Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Cereb Blood Flow Metab ; 42(7): 1294-1308, 2022 07.
Article in English | MEDLINE | ID: mdl-35107038

ABSTRACT

Perinatal hypoxic/ischemic (HI) brain injury is a major clinical problem with devastating neurodevelopmental outcomes in neonates. During HI brain injury, dysregulated factor production contributes to microvascular impairment. Glycolysis-derived lactate accumulated during ischemia has been proposed to protect against ischemic injury, but its mechanism of action is poorly understood. Herein, we hypothesize that lactate via its G-protein coupled receptor (GPR81) controls postnatal brain angiogenesis and plays a protective role after HI injury. We show that GPR81 is predominantly expressed in neurons of the cerebral cortex and hippocampus. GPR81-null mice displayed a delay in cerebral microvascular development linked to reduced levels of various major angiogenic factors and augmented expression of anti-angiogenic Thrombospondin-1 (TSP-1) in comparison to their WT littermates. Coherently, lactate stimulation induced an increase in growth factors (VEGF, Ang1 and 2, PDGF) and reduced TSP-1 expression in neurons, which contributed to accelerating angiogenesis. HI injury in GPR81-null animals curtailed vascular density and consequently increased infarct size compared to changes seen in WT mice; conversely intracerebroventricular lactate injection increased vascular density and diminished infarct size in WT but not in GPR81-null mice. Collectively, we show that lactate acting via GPR81 participates in developmental brain angiogenesis, and attenuates HI injury by restoring compromised microvasculature.


Subject(s)
Brain Injuries , Hypoxia-Ischemia, Brain , Neovascularization, Physiologic , Receptors, G-Protein-Coupled , Animals , Animals, Newborn , Brain/metabolism , Brain Injuries/metabolism , Female , Hypoxia-Ischemia, Brain/metabolism , Infarction , Ischemia/metabolism , Lactic Acid/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Pregnancy , Receptors, G-Protein-Coupled/genetics , Thrombospondin 1/metabolism
2.
J Cell Mol Med ; 24(10): 5718-5730, 2020 05.
Article in English | MEDLINE | ID: mdl-32279433

ABSTRACT

δ-opioid receptor (DOPr) agonists have analgesic efficacy in chronic pain models but development of tolerance limits their use for long-term pain management. Although agonist potential for inducing acute analgesic tolerance has been associated with distinct patterns of DOPr internalization, the association between trafficking and chronic tolerance remains ill-defined. In a rat model of streptozotocin (STZ)-induced diabetic neuropathy, deltorphin II and TIPP produced sustained analgesia  following daily (intrathecal) i.t. injections over six days, whereas similar treatment with SNC-80 or SB235863 led to progressive tolerance and loss of the analgesic response. Trafficking assays in murine neuron cultures showed no association between the magnitude of ligand-induced sequestration and development of chronic tolerance. Instead, ligands that supported DOPr recycling were also the ones producing sustained analgesia over 6-day treatment. Moreover, endosomal endothelin-converting enzyme 2 (ECE2) blocker 663444 prevented DOPr recycling by deltorphin II and TIPP and precipitated tolerance by these ligands. In conclusion, agonists, which support DOPr recycling, avoid development of analgesic tolerance over repeated administration.


Subject(s)
Analgesics/pharmacology , Drug Tolerance , Receptors, Opioid, delta/metabolism , Animals , Cells, Cultured , Chronic Pain/diagnosis , Chronic Pain/drug therapy , Chronic Pain/etiology , Chronic Pain/metabolism , Diabetic Nephropathies/complications , Disease Models, Animal , Ligands , Male , Neurons/drug effects , Neurons/metabolism , Pain Measurement , Protein Binding , Rats
3.
Cell Mol Life Sci ; 75(12): 2257-2271, 2018 06.
Article in English | MEDLINE | ID: mdl-29288293

ABSTRACT

Soon after internalization delta opioid receptors (DOPrs) are committed to the degradation path by G protein-coupled receptor (GPCR)-associated binding protein. Here we provide evidence that this classical post-endocytic itinerary may be rectified by downstream sorting decisions which allow DOPrs to regain to the membrane after having reached late endosomes (LE). The LE sorting mechanism involved ESCRT accessory protein Alix and the TIP47/Rab9 retrieval complex which supported translocation of the receptor to the TGN, from where it subsequently regained the cell membrane. Preventing DOPrs from completing this itinerary precipitated acute analgesic tolerance to the agonist DPDPE, supporting the relevance of this recycling path in maintaining the analgesic response by this receptor. Taken together, these findings reveal a post-endocytic itinerary where GPCRs that have been sorted for degradation can still recycle to the membrane.


Subject(s)
Cell Membrane/metabolism , Receptors, Opioid, delta/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Neurons/metabolism , Protein Transport , Proteolysis , Rats , Rats, Sprague-Dawley
4.
Cell Mol Life Sci ; 72(18): 3543-57, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25900661

ABSTRACT

Kir3 channels control excitability in the nervous system and the heart. Their surface expression is strictly regulated, but mechanisms responsible for channel removal from the membrane remain incompletely understood. Using transfected cells, we show that Kir3.1/3.2 channels and delta opioid receptors (DORs) associate in a complex which persists during receptor activation, behaving as a scaffold that allows beta-arrestin (ßarr) to interact with both signaling partners. This organization favored co-internalization of DORs and Kir3 channels in a ßarr-dependent manner via a clathrin/dynamin-mediated endocytic path. Taken together, these findings identify a new way of modulating Kir3 channel availability at the membrane and assign a putatively novel role for ßarrs in regulating canonical effectors for G protein-coupled receptors.


Subject(s)
Arrestins/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Receptors, Opioid, delta/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Clathrin/metabolism , Dynamins/metabolism , Endocytosis/physiology , HEK293 Cells , Humans , Rats , Signal Transduction/physiology , beta-Arrestins
5.
Cell Mol Life Sci ; 71(8): 1529-46, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24022593

ABSTRACT

Signaling bias refers to G protein-coupled receptor ligand ability to preferentially activate one type of signal over another. Bias to evoke signaling as opposed to sequestration has been proposed as a predictor of opioid ligand potential for generating tolerance. Here we measured whether delta opioid receptor agonists preferentially inhibited cyclase activity over internalization in HEK cells. Efficacy (τ) and affinity (KA) values were estimated from functional data and bias was calculated from efficiency coefficients (log τ/KA). This approach better represented the data as compared to alternative methods that estimate bias exclusively from τ values. Log (τ/KA) coefficients indicated that SNC-80 and UFP-512 promoted cyclase inhibition more efficiently than DOR internalization as compared to DPDPE (bias factor for SNC-80: 50 and for UFP-512: 132). Molecular determinants of internalization were different in HEK293 cells and neurons with ßarrs contributing to internalization in both cell types, while PKC and GRK2 activities were only involved in neurons. Rank orders of ligand ability to engage different internalization mechanisms in neurons were compared to rank order of E max values for cyclase assays in HEK cells. Comparison revealed a significant reversal in rank order for cyclase E max values and ßarr-dependent internalization in neurons, indicating that these responses were ligand-specific. Despite this evidence, and because kinases involved in internalization were not the same across cellular backgrounds, it is not possible to assert if the magnitude and nature of bias revealed by rank orders of maximal responses is the same as the one measured in HEK cells.


Subject(s)
Chronic Pain/drug therapy , Cyclic AMP/metabolism , Drug Tolerance/physiology , Neurons/metabolism , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Analysis of Variance , Benzamides/pharmacology , Benzimidazoles/pharmacology , Blotting, Western , Dose-Response Relationship, Drug , Enkephalin, D-Penicillamine (2,5)- , Fluorescent Antibody Technique , G-Protein-Coupled Receptor Kinase 2/metabolism , Green Fluorescent Proteins , HEK293 Cells , Humans , Ligands , Oligopeptides/pharmacology , Piperazines/pharmacology
6.
J Neurosci ; 32(14): 4827-40, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22492038

ABSTRACT

Opioid tendency to generate analgesic tolerance has been previously linked to biased internalization. Here, we assessed an alternative possibility; whether tolerance of delta opioid receptor agonists (DORs) could be related to agonist-specific recycling. A first series of experiments revealed that DOR internalization by DPDPE and SNC-80 was similar, but only DPDPE induced recycling. We then established that the non-recycling agonist SNC-80 generated acute analgesic tolerance that was absent in mice treated with DPDPE. Furthermore, both agonists stabilized different conformations, whose distinct interaction with Gßγ subunits led to different modalities of ß-arrestin2 (ßarr2) recruitment. In particular, bioluminescence resonance energy transfer (BRET) assays revealed that sustained activation by SNC-80 drew the receptor C terminus in close proximity of the N-terminal domain of Gγ2, causing ßarr2 to interact with receptors and Gßγ subunits. DPDPE moved the receptor C-tail away from the Gßγ dimer, resulting in ßarr2 recruitment to the receptor but not in the vicinity of Gγ2. These differences were associated with stable DOR-ßarr2 association, poor recycling, and marked desensitization following exposure to SNC-80, while DPDPE promoted transient receptor interaction with ßarr2 and effective recycling, which conferred protection from desensitization. Together, these data indicate that DORs may adopt ligand-specific conformations whose distinct recycling properties determine the extent of desensitization and are predictive of analgesic tolerance. Based on these findings, we propose that the development of functionally selective DOR ligands that favor recycling could constitute a valid strategy for the production of longer acting opioid analgesics.


Subject(s)
Analgesics, Opioid/metabolism , Arrestins/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Analgesics, Opioid/pharmacology , Animals , Animals, Newborn , Arrestins/physiology , Cell Line, Transformed , Cells, Cultured , Drug Tolerance/physiology , GTP-Binding Protein beta Subunits/physiology , GTP-Binding Protein gamma Subunits/physiology , HEK293 Cells , Humans , Male , Mice , Protein Binding/drug effects , Protein Binding/physiology , Protein Transport/drug effects , Protein Transport/physiology , Rats , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL