Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Neural Circuits ; 15: 639900, 2021.
Article in English | MEDLINE | ID: mdl-33897379

ABSTRACT

A key function of the mesencephalic locomotor region (MLR) is to control the speed of forward symmetrical locomotor movements. However, the ability of freely moving mammals to integrate environmental cues to brake and turn during MLR stimulation is poorly documented. Here, we investigated whether freely behaving mice could brake or turn, based on environmental cues during MLR stimulation. We photostimulated the cuneiform nucleus (part of the MLR) in mice expressing channelrhodopsin in Vglut2-positive neurons in a Cre-dependent manner (Vglut2-ChR2-EYFP) using optogenetics. We detected locomotor movements using deep learning. We used patch-clamp recordings to validate the functional expression of channelrhodopsin and neuroanatomy to visualize the stimulation sites. In the linear corridor, gait diagram and limb kinematics were similar during spontaneous and optogenetic-evoked locomotion. In the open-field arena, optogenetic stimulation of the MLR evoked locomotion, and increasing laser power increased locomotor speed. Mice could brake and make sharp turns (~90°) when approaching a corner during MLR stimulation in the open-field arena. The speed during the turn was scaled with the speed before the turn, and with the turn angle. Patch-clamp recordings in Vglut2-ChR2-EYFP mice show that blue light evoked short-latency spiking in MLR neurons. Our results strengthen the idea that different brainstem neurons convey braking/turning and MLR speed commands in mammals. Our study also shows that Vglut2-positive neurons of the cuneiform nucleus are a relevant target to increase locomotor activity without impeding the ability to brake and turn when approaching obstacles, thus ensuring smooth and adaptable navigation. Our observations may have clinical relevance since cuneiform nucleus stimulation is increasingly considered to improve locomotion function in pathological states such as Parkinson's disease, spinal cord injury, or stroke.


Subject(s)
Mesencephalon , Optogenetics , Animals , Brain Stem , Electric Stimulation , Locomotion , Mice , Neurons
2.
Behav Brain Res ; 405: 113189, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33607165

ABSTRACT

The endogenous tridecapeptide neurotensin (NT) has emerged as an important inhibitory modulator of pain transmission, exerting its analgesic action through the activation of the G protein-coupled receptors, NTS1 and NTS2. Whereas both NT receptors mediate the analgesic effects of NT, NTS1 activation also produces hypotension and hypothermia, which may represent obstacles for the development of new pain medications. In the present study, we implemented various chemical strategies to improve the metabolic stability of the biologically active fragment NT(8-13) and assessed their NTS1/NTS2 relative binding affinities. We then determined their ability to reduce the nociceptive behaviors in acute, tonic, and chronic pain models and to modulate blood pressure and body temperature. To this end, we synthesized a series of NT(8-13) analogs carrying a reduced amide bond at Lys8-Lys9 and harboring site-selective modifications with unnatural amino acids, such as silaproline (Sip) and trimethylsilylalanine (TMSAla). Incorporation of Sip and TMSAla respectively in positions 10 and 13 of NT(8-13) combined with the Lys8-Lys9 reduced amine bond (JMV5296) greatly prolonged the plasma half-life time over 20 h. These modifications also led to a 25-fold peptide selectivity toward NTS2. More importantly, central delivery of JMV5296 was able to induce a strong antinociceptive effect in acute (tail-flick), tonic (formalin), and chronic inflammatory (CFA) pain models without inducing hypothermia. Altogether, these results demonstrate that the chemically-modified NT(8-13) analog JMV5296 exhibits a better therapeutic profile and may thus represent a promising avenue to guide the development of new stable NT agonists and improve pain management.


Subject(s)
Acute Pain/drug therapy , Analgesia , Analgesics/pharmacology , Behavior, Animal/drug effects , Chronic Pain/drug therapy , Neurotensin/pharmacology , Nociceptive Pain/drug therapy , Analgesics/chemistry , Animals , Disease Models, Animal , Male , Neurotensin/analysis , Rats , Rats, Sprague-Dawley
3.
J Med Chem ; 63(21): 12929-12941, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32902268

ABSTRACT

Fusion of nonopioid pharmacophores, such as neurotensin, with opioid ligands represents an attractive approach for pain treatment. Herein, the µ-/δ-opioid agonist tetrapeptide H-Dmt-d-Arg-Aba-ß-Ala-NH2 (KGOP01) was fused to NT(8-13) analogues. Since the NTS1 receptor has been linked to adverse effects, selective MOR-NTS2 ligands are preferred. Modifications were introduced within the native NT sequence, particularly a ß3-homo amino acid in position 8 and Tyr11 substitutions. Combination of ß3hArg and Dmt led to peptide 7, a MOR agonist, showing the highest NTS2 affinity described to date (Ki = 3 pM) and good NTS1 affinity (Ki = 4 nM), providing a >1300-fold NTS2 selectivity. The (6-OH)Tic-containing analogue 9 also exhibited high NTS2 affinity (Ki = 1.7 nM), with low NTS1 affinity (Ki = 4.7 µM), resulting in an excellent NTS2 selectivity (>2700). In mice, hybrid 7 produced significant and prolonged antinociception (up to 8 h), as compared to the KGOP01 opioid parent compound.


Subject(s)
Drug Design , Peptides/chemistry , Receptors, Neurotensin/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Amino Acid Sequence , Animals , Disease Models, Animal , Humans , Male , Mice , Oligopeptides/chemistry , Oligopeptides/metabolism , Oligopeptides/therapeutic use , Pain/drug therapy , Pain/pathology , Peptides/metabolism , Peptides/therapeutic use , Protein Binding , Receptors, Neurotensin/chemistry , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Structure-Activity Relationship
4.
Front Chem ; 8: 406, 2020.
Article in English | MEDLINE | ID: mdl-32582624

ABSTRACT

Therapeutic hypothermia represents a brain-protective strategy for multiple emergency situations, such as stroke or traumatic injury. Neurotensin (NT), which exerts its effects through activation of two G protein-coupled receptors, namely NTS1 and NTS2, induces a strong and long-lasting decrease in core body temperature after its central administration. Growing evidence demonstrates that NTS1 is the receptor subtype mediating the hypothermic action of NT. As such, potent NTS1 agonists designed on the basis of the minimal C-terminal NT(8-13) bioactive fragment have been shown to produce mild hypothermia and exert neuroprotective effects under various clinically relevant conditions. The high susceptibility of NT(8-13) to protease degradation (half-life <2 min) represents, however, a serious limitation for its use in pharmacological therapy. In light of this, we report here a structure-activity relationship study in which pairs of NT(8-13) analogs have been developed, based on the incorporation of a reduced Lys8-Lys9 bond. To further stabilize the peptide bonds, a panel of backbone modifications was also inserted along the peptide sequence, including Sip10, D-Trp11, Dmt11, Tle12, and TMSAla13. Our results revealed that the combination of appropriate chemical modifications leads to compounds exhibiting improved resistance to proteolytic cleavages (>24 h; 16). Among them, the NT(8-13) analogs harboring the reduced amine bond combined with the unnatural amino acids TMSAla13 (4) and Sip10 (6) or the di-substitution Lys11 - TMSAla13 (12), D-Trp11-TMSAla13 (14), and Dmt11-Tle12 (16) produced sustained hypothermic effects (-3°C for at least 1 h). Importantly, we observed that hypothermia was mainly driven by the increased stability of the NT(8-13) derivatives, instead of the high binding-affinity at NTS1. Altogether, these results reveal the importance of the reduced amine bond in optimizing the metabolic properties of the NT(8-13) peptide and support the development of stable NTS1 agonists as first drug candidate in neuroprotective hypothermia.

5.
J Med Chem ; 61(16): 7103-7115, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30035538

ABSTRACT

The neurotensin receptors are attractive targets for the development of new analgesic compounds. They represent potential alternatives or adjuvants to opioids. Herein, we report the structural optimization of our recently reported macrocyclic peptide analogues of NT(8-13). The macrocycle was formed via ring-closing metathesis (RCM) between an ortho allylated tyrosine residue in position 11 and the side chain of alkene-functionalized amino acid in position 8 of NT(8-13). Minute modifications led to significant binding affinity improvement ( Ki improved from 5600 to 15 nM) with greatly improved plasma stability compared to NT(8-13). This study also delineates the structural features influencing these parameters. The signaling profiles of the new macrocycles were determined on the NTS1 receptor, and the physiological effects of the two most potent and stable analogues were assessed in vivo using rodent models. Both compounds displayed strong analgesic effects.


Subject(s)
Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Neurotensin/pharmacology , Peptide Fragments/pharmacology , Peptides, Cyclic/chemistry , Receptors, Neurotensin/metabolism , Animals , Binding, Competitive , Blood Pressure/drug effects , Body Temperature/drug effects , CHO Cells , Cricetulus , Cyclization , Drug Evaluation, Preclinical/methods , Drug Stability , Male , Molecular Docking Simulation , Neurotensin/agonists , Neurotensin/chemistry , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptides, Cyclic/blood , Peptides, Cyclic/pharmacology , Rats, Sprague-Dawley , Receptors, Neurotensin/chemistry , Structure-Activity Relationship , Tyrosine/chemistry
6.
J Med Chem ; 60(8): 3303-3313, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28368584

ABSTRACT

Neurotensin exerts potent analgesia by acting at both NTS1 and NTS2 receptors, whereas NTS1 activation also results in other physiological effects such as hypotension and hypothermia. Here, we used molecular modeling approach to design highly selective NTS2 ligands by investigating the docking of novel NT[8-13] compounds at both NTS1 and NTS2 sites. Molecular dynamics simulations revealed an interaction of the Tyr11 residue of NT[8-13] with an acidic residue (Glu179) located in the ECL2 of hNTS2 or with a basic residue (Arg212) at the same position in hNTS1. The importance of the residue at position 11 for NTS1/NTS2 selectivity was further demonstrated by the design of new NT analogues bearing basic (Lys, Orn) or acid (Asp or Glu) function. As predicted by the molecular dynamics simulations, binding of NT[8-13] analogues harboring a Lys11 exhibited higher affinity toward the hNTS1-R212E mutant receptor, in which Arg212 was substituted by the negatively charged Glu residue.


Subject(s)
Drug Design , Molecular Dynamics Simulation , Neurotensin/analogs & derivatives , Amino Acid Sequence , Neurotensin/metabolism , Receptors, Neurotensin/chemistry , Receptors, Neurotensin/metabolism
7.
ACS Chem Neurosci ; 7(9): 1225-31, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27359371

ABSTRACT

Neurotensin receptor type 2 (NTS2) compounds display analgesic activity in animal pain models. We have identified the first high-affinity NTS2-selective antagonist (8) that is active in vivo. This study also revealed that the NTS2 FLIPR assay designation for a compound, agonist, partial agonist, and so forth, did not correlate with its in vivo activity as observed in the thermal tail-flick acute model of pain. This suggests that calcium mobilization is not the signaling pathway involved in NTS2-mediated analgesia as assessed by the thermal tail-flick model. Finally, we found a significant bias between rat and human for compound 9 in the NTS2 binding assay.


Subject(s)
Analgesics/therapeutic use , Carboxylic Acids/chemistry , Neurotransmitter Agents/pharmacology , Pain/drug therapy , Piperidines/chemistry , Receptors, Neurotensin/antagonists & inhibitors , Receptors, Neurotensin/metabolism , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacology , Analysis of Variance , Animals , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Calcium/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Hindlimb Suspension , Humans , Injections, Spinal , Male , Neurotransmitter Agents/chemical synthesis , Neurotransmitter Agents/chemistry , Pain/physiopathology , Protein Binding/drug effects , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Radioligand Assay , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects
8.
Can Vet J ; 56(7): 730-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26130835

ABSTRACT

Meloxicam, a non-steroidal anti-inflammatory drug, is approved for use in horses in several countries, but an equine formulation is not available in North America. However, meloxicam is being used in an extra-label manner in horses in Canada. The purpose of this study, therefore, was to assess the bioequivalence of an approved oral meloxicam suspension (Metacam 15 mg/mL for horses; Boehringer Ingelheim Vetmedica GmBH, Ingelheim, Germany) from the European Union with human meloxicam tablets (Meloxicam 15 mg tablets; TEVA Canada, Toronto, Ontario) compounded with molasses to improve palatability and administration. The geometric mean ratios (GMR test/reference) and the 90% confidence intervals of the pivotal pharmacokinetic parameters (area under the curve and maximum concentration) were within the defined limits of 80% to 125% generally accepted for products to be considered bioequivalent. Therefore, use of human meloxicam tablets compounded with molasses would be expected to produce a similar clinical response in horses as the approved oral product from the European Union.


Pharmacocinétique et bioéquivalence de 2 formulations de posologie orale de méloxicam chez des chevaux adultes en santé. Le méloxicam, un médicament anti-inflammatoire non stéroïdien, est approuvé pour utilisation chez les chevaux dans plusieurs pays, mais une formulation équine n'est pas disponible en Amérique du Nord. Cependant, le méloxicam est utilisé en dérogation des directives de l'étiquette chez les chevaux du Canada. Par conséquent, le but de la présente étude était d'évaluer la bioéquivalence d'une suspension orale approuvée de méloxicam (Metacam 15 mg/ml pour les chevaux; Boehringer Ingelheim Vetmedica GmBH, Ingelheim, Allemagne) de l'Union européenne avec celle des comprimés de méloxicam pour les humains (comprimés de 15 mg de méloxicam; TEVA Canada, Toronto, Ontario) préparés avec de la mélasse pour améliorer la sapidité et l'administration. Les ratios géométriques moyens (test RGM/référence) et les intervalles de confiance de 90 % des paramètres phamacocinétiques clés (secteur sous la courbe et concentration maximale) se situaient dans les limites définies de 80 % à 125 % généralement attendues pour des produits considérés comme bioéquivalents. Par conséquent, l'utilisation des comprimés de méloxicam pour humains préparés avec de la mélasse devrait produire une réponse clinique semblable chez les chevaux à celle du produit oral approuvé provenant de l'Union européenne.(Traduit par Isabelle Vallières).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Horses/metabolism , Thiazines/pharmacokinetics , Thiazoles/pharmacokinetics , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Area Under Curve , Cross-Over Studies , Dosage Forms , Female , Half-Life , Horses/blood , Male , Meloxicam , Therapeutic Equivalency , Thiazines/administration & dosage , Thiazines/chemistry , Thiazoles/administration & dosage , Thiazoles/chemistry
9.
BMC Cancer ; 15: 479, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26104798

ABSTRACT

BACKGROUND: Valosin containing protein (VCP) is a critical mediator of protein homeostasis and may represent a valuable therapeutic target for several forms of cancer. Overexpression of VCP occurs in many cancers, and often in a manner correlating with malignancy and poor outcome. Here, we analyzed VCP expression in canine lymphoma and assessed its potential as a therapeutic target for this disease. METHODS: VCP expression in canine lymphomas was evaluated by immunoblotting and immunohistochemistry. The canine lymphoma cell lines CLBL-1, 17-71 and CL-1 were treated with the VCP inhibitor Eeyarestatin 1 (EER-1) at varying concentrations and times and were assessed for viability by trypan blue exclusion, apoptosis by TUNEL and caspase activity assays, and proliferation by propidium iodide incorporation and FACS. The mechanism of EER-1 action was determined by immunoblotting and immunofluorescence analyses of Lys48 ubiquitin and markers of ER stress (DDIT3), autophagy (SQSTM1, MAP1LC3A) and DNA damage (γH2AFX). TRP53/ATM-dependent signaling pathway activity was assessed by immunoblotting for TRP53 and phospho-TRP53 and real-time RT-PCR measurement of Cdkn1a mRNA. RESULTS: VCP expression levels in canine B cell lymphomas were found to increase with grade. EER-1 treatment killed canine lymphoma cells preferentially over control peripheral blood mononuclear cells. EER-1 treatment of CLBL-1 cells was found to both induce apoptosis and cell cycle arrest in G1. Unexpectedly, EER-1 did not appear to act either by inducing ER stress or inhibiting the aggresome-autophagy pathway. Rather, a rapid and dramatic increase in γH2AFX expression was noted, indicating that EER-1 may act by promoting DNA damage accumulation. Increased TRP53 phosphorylation and Cdkn1a mRNA levels indicated an activation of the TRP53/ATM DNA damage response pathway in response to EER-1, likely contributing to the induction of apoptosis and cell cycle arrest. CONCLUSIONS: These results correlate VCP expression with malignancy in canine B cell lymphoma. The selective activity of EER-1 against lymphoma cells suggests that VCP will represent a clinically useful therapeutic target for the treatment of lymphoma. We further suggest a mechanism of EER-1 action centered on the DNA repair response that may be of central importance for the design and characterization of VCP inhibitory compounds for therapeutic use.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , DNA Damage/drug effects , Lymphoma/genetics , Lymphoma/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Gene Expression , Lymph Nodes/pathology , Lymphoma/pathology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Valosin Containing Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...