and Development Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | |
Collapse
|
28
|
Nattie E, Li A. Neurokinin-1 receptor-expressing neurons in the ventral medulla are essential for normal central and peripheral chemoreception in the conscious rat. J Appl Physiol (1985) 2006; 101:1596-606. [PMID: 16902062 DOI: 10.1152/japplphysiol.00347.2006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurokinin-1 receptor immunoreactive (NK1R-ir) neurons and processes are widely distributed within the medulla, prominently at central chemoreceptor sites. Focal lesions of NK1R-ir neurons in the medullary raphe or the retrotrapezoid nucleus partially reduced the CO(2) response in conscious rats. We ask if NK1R-ir cells and processes over a wide region of the ventral medulla are essential for central and peripheral chemoreception by cisterna magna injection of SSP-SAP, a high-affinity version of substance P-saporin. After 22 days, NK1R-ir cell loss was -79% in the retrotrapezoid nucleus and -65% in the A5 region, which lie close to the ventral surface, and -38% in the medullary raphe and -49% in the pre-Bötzinger complex/rostral ventral respiratory group, which lie deeper. Dorsal chemoreceptor sites, the caudal nucleus tractus solitarius and the A6 region, were unaffected. At 8 and 22 days, these lesions produced 1) hypoventilation during air breathing in wakefulness ( approximately 8%) and in non-rapid eye movement (NREM) ( approximately 9%) and rapid eye movement ( approximately 14%) sleep, as measured over a 4-h period; 2) a substantially reduced ventilatory response to 7% CO(2) by 61% in wakefulness and 46-57% in NREM sleep; and 3) a decreased ventilatory response to 12% O(2) by 40% in wakefulness and 35% in NREM sleep at 8 days, with partial recovery by 22 days. NK1R-ir neurons in the ventral medulla are essential for normal central chemoreception, provide a drive to breathe, and modulate the peripheral chemoreceptor responses. These effects are not state dependent.
Collapse
Affiliation(s)
- Eugene Nattie
- Department of Physiology, Dartmouth Medical School, Borwell Bldg., Lebanon, NH 03756-0001, USA.
| | | |
Collapse
|
29
|
Czéh B, Fuchs E, Simon M. NK1 receptor antagonists under investigation for the treatment of affective disorders. Expert Opin Investig Drugs 2006; 15:479-86. [PMID: 16634686 DOI: 10.1517/13543784.15.5.479] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Substance P-neurokinin-1 (NK1) receptor pathways have been repeatedly implicated in the pathophysiology of affective disorders. Anatomical studies in humans have shown a high expression of NK1 receptors in brain regions that are important for the regulation of affective behaviours and stress responses. A large body of evidence that has been generated from animal experiments indicates that treatment with a selective NK1 receptor antagonist might be effective in the treatment of certain forms of anxiety and depressive disorders. Accordingly, numerous NK1 receptor antagonists have either been synthesised and are under clinical development, or have already been tested in clinical trials. However, the initial encouraging clinical results were followed by repeated demonstrations of a lack of effectiveness, thus disappointment and doubt currently surrounds the idea that these compounds may become effective antidepressants. Research continues and novel molecules may show better pharmacokinetic and pharmacodynamic properties and, therefore, may achieve therapeutic success. Furthermore, NK1 receptor antagonists that are ineffective in the treatment of mood disorders may still prove to be effective in the treatment of anxiety problems.
Collapse
Affiliation(s)
- Boldizsár Czéh
- German Primate Center, Clinical Neurobiology Laboratory, Kellnerweg 4, 37077 Göttingen, Germany.
| | | | | |
Collapse
|
30
|
Ebner K, Singewald N. The role of substance P in stress and anxiety responses. Amino Acids 2006; 31:251-72. [PMID: 16820980 DOI: 10.1007/s00726-006-0335-9] [Citation(s) in RCA: 216] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Accepted: 02/21/2006] [Indexed: 12/18/2022]
Abstract
Substance P (SP) is one of the most abundant peptides in the central nervous system and has been implicated in a variety of physiological and pathophysiological processes including stress regulation, as well as affective and anxiety-related behaviour. Consistent with these functions, SP and its preferred neurokinin 1 (NK1) receptor has been found within brain areas known to be involved in the regulation of stress and anxiety responses. Aversive and stressful stimuli have been shown repeatedly to change SP brain tissue content, as well as NK1 receptor binding. More recently it has been demonstrated that emotional stressors increase SP efflux in specific limbic structures such as amygdala and septum and that the magnitude of this effect depends on the severity of the stressor. Depending on the brain area, an increase in intracerebral SP concentration (mimicked by SP microinjection) produces mainly anxiogenic-like responses in various behavioural tasks. Based on findings that SP transmission is stimulated under stressful or anxiety-provoking situations it was hypothesised that blockade of NK1 receptors may attenuate stress responses and exert anxiolytic-like effects. Preclinical and clinical studies have found evidence in favour of such an assumption. The status of this research is reviewed here.
Collapse
Affiliation(s)
- K Ebner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | | |
Collapse
|
31
|
Renoldi G, Invernizzi RW. Blockade of tachykinin NK1 receptors attenuates stress-induced rise of extracellular noradrenaline and dopamine in the rat and gerbil medial prefrontal cortex. J Neurosci Res 2006; 84:961-8. [PMID: 16862563 DOI: 10.1002/jnr.20997] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Substance P receptor antagonists cause antidepressant- and anxiolytic-like effects in rodents that are thought to involve brain monoamines. In the present study, we examined the effects of the NK1 receptor antagonist GR-205,171 on basal and stress-induced rise of extracellular noradrenaline (NA) and dopamine (DA) in the medial prefrontal cortex (mPFC) of conscious rats and gerbils with the in vivo microdialysis technique. GR-205,171 given intraperitoneally to rats (10 and 30 mg/kg) and gerbils (0.3 and 1 mg/kg) did not affect extracellular NA in either species and increased extracellular DA in rats. Forty minutes of immobilization increased extracellular NA and DA by, respectively, 179% and 188% of baseline values in rats and 222% and 316% of baseline values in gerbils. At 10 mg/kg, GR-205,171 attenuated the stress-induced increase of extracellular NA in the rat. At 30 mg/kg, GR-205,171 suppressed the effect of stress on extracellular DA but had no effect on NA. A lower dose (1 mg/kg) attenuated the stress-induced rise of extracellular NA and DA in the mPFC of gerbils. The results show that blockade of NK1 receptors marginally increased basal extracellular DA in rats but had no effect in gerbils, whereas the stress-induced rise of extracellular NA and DA was markedly attenuated in both species. It is suggested that catecholamines may contribute to the functional effects of GR-205,171.
Collapse
|
32
|
Guiard BP, Froger N, Hamon M, Gardier AM, Lanfumey L. Sustained pharmacological blockade of NK1 substance P receptors causes functional desensitization of dorsal raphe 5-HT 1A autoreceptors in mice. J Neurochem 2005; 95:1713-23. [PMID: 16219031 DOI: 10.1111/j.1471-4159.2005.03488.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Antagonists at NK1 substance P receptors have demonstrated similar antidepressant properties in both animal paradigms and in human as selective serotonin reuptake inhibitors (SSRIs) that induce desensitization of 5-HT 1A autoreceptors within the dorsal raphe nucleus (DRN). We investigated whether this receptor adaptation also occurs upon NK1 receptor blockade. C57B/L6J mice were treated for 21 days with the selective NK1 receptor antagonist GR 205171 (10 mg/kg daily) through subcutaneously implanted osmotic mini pumps, and DRN 5-HT 1A autoreceptor functioning was assessed using various approaches. Recording of DRN serotonergic neurons in brainstem slices showed that GR 205171 treatment reduced (by approximately 1.5 fold) the potency of the 5-HT 1A receptor agonist, ipsapirone, to inhibit cell firing. In parallel, the 5-HT 1A autoreceptor-mediated [35S]GTP-gamma-S binding induced by 5-carboxamidotryptamine onto the DRN in brainstem sections was significantly decreased in GR 205171-treated mice. In vivo microdialysis showed that the cortical 5-HT overflow caused by acute injection of the SSRI paroxetine (1 mg/kg) was twice as high in GR 205171-treated as in vehicle-treated controls. In the DRN, basal 5-HT outflow was significantly enhanced by GR 205171 treatment. These data supported the hypothesis that chronic NK1 receptor blockade induces a functional desensitization of 5-HT 1A autoreceptors similar to that observed with SSRIs.
Collapse
Affiliation(s)
- Bruno P Guiard
- INSERM/UPMC, Neuropsychopharmacologie, CHU Pitié-Salpêtrière, Paris, France
| | | | | | | | | |
Collapse
|
33
|
Abstract
Increasing evidence suggests that substance P (SP) and its receptor (neurokinin [NK]-1 receptor [NK1R]) might play an important role in the modulation of stress-related, affective and/or anxious behaviour. First, SP and NK1R are expressed in brain regions that are involved in stress, fear and affective response (e.g. amygdala, hippocampus, hypothalamus and frontal cortex). Second, the SP content in these areas changes upon application of stressful stimuli. Third, the central administration of SP produces a range of fear-related behaviours. In addition, the SP/NK1R system shows significant spatial overlap with neurotransmitters such as serotonin and noradrenaline (norepinephrine), which are known to be involved in the regulation of stress, mood and anxiety. Therefore, it was hypothesised that blockade of the NK1R might have anxiolytic as well as antidepressant effects. Preclinical studies investigating the effects of genetic or pharmacological NK1R inactivation on animal behaviour in assays relevant to depression and anxiety revealed that the behavioural changes resemble those seen with reference antidepressant or anxiolytic drugs. Furthermore, antagonism or genetic inactivation of the NK1R causes alterations in serotonin and norepinephrine neuronal transmission that are likely to contribute to the antidepressant/anxiolytic activity of NK1R antagonists but that are--at least partially--distinct from those produced by established antidepressant drugs. This underlines the conceivable unique mechanism of action of this new class of compounds. In three independent clinical trials with three different compounds (aprepitant [MK-869], L-759274 and CP-122721), an antidepressant effect of NK1R antagonists could be demonstrated. These results, however, have been challenged by recent failed studies with aprepitant. There are numerous indications from preclinical studies that, in addition to SP and NK1R, other neurokinins and/or neurokinin receptors might also be involved in the modulation of stress-related behaviour and that exclusive blockade of the NK1R might not be sufficient to produce consistent anxiolytic and antidepressant effects. One such candidate is the neurokinin-2 receptor (NK2R), and clinical trials to assess the antidepressant effects of NK2R antagonists are currently underway. Of special interest might also be substances that block more than one receptor type such as NK1/2R antagonists or NK1/2/3R antagonists. These compounds may be more efficacious in antagonising the effects of SP than compounds that only block the NK1R.
