Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Neuropharmacology ; 143: 113-121, 2018 12.
Article in English | MEDLINE | ID: mdl-30248304

ABSTRACT

Neuropeptides are often co-expressed in neurons, and may therefore be working together to coordinate proper neural circuit function. However, neurophysiological effects of neuropeptides are commonly studied individually possibly underestimating their modulatory roles. Here, we triggered the release of endogenous neuropeptides in brain slices from male mice to better understand their modulation of central amygdala (CeA) inhibitory inputs onto oval (ov) BNST neurons. We found that locally-released neurotensin (NT) and dynorphin (Dyn) antagonistically regulated CeA inhibitory inputs onto ovBNST neurons. NT and Dyn respectively increased and decreased CeA-toovBNST inhibitory inputs through NT receptor 1 (NTR1) and kappa opioid receptor (KOR). Additionally, NT and Dyn mRNAs were highly co-localized in ovBNST neurons suggesting that they may be released from the same cells. Together, we showed that NT and Dyn are key modulators of CeA inputs to ovBNST, paving the way to determine whether different conditions or states can alter the neuropeptidergic regulation of this particular brain circuit.


Subject(s)
Central Amygdaloid Nucleus/metabolism , Dynorphins/metabolism , Neural Inhibition/physiology , Neurons/metabolism , Neurotensin/metabolism , Septal Nuclei/metabolism , Animals , Central Amygdaloid Nucleus/cytology , Central Amygdaloid Nucleus/drug effects , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Neurons/cytology , Neurons/drug effects , Neurotransmitter Agents/pharmacology , RNA, Messenger/metabolism , Receptors, GABA-A/metabolism , Receptors, Neurotensin/metabolism , Receptors, Opioid, kappa/metabolism , Septal Nuclei/cytology , Septal Nuclei/drug effects , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
2.
Pain Res Manag ; 2017: 8123812, 2017.
Article in English | MEDLINE | ID: mdl-28280406

ABSTRACT

The Quebec Pain Registry (QPR) is a large research database of patients suffering from various chronic pain (CP) syndromes who were referred to one of five tertiary care centres in the province of Quebec (Canada). Patients were monitored using common demographics, identical clinical descriptors, and uniform validated outcomes. This paper describes the development, implementation, and research potential of the QPR. Between 2008 and 2013, 6902 patients were enrolled in the QPR, and data were collected prior to their first visit at the pain clinic and six months later. More than 90% of them (mean age ± SD: 52.76 ± 4.60, females: 59.1%) consented that their QPR data be used for research purposes. The results suggest that, compared to patients with serious chronic medical disorders, CP patients referred to tertiary care clinics are more severely impaired in multiple domains including emotional and physical functioning. The QPR is also a powerful and comprehensive tool for conducting research in a "real-world" context with 27 observational studies and satellite research projects which have been completed or are underway. It contains data on the clinical evolution of thousands of patients and provides the opportunity of answering important research questions on various aspects of CP (or specific pain syndromes) and its management.


Subject(s)
Chronic Pain/epidemiology , Chronic Pain/therapy , Health Plan Implementation , Pain Clinics/statistics & numerical data , Pain Management/methods , Registries , Adult , Aged , Chronic Pain/diagnosis , Female , Health Plan Implementation/methods , Health Plan Implementation/standards , Humans , Male , Middle Aged , Pain Measurement , Quebec/epidemiology , Registries/standards , Registries/statistics & numerical data , Retrospective Studies , Surveys and Questionnaires , Time Factors
3.
Neuroscience ; 315: 70-8, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26691963

ABSTRACT

Clinical evidences suggest that an imbalance between descending inhibition and facilitation drives the development of chronic pain. However, potential mechanisms promoting the establishment of a persistent pain state and the increased pain vulnerability remain unknown. This preclinical study was designed to evaluate temporal changes in descending pain modulation at specific experimental endpoints (12, 28, 90 and 168 days) using a novel double-hit model of chronic/tonic pain (first hit: chronic constriction injury (CCI) model; second hit: tonic formalin pain in the contralateral hindpaw). Basal activity of bulbo-spinal monoaminergic systems was further assessed through liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) screening of cerebrospinal fluid (CSF). We found that CCI-operated rats exhibited a reduced nociceptive response profile, peaking on day 28, when subjected to tonic pain. This behavioral response was accompanied by a rapid increase in basal CSF serotonin and norepinephrine levels 12 days after neuropathy, followed by a return to sham levels on day 28. These molecular and behavioral adaptive changes in descending pain inhibition seemed to slowly fade over time. We therefore suggest that chronic neuropathic pain produces a transient hyperactivation of bulbo-spinal monoaminergic drive when previously primed using a tonic pain paradigm (i.e., formalin test), translating into inhibition of subsequent nociceptive behaviors. Altogether, we propose that early hyperactivation of descending pain inhibitory mechanisms, and its potential ensuing exhaustion, could be part of the temporal neurophysiological chain of events favoring chronic neuropathic pain establishment.


