Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 526
Filter
1.
Biomed Pharmacother ; 174: 116524, 2024 May.
Article in English | MEDLINE | ID: mdl-38574622

ABSTRACT

This study evaluates the antiallodynic and antihyperalgesic effects of LMH-2, a new haloperidol (HAL) analog that acts as sigma-1 receptor (σ1 R) antagonist, in diabetic mice using a model of neuropathic pain induced by chronic hyperglycemia. Additionally, we compared its effects with those of HAL. Hyperglycemia was induced in mice by nicotinamide-streptozotocin administration (NA-STZ, 50-130 mg/kg). Four weeks later, mechanical allodynia was assessed using the up-down method, and hyperalgesia was evoked with formalin 0.5%. We evaluated antiallodynic and antihyperalgesic effects of LMH-2 (5.6-56.2 mg/kg), HAL (0.018-0.18 mg/kg) and gabapentin (GBP, 5.6-56.2 mg/kg). The results showed that LMH-2 had a more significant antiallodynic effect compared to HAL and GBP (90.4±8.7 vs 75.1±3.1 and 41.9±2.3%, respectively; P<0.05), as well as an antihyperalgesic effect (96.3±1.2 vs 86.9±7.41 and 86.9±4.8%, respectively; P<0.05). Moreover, the antiallodynic and antihyperalgesic effect of both LMH-2 and HAL were completely abolished by PRE-084 (σ1 R agonist); and partially by pramipexole (a D2-like receptor agonist). Finally, the effect of all treatments on the rotarod test, barra, open field and exploratory behaviors showed that LMH-2 did not alter the animals' balance or the exploratory behavior, unlike as HAL or GBP. The molecular docking included indicate that LMH-2 has lower affinity to the D2R than HAL. These results provide evidence that LMH-2 exerts its antinociceptive effects as a σ1 R antagonist without the adverse effects induced by HAL or GBP. Consequently, LMH-2 can be considered a good and safe strategy for treating neuropathic pain caused by hyperglycemia in patients with diabetes.


Subject(s)
Analgesics , Diabetes Mellitus, Experimental , Haloperidol , Hyperalgesia , Neuralgia , Receptors, sigma , Sigma-1 Receptor , Animals , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism , Haloperidol/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Male , Mice , Analgesics/pharmacology , Neuralgia/drug therapy , Hyperalgesia/drug therapy , Diabetic Neuropathies/drug therapy , Molecular Docking Simulation , Streptozocin , Dose-Response Relationship, Drug , Gabapentin/pharmacology
2.
Eur J Neurosci ; 59(10): 2436-2449, 2024 May.
Article in English | MEDLINE | ID: mdl-38444104

ABSTRACT

Psychostimulant use disorders (PSUD) are prevalent; however, no FDA-approved medications have been made available for treatment. Previous studies have shown that dual inhibitors of the dopamine transporter (DAT) and sigma receptors significantly reduce the behavioral/reinforcing effects of cocaine, which have been associated with stimulation of extracellular dopamine (DA) levels resulting from DAT inhibition. Here, we employ microdialysis and fast scan cyclic voltammetry (FSCV) procedures to investigate the effects of dual inhibitors of DAT and sigma receptors in combination with cocaine on nucleus accumbens shell (NAS) DA dynamics in naïve male Sprague Dawley rats. In microdialysis studies, administration of rimcazole (3, 10 mg/kg; i.p.) or its structural analog SH 3-24 (1, 3 mg/kg; i.p.), compounds that are dual inhibitors of DAT and sigma receptors, significantly reduced NAS DA efflux stimulated by increasing doses of cocaine (0.1, 0.3, 1.0 mg/kg; i.v.). Using the same experimental conditions, in FSCV tests, we show that rimcazole pretreatments attenuated cocaine-induced stimulation of evoked NAS DA release but produced no additional effect on DA clearance rate. Under the same conditions, JJC8-091, a modafinil analog and dual inhibitor of DAT and sigma receptors, similarly attenuated cocaine-induced stimulation of evoked NAS DA release but produced no additional effect on DA clearance rate. Our results provide the neurochemical groundwork towards understanding actions of dual inhibitors of DAT and sigma receptors on DA dynamics that likely mediate the behavioral effects of psychostimulants like cocaine.


Subject(s)
Cocaine , Dopamine Plasma Membrane Transport Proteins , Dopamine Uptake Inhibitors , Dopamine , Nucleus Accumbens , Rats, Sprague-Dawley , Receptors, sigma , Animals , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Receptors, sigma/metabolism , Receptors, sigma/antagonists & inhibitors , Male , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine/metabolism , Cocaine/pharmacology , Rats , Dopamine Uptake Inhibitors/pharmacology , Piperidines/pharmacology , Benzhydryl Compounds/pharmacology , Microdialysis/methods , Modafinil/pharmacology
3.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36768323

ABSTRACT

Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.


Subject(s)
Cardiovascular Diseases , Kidney Diseases , Receptors, sigma , Humans , Cardiomegaly , COVID-19/complications , Heart Failure/complications , Ligands , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/genetics , Receptors, sigma/metabolism , Signal Transduction/physiology , Cardiovascular Diseases/complications , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Kidney Diseases/complications , Kidney Diseases/genetics , Kidney Diseases/metabolism , Sigma-1 Receptor
4.
Int J Mol Sci ; 23(19)2022 Oct 08.
Article in English | MEDLINE | ID: mdl-36233233

ABSTRACT

Sigma-1 receptor (σ1R) ligands have been shown to be effective at relieving neuropathic and inflammatory pain, but have not yet been tested in experimental models of fibromyalgia. The objective of this study was to evaluate the effect of a σ1R antagonist (BD1063) compared to pregabalin. ICR-CD1 female mice were subjected to either six repeated injections of reserpine, to cause reserpine-induced myalgia (RIM6), or acidified saline intramuscular injections (ASI). In these two models, we evaluated the effect of BD1063 and pregabalin on thermal hypersensitivity, anxiety-like and depression-like behaviors, and on spinal cord gliosis. BD1063 exerted an antinociceptive effect on both reflexive (thermal hyperalgesia) and nonreflexive (anxiety- and depression-like) pain behaviors, and reduced spinal astroglial and microglial reactivity, following repeated treatment for 2 weeks. Interestingly, the effects of BD1063 were long-term, lasting several weeks after treatment discontinuation in both fibromyalgia-like models. Similar results were obtained with pregabalin, but the effects on pain behaviors lasted for a shorter length of time, and pregabalin did not significantly modulate spinal glial reactivity. The inhibitory and long-lasting effect of pharmacological blockade of σ1Rs on both sensory and affective dimensions of nociplastic-like pain and spinal cord gliosis in two experimental models of fibromyalgia support the application of this therapeutic strategy to treat fibromyalgia.


Subject(s)
Analgesics , Chronic Pain , Fibromyalgia , Receptors, sigma , Analgesics/therapeutic use , Animals , Chronic Pain/drug therapy , Disease Models, Animal , Female , Fibromyalgia/drug therapy , Gliosis , Hyperalgesia/drug therapy , Mice , Mice, Inbred ICR , Pregabalin/therapeutic use , Receptors, sigma/antagonists & inhibitors , Reserpine/adverse effects , Sigma-1 Receptor
5.
Eur J Med Chem ; 241: 114649, 2022 Nov 05.
Article in English | MEDