Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1870(5): 119476, 2023 06.
Article in English | MEDLINE | ID: mdl-37059189

ABSTRACT

Endosomal trafficking is intricately linked to G protein-coupled receptors (GPCR) fate and signaling. Extracellular uridine diphosphate (UDP) acts as a signaling molecule by selectively activating the GPCR P2Y6. Despite the recent interest for this receptor in pathologies, such as gastrointestinal and neurological diseases, there is sparse information on the endosomal trafficking of P2Y6 receptors in response to its endogenous agonist UDP and synthetic selective agonist 5-iodo-UDP (MRS2693). Confocal microscopy and cell surface ELISA revealed delayed internalization kinetics in response to MRS2693 vs. UDP stimulation in AD293 and HCT116 cells expressing human P2Y6. Interestingly, UDP induced clathrin-dependent P2Y6 internalization, whereas receptor stimulation by MRS2693 endocytosis appeared to be associated with a caveolin-dependent mechanism. Internalized P2Y6 was associated with Rab4, 5, and 7 positive vesicles independent of the agonist. We have measured a higher frequency of receptor expression co-occurrence with Rab11-vesicles, the trans-Golgi network, and lysosomes in response to MRS2693. Interestingly, a higher agonist concentration reversed the delayed P2Y6 internalization and recycling kinetics in the presence of MRS2693 stimulation without changing its caveolin-dependent internalization. This work showed a ligand-dependent effect affecting the P2Y6 receptor internalization and endosomal trafficking. These findings could guide the development of bias ligands that could influence P2Y6 signaling.


Subject(s)
Receptors, G-Protein-Coupled , Uridine Diphosphate , Humans , Ligands , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Uridine Diphosphate/metabolism , GTP-Binding Proteins/metabolism
2.
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
3.
J Cell Physiol ; 235(12): 9676-9690, 2020 12.
Article in English | MEDLINE | ID: mdl-32420639

ABSTRACT

Cell migration is a ubiquitous process necessary to maintain and restore tissue functions. However, in cancer, cell migration leads to metastasis development and thus worsens the prognosis. Although the mechanism of cell migration is well understood, the identification of new targets modulating cell migration and deciphering their signaling events could lead to new therapies to restore tissue functions in diseases, such as inflammatory bowel disease, or to block metastatic development in different forms of cancer. Previous research has identified the G-protein-coupled P2Y6 receptor as an innovative target that could dictate cell migration under normal and pathological conditions. Surprisingly, there is little information on the cellular events triggered by activated P2Y6 during cell migration. Here, we demonstrated that P2Y6 activation stimulated A549 human lung cancer cells and Caco-2 colorectal cancer cell migration. Activated P2Y6 increased the number of filopodia and focal adhesions; two migratory structures required for cell migration. The generation of these structures involved Gαq /calcium/protein kinases C (PKC) and Gα13 /RHO-associated protein kinase-dependent pathways that dictate the formation of the migratory structures. These pathways led to the stabilization of the actin cytoskeleton through a PKC-dependent phosphorylation of cofilin. These results support the idea that the P2Y6 receptor represents a target of interest to modulate cell migration and revealed an intricate dialogue between two Gα-protein signaling pathways.


Subject(s)
Cell Movement/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Protein Kinase C-alpha/genetics , Receptors, Purinergic P2/genetics , A549 Cells , Actins/genetics , Caco-2 Cells , Calcium/metabolism , Cell Surface Extensions/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial Cells/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Gene Expression Regulation, Neoplastic , Humans , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , rho-Associated Kinases/genetics
4.
J Pharmacol Exp Ther ; 374(1): 52-61, 2020 07.
Article in English | MEDLINE | ID: mdl-32327529

ABSTRACT

The analgesic potency of morphine-6-glucuronide (M6G) has been shown to be 50-fold higher than morphine after intracerebral injection. However, the brain penetration of M6G is significantly lower than morphine, thus limiting its usefulness in pain management. Here, we created new entities by the conjugation of the angiopep-2 peptide (An2) that crosses the blood-brain barrier (BBB) by low-density lipoprotein receptor-related protein 1 receptor-mediated transcytosis with either morphine or M6G. We demonstrated improvement of BBB permeability of these new entities compared with that of unconjugated M6G and morphine. Intravenous or subcutaneous administration of the An2-M6G conjugate exerted greater and more sustained analgesic activity than equivalent doses of either morphine or M6G. Likewise, subcutaneous An2-morphine induced a delayed but prolonged antinociceptive effect. The effects of these conjugates on the gastrointestinal tract motility were also evaluated. An2-morphine significantly reduced the intestinal transit time, whereas An2-M6G exhibited a reduced constipation profile, as compared with an equimolar dose of morphine. In summary, we have developed new brain-penetrant opioid conjugates exhibiting improved analgesia to side effect ratios. These results thus support the use of An2-carrier peptides as an innovative BBB-targeting technology to deliver effective drugs, such as M6G, for pain management. SIGNIFICANCE STATEMENT: The metabolite morphine-6-glucuronide (M6G) does not efficiently cross the blood-brain barrier. The low-density lipoprotein receptor-related protein 1 peptide ligand angiopep-2 may serve as an effective drug delivery system to the brain. Here, we demonstrated that the coupling of M6G to angiopep-2 peptide (An2) improves its brain penetration and significantly increases its analgesic potency. The An2-M6G conjugate has a favorable side effect profile that includes reduction of developing constipation. An2-M6G exhibits a unique pharmacodynamic profile with a better therapeutic window than morphine.


Subject(s)
Analgesics, Opioid/chemistry , Analgesics, Opioid/metabolism , Blood-Brain Barrier/metabolism , Morphine Derivatives/chemistry , Morphine Derivatives/metabolism , Peptides/chemistry , Administration, Intravenous , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Biological Transport , Gastrointestinal Motility/drug effects , Male , Mice , Morphine Derivatives/administration & dosage , Morphine Derivatives/pharmacology , Nociception/drug effects
5.
ACS Chem Neurosci ; 10(11): 4535-4544, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31589400

ABSTRACT

Neurotensin (NT) exerts its analgesic effects through activation of the G protein-coupled receptors NTS1 and NTS2. This opioid-independent antinociception represents a potential alternative for pain management. While activation of NTS1 also induces a drop in blood pressure and body temperature, NTS2 appears to be an analgesic target free of these adverse effects. Here, we report modifications of NT at Tyr11 to increase selectivity toward NTS2, complemented by modifications at the N-terminus to impair proteolytic degradation of the biologically active NT(8-13) sequence. Replacement of Tyr11 by either 6-OH-Tic or 7-OH-Tic resulted in a significant loss of binding affinity to NTS1 and subsequent NTS2 selectivity. Incorporation of the unnatural amino acid ß3hLys at position 8 increased the half-life to over 24 h in plasma. Simultaneous integration of both ß3hLys8 and 6-OH-Tic11 into NT(8-13) produced a potent and NTS2-selective analogue with strong analgesic action after intrathecal delivery in the rat formalin-induced pain model with an ED50 of 1.4 nmol. Additionally, intravenous administration of this NT analogue did not produce persistent hypotension or hypothermia. These results demonstrate that NT analogues harboring unnatural amino acids at positions 8 and 11 can enhance crucial pharmacokinetic and pharmacodynamic features for NT(8-13) analogues, i.e., proteolytic stability, NTS2 selectivity, and improved analgesic/adverse effect ratio.


Subject(s)
Analgesia/methods , Hypotension/metabolism , Hypothermia/metabolism , Neurotensin/analogs & derivatives , Receptors, Neurotensin/metabolism , Tyrosine/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Hypotension/chemically induced , Hypothermia/chemically induced , Male , Neurotensin/toxicity , Pain Measurement/drug effects , Pain Measurement/methods , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Receptors, Neurotensin/agonists , Tyrosine/genetics
6.
Eur J Pharmacol ; 848: 80-87, 2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30707956

ABSTRACT

Opioid and neurotensin (NT) receptors are expressed in both central and peripheral nervous systems where they modulate nociceptive responses. Nowadays, opioid analgesics like morphine remain the most prescribed drugs for the treatment of moderate to severe pain. However, despite their daily used, opioids can produce life-threatening side effects, such as constipation or respiratory depression. Besides, NT analogs exert strong opioid-independent analgesia. Here, we thus hypothesized that the combined use of opioid and NT agonists would require lower doses to produce significant analgesic effects, hence decreasing opioid-induced adverse effects. We used isobologram analyses to determine if the combination of a NT brain-penetrant analog, An2-NT(8-13) with morphine results in an inhibitory, synergistic or additive analgesic response. We found that intravenous administration of An2-NT(8-13) reduced by 90% the nocifensive behaviors induced by formalin injection, at the dose of 0.018 mg/kg. Likewise, subcutaneous morphine reduced pain by 90% at 1.8 mg/kg. Importantly, isobologram analyses revealed that the co-injection of An2-NT(8-13) with morphine induced an additive analgesic response. We finally assessed the effects of morphine and An2-NT(8-13) on the gastrointestinal tract motility using the charcoal meal test. As opposed to morphine which significantly reduced the intestinal motility at the analgesic effective dose of 1.8 mg/kg, An2-NT(8-13) did not affect the charcoal meal intestinal transit at 0.018 mg/kg. Interestingly, at the dose providing 90% pain relief, the co-administration of morphine with An2-NT(8-13) had a reduced effect on constipation. Altogether, these results suggest that combining NT agonists with morphine may improve its analgesic/adverse effect ratio.


Subject(s)
Analgesics, Opioid/administration & dosage , Neurotensin/administration & dosage , Pain Measurement/drug effects , Peptide Fragments/administration & dosage , Peptides/administration & dosage , Receptors, Neurotensin/agonists , Receptors, Opioid, mu/agonists , Animals , Dose-Response Relationship, Drug , Drug Therapy, Combination , Male , Morphine/administration & dosage , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Receptors, Neurotensin/metabolism , Receptors, Opioid, mu/metabolism
8.
Bioorg Med Chem Lett ; 28(13): 2320-2323, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29853330

ABSTRACT

In this study, affinities and activities of derivatized analogues of Dmt-dermorphin[1-4] (i.e. Dmt-d-Ala-Phe-GlyNH2, Dmt = 2',6'-dimethyl-(S)-tyrosine) for the µ opioid receptor (MOP) and δ opioid receptor (DOP) were evaluated using radioligand binding studies, functional cell-based assays and isolated organ bath experiments. By means of solid-phase or solution-phase Suzuki-Miyaura cross-couplings, various substituted regioisomers of the phenylalanine moiety in position 3 of the sequence were prepared. An 18-membered library of opioid tetrapeptides was generated via screening of the chemical space around the Phe3 side chain. These substitutions modulated bioactivity, receptor subtype selectivity and highly effective ligands with subnanomolar binding affinities, contributed to higher functional activities and potent analgesic actions. In search of selective peptidic ligands, we show here that the Suzuki-Miyaura reaction is a versatile and robust tool which could also be deployed elsewhere.


Subject(s)
Analgesics, Opioid/therapeutic use , Oligopeptides/therapeutic use , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Guinea Pigs , HEK293 Cells , Humans , Ligands , Male , Mice , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/pharmacology , Rats, Sprague-Dawley
9.
ACS Med Chem Lett ; 8(11): 1177-1182, 2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29152051

ABSTRACT

Herein, the synthesis of novel conformationally constrained amino acids, 4-amino-8-bromo-2-benzazepin-3-one (8-Br-Aba), 3-amino-3,4-dihydroquinolin-2-one, and regioisomeric 4-amino-naphthoazepinones (1- and 2-Ana), is described. Introduction of these constricted scaffolds into the N-terminal tetrapeptide of dermorphin (i.e., H-Tyr-d-Ala-Phe-Gly-NH2) induced significant shifts in binding affinity, selectivity, and in vitro activity at the µ- and δ-opioid receptors (MOP and DOP, respectively). A reported constrained µ-/δ-opioid lead tetrapeptide H-Dmt-d-Arg-Aba-Gly-NH2 was modified through application of various constrained building blocks to identify optimal spatial orientations in view of activity at the opioid receptors. Interestingly, when the aromatic moieties were turned toward the C-terminus of the peptide sequences, (partial) (ant)agonism at MOP and weak (ant)agonism at DOP were noticed, whereas the incorporation of the 1-Ana residue led toward balanced low nanomolar MOP/DOP binding and in vitro agonism.

10.
Hum Mol Genet ; 24(12): 3440-56, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25765661

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that primarily affects motoneurons in the brain and spinal cord. Astrocyte and microglia activation as well as skeletal muscle atrophy are also typical hallmarks of the disease. However, the functional relationship between astrocytes, microglia and skeletal muscle in the pathogenic process remains unclear. Here, we report that the tumor necrosis factor-like weak inducer of apoptosis (Tweak) and its receptor Fn14 are aberrantly expressed in spinal astrocytes and skeletal muscle of SOD1(G93A) mice. We show that Tweak induces motoneuron death, stimulates astrocytic interleukin-6 release and astrocytic proliferation in vitro. The genetic ablation of Tweak in SOD1(G93A) mice significantly reduces astrocytosis, microgliosis and ameliorates skeletal muscle atrophy. The peripheral neutralization of Tweak through antagonistic anti-Tweak antibody ameliorates muscle pathology and notably, decreases microglial activation in SOD1(G93A) mice. Unexpectedly, none of these approaches improved motor function, lifespan and motoneuron survival. Our work emphasizes the multi-systemic aspect of ALS, and suggests that a combinatorial therapy targeting multiple cell types will be instrumental to halt the neurodegenerative process.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Gliosis/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Tumor Necrosis Factors/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Cell Death , Cell Proliferation , Cytokine TWEAK , Disease Models, Animal , Gene Deletion , Gene Expression Regulation , Interleukin-6/biosynthesis , Life Expectancy , Mice , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Motor Endplate/genetics , Motor Endplate/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , TWEAK Receptor , Tumor Necrosis Factors/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...