Subject(s)
Chronic Pain/physiopathology , Neural Inhibition/physiology , Nociceptive Pain/physiopathology , Animals , Chromatography, Liquid , Disease Models, Animal , Formaldehyde , Hyperalgesia/physiopathology , Male , Norepinephrine/cerebrospinal fluid , Physical Stimulation , Random Allocation , Rats, Sprague-Dawley , Serotonin/cerebrospinal fluid , Tandem Mass Spectrometry , Touch
4.
Neuroscience ; 272: 261-70, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24813434

ABSTRACT

Some pathophysiological models of schizophrenia posit that prenatal inflammation sensitizes the developing brain to second insults in early life and enhances brain vulnerability, thereby increasing the risk of developing the disorder during adulthood. We previously developed a two-hit animal model, based on the well-established prenatal immune challenge with poly-inosinic/cytidylic acid (polyI:C), followed by juvenile restraint stress (RS). We observed an additive disruption of prepulse inhibition (PPI) of acoustic startle in juvenile mice submitted to both insults. Previous studies have also reported that oxidative stress is associated with pathophysiological mechanisms of psychiatric disorders, including schizophrenia. We report here that PPI disruption in our two-hit animal model of schizophrenia is associated with an increase in oxidative stress. These findings led us to assess whether α-lipoic acid, an antioxidant, can prevent both increase in oxidative status and PPI deficits in our juvenile in vivo model of schizophrenia. In the offspring submitted to prenatal injection of polyI:C and to RS, treatment with α-lipoic acid prevented the development of PPI deficits 24h after the last period of RS. α-Lipoic acid also improved PPI performance in control mice. The reversal effect of α-lipoic acid pretreatment on these behavioral alterations was further accompanied by a normalization of the associated oxidative status and dopaminergic and GABAergic abnormalities in the prefrontal cortex. Based on our double insult paradigm, these results support the hypothesis that oxidative stress plays an important role in the development of PPI deficits, a well-known behavior associated with schizophrenia. These findings form the basis of future studies aiming to unravel mechanistic insights of the putative role of antioxidants in the treatment of schizophrenia, especially during the prodromal stage.


Subject(s)
Prefrontal Cortex/drug effects , Receptors, Dopamine/drug effects , Reflex, Startle/drug effects , Schizophrenia/drug therapy , Thioctic Acid/pharmacology , Aging , Animals , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Prefrontal Cortex/physiopathology , Pregnancy
5.
Eur J Pain ; 18(7): 923-31, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24343901

ABSTRACT

BACKGROUND: The difficulty in identifying the onset of low back pain (LBP) limits the capacity to determine the incidence of LBP at the population level and, further, to identify risk factors. In the literature, incidence cohorts have been built with patients initially considered LBP-free for 6-12 months prior to their selection. This 'clearance period' might not be sufficient to exclude recurrent patients having experienced previous LBP episodes and might result in a misclassification bias. METHODS: Using the Canadian province of Quebec's medical administrative physicians' claims database, a cohort of prevalent claims-based recurrent LBP patients was built for 2007. The medical history of 81,329 patients was screened for a period of 11 years. Positive predictive values (PPVs), kappa statistics and a survival function were calculated to determine the optimal clearance period for capturing first-time events. RESULTS: The 2007 annual incidence of adult claims-based recurrent LBP was estimated at 242 per 100,000 persons. Men between 18 and 34 years of age were found to be 1.18 times more at risk than their counterparts. Altogether, the elderly (over 80 years) had 52% more new cases than the 18-34 age group. A very good convergence for PPV and kappa was found for a 7-year clearance period. This allowed determining the annual incidence from 2000 to 2007, showing a decrease of 26%. CONCLUSION: Screening the medical history of LBP patients can provide more accurate incidence estimates by limiting the over-ascertainment of first-time LBP patients. A 4- to 7-year clearance period should be considered.


Subject(s)
Low Back Pain/epidemiology , Adolescent , Adult , Age Factors , Age of Onset , Aged , Aged, 80 and over , Canada , Female , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Risk Factors , Young Adult
6.
Eur J Pain ; 16(4): 473-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22396077

ABSTRACT

Chronic neuropathic pain arising from peripheral nerve damage is a severe clinical issue where there is a major unmet medical need. We previously demonstrated that both neurotensin (NT) receptor subtypes 1 (NTS1) and 2 (NTS2) are involved in mediating the naloxone-insensitive antinociceptive effects of neurotensin in different analgesic tests including hotplate, tail-flick, and tonic pain. However, the role of these receptors in neuropathic pain management has been poorly investigated. In the present study, we therefore examined whether intrathecal delivery of NTS1 agonists was effective in reducing neuropathic pain symptoms in rats. Neuropathy was induced by sciatic nerve constriction (CCI model), and the development of mechanical allodynia and thermal hyperalgesia on the ipsi- and contralateral hind paws was examined 3, 7, 14, 21, and 28 days post-surgery. CCI-operated rats exhibited significant increases in thermal and mechanical hypersensitivities over a 28-day testing period. Spinal injection of NT to CCI rats alleviated the behavioral responses to radiant heat and mechanical stimuli, with a maximal reversal of 91% of allodynia at 6 µg/kg. Intrathecal administration of the NTS1-selective agonist, PD149163 (30-90 µg/kg) also produced potent anti-allodynic and anti-hyperalgesic effects in nerve-injured rats. Likewise, heat hyperalgesia and tactile allodynia produced by CCI of the sciatic nerve were fully reversed by the NTS1 agonist, NT69L (5-25 µg/kg). Altogether, these results support the idea that the NTS1 receptor subtype is involved in pain modulation, and the potential use of NTS1 agonists for the treatment of painful neuropathies.


Subject(s)
Behavior, Animal/drug effects , Neuralgia/drug therapy , Neuralgia/psychology , Nociception/drug effects , Receptors, Neurotensin/agonists , Animals , Hot Temperature , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/psychology , Injections, Spinal , Male , Neurotensin/analogs & derivatives , Neurotensin/pharmacology , Pain Measurement/drug effects , Peptide Fragments/pharmacology , Physical Stimulation , Rats , Rats, Sprague-Dawley , Sciatica/drug therapy , Sciatica/psychology
7.
Neuroscience ; 183: 221-9, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21458544

ABSTRACT

Over the past few years, significant progress has been made in cancer therapy. Indeed, the lifespan of cancer patients has significantly increased. Although patients live longer, cancer-related pain remains a daily problem affecting their quality of life, especially when metastases reach the bone. In patients coping with cancer-induced bone pain, morphine and NSAIDs, often used in combination with other medications, are the most commonly used drugs to alleviate pain. However, these drugs have dose-limiting side effects. Morphine and other routinely used opioids are mu opioid receptor (MOPR) agonists. The MOPR is responsible for most opioid-related adverse effects. In the present study, we revealed potent analgesic effects of an intrathecally-administered selective delta opioid receptor (DOPR) agonist, deltorphin II, in a recently developed rat bone cancer model. Indeed, we found that deltorphin II dose-dependently reversed mechanical allodynia 14 days post-surgery in this cancer pain model, which is based on the implantation of mammary MRMT-1 cells in the femur. This effect was DOPR-mediated as it was completely blocked by naltrindole, a selective DOPR antagonist. Using the complete Freund's adjuvant model of inflammatory pain, we further demonstrated that deltorphin II was equipotent at alleviating inflammatory and cancer pain (i.e. similar ED50 values). Altogether, the present results show, for the first time, that activation of spinal DOPRs causes significant analgesia at doses sufficient to reduce inflammatory pain in a rat bone cancer pain model. Our results further suggest that DOPR represents a potential target for the development of novel analgesic therapies to be used in the treatment of cancer-related pain.


Subject(s)
Analgesics, Opioid/therapeutic use , Bone Neoplasms/complications , Oligopeptides/therapeutic use , Pain Management , Pain/etiology , Receptors, Opioid, delta/metabolism , Animals , Bone Neoplasms/physiopathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Inflammation/chemically induced , Inflammation/complications , Inflammation/drug therapy , Injections, Spinal/methods , Male , Pain Measurement , Rats , Rats, Sprague-Dawley , Time Factors
8.
Neuroscience ; 170(4): 1286-94, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-20727387

ABSTRACT

Both neurotensin (NT) and opioid agonists have been shown to induce antinociception in rodents after central administration. Besides, previous studies have revealed the existence of functional interactions between NT and opioid systems in the regulation of pain processing. We recently demonstrated that NTS1 receptors play a key role in the mediation of the analgesic effects of NT in long-lasting pain. In the present study, we therefore investigated whether NTS1 gene deletion affected the antinociceptive action of mu opioid drugs. To this end, pain behavioral responses to formalin were determined following systemic administration of morphine in both male and female NTS1 knockout mice. Acute injection of morphine (2 or 5 mg/kg) produced strong antinociceptive effects in both male and female wild-type littermates, with no significant sex differences. On the other hand, morphine analgesia was considerably reduced in NTS1-deficient mice of both sexes compared to their respective controls, indicating that the NTS1 receptor actively participates in mu opioid alleviating pain. By examining specifically the flinching, licking and biting nociceptive behaviors, we also showed that the functional crosstalk between NTS1 and mu opioid receptors influences the supraspinally-mediated behaviors. Interestingly, sexual dimorphic action of morphine-induced pain inhibition was found in NTS1 null mice in the formalin test, suggesting that the endogenous NT system interacts differently with the opioid network in male and female mice. Altogether, these results demonstrated that NTS1 receptor activation operates downstream to the opioidergic transmission and that NTS1-selective agonists combined with morphine may act synergistically to reduce persistent pain.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Pain/physiopathology , Receptors, Neurotensin/physiology , Receptors, Opioid, mu/physiology , Animals , Female , Male , Mice , Mice, Knockout , Pain/psychology , Pain Measurement , Receptor Cross-Talk , Receptors, Neurotensin/genetics , Receptors, Opioid, mu/agonists , Sex Factors , Signal Transduction
9.
Neuroscience ; 166(2): 639-52, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20035838

ABSTRACT

Stress activates multiple neural systems that suppress pain sensation. This adaptive phenomenon referred as stress-induced analgesia (SIA) is mediated by the activation of endogenous pain inhibitory systems. Both opioid and non-opioid forms of SIA have been elicited in rodents according to stressor parameters and duration. There is accumulating evidence that the endogenous neurotensin (NT) system plays an important role in SIA. Especially, NT-deficient mice were shown to exhibit reduced SIA following water avoidance or restraint stress. Since central NT produces naloxone-insensitive analgesic effects by acting on spinal and supraspinal NTS2 receptors, we hypothesized that NT might mediate non-opioid SIA through NTS2 activation. Here, we evaluated the influence of an opioid-independent severe stress produced by a cold-water swim for 3 min at 15 degrees C on rodent offspring's pain perception. Our results demonstrated that mice lacking NTS2 exhibit significantly reduced SIA following cold-water swim stress. Indeed, NTS2 knockout mice submitted to both acute (plantar test) and tonic (formalin test) pain stimuli show a greater sensitivity to pain in comparison to wild-type littermates. Accordingly, pretreatment with the NT receptor antagonist SR142948A results in a hyperalgesic response to stress induced by cold-water swim. Endogenous NT regulates hypothalamic-pituitary-adrenal axis activity in stress condition by increasing corticosterone plasma levels. Accordingly, the plasma levels of corticosterone measured by radioimmunoassay are significantly reduced in non-stressed and stressed NTS2-deficient mice in comparison with wild-type mice. To further investigate the site of action of NT in mediating SIA, we microinjected NTS2 agonists in lumbar spinal cord and quantified post-stress sensitivity to pain in rats using the plantar test. Exogenously administered NTS2 analogs, JMV-431, beta-lactotensin and NT69L markedly enhance the magnitude and duration of stress antinociception in both 25- and 60-day-old rats. In sum, by using genetic and pharmacological approaches, we demonstrated here that NTS2 receptors mediate non-opioid SIA. Our results also revealed that the release of endogenous NT in response to stress requires the presence of NTS2 to stimulate corticotropin-releasing factor (CRF)-induced elevation of plasma corticosterone, and that NTS2 receptors localized at the lumbar spinal cord participate to the disinhibition of descending pain control pathways. Therefore, these data highlight the significance of NTS2 as a novel target for the treatment of pain and stress-related disorders.


Subject(s)
Pain/genetics , Receptors, Neurotensin/metabolism , Stress, Physiological/genetics , Stress, Psychological/genetics , Analgesia , Analysis of Variance , Animals , Corticosterone/blood , Mice , Mice, Knockout , Neurotensin/metabolism , Pain/blood , Pain/chemically induced , Pain Measurement , Rats , Receptors, Neurotensin/genetics , Spinal Cord/metabolism , Stress, Psychological/blood , Swimming
10.
Neuroscience ; 158(2): 673-82, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19010395

ABSTRACT

A critical issue in animal models of perinatal brain injury is to adapt the pertinent pathophysiological scenarios to their corresponding developmental window in order to induce neuropathological and behavioral characteristics reminiscent to perinatal cerebral palsy (CP). A major problem in most of these animal models designed up to now is that they do not present motor deficits characteristic of CP. Using a unique rat paradigm of prenatal inflammation combined to an early postnatal hypoxia-ischemia pertinent to the context of very early premature human newborns, we were interested in finding out if such experimental conditions might reproduce both histological damages and behavioral deficits previously described in the human context. We showed that exposure to lipopolysaccharide (LPS) or hypoxia-ischemia (H/I) induced behavioral alterations in animals subjected to forced motor activity. When both LPS and H/I aggressions were combined, the motor deficits reached their highest intensity and affected both spontaneous and forced motor activities. LPS+H/I-exposed animals also showed extensive bilateral cortical and subcortical lesions of the motor networks affecting the frontal cortices and underlying white matters fascicles, lenticular nuclei and the substantia nigra. These neuropathological lesions and their associated motor behavioral deficits are reminiscent of those observed in very preterm human neonates affected by subsequent CP and validate the value of the present animal model to test new therapeutic strategies which might open horizons for perinatal neuroprotection.


Subject(s)
Cerebral Palsy/etiology , Hypoxia-Ischemia, Brain/complications , Lipopolysaccharides , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Age Factors , Animals , Animals, Newborn , Behavior, Animal , Calcium-Binding Proteins/metabolism , Cell Count , Disease Models, Animal , Female , Functional Laterality , Glial Fibrillary Acidic Protein/metabolism , Hypoxia-Ischemia, Brain/pathology , Immobility Response, Tonic/physiology , Locomotion/physiology , Male , Microfilament Proteins , Pregnancy , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Inbred Lew , Tyrosine 3-Monooxygenase/metabolism
11.
Curr Med Chem ; 15(27): 2866-75, 2008.
Article in English | MEDLINE | ID: mdl-18991641

ABSTRACT

Physical insults including but not limited to nerve damage, inflammation, visceral pathologies and cancer generate long lasting pain commonly referred as chronic pain. Recently, members of the chemokine family and their receptors emerged as key modulators in nociceptive influx transmission in neuropathic and inflammatory chronic pain models. To this day, rodents defective in specific chemokine receptors have provided evidence of the implication of chemokine in pain sensitivity. In addition, up-regulation of chemokines and their receptors at multiple levels in the central nervous (CNS) and peripheral (PNS) systems is associated in the development of chronic pain. Indeed, we point out the fact that chemokines are synthesized and released by both neuronal and non-neuronal cells and act as neuromodulators. Even if their functional roles in the CNS remain largely unknown, chemokines participate in the glial activation and modulation of neuronal excitability as well as neurotransmitter release. This review focuses on three chemokines (i.e. CCL2, CXCL12, CX3CL1) recently identified as important mediators of the initiation and maintenance of pain hypersensitivity, thus broadening the panel of new strategies for the management of chronic pain.


Subject(s)
Chemokines/physiology , Nervous System Physiological Phenomena/physiology , Pain/physiopathology , Analgesics/chemistry , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Chemokines/antagonists & inhibitors , Humans , Molecular Structure , Nervous System Physiological Phenomena/drug effects , Nociceptors/drug effects , Nociceptors/physiology , Pain/drug therapy , Receptors, Cytokine/antagonists & inhibitors , Receptors, Cytokine/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
12.
FEBS Lett ; 495(1-2): 100-5, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11322955

ABSTRACT

We recently reported the molecular identification of a new type of receptor for the neuropeptide neurotensin (NT), the neurotensin receptor 3 (NTR3), identical to sortilin, which binds receptor-associated protein. Here, we demonstrate that the cloned mouse NTR3 is expressed on the plasma membrane of transfected COS-7 cells. The mouse NTR3 is detectable by photoaffinity labeling and immunoblotting at the cell surface as a 100 kDa N-glycosylated protein. Biochemical analysis and confocal microscopic imaging clearly indicate that NT is efficiently internalized after binding to NTR3, and that despite this internalization, the amount of receptor present on the cell surface is maintained.


Subject(s)
Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Neurotensin/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Biological Transport , Blotting, Western , COS Cells , Cell Membrane/metabolism , Gene Expression , Immunoblotting , Iodine Radioisotopes , Membrane Glycoproteins/genetics , Mice , Microscopy, Confocal , Molecular Weight , Nerve Tissue Proteins/genetics , Neurotensin/pharmacokinetics , Photoaffinity Labels , Radioligand Assay , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Subcellular Fractions/metabolism , Substrate Specificity , Transfection
13.
Int J Cancer ; 92(4): 503-9, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11304684

ABSTRACT

The expression of the 3 currently known neurotensin receptors was studied in human cancer cells of prostatic, colonic or pancreatic origin by means of RT-PCR analysis and binding experiments. All the cells selected for this work have been shown to exhibit a growth response to neurotensin. We found that the 7 transmembrane domain, levocabastine insensitive receptor (NTR1) is expressed in most but not all of the cells studied whereas the 7 transmembrane domain, levocabastine sensitive receptor (NTR2) is present in none of these cells. The 100 kDa-type I neurotensin receptor (NTR3) is expressed in all the cells assayed. Moreover, we demonstrated that neurotensin can stimulate the growth of CHO cells stably transfected with the NTR3. Taken together, our results strongly suggest that the NTR3 subtype could be involved in the growth response of human cancer cells to neurotensin.


Subject(s)
Neurotensin/metabolism , Neurotensin/pharmacology , Receptors, Neurotensin/biosynthesis , Animals , CHO Cells , Cell Membrane/metabolism , Cholic Acids/pharmacology , Colonic Neoplasms/metabolism , Cricetinae , Drug Resistance, Neoplasm , Electrophoresis, Polyacrylamide Gel , Humans , Kinetics , Male , Pancreatic Neoplasms/metabolism , Piperidines/pharmacology , Prostatic Neoplasms/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Receptors, Neurotensin/chemistry , Receptors, Neurotensin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Tumor Cells, Cultured
14.
J Neurosci ; 20(16): 5932-9, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10934240

ABSTRACT

To investigate the effects of somatostatin (somatotropin release-inhibiting factor, SRIF) on the regulation of SST(2A) receptors in mammalian brain, we examined how blockade of SRIF release or stimulation by the SRIF analog [d-Trp(8)]-SRIF would affect the expression and cell surface availability of SST(2A) receptors in rat brain slices. First, we measured the intensity of SST(2A) immunoreactivity, using quantitative light microscopic immunocytochemistry, and levels of SST(2A) mRNA, using semiquantitative RT-PCR, under conditions of acute SRIF release blockade. Incubation of slices from the claustrum or basolateral amygdala, two regions previously shown to contain high concentrations of SST(2A) receptors, in Ca(2+)-free Ringer's for 40 min induced a decrease in the intensity of SST(2A) receptor immunoreactivity and concentration of SST(2A) mRNA as compared with control values obtained in Ca(2+)-supplemented Ringer's. These effects were counteracted in a dose-dependent manner by the addition of 10-100 nm [d-Trp(8)]-SRIF to the Ca(2+)-free medium. Furthermore, both of these effects were abolished in the presence of the endocytosis inhibitors phenylarsine oxide or hyperosmolar sucrose, suggesting that they were dependent on receptor internalization. Electron microscopic immunogold labeling confirmed the existence of an agonist-induced internalization of SST(2A) receptors in central neurons. At a high (10 microm), but not at a low (10 nm), concentration of agonist this internalization resulted in a significant decrease in cell surface receptor density, irrespective of the presence of Ca(2+) in the medium. Taken together, these results suggest that ligand-induced endocytosis is responsible for rapid transcriptional (increase in SST(2A) expression) and trafficking (loss of cell surface receptors) events involved in the control of the somatostatinergic signal.


Subject(s)
Brain/drug effects , Neurons/drug effects , Receptors, Cell Surface/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Amygdala/drug effects , Amygdala/metabolism , Amygdala/ultrastructure , Animals , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Basal Ganglia/ultrastructure , Brain/metabolism , Brain/ultrastructure , In Vitro Techniques , Male , Neurons/metabolism , Neurons/ultrastructure , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/drug effects , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/genetics , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
15.
Endocrinology ; 141(1): 354-65, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10614658

ABSTRACT

Internalization of G protein-coupled receptors is crucial for resensitization of phosphorylation-desensitized receptors, but also for their long term desensitization through sequestration. To elucidate the mechanisms regulating cell surface availability of the somatostatin (SRIF) receptor subtype sst5, we characterized its internalization properties in transfected COS-7 cells using biochemical, confocal microscopic, and electron microscopic techniques. Our results demonstrated rapid and efficient sequestration of specifically bound [125I]Tyr0-D-Trp8-SRIF (up to 45% of bound radioactivity). Combined immunocytochemical detection of sst5 and visualization of a fluorescent SRIF analog by confocal microscopy revealed that whereas the internalized ligand progressively clustered toward the cell center with time, immunoreactive receptors remained predominantly associated with the plasma membrane. The preservation of cell surface receptors was confirmed by binding experiments on whole cells revealing a lack of saturability of [125I]Tyr0-D-Trp8-SRIF binding at 37 C. Binding was rendered saturable by the drug monensin, showing that receptor recycling played a key role in the preservation of cell surface receptors. Electron microscopy demonstrated that in addition to receptor recycling, internalization of receptor-ligand complexes triggered a massive recruitment of sst5 receptor molecules from intracellular stores to the membrane. This combination of recycling and recruitment of spare receptors may protect sst5 from long term down-regulation through sequestration and, therefore, facilitate extended SRIF signaling.


Subject(s)
Endocytosis/physiology , Receptors, Somatostatin/metabolism , Transfection/genetics , Animals , COS Cells , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Endocytosis/drug effects , Endocytosis/genetics , Kinetics , Ligands , Microscopy, Confocal , Microscopy, Electron , Somatostatin/analogs & derivatives , Somatostatin/metabolism
16.
J Biol Chem ; 274(27): 19294-300, 1999 Jul 02.
Article in English | MEDLINE | ID: mdl-10383439

ABSTRACT

The inhibitory effect of the neuropeptide somatostatin on the expression of growth hormone was measured by quantitative polymerase chain reaction in the pituitary cell line AtT-20. We demonstrate that this effect is dependent on the internalization of somatostatin-receptor complexes and that it is totally independent from the peptide-induced inhibition of adenylate cyclase. Indeed, the inhibitory effect of the peptide on growth hormone mRNA levels was totally insensitive to pertussis toxin treatment but was totally abolished under conditions which block somatostatin receptor internalization. Comparative confocal microscopic imaging of fluorescent somatostatin sequestration and fluorescence immunolabeling of sst1, sst2A, and sst5 receptors suggests that sst2A is most probably responsible of the inhibitory effect of somatostatin on growth hormone expression.


Subject(s)
Growth Hormone/biosynthesis , Pituitary Gland/metabolism , Receptors, Somatotropin/metabolism , Somatostatin/metabolism , Animals , Cyclic AMP/metabolism , Ligands , Mice , Pituitary Neoplasms/metabolism , RNA, Messenger/metabolism , Tumor Cells, Cultured
17.
J Neurosci ; 19(1): 503-10, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9870978

ABSTRACT

The neuropeptide neurotensin (NT) elicits hypothermic and naloxone-insensitive analgesic responses after brain injection. Recent pharmacological evidence obtained with NT agonists and antagonists suggests that these effects are mediated by a receptor distinct from the initially cloned high-affinity NT receptor (NTR1). The recent cloning of a second NT receptor (NTR2) prompted us to evaluate its role in NT-induced analgesia. Intracerebroventricular injections in mice of two different antisense oligodeoxynucleotides from the NTR2 markedly decreased NTR2 mRNA and protein and reduced NT-induced analgesia. This effect was specific, because NTR1 levels were unaffected, and sense or scramble oligodeoxynucleotides had no effect. Structure-activity studies revealed a close correlation between the analgesic potency of NT analogs and their affinity for the NTR2 and disclosed potent and selective agonists of this receptor. These data confirm that NTR1 is involved in the NT-elicited turning behavior and demonstrate that the NTR2 mediates NT-induced analgesia.


Subject(s)
Analgesics/pharmacology , Neurotensin/pharmacology , Receptors, Neurotensin/drug effects , Analysis of Variance , Animals , Body Temperature Regulation/drug effects , CHO Cells , Cricetinae , Injections, Intraventricular , Male , Mice , Neurotensin/metabolism , Oligodeoxyribonucleotides, Antisense , RNA, Messenger/biosynthesis , Receptors, Neurotensin/metabolism , Structure-Activity Relationship
18.
Neuroscience ; 94(4): 1189-97, 1999.
Article in English | MEDLINE | ID: mdl-10625058

ABSTRACT

The pharmacological properties, molecular identity and physiopathological regulation of neurotensin receptors expressed by central astrocytes were investigated in primary glial cultures and sections from the adult rat brain. Binding experiments carried out on astrocytes in culture revealed the presence of a single apparent class of neurotensin binding sites. These sites bound [125]neurotensin with an affinity (6 nM) comparable to that of the recently cloned NT2 low-affinity receptor expressed in transfected cells. The glial receptor was sensitive to the antihistamine, levocabastine, but less so than the NT2 site expressed in heterologous expression systems, suggesting the presence of an additional site or a differential coupling of the NT2 receptor in glia. Reverse transcription-polymerase chain reaction experiments demonstrated that both NT2 and NT3 neurotensin receptor sub-types were in fact expressed by cortical glial cells in culture. Confocal microscopic visualization of specifically bound fluorescent neurotensin indicated that this expression concerned only a sub-population of astrocytes in culture, in conformity with earlier reports of a heterogeneous expression of neuropeptides and their receptors by glial cells. To further investigate the functionality of NT2 receptors expressed in astrocytes, dual immunohistochemical labeling of glial fibrillary acidic protein and in situ hybridization of NT2 messenger RNA was performed on sections of normal and lesioned rat brain. In sections from normal brain, only a small subset of immunolabeled astrocytes hybridized NT2 messenger RNA. By contrast, in sections of stab-wounded rat brains, there was a marked increase in the number of NT2-hybridizing astrocytes in the surround of the lesion. Furthermore, NT2 expression within immunopositive reactive astrocytes was significantly enhanced as compared to immunolabeled glial cells in the brain of control animals. These results indicate that NT2 receptor expression is up-regulated during astrocytic reaction, suggesting that NT2 receptors may play a role in regulating glial response to injury.


Subject(s)
Neuroglia/metabolism , Receptors, Neurotensin/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cell Division , Cells, Cultured , Glial Fibrillary Acidic Protein/metabolism , In Situ Hybridization , Microscopy, Confocal , RNA, Messenger/metabolism , Rats , Receptors, Neurotensin/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
Neuroendocrinology ; 68(1): 37-43, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9695937

ABSTRACT

The expression of sst2A and sst2B somatostatin (SRIH) receptor splice variants was examined by polymerase chain reaction (PCR) in rat brain and pituitary. Two sets of primers chosen to recognize either both sst2A and sst2B or only the sst2B transcript yielded bands of the size predicted from the cloned sst2A and sst2B sequences in both mouse and rat tissue extracts. Accordingly, experiments carried out on mouse brain extracts and on AtT-20 pituitary cells yielded sst2A/sst2B expression ratios comparable to those previously published. As in the mouse, rat pituitary extracts contained both sst2A and sst2B transcripts. By contrast, various cerebral regions in which both sst2A and sst2B forms were detected in the mouse contained only sst2A form in rat brain, with the exception of the cerebral cortex which also showed weak sst2B expression. No sst2B mRNA was detected in the arcuate nucleus/median eminence complex, indicating that the sst2A form is the one that is associated with growth-hormone-releasing-hormone-containing neurons documented to express sst2 receptors in this region. Finally, only sst2A transcripts were detected in rat astrocytes in culture, suggesting that this form of the receptor is responsible for sst2-mediated effects of SRIH on these cells.


Subject(s)
Brain/metabolism , Gene Expression , Genetic Variation , Pituitary Gland/metabolism , RNA Splicing , Receptors, Somatostatin/genetics , Animals , Base Sequence , Cloning, Molecular , DNA/chemistry , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Tissue Distribution
20.
J Comp Neurol ; 394(3): 344-56, 1998 May 11.
Article in English | MEDLINE | ID: mdl-9579398

ABSTRACT

Levocabastine-sensitive neurotensin receptor (NTRL) mRNAs were localized by in situ hybridization in adult and developing mouse brain. NTRL hybridization signal was widely distributed throughout the neuraxis. The highest concentrations of NTRL mRNA were detected in the olfactory system, olfactory tubercle, cerebral and cerebellar cortices, hippocampal formation, and selective hypothalamic nuclei. Moderate to dense hybridization signal was also observed in association with a variety of auditory, visual, and somatosensory relay nuclei, suggesting that the NTRL might be involved in a widespread modulation of primary afferent pathways. Finally a high expression of NTRL was evident in brainstem structures implicated in descending antinociceptive influences (e.g., the periaqueductal gray, nucleus raphe magnus, gigantocellular reticular nucleus, pars alpha, and lateral paragigantocellular nucleus) consistent with the proposed mediation of NT-induced analgesia by the NTRL. Although most of the regions found here to express NTRL mRNA were previously reported to be devoid of mRNA encoding the high affinity NT receptor (NTRH), a few areas (e.g., the anterior olfactory nucleus, medial septum, diagonal band of Broca, reticular thalamic nucleus, suprachiasmatic hypothalamic nucleus, and pontine nucleus) were enriched in both receptor subtypes, suggesting a possible coexpression of these receptors by the same cells. Ontogenic studies revealed that in the mouse brain, NTRL mRNA was detected only from postnatal day 14 and did not reach adulthood concentrations before day 30. In cerebral cortex, the developmental increase in NTRL expression was correlated over time with the decrease in NTRH expression previously documented in the rat, suggesting a progressive takeover of the latter by the former for transduction of the effects of NT in this structure.


Subject(s)
Brain/metabolism , RNA, Messenger/analysis , Receptors, Neurotensin/genetics , Animals , Brain/growth & development , Female , In Situ Hybridization , Male , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...