Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters










Publication year range
1.
J Neurochem ; 168(4): 428-440, 2024 Apr.
Article in English | MEDLINE | ID: mdl-36912731

ABSTRACT

People bitten by Alpine vipers are usually treated with antivenom antisera to prevent the noxious consequences caused by the injected venom. However, this treatment suffers from a number of drawbacks and additional therapies are necessary. The venoms of Vipera ammodytes and of Vipera aspis are neurotoxic and cause muscle paralysis by inducing neurodegeneration of motor axon terminals because they contain a presynaptic acting sPLA2 neurotoxin. We have recently found that any type of damage to motor axons is followed by the expression and activation of the intercellular signaling axis consisting of the CXCR4 receptor present on the membrane of the axon stump and of its ligand, the chemokine CXCL12 released by activated terminal Schwann cells. We show here that also V. ammodytes and V. aspis venoms cause the expression of the CXCL12-CXCR4 axis. We also show that a small molecule agonist of CXCR4, dubbed NUCC-390, induces a rapid regeneration of the motor axon terminal with functional recovery of the neuromuscular junction. These findings qualify NUCC-390 as a promising novel therapeutics capable of improving the recovery from the paralysis caused by the snakebite of the two neurotoxic Alpine vipers.


Subject(s)
Indazoles , Receptors, CXCR4 , Viper Venoms , Viperidae , Animals , Paralysis/chemically induced , Receptors, CXCR4/agonists , Viper Venoms/antagonists & inhibitors , Viper Venoms/toxicity , Vipera/metabolism , Viperidae/metabolism , Mice , Indazoles/pharmacology , Indazoles/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Snake Bites/drug therapy
2.
Biochem Pharmacol ; 216: 115764, 2023 10.
Article in English | MEDLINE | ID: mdl-37634595

ABSTRACT

Development of specific therapies that target and accelerate diabetic wound repair is an urgent need to alleviate pain and suffering and the huge socioeconomic burden of this debilitating disease. C-X-C Motif Chemokine Ligand 12 (CXCL12) also know an stromal cell-derived factor 1α (SDF-1α) is a chemokine that binds the CXC chemokine receptor type 4 (CXCR4) and activates downstream signaling resulting in recruitment of hematopoietic cells to locations of tissue injury and promotes tissue repair. In diabetes, low expression of CXCL12 correlates with impaired wound healing. Activation of CXCR4 receptor signaling with agonists or positive allosteric modulators (PAMs) provides a potential for small molecule therapeutic discovery and development. We recently reported high throughput screening and identification of the CXCR4 partial agonist UCUF-728, characterization of in vitro activity and reduced wound closure time in diabetic mice at 100 µM as a proof-of-concept study. We report here, the discovery of a second chemical scaffold demonstrating increased agonist potency and represented by thiadiazine derivative, UCUF-965. UCUF-965 is a potent partial agonist of ß-arrestin recruitment in CXCR4 receptor overexpressing cell line. Furthermore, UCUF-965 potentiates the CXCL12 maximal response in cAMP signaling pathway, activates CXCL12 stimulated migration in lymphoblast cells and modulates the levels of specific microRNA involved in the complex wound repair process, specifically in mouse fibroblasts. Our results indicate that UCUF-965 acts as a PAM agonist of the CXCR4 receptor. Furthermore, UCUF-965 enhanced angiogenesis markers and reduced wound healing time by 36% at 10.0 µM in diabetic mice models compared to untreated control.


Subject(s)
Diabetes Mellitus, Experimental , Receptors, CXCR4 , Wound Healing , Animals , Mice , Cell Movement/physiology , Chemokine CXCL12/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Hematopoietic Stem Cells , Receptors, CXCR4/agonists , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction , Wound Healing/drug effects , Wound Healing/genetics , Wound Healing/physiology
3.
Int J Mol Sci ; 23(4)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35216311

ABSTRACT

Diabetes produces a chronic inflammatory state that contributes to the development of vascular disease and impaired wound healing. Despite the known individual and societal impacts of diabetic ulcers, there are limited therapies effective at improving healing. Stromal cell-derived factor 1α (SDF-1α) is a CXC chemokine that functions via activation of the CXC chemokine receptor type 4 (CXCR4) receptor to recruit hematopoietic cells to locations of tissue injury and promote tissue repair. The expression of SDF-1α is reduced in diabetic wounds, suggesting a potential contribution to wound healing impairment and presenting the CXCR4 receptor as a target for therapeutic investigations. We developed a high-throughput ß-arrestin recruitment assay and conducted structure-activity relationship (SAR) studies to screen compounds for utility as CXCR4 agonists. We identified CXCR4 agonist UCUF-728 from our studies and further validated its activity in vitro in diabetic fibroblasts. UCUF-728 reduced overexpression of miRNA-15b and miRNA-29a, negative regulators of angiogenesis and type I collagen production, respectively, in diabetic fibroblasts. In vivo, UCUF-728 reduced the wound closure time by 36% and increased the evidence of angiogenesis in diabetic mice. Together, this work demonstrates the clinical potential of small molecule CXCR4 agonists as novel therapies for pathologic wound healing in diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Receptors, CXCR4 , Wound Healing , Animals , Chemokine CXCL12/metabolism , Diabetes Mellitus, Experimental/drug therapy , Mice , MicroRNAs , Neovascularization, Physiologic , Receptors, CXCR4/agonists , Receptors, CXCR4/metabolism
5.
PLoS Negl Trop Dis ; 14(9): e0008547, 2020 09.
Article in English | MEDLINE | ID: mdl-32898186

ABSTRACT

Envenomation by snakes is a major neglected human disease. Hospitalization and use of animal-derived antivenom are the primary therapeutic supports currently available. There is consensus that additional, not expensive, treatments that can be delivered even long after the snake bite are needed. We recently showed that the drug dubbed NUCC-390 shortens the time of recovery from the neuroparalysis caused by traumatic or toxic degeneration of peripheral motor neurons. These syndromes are characterized by the activation of a pro-regenerative molecular axis, consisting of the CXCR4 receptor expressed at the damaged site in neuronal axons and by the release of its ligand CXCL12α, produced by surrounding Schwann cells. This intercellular signaling axis promotes axonal growth and functional recovery from paralysis. NUCC-390 is an agonist of CXCR4 acting similarly to CXCL12α. Here, we have tested its efficacy in a murine model of neuroparalytic envenoming by a Papuan Taipan (Oxyuranus scutellatus) where a degeneration of the motor axon terminals caused by the presynaptic PLA2 toxin Taipoxin, contained in the venom, occurs. Using imaging of the neuromuscular junction and electrophysiological analysis, we found that NUCC-390 administration after injection of either the purified neuroparalytic Taipoxin or the whole Taipan venom, significantly accelerates the recovery from paralysis. These results indicate that NUCC-390, which is non-toxic in mice, should be considered for trials in humans to test its efficacy in accelerating the recovery from the peripheral neuroparalysis induced by Taipans. NUCC-390 should be tested as well in the envenomation by other snakes that cause neuroparalytic syndromes in humans. NUCC-390 could become an additional treatment, common to many snake envenomings, that can be delivered after the bite to reduce death by respiratory deficits and to shorten and improve functional recovery.


Subject(s)
Elapid Venoms/toxicity , Indazoles/pharmacology , Neuromuscular Junction/drug effects , Paralysis/therapy , Piperidines/pharmacology , Pyridines/pharmacology , Receptors, CXCR4/agonists , Action Potentials/drug effects , Animals , Mice , Mice, Inbred C57BL , Motor Neurons/drug effects
6.
Sci Rep ; 10(1): 11359, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32647374

ABSTRACT

We compared therapeutic properties of natural and engineered chemokine (C-X-C motif) receptor 4 (CXCR4) agonists in a rat acute respiratory distress syndrome (ARDS) model utilizing the PaO2/FiO2-ratio as a clinically relevant primary outcome criterion. Ventilated rats underwent unilateral lung ischemia from t = 0-70 min plus hemorrhage to a mean arterial blood pressure (MAP) of 30 mmHg from t = 40-70 min, followed by reperfusion/fluid resuscitation until t = 300 min. Natural CXCR4 agonists (CXCL12, ubiquitin) and engineered CXCL12 variants (CXCL121, CXCL22, CXCL12K27A/R41A/R47A, CXCL12 (3-68)) were administered within 5 min of fluid resuscitation. Animals treated with vehicle or CXCL12 (3-68) reached criteria for mild and moderate ARDS between t = 90-120 min and t = 120-180 min, respectively, and remained in moderate ARDS until t = 300 min. Ubiquitin, CXCL12, CXCL121 and CXCL122 prevented ARDS development. Potencies of CXCL12/CXCL121/CXCL122 were higher than the potency of ubiquitin. CXCL12K27A/R41A/R47A was inefficacious. CXCL121 > CXCL12 stabilized MAP and reduced fluid requirements. CXCR4 agonists at doses that preserved lung function reduced histological injury of the post-ischemic lung and reduced mortality from 55 to 9%. Our findings suggest that CXCR4 protein agonists prevent development of ARDS and reduce mortality in a rat model, and that development of new engineered protein therapeutics with improved pharmacological properties for ARDS is possible.


Subject(s)
Receptors, CXCR4/agonists , Reperfusion Injury/prevention & control , Respiratory Distress Syndrome/prevention & control , Resuscitation/methods , Shock, Hemorrhagic/therapy , Wounds and Injuries/therapy , Animals , Chemokine CXCL12/administration & dosage , Chemokine CXCL12/genetics , Disease Models, Animal , Fluid Therapy/methods , Humans , Lung/blood supply , Lung/pathology , Male , Protein Engineering , Rats , Reperfusion Injury/etiology , Reperfusion Injury/mortality , Reperfusion Injury/pathology , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/mortality , Respiratory Distress Syndrome/pathology , Shock, Hemorrhagic/etiology , Shock, Hemorrhagic/mortality , Shock, Hemorrhagic/pathology , Thoracotomy/adverse effects , Ubiquitin/administration & dosage , Wounds and Injuries/complications , Wounds and Injuries/mortality
7.
Chemistry ; 26(44): 10113-10125, 2020 Aug 06.
Article in English | MEDLINE | ID: mdl-32603023

ABSTRACT

Here we investigated the structural and biological effects ensuing from the disulfide bond replacement of a potent and selective C-X-C chemokine receptor type 4 (CXCR4) peptide antagonist, with 1,4- and 1,5- disubstituted 1,2,3-triazole moieties. Both strategies produced candidates that showed high affinity and selectivity against CXCR4. Notably, when assessed for their ability to modulate the CXCL12-mediated cell migration, the 1,4-triazole variant conserved the antagonistic effect in the low-mid nanomolar range, while the 1,5-triazole one displayed the ability to activate the migration, becoming the first in class low-molecular-weight CXCR4 peptide agonist. By combining NMR and computational studies, we provided a valuable model that highlighted differences in the interactions of the two peptidomimetics with the receptor that could account for their different functional profile. Finally, we envisage that our findings could be translated to different GPCR-interacting peptides for the pursuit of novel chemical probes that could assist in dissecting the complex puzzle of this fundamental class of transmembrane receptors.


Subject(s)
Disulfides/chemistry , Peptides/chemistry , Peptides/pharmacology , Receptors, CXCR4/chemistry , Triazoles/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , Humans , Ligands , Peptidomimetics , Receptors, CXCR4/agonists
8.
Chembiochem ; 21(4): 500-507, 2020 02 17.
Article in English | MEDLINE | ID: mdl-31418992

ABSTRACT

Deep convolutional neural networks (CNNs) are a method of choice for image recognition. Herein a hybrid CNN approach is presented for molecular pattern recognition in drug discovery. Using self-organizing map images of molecular pharmacophores as input, CNN models were trained to identify chemokine receptor CXCR4 modulators with high accuracy. This machine learning classifier identified first-in-class synthetic CXCR4 full agonists. The receptor-activating effects were confirmed by intracellular cAMP response and in a phenotypic spheroid invasion assay of medulloblastoma cell invasion. Additional macromolecular targets of the small molecules were predicted in silico and tested in vitro, revealing modulatory effects on dopamine receptors and CCR1. These results positively advocate the applicability of molecular image recognition by CNNs to ligand-based virtual compound screening, and demonstrate the complementarity of machine intelligence and human expert knowledge.


Subject(s)
Cell Movement , Deep Learning , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors , Cell Line, Tumor , Drug Design , Humans
9.
Ann Clin Transl Neurol ; 6(12): 2395-2402, 2019 12.
Article in English | MEDLINE | ID: mdl-31725979

ABSTRACT

OBJECTIVE: To test whether the signaling axis CXCL12α-CXCR4 is activated upon crush/cut of the sciatic nerve and to test the activity of NUCC-390, a new CXCR4 agonist, in promoting nerve recovery from damage. METHODS: The sciatic nerve was either crushed or cut. Expression and localization of CXCL12α and CXCR4 were evaluated by imaging with specific antibodies. Their functional involvement in nerve regeneration was determined by antibody-neutralization of CXCL12α, and by the CXCR4 specific antagonist AMD3100, using as quantitative read-out the compound muscle action potential (CMAP). NUCC-390 activity on nerve regeneration was determined by imaging and CMAP recordings. RESULTS: CXCR4 is expressed at the injury site within the axonal compartment, whilst its ligand CXCL12α is expressed in Schwann cells. The CXCL12α-CXCR4 axis is involved in the recovery of neurotransmission of the injured nerve. More importantly, the small molecule NUCC-390 is a strong promoter of the functional and anatomical recovery of the nerve, by acting very similarly to CXCL12α. This pharmacological action is due to the capability of NUCC-390 to foster elongation of motor neuron axons both in vitro and in vivo. INTERPRETATION: Imaging and electrophysiological data provide novel and compelling evidence that the CXCL12α-CXCR4 axis is involved in sciatic nerve repair after crush/cut. This makes NUCC-390 a strong candidate molecule to stimulate nerve repair by promoting axonal elongation. We propose this molecule to be tested in other models of neuronal damage, to lay the basis for clinical trials on the efficacy of NUCC-390 in peripheral nerve repair in humans.


Subject(s)
Axons/drug effects , Chemokine CXCL12/metabolism , Indazoles/pharmacology , Nerve Regeneration/drug effects , Piperidines/pharmacology , Pyridines/pharmacology , Receptors, CXCR4/agonists , Receptors, CXCR4/metabolism , Schwann Cells/metabolism , Sciatic Nerve/injuries , Sciatic Neuropathy/drug therapy , Animals , Benzylamines , Cyclams , Disease Models, Animal , Heterocyclic Compounds/pharmacology , Mice , Mice, Inbred C57BL , Receptors, CXCR4/antagonists & inhibitors
10.
Cells ; 8(10)2019 09 30.
Article in English | MEDLINE | ID: mdl-31575088

ABSTRACT

The activation of the G-protein coupled receptor CXCR4 by its ligand CXCL12α is involved in a large variety of physiological and pathological processes, including the growth of B cells precursors and of motor axons, autoimmune diseases, stem cell migration, inflammation, and several neurodegenerative conditions. Recently, we demonstrated that CXCL12α potently stimulates the functional recovery of damaged neuromuscular junctions via interaction with CXCR4. This result prompted us to test the neuroregeneration activity of small molecules acting as CXCR4 agonists, endowed with better pharmacokinetics with respect to the natural ligand. We focused on NUCC-390, recently shown to activate CXCR4 in a cellular system. We designed a novel and convenient chemical synthesis of NUCC-390, which is reported here. NUCC-390 was tested for its capability to induce the regeneration of motor axon terminals completely degenerated by the presynaptic neurotoxin α-Latrotoxin. NUCC-390 was found to strongly promote the functional recovery of the neuromuscular junction, as assayed by electrophysiology and imaging. This action is CXCR4 dependent, as it is completely prevented by AMD3100, a well-characterized CXCR4 antagonist. These data make NUCC-390 a strong candidate to be tested in human therapy to promote nerve recovery of function after different forms of neurodegeneration.


Subject(s)
Indazoles/pharmacology , Motor Neurons/drug effects , Nerve Degeneration/drug therapy , Nerve Regeneration/drug effects , Piperidines/pharmacology , Presynaptic Terminals/drug effects , Pyridines/pharmacology , Receptors, CXCR4/agonists , Animals , Benzylamines , Cyclams , Heterocyclic Compounds/pharmacology , Mice , Mice, Inbred Strains , Motor Neurons/pathology , Motor Neurons/physiology , Nerve Degeneration/chemically induced , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiology , Presynaptic Terminals/pathology , Presynaptic Terminals/physiology , Primary Cell Culture , Rats , Spider Venoms/toxicity
11.
Mol Pharmacol ; 96(6): 737-752, 2019 12.
Article in English | MEDLINE | ID: mdl-31548340

ABSTRACT

The two G protein-coupled receptors (GPCRs) C-X-C chemokine receptor type 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) are part of the class A chemokine GPCR family and represent important drug targets for human immunodeficiency virus (HIV) infection, cancer, and inflammation diseases. CXCR4 is one of only three chemokine receptors with a US Food and Drug Administration approved therapeutic agent, the small-molecule modulator AMD3100. In this review, known modulators of the two receptors are discussed in detail. Initially, the structural relationship between receptors and ligands is reviewed on the basis of common structural motifs and available crystal structures. To date, no atypical chemokine receptor has been crystallized, which makes ligand design and predictions for these receptors more difficult. Next, the selectivity, receptor activation, and the resulting ligand-induced signaling output of chemokines and other peptide ligands are reviewed. Binding of pepducins, a class of lipid-peptides whose basis is the internal loop of a GPCR, to CXCR4 is also discussed. Finally, small-molecule modulators of CXCR4 and ACKR3 are reviewed. These modulators have led to the development of radio- and fluorescently labeled tool compounds, enabling the visualization of ligand binding and receptor characterization both in vitro and in vivo. SIGNIFICANCE STATEMENT: To investigate the pharmacological modulation of CXCR4 and ACKR3, significant effort has been focused on the discovery and development of a range of ligands, including small-molecule modulators, pepducins, and synthetic peptides. Imaging tools, such as fluorescent probes, also play a pivotal role in the field of drug discovery. This review aims to provide an overview of the aforementioned modulators that facilitate the study of CXCR4 and ACKR3 receptors.


Subject(s)
Receptors, CXCR4/physiology , Receptors, CXCR/physiology , Amino Acid Sequence , Animals , Benzylamines , Cyclams , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacology , Humans , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, CXCR/agonists , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors
12.
Physiol Res ; 68(4): 675-679, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31177801

ABSTRACT

Chemokine (C-X-C motif) receptor 4 (CXCR4) agonists have been shown to protect lung endothelial barrier function in vitro. In vivo effects of CXCR4 modulation on lung endothelial permeability are unknown. Here we tested the effects of the CXCR4 agonist ubiquitin and the antagonist AMD3100 on lung vascular permeability and cytokine concentrations in a rat hemorrhage model. Animals were hemorrhaged (mean arterial blood pressure 30 mmHg for 30 min), treated with vehicle, ubiquitin (0.7 and 3.5 µmol/kg) or AMD3100 (3.5 µmol/kg), and resuscitated with crystalloids. Evans blue extravasation was employed to quantify lung vascular permeability. Ubiquitin dose-dependently reduced Evans blue extravasation into the lung. AMD3100 increased Evans blue extravasation. With AMD3100, TNFalpha levels in lung homogenates were increased; while TNFalpha levels were lower with ubiquitin, these differences did not reach statistical significance. Our findings suggest that CXCR4 regulates lung vascular permeability and further point towards CXCR4 as a drug target to confer lung protection during resuscitation from traumatic-hemorrhagic shock.


Subject(s)
Capillary Permeability/physiology , Receptors, CXCR4/physiology , Respiratory Mucosa/metabolism , Resuscitation , Shock, Hemorrhagic/metabolism , Shock, Hemorrhagic/therapy , Animals , Benzylamines , Capillary Permeability/drug effects , Cyclams , Dose-Response Relationship, Drug , Heterocyclic Compounds/pharmacology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors , Respiratory Mucosa/drug effects , Resuscitation/trends , Ubiquitin/pharmacology
13.
Neurochem Int ; 126: 59-63, 2019 06.
Article in English | MEDLINE | ID: mdl-30858017

ABSTRACT

We investigated the impact of the prolonged exposure of rat hippocampal synaptosomes to CXCL12 (3 nM) on the NMDA-mediated release of [3H]D-aspartate ([3H]D-Asp) or [3H]noradrenaline ([3H]NA). Synaptosomes were stimulated twice with NMDA/CXCL12 and the amount of the NMDA-evoked tritium release (S1 and S2) quantified to calculate the S2/S1 ratio. The S2/S1 ratio for both transmitters was drastically decreased by 3 nM CXCL12 between the two stimuli (CXCL12-treated synaptosomes) in a AMD3100-sensitive manner. The phosphorylation of the GluN1 subunit in Ser 896 was reduced in CXCL12-treated synaptosomes, while the overall amount of GluN1 and GluN2B proteins as well as the GluN2B insertion in synaptosomal plasmamembranes were unchanged. We conclude that the CXCR4/NMDA cross-talk is dynamically regulated by the time of activation of the CXCR4s. Our results unveil a functional cross-talk that might account for the severe impairments of central transmission that develop in pathological conditions characterized by CXCL12 overproduction.


Subject(s)
Hippocampus/metabolism , Presynaptic Terminals/metabolism , Receptors, CXCR4/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptosomes/metabolism , Animals , Chemokine CXCL12/pharmacology , Hippocampus/drug effects , N-Methylaspartate/pharmacology , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptosomes/drug effects
15.
Hypertension ; 72(4): 909-917, 2018 10.
Article in English | MEDLINE | ID: mdl-30354710

ABSTRACT

SDF-1α (stromal cell-derived factor-1α) is a CXCR4-receptor agonist and DPP4 (dipeptidyl peptidase 4) substrate. SDF-1α, particularly when combined with sitagliptin to block the metabolism of SDF-1α by DPP4, stimulates proliferation of cardiac fibroblasts via the CXCR4 receptor; this effect is greater in cells from spontaneously hypertensive rats versus Wistar-Kyoto normotensive rats. Emerging evidence indicates that ubiquitin(1-76) exists in plasma and is a potent CXCR4-receptor agonist. Therefore, we hypothesized that ubiquitin(1-76), similar to SDF-1α, should increase proliferation of cardiac fibroblasts. Contrary to our working hypothesis, ubiquitin(1-76) did not stimulate cardiac fibroblast proliferation, yet unexpectedly antagonized the proproliferative effects of SDF-1α combined with sitagliptin. In this regard, ubiquitin(1-76) was more potent in spontaneously hypertensive versus Wistar-Kyoto cells. In the presence of 6bk (selective inhibitor of insulin-degrading enzyme [IDE]; an enzyme known to convert ubiquitin(1-76) to ubiquitin(1-74)), ubiquitin(1-76) no longer antagonized the proproliferative effects of SDF-1α/sitagliptin. Ubiquitin(1-74) also antagonized the proproliferative effects of SDF-1α/sitagliptin, and this effect of ubiquitin(1-74) was not blocked by 6bk and was >10-fold more potent compared with ubiquitin(1-76). Neither ubiquitin(1-76) nor ubiquitin(1-74) inhibited the proproliferative effects of the non-CXCR4 receptor agonist neuropeptide Y (activates Y1 receptors). Cardiac fibroblasts expressed IDE mRNA, protein, and activity and converted ubiquitin(1-76) to ubiquitin(1-74). Spontaneously hypertensive fibroblasts expressed greater IDE activity. Extracellular ubiquitin(1-76) blocks the proproliferative effects of SDF-1α/sitagliptin via its conversion by IDE to ubiquitin(1-74), a potent CXCR4 antagonist. Thus, IDE inhibitors, particularly when combined with DPP4 inhibitors or hypertension, could increase the risk of cardiac fibrosis.


Subject(s)
Cell Proliferation , Chemokine CXCL12/metabolism , Fibroblasts , Hypertension/metabolism , Insulysin , Myocardium/pathology , Receptors, CXCR4 , Animals , Blood Pressure/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Insulysin/antagonists & inhibitors , Insulysin/metabolism , Neuropeptide Y/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, CXCR4/agonists , Receptors, CXCR4/metabolism , Signal Transduction , Sitagliptin Phosphate/pharmacology , Ubiquitin/metabolism
16.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3837-3846, 2018 11.
Article in English | MEDLINE | ID: mdl-30251699

ABSTRACT

Chemokine receptor CXCR4 was involved in the progression of breast cancer to a metastatic phenotype, leading to the major cause of death in patients. A more in-depth understanding of signaling mechanism underlying CXCR4 is critical to develop effective therapies toward metastasis. Recently, the role of antimicrobial peptide LL-37 in contributing to the metastasis of breast cancer cells was observed. Clinical analysis of data herein demonstrated for the first time that overexpression of LL-37 and CXCR4 co-existed in human primary breast tumors with lymph node metastases. Further study disclosed that forced expression of CXCR4 led to the enhancement of pro-migratory signaling and migration rate induced by LL-37 in breast cancer cells. Moreover, LL-37 affected tumor microenvironment including induction of migration of mesenchymal stem cells and CXCR4-dependent capillary-like tubule formation. Functional analysis showed that LL-37 induced the internalization of CXCR4 through approaching Glu268, the residue of CXCR4, independent of the binding pocket (Asp171, Asp262, and Glu288) for CXCR4 inhibitor AMD3100, signifying that LL-37 is a distinct agonist of CXCR4. These results suggest the reciprocal roles of LL-37 and CXCR4 in promoting breast cancer cell migration and provide new insight into the design of CXCR4 inhibitor for intervention of metastatic breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cathelicidins/metabolism , Cell Movement , Receptors, CXCR4/metabolism , Adult , Aged , Antimicrobial Cationic Peptides , Benzylamines , Binding Sites , Breast/pathology , Cathelicidins/chemistry , Cell Line, Tumor , Cell Proliferation , Cyclams , Female , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Lymphatic Metastasis , Mesenchymal Stem Cells , Middle Aged , Molecular Docking Simulation , Protein Binding/drug effects , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/chemistry , Signal Transduction , Tumor Microenvironment , Up-Regulation
17.
PLoS One ; 13(9): e0204041, 2018.
Article in English | MEDLINE | ID: mdl-30248140

ABSTRACT

We observed in PRESTO-Tango ß-arrestin recruitment assays that the α1-adrenergic receptor (AR) antagonist prazosin activates chemokine (C-X-C motif) receptor (CXCR)4. This prompted us to further examine this unexpected pharmacological behavior. We screened a panel of 14 α1/2- and ß1/2/3-AR antagonists for CXCR4 and atypical chemokine receptor (ACKR)3 agonist activity in PRESTO-Tango assays against the cognate agonist CXCL12. We observed that multiple α1-AR antagonists activate CXCR4 (CXCL12 = prazosin = cyclazosin > doxazosin) and ACKR3 (CXCL12 = prazosin = cyclazosin > alfuzosin = doxazosin = phentolamine > terazosin = silodosin = tamsulosin). The two strongest CXCR4/ACKR3 activators, prazosin and cyclazosin, were selected for a more detailed evaluation. We found that the drugs dose-dependently activate both receptors in ß-arrestin recruitment assays, stimulate ERK1/2 phosphorylation in HEK293 cells overexpressing each receptor, and that their effects on CXCR4 could be inhibited with AMD3100. Both α1-AR antagonists induced significant chemical shift changes in the 1H-13C-heteronuclear single quantum correlation spectrum of CXCR4 and ACKR3 in membranes, suggesting receptor binding. Furthermore, prazosin and cyclazosin induced internalization of endogenous CXCR4/ACKR3 in human vascular smooth muscle cells (hVSMC). While these drugs did not in induce chemotaxis in hVSMC, they inhibited CXCL12-induced chemotaxis with high efficacy and potency (IC50: prazosin-4.5 nM, cyclazosin 11.6 pM). Our findings reveal unexpected pharmacological properties of prazosin, cyclazosin, and likely other α1-AR antagonists. The results of the present study imply that prazosin and cyclazosin are biased or partial CXCR4/ACKR3 agonists, which function as potent CXCL12 antagonists. Our findings could provide a mechanistic basis for previously observed anti-cancer properties of α1-AR antagonists and support the concept that prazosin could be re-purposed for the treatment of disease processes in which CXCR4 and ACKR3 are thought to play significant pathophysiological roles, such as cancer metastases or various autoimmune pathologies.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Receptors, CXCR4/agonists , Receptors, CXCR/agonists , Binding Sites , Cells, Cultured , Chemokine CXCL12/antagonists & inhibitors , Chemotaxis/drug effects , Drug Evaluation, Preclinical , Drug Repositioning , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nuclear Magnetic Resonance, Biomolecular , Prazosin/pharmacology , Quinazolines/pharmacology , Quinoxalines/pharmacology , Receptors, CXCR/chemistry , Receptors, CXCR4/chemistry , beta-Arrestins/metabolism
18.
J Comput Aided Mol Des ; 32(4): 573-582, 2018 04.
Article in English | MEDLINE | ID: mdl-29582229

ABSTRACT

Antagonism of CCR9 is a promising mechanism for treatment of inflammatory bowel disease, including ulcerative colitis and Crohn's disease. There is limited experimental data on CCR9 and its ligands, complicating efforts to identify new small molecule antagonists. We present here results of a successful virtual screening and rational hit-to-lead campaign that led to the discovery and initial optimization of novel CCR9 antagonists. This work uses a novel data fusion strategy to integrate the output of multiple computational tools, such as 2D similarity search, shape similarity, pharmacophore searching, and molecular docking, as well as the identification and incorporation of privileged chemokine fragments. The application of various ranking strategies, which combined consensus and parallel selection methods to achieve a balance of enrichment and novelty, resulted in 198 virtual screening hits in total, with an overall hit rate of 18%. Several hits were developed into early leads through targeted synthesis and purchase of analogs.


Subject(s)
Computer Simulation , Molecular Docking Simulation/methods , Receptors, CCR/agonists , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Ligands , Molecular Structure , Principal Component Analysis , Receptors, CXCR4/agonists , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
19.
ACS Chem Biol ; 13(4): 881-886, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29461034

ABSTRACT

CXC chemokine receptor 4 (CXCR4) is involved in multiple physiological and pathological processes, notably as a coreceptor for human immunodeficiency virus (HIV) cell entry. Its broad expression pattern and vital biological importance make CXCR4 a troublesome drug target, as disruption of the interaction with its endogenous ligand, CXC chemokine ligand 12 (CXCL12), has severe consequences. In fact, only one CXCR4 drug, the bicyclam antagonist and HIV entry inhibitor AMD3100 (Plerixafor/Mozobil), has been approved for clinical use, however only for stem cell mobilization-a consequence of CXCR4 antagonism. Here, we report the engineering of an efficacy switch mutation in CXCR4-F292A7.43 in the middle of transmembrane helix 7-that converted the antagonists AMD3100 and AMD11070 into partial agonists. As agonists on F292A CXCR4, AMD3100 and AMD11070 were less disruptive to CXCR4 signaling while they remained efficient inhibitors of HIV fusion. This demonstrates that small molecule CXCR4 agonists can have a therapeutic potential as HIV entry inhibitors.


Subject(s)
HIV Fusion Inhibitors/chemistry , HIV-1/drug effects , Receptors, CXCR4/drug effects , Anti-HIV Agents , Humans , Protein Engineering/methods , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors
20.
Mol Cancer Res ; 16(4): 728-739, 2018 04.
Article in English | MEDLINE | ID: mdl-29330286

ABSTRACT

G-protein-coupled receptor (GPCR) heterodimerization has emerged as a means by which alternative signaling entities can be created; yet, how receptor heterodimers affect receptor pharmacology remains unknown. Previous observations suggested a biochemical antagonism between GPCRs, CXCR4 and CB2 (CNR2), where agonist-bound CXCR4 and agonist-bound CB2 formed a physiologically nonfunctional heterodimer on the membrane of cancer cells, inhibiting their metastatic potential in vitro However, the reduced signaling entities responsible for the observed functional outputs remain elusive. This study now delineates the signaling mechanism whereby heterodimeric association between CXCR4 and CB2, induced by simultaneous agonist treatment, results in decreased CXCR4-mediated cell migration, invasion, and adhesion through inhibition of the Gα13/RhoA signaling axis. Activation of CXCR4 by its cognate ligand, CXCL12, stimulates Gα13 (GNA13), and subsequently, the small GTPase RhoA, which is required for directional cell migration and the metastatic potential of cancer cells. These studies in prostate cancer cells demonstrate decreased protein expression levels of Gα13 and RhoA upon simultaneous CXCR4/CB2 agonist stimulation. Furthermore, the agonist-induced heterodimer abrogated RhoA-mediated cytoskeletal rearrangement resulting in the attenuation of cell migration and invasion of an endothelial cell barrier. Finally, a reduction was observed in the expression of integrin α5 (ITGA5) upon heterodimerization, supported by decreased cell adhesion to extracellular matrices in vitro Taken together, the data identify a novel pharmacologic mechanism for the modulation of tumor cell migration and invasion in the context of metastatic disease.Implications: This study investigates a signaling mechanism by which GPCR heterodimerization inhibits cancer cell migration. Mol Cancer Res; 16(4); 728-39. ©2018 AACR.


Subject(s)
Chemokine CXCL12/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Heterocyclic Compounds/pharmacology , Neoplasms/metabolism , rhoA GTP-Binding Protein/metabolism , Benzylamines , Cannabinoids/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cyclams , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Male , PC-3 Cells , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Receptors, CXCR4/agonists , Receptors, CXCR4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...