Collapse
Affiliation(s)
- Inga Herpfer
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical School, Freiburg, Germany
| | | |
Collapse
|
34
|
Gobbi G, Blier P. Effect of neurokinin-1 receptor antagonists on serotoninergic, noradrenergic and hippocampal neurons: comparison with antidepressant drugs. Peptides 2005; 26:1383-93. [PMID: 16042978 DOI: 10.1016/j.peptides.2005.03.032] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Neurokinin-1 (NK1) receptor antagonists have been reported to possess antidepressant and anxiolytic properties in controlled trials. Since antidepressant and anxiolytic drugs act mainly by enhancing serotonin (5-HT) and norepinephrine (NE) neurotransmission in forebrain areas, the main focus of the present review is to critically examine the electrophysiological effects of NK1 receptor antagonists on serotoninergic and noradrenergic neurons, and then hippocampal neurons. It is concluded that NK1 antagonists increase the firing and burst activity of 5-HT neurons, increase burst activity of NE neurons, and modulate postsynaptic transmission at the hippocampus level. Further research is needed in order to develop more selective ligands for the human NK1 receptor and to gain better knowledge of required brain penetration and optimal pharmacodynamic conditions for their use in patients.
Collapse
Affiliation(s)
- Gabriella Gobbi
- Department of Psychiatry, Univ. de Montreal and McGill University, 1033, Av. des Pins Ouest, Montreal, Canada H3A 1A1.
| | | |
Collapse
|
35
|
Herpfer I, Hunt SP, Stanford SC. A comparison of neurokinin 1 receptor knock-out (NK1−/−) and wildtype mice: exploratory behaviour and extracellular noradrenaline concentration in the cerebral cortex of anaesthetised subjects. Neuropharmacology 2005; 48:706-19. [PMID: 15814105 DOI: 10.1016/j.neuropharm.2004.12.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2004] [Revised: 11/22/2004] [Accepted: 12/17/2004] [Indexed: 11/17/2022]
Abstract
In behavioural screens, mice lacking functional NK1 receptors (NK1-/-) resemble wildtypes (NK1+/+) that have been given an antianxiety/antidepressant drug. Most, if not all, antidepressants increase noradrenergic transmission in the brain. Here, we have used in vivo microdialysis to compare the concentrations of extracellular noradrenaline ('efflux') in the cerebral cortex of anaesthetised NK1-/- and NK1+/+ mice. The effects of systemic administration of the antidepressant, desipramine, with and without local infusion of the alpha(2)-adrenoceptor antagonist, RX821002, were also evaluated. Finally, we compared the effects of desipramine on behaviour of NK1+/+ and NK1-/- mice in an activity chamber and in a light/dark exploration box. Basal noradrenaline efflux was increased 2 to 4-fold in NK1-/- mice compared with NK1+/+ mice but there was no difference in the effects of desipramine. RX821002 increased noradrenaline efflux in all vehicle-injected mice but, in desipramine-pretreated mice, noradrenaline efflux was increased in NK1+/+ mice, only. All behaviours in the light/dark exploration box differed in the two genotypes. Furthermore, with the exception of 'grooming', the effects of desipramine on behaviour of NK1-/- mice could be explained by the effects of this antidepressant on locomotor activity. Finally, alpha(2)-adrenoceptors are possibly desensitised in NK1-/- mice. We have yet to establish whether this is a cause or a consequence of the increased noradrenaline efflux.
Collapse
Affiliation(s)
- Inga Herpfer
- Department of Pharmacology, University College London, UK
| | | | | |
Collapse
|
36
|
Sartori SB, Burnet PWJ, Sharp T, Singewald N. Evaluation of the effect of chronic antidepressant treatment on neurokinin-1 receptor expression in the rat brain. Neuropharmacology 2004; 46:1177-1183. [PMID: 15111024 DOI: 10.1016/j.neuropharm.2004.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2003] [Revised: 01/23/2004] [Accepted: 02/11/2004] [Indexed: 12/27/2022]
Abstract
Clinically effective antidepressants are thought to exert their therapeutic effects by facilitating central monoamine neurotransmission. However, recent data showing that neurokinin-1 receptor (NK1R) antagonists have antidepressant properties in both animal and clinical studies raise the possibility that classical antidepressants may also influence NK1R expression in the brain. To test this hypothesis, rats were treated with desipramine, paroxetine, venlafaxine, tranylcypromine or vehicle for 14-42 days. NK1R binding sites and mRNA were determined in a wide variety of brain areas using in situ hybridization and quantitative receptor autoradiography. In all areas examined, the abundance of NK1R binding sites was unchanged after 14 days of treatment. None of the treatments altered the number of NK1R binding sites following 42 days treatment with the exception that an increase was found in the locus coeruleus with tranylcypromine. Taken together, we report that repeated treatment with antidepressants of different classes does not cause significant changes in NK1R expression.
Collapse
Affiliation(s)
- S B Sartori
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Peter-Mayr-Strasse 1, A-6020 Innsbruck, Austria
| | | | | | | |
Collapse
|
37
|
Guiard BP, Przybylski C, Guilloux JP, Seif I, Froger N, De Felipe C, Hunt SP, Lanfumey L, Gardier AM. Blockade of substance P (neurokinin 1) receptors enhances extracellular serotonin when combined with a selective serotonin reuptake inhibitor: an in vivo microdialysis study in mice. J Neurochem 2004; 89:54-63. [PMID: 15030389 DOI: 10.1046/j.1471-4159.2003.02304.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract Substance P antagonists of the neurokinin-1 receptor type (NK1) are gaining growing interest as new antidepressant therapies. It has been postulated that these drugs exert this putative therapeutic effect without direct interactions with serotonin (5-HT) neurones. Our recent microdialysis experiment performed in NK1 receptor knockout mice suggested evidence of changes in 5-HT neuronal function (Froger et al. 2001). The aim of the present study was to evaluate the effects of coadministration of the selective 5-HT reuptake inhibitor (SSRI) paroxetine with a NK1 receptor antagonist (GR205171 or L733060), given either intraperitoneally (i.p.) or locally into the dorsal raphe nucleus, on extracellular levels of 5-HT ([5-HT]ext) in the frontal cortex and the dorsal raphe nucleus using in vivo microdialysis in awake, freely moving mice. The systemic or intraraphe administration of a NK1 receptor antagonist did not change basal cortical [5-HT]ext in mice. A single systemic dose of paroxetine (4 mg/kg; i.p.) resulted in a statistically significant increase in [5-HT]ext with a larger extent in the dorsal raphe nucleus (+ 138% over basal AUC values), than in the frontal cortex (+ 52% over basal AUC values). Co-administration of paroxetine (4 mg/kg; i.p.) with the NK1 receptor antagonists, GR205171 (30 mg/kg; i.p.) or L733060 (40 mg/kg; i.p.), potentiated the effects of paroxetine on cortical [5-HT]ext in wild-type mice, whereas GR205171 (30 mg/kg; i.p.) had no effect on paroxetine-induced increase in cortical [5-HT]ext in NK1 receptor knock-out mice. When GR205171 (300 micro mol/L) was perfused by 'reverse microdialysis' into the dorsal raphe nucleus, it potentiated the effects of paroxetine on cortical [5-HT]ext, and inhibited paroxetine-induced increase in [5-HT]ext in the dorsal raphe nucleus. Finally, in mice whose 5-HT transporters were first blocked by a local perfusion of 1 micro mol/L of citalopram into the frontal cortex, a single dose of paroxetine (4 mg/kg i.p.) decreased cortical 5-HT release, and GR205171 (30 mg/kg i.p.) reversed this effect. The present findings suggest that NK1 receptor antagonists, when combined with a SSRI, augment 5-HT release by modulating substance P/5-HT interactions in the dorsal raphe nucleus.
Collapse
Affiliation(s)
- Bruno P Guiard
- Laboratoire de Neuropharmacologie EA 3544 MJENR, Faculté de Pharmacie IFR75 - Institut de Signalisation et d'Innovation Thérapeutique, Université Paris-Sud, Châtenay-Malabry, France
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Nattie EE, Li A, Richerson GB, Richerson G, Lappi DA. Medullary serotonergic neurones and adjacent neurones that express neurokinin-1 receptors are both involved in chemoreception in vivo. J Physiol 2004; 556:235-53. [PMID: 14724193 PMCID: PMC1664900 DOI: 10.1113/jphysiol.2003.059766] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Neurokinin-1 receptor (NK1R)-expressing neurones that are involved in chemoreception at the retrotrapezoid nucleus (Nattie & Li, 2002b) are also prominent at locations that contain medullary serotonergic neurones, which are chemosensitive in vitro. In medullary regions containing both types, we evaluated their role in central chemoreception by specific cell killing. We injected (2 x 100 nl) (a) substance P-saporin (SP-SAP; 1 microm) to kill NK1R-expressing neurones, (b) a novel conjugate of a monoclonal antibody to the serotonin transporter (SERT) and saporin (anti-SERT-SAP; 1 microm) to kill serotonergic neurones, or (c) SP-SAP and anti-SERT-SAP together to kill both types. Controls received IgG-SAP injections (1 microm). There was no double-labelling of NK1R-immunoreactive (ir) and tryptophan-hydroxylase (TPOH)-ir neurones. Cell (somatic profile) counts showed that NK1R-ir neurones in the SP-SAP group were reduced by 31%; TPOH-ir neurones in the anti-SERT-SAP group by 28%; and NK1R-ir and TPOH-ir neurones, respectively, in the combined lesion group by 55% and 31% (P < 0.001; two-way ANOVA; P < 0.05, Tukey's post hoc test). The treatments had no significant effect on sleep/wake time, body temperature, or oxygen consumption but all three reduced the ventilatory response to 7% inspired CO(2) in wakefulness and sleep by a similar amount. SP-SAP treatment decreased the averaged CO(2) responses (3, 7 and 14 days after lesions) in wakefulness and sleep by 21% and 16%, anti-SERT-SAP decreased the responses by 15% and 18%, and the combined treatment decreased the responses by 12% and 12% (P < 0.001; two-way ANOVA; P < 0.05, Tukey's post hoc test). We conclude that separate populations of serotonergic and adjacent NK1R-expressing neurones in the medulla are both involved in central chemoreception in vivo.
Collapse
Affiliation(s)
- Eugene E Nattie
- Department of Physiology, Dartmouth Medical School, Borwell Bldg, Lebanon, NH 03756-0001, USA.
| | | | | | | | | |
Collapse
|
39
|
Neurokinin-1 receptor-expressing neurons in the amygdala modulate morphine reward and anxiety behaviors in the mouse. J Neurosci 2003. [PMID: 12967989 DOI: 10.1523/jneurosci.23-23-08271.2003] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mice lacking the neurokinin-1 (NK1) receptor, the preferred receptor for the neuropeptide substance P (SP), do not show many of the behaviors associated with morphine reward. To identify the areas of the brain that might contribute to this effect, we assessed the behavioral effects of ablation of neurons expressing the NK1 receptor in specific regions of the mouse brain using the neurotoxin substance P-saporin. In a preliminary investigation, bilateral ablation of these neurons from the amygdala, but not the nucleus accumbens and dorsomedial caudate putamen, brought about reductions in morphine reward behavior. Subsequently, the effect of ablation of these neurons in the amygdala on anxiety behavior was assessed using the elevated plus maze (EPM), before conditioned place preference (CPP), and locomotor responses to morphine were measured. Loss of NK1 receptor-expressing neurons in the amygdala caused an increase in anxiety-like behavior on the EPM. It also brought about a reduction in morphine CPP scores and the stimulant effect of acute morphine administration relative to saline controls, without affecting CPP to cocaine. NK1 receptor-expressing neurons in the mouse amygdala therefore modulate morphine reward behaviors. These observations mirror those observed in NK1 receptor knock-out (NK1-/-) mice and suggest that the amygdala is an important area for the effects of SP and the NK1 receptor in the motivational properties of opiates, as well as the control of behaviors related to anxiety.
Collapse
|
40
|
Morcuende S, Gadd CA, Peters M, Moss A, Harris EA, Sheasby A, Fisher AS, De Felipe C, Mantyh PW, Rupniak NMJ, Giese KP, Hunt SP. Increased neurogenesis and brain-derived neurotrophic factor in neurokinin-1 receptor gene knockout mice. Eur J Neurosci 2003; 18:1828-36. [PMID: 14622216 DOI: 10.1046/j.1460-9568.2003.02911.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
It has previously been shown that chronic treatment with antidepressant drugs increases neurogenesis and levels of brain-derived neurotrophic factor in the hippocampus. These changes have been correlated with changes in learning and long-term potentiation and may contribute to the therapeutic efficacy of antidepressant drug treatment. Recently, antagonists at the neurokinin-1 receptor, the preferred receptor for the neuropeptide substance P, have been shown to have antidepressant activity. Mice with disruption of the neurokinin-1 receptor gene are remarkably similar both behaviourally and neurochemically to mice maintained chronically on antidepressant drugs. We demonstrate here that there is a significant elevation of neurogenesis but not cell survival in the hippocampus of neurokinin-1 receptor knockout mice. Neurogenesis can be increased in wild-type but not neurokinin-1 receptor knockout mice by chronic treatment with antidepressant drugs which preferentially target noradrenergic and serotonergic pathways. Hippocampal levels of brain-derived neurotrophic factor are also two-fold higher in neurokinin-1 receptor knockout mice, whereas cortical levels are similar. Finally, we examined hippocampus-dependent learning and memory but found no clear enhancement in neurokinin-1 receptor knockout mice. These data argue against a simple correlation between increased levels of neurogenesis or brain-derived neurotrophic factor and mnemonic processes in the absence of increased cell survival. They support the hypothesis that increased neurogenesis, perhaps accompanied by higher levels of brain-derived neurotrophic factor, may contribute to the efficacy of antidepressant drug therapy.
Collapse
Affiliation(s)
- Sara Morcuende
- Department of Anatomy and Developmental Biology, Medawar Building, UCL, Gower Street, London WC1E 6BT, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chen LW, Cao R, Liu HL, Ju G, Chan YS. The striatal GABA-ergic neurons expressing substance P receptors in the basal ganglia of mice. Neuroscience 2003; 119:919-25. [PMID: 12831852 DOI: 10.1016/s0306-4522(03)00223-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
By using a double immunofluorescence, we have examined the distribution of striatal GABAergic neurons that expressed substance P receptor (SPR) in the basal ganglia of adult C57 mice. The distribution of GABA-immunoreactive neurons completely or partially overlapped with that of SPR-immunoreactive neurons in the striatum (i.e. the caudate-putamen), globus pallidus, ventral pallidum, and nucleus accumbens. Neurons showing both GABA- and SPR-immunoreactivities were, however, predominantly found in the caudate-putamen, and most of them were characterized by their large-sized aspiny neuronal profile. Semi-quantification indicated that only about 13% of the total GABA-immunoreactive neurons (including large and medium-sized) displayed SPR-immunoreactivity, and these double-labeled neurons constituted about 31% of the total SPR-immunoreactive cells in the striatum. Neurons double-labeled with GABA- and SPR-immunoreactivities were hardly detected in other aforementioned regions of the basal ganglia. In addition, double immunofluorescence also showed co-localization of SPR- with glutamic acid decarboxylase-immunoreactivity, but not with parvalbumin-immunoreactivity, in the striatal neurons. Taken together with previous reports, the present study has suggested that a sub-population of striatal GABA-ergic neurons, most possibly GABA-ergic interneurons, may also receive direct physiological modulation by tachykinins through SPR in the basal ganglia of mammals.
Collapse
Affiliation(s)
- L-W Chen
- Institute of Neurosciences, Fourth Military Medical University, 710032, Xi'an, China.
| | | | | | | | | |
Collapse
|
42
|
Abstract
Fear is an adaptive component of the acute "stress" response to potentially-dangerous (external and internal) stimuli which threaten to perturb homeostasis. However, when disproportional in intensity, chronic and/or irreversible, or not associated with any genuine risk, it may be symptomatic of a debilitating anxious state: for example, social phobia, panic attacks or generalized anxiety disorder. In view of the importance of guaranteeing an appropriate emotional response to aversive events, it is not surprising that a diversity of mechanisms are involved in the induction and inhibition of anxious states. Apart from conventional neurotransmitters, such as monoamines, gamma-amino-butyric acid (GABA) and glutamate, many other modulators have been implicated, including: adenosine, cannabinoids, numerous neuropeptides, hormones, neurotrophins, cytokines and several cellular mediators. Accordingly, though benzodiazepines (which reinforce transmission at GABA(A) receptors), serotonin (5-HT)(1A) receptor agonists and 5-HT reuptake inhibitors are currently the principle drugs employed in the management of anxiety disorders, there is considerable scope for the development of alternative therapies. In addition to cellular, anatomical and neurochemical strategies, behavioral models are indispensable for the characterization of anxious states and their modulation. Amongst diverse paradigms, conflict procedures--in which subjects experience opposing impulses of desire and fear--are of especial conceptual and therapeutic pertinence. For example, in the Vogel Conflict Test (VCT), the ability of drugs to release punishment-suppressed drinking behavior is evaluated. In reviewing the neurobiology of anxious states, the present article focuses in particular upon: the multifarious and complex roles of individual modulators, often as a function of the specific receptor type and neuronal substrate involved in their actions; novel targets for the management of anxiety disorders; the influence of neurotransmitters and other agents upon performance in the VCT; data acquired from complementary pharmacological and genetic strategies and, finally, several open questions likely to orientate future experimental- and clinical-research. In view of the recent proliferation of mechanisms implicated in the pathogenesis, modulation and, potentially, treatment of anxiety disorders, this is an opportune moment to survey their functional and pathophysiological significance, and to assess their influence upon performance in the VCT and other models of potential anxiolytic properties.
Collapse
Affiliation(s)
- Mark J Millan
- Psychopharmacology Department, Centre de Rescherches de Croissy, Institut de Recherches (IDR) Servier, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, Paris, France.
| |
Collapse
|
43
|
Caberlotto L, Hurd YL, Murdock P, Wahlin JP, Melotto S, Corsi M, Carletti R. Neurokinin 1 receptor and relative abundance of the short and long isoforms in the human brain. Eur J Neurosci 2003; 17:1736-46. [PMID: 12752772 DOI: 10.1046/j.1460-9568.2003.02600.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Substance P exerts its various biochemical effects mainly via interactions through neurokinin-1 receptors (NK1). Recently, the NK1 receptor has attracted considerable interest for its possible role in a variety of psychiatric disorders including depression and anxiety. However, little is known regarding the anatomical distribution of NK1 in the human central nervous system (CNS). Riboprobe in situ hybridization, quantitative PCR and in vitro autoradiography were performed. Highest NK1 mRNA levels were localized in the locus coeruleus and ventral striatum, while moderate hybridization signals were observed in the cerebral cortex (most abundant in the visual cortex), hippocampus and different amygdaloid nuclei. Very low levels of the NK1 mRNA were detected in the cerebellum and thalamus. In view of the existence of a long and short isoform of the NK1 receptor, it was of interest to assess whether there was a differential distribution of the two splice variants in the human CNS and peripheral tissues. A quantitative TaqMan PCR analysis showed that the long NK1 isoform was the most prevalent throughout the human brain, while in peripheral tissues the truncated form was the most represented. 3H-Substance P autoradiography revealed a good correlation between receptor binding sites and NK1 mRNA expression throughout the brain, with the highest levels of binding in the locus coeruleus. These results provide the anatomical evidence that the NK1 receptors have a strong association with neuronal systems relevant to mood regulation and stress in the human brain, but do not suggest a region-specific role of the two isoforms in the CNS.
Collapse
Affiliation(s)
- Laura Caberlotto
- Department of Biology, Psychiatry CEDD, GlaxoSmithKline Medicine Research Centre, Verona, Italy.
| | | | | | | | | | | | | |
Collapse
|
44
|
Steinberg R, Alonso R, Rouquier L, Desvignes C, Michaud JC, Cudennec A, Jung M, Simiand J, Griebel G, Emonds-Alt X, Le Fur G, Soubrié P. SSR240600 [(R)-2-(1-[2-[4-[2-[3,5-bis(trifluoromethyl)phenyl]acetyl]-2-(3,4-dichlorophenyl)-2-morpholinyl]ethyl]-4-piperidinyl)-2-methylpropanamide], a centrally active nonpeptide antagonist of the tachykinin neurokinin 1 receptor: II. Neurochemical and behavioral characterization. J Pharmacol Exp Ther 2002; 303:1180-8. [PMID: 12438542 DOI: 10.1124/jpet.102.040279] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
SSR240600 [(R)-2-(1-[2-[4-[2-[3,5-bis(trifluoromethyl)phenyl]acetyl]-2-(3,4-dichlorophenyl)-2-morpholinyl]ethyl]-4-piperidinyl)-2-methylpropanamide], a new nonpeptide tachykinin neurokinin 1 (NK1) receptor antagonist, was evaluated against the neurochemical, electrophysiological, and behavioral effects provoked by direct activation of brain tachykinin NK1 receptors or by stress in guinea pigs. SSR240600 (0.1-10 mg/kg i.p. or p.o.) antagonized the excitatory effect of i.c.v. infusion of [Sar(9),Met(O2)(11)]substance P (SP) on the release of acetylcholine in the striatum of anesthetized and awake guinea pigs. This antagonistic action was still observed after repeated administration of SSR240600 (5 days, 10 mg/kg p.o., once a day). SSR240600 (10 mg/kg i.p.) inhibited the phosphorylation of the cAMP response element-binding protein in various brain regions induced by i.c.v. administration of [Sar9,Met(O2)(11)]SP. In slice preparations, neuronal firing of the locus coeruleus (LC) neurons elicited by the application of [Sar9,Met(O2)(11)]SP was suppressed by SSR240600 at 100 nM. Norepinephrine release in the prefrontal cortex, elicited either by an intra-LC application of [Sar9,Met(O2)(11)]SP or by an i.c.v administration of corticotropin-releasing factor, was reduced by SSR240600 (0.3-1 mg/kg and 1-10 mg/kg i.p., respectively). SSR240600 (1-10 mg/kg i.p.) inhibited vocalizations induced in adult guinea pigs by an i.c.v. administration of the NK1 receptor agonist, GR73632 [D-Ala-[L-Pro9,Me-Leu8]substance P(7-11)]. Furthermore, SSR240600 (1-10 mg/kg i.p.) inhibited distress vocalizations produced in guinea pig pups by maternal separation. SSR240600 also reduced maternal separation-induced increase in the number of neurons displaying NK1 receptor internalization in the amygdala. Finally, SSR240600 counteracted the increase in body temperature induced by isolation stress. In conclusion, SSR240600 is able to antagonize various NK1 receptor-mediated as well as stress-mediated effects in the guinea pig.
Collapse
Affiliation(s)
- Régis Steinberg
- C.N.S. Research Department, Sanofi-Synthélabo Recherche, Montpellier, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Holden JE, Van Poppel AY, Thomas S. Antinociception from lateral hypothalamic stimulation may be mediated by NK(1) receptors in the A7 catecholamine cell group in rat. Brain Res 2002; 953:195-204. [PMID: 12384253 DOI: 10.1016/s0006-8993(02)03285-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Stimulation of the lateral hypothalamus (LH) produces antinociception that is modified by intrathecal alpha-adrenergic antagonists. Spinally-projecting noradrenergic neurons in the LH have not been identified, suggesting that the LH may innervate brainstem noradrenergic neurons, such as the A7 catecholamine cell group in the dorsolateral pontine tegmentum, that modify nociception at the level of the spinal cord dorsal horn. Recently we demonstrated in neuroanatomical studies that substance P-immunoreactive neurons in the LH project the A7 area. To identify a functional connection between substance P neurons in the LH and the A7 cell group, the cholinergic agonist carbachol (125 nmol) was microinjected into the LH of female Sprague-Dawley rats and antinociception was obtained on the tail flick or foot withdrawal test. Cobalt chloride (100 nM) was then microinjected near the A7 cell group to block synaptic activation of spinally-projecting A7 neurons, which were identified using tyrosine-hydroxylase immunoreactivity. Within 5 min of the cobalt chloride injection, the antinociceptive effect of carbachol stimulation was blocked. In another set of experiments, the NK(1) receptor antagonist L-703-606 (5 microg) was microinjected near the A7 cell group following LH stimulation with carbachol. L-703-606 also abolished LH-induced antinociception. These results support the conclusion that antinociception produced by activating substance P neurons in the LH is mediated in part by the subsequent activation of spinally-projecting noradrenergic neurons in the A7 cell group.
Collapse
Affiliation(s)
- Janean E Holden
- Department of Medical-Surgical Nursing, The University of Illinois at Chicago, 718 College of Nursing (M/C 802), 845 S. Damen Avenue, Chicago, IL 60612-7350, USA.
| | | | | |
Collapse
|
46
|
Liu HL, Cao R, Jin L, Chen LW. Immunocytochemical localization of substance P receptor in hypothalamic oxytocin-containing neurons of C57 mice. Brain Res 2002; 948:175-9. [PMID: 12383972 DOI: 10.1016/s0006-8993(02)03146-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
With the use of double immunofluorescence, we have examined the distribution of oxytocin-containing neurons that express substance P receptor (SPR) in the hypothalamus of C57 mice. The distribution of oxytocin-like immunoreactive neurons overlapped with that of SPR-like immunoreactive neurons in the paraventricular nucleus and supraoptic nucleus of the hypothalamus. Neurons showing both oxytocin- and SPR-like immunoreactivities were predominantly found in both nuclei. A few neurons that were double-labeled with oxytocin- and SPR-like immunoreactivities were also scattered in the hypothalamic periventricular and preoptic regions. Semi-quantitative analysis indicated that about 94% of the oxytocin-like neurons displayed SPR-like immunoreactivity. These double-labeled cells constituted about 91% of the SPR-like neurons in the aforementioned regions. The present study provides morphological evidence for tachykinin-induced modulation of oxytocin-containing neurons as mediated by substance P receptor in the hypothalamus of mammals.
Collapse
Affiliation(s)
- Hui-Ling Liu
- Institute of Neurosciences, The Fourth Military Medical University, Xi'an 710032, PR China
| | | | | | | |
Collapse
|
47
|
Léger L, Gay N, Cespuglio R. Neurokinin NK1- and NK3-immunoreactive neurons in serotonergic cell groups in the rat brain. Neurosci Lett 2002; 323:146-50. [PMID: 11950514 DOI: 10.1016/s0304-3940(01)02543-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interactions are known to occur in the brain between serotonin (5-HT) and substance P (SP). To investigate whether SP can directly influence serotonergic neurons, double immunohistochemical labelings were performed on rat brain sections with NK1 or NK3 affinity-purified antibodies and a 5-HT monoclonal antibody. It was found that the vast majority of serotonergic cell bodies do not colocalize NK1 or NK3 labeling. Only in the central linear nucleus and ventral part of the dorsal raphe nucleus were a few serotonergic neurons double-labeled for NK1 receptors (15 and 0.8% of serotonergic neurons, respectively). It is suggested that serotonergic neurons are not major direct targets for SP in the rat brain.
Collapse
Affiliation(s)
- Lucienne Léger
- INSERM U480, Institut Fédératif des Neurosciences de Lyon (IFNL 19), Faculté de Médecine, 8, Avenue Rockefeller, 69373 Lyon Cedex 08, France.
| | | | | |
Collapse
|
48
|
Schwarz MJ, Ackenheil M. The role of substance P in depression: therapeutic implications. DIALOGUES IN CLINICAL NEUROSCIENCE 2002. [PMID: 22033776 PMCID: PMC3181667 DOI: 10.31887/dcns.2002.4.1/mschwarz] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Substance P (for "powder"), identified as a gut tachykinin in 1931 and involved in the control of multiple other autonomic functions, notably pain transmission, is the focus of intense fundamental and clinical psychiatric research as a central neurotransmitter, neuromodulator, and immunomodulator, along with sister neurokinins A and B (NKA and NKB), discovered in 1984. Substance P is widely distributed throughout the central nervous system, where if is often colocalized with serotonin, norepinephrine, and dopamine. Many neurokinin (NK) receptor antagonists and agonists have been synthesized and some clinically tested. A double-blind study of MK869, a selective NK1 receptor antagonist that blocks the action of substance P, showed significant activity versus placebo and fewer sexual side effects than paroxetine in outpatients with major depression and moderate anxiety. Substance P, which is degraded by the angiotensin-converting enzyme (ACE), may mediate modulation of therapeutic outcome in affective disorders by functional polymorphism within the ACE gene: the D allele is associated with higher ACE levels and increased neuropeptide degradation, with the result that patients with major depression who carry the D allele have lower depression scores and shorter hospitalization. ACE polymorphism genotypinq might thus identify those patients with major depression likely to benefit from NK1 receptor antagonist therapy.
Collapse
Affiliation(s)
- Markus J. Schwarz
- Department of Neurochemistry, Psychiatric Hospital, University of Munich, Germany
| | | |
Collapse
|
49
|
Bert L, Rodier D, Bougault I, Allouard N, Le-Fur G, Soubrié P, Steinberg R. Permissive role of neurokinin NK(3) receptors in NK(1) receptor-mediated activation of the locus coeruleus revealed by SR 142801. Synapse 2002; 43:62-9. [PMID: 11746734 DOI: 10.1002/syn.10021] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The present experiments investigated the role of neurokinin-1 (NK(1)) and neurokinin-3 (NK(3)) receptors on the activity of the locus coeruleus (LC)-noradrenergic system by using a dual probe microdialysis technique in anesthetized guinea pigs. The local application in the LC of the selective NK(1) receptor agonists [SAR(9),Met(O(2))(11)]-SP (10 microM) and septide (1 microM) as well as the selective NK(3) receptor agonist senktide (1 microM), enhanced the extracellular norepinephrine (NE) levels in the prefrontal cortex. The enhancing effect of [SAR(9),Met(O(2))(11)]-SP was completely blocked by the peripheral administration of the selective non peptide NK(1) and NK(3) receptor antagonists, GR 205171 (1 mg/kg, i.p.) and SR 142801 (0.1 mg/kg, i.p.), respectively, whereas SR 142806 (0.1 mg/kg, i.p.) the inactive enantiomer of SR 142801 had no effect. Moreover, the [SAR(9),Met(O(2))(11)]-SP-induced increase in LC DOPAC concentrations, is only antagonized by GR 205171. In contrast, only SR 142801 (0.3 mg/kg, i.p.) could block stereoselectively the senktide-evoked increase in NE levels. Both [SAR(9),Met(O(2))(11)]-SP and senktide effects were blocked by local infusion into the LC of SR 142801 (10(-9) M). These results demonstrate that stimulation of NK(1) and NK(3) receptors located in the LC area modulates the activity of the LC-NE system, and that the excitatory effects of NK(1) receptor agonists require NKB/NK(3) receptor activation in the LC.
Collapse
Affiliation(s)
- Lionel Bert
- Sanofi-Synthélabo, Central Nervous System Department, Montpellier, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Makeham JM, Goodchild AK, Pilowsky PM. NK1 receptor and the ventral medulla of the rat: bulbospinal and catecholaminergic neurons. Neuroreport 2001; 12:3663-7. [PMID: 11726770 DOI: 10.1097/00001756-200112040-00012] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Bulbospinal C1 neurons are sympathoexcitatory and excited by substance P. However the substance P receptor (NK1 receptor), has been reported to be absent from the somata of C1 neurons. In this study, using double and triple labelling immunofluorescence and retrograde tracing, we provide evidence that the NK1 receptor is present on 5.3% of C1 neurons, and that 4.7% of C1 neurons receive close oppositions from NK1 receptor immunoreactive terminals, indicating a pre-synaptic and post-synaptic site for the action of substance P. These results provide support for the sympathoexcitatory actions of substance P on C1 neurons. We also demonstrate the NK1 receptor on bulbospinal neurons of the ventral respiratory group, in a region overlapping the pre-Bötzinger Complex.
Collapse
Affiliation(s)
- J M Makeham
- Hypertension and Stroke Research Laboratory, Department of Physiology, University of Sydney, Australia
| | | | | |
Collapse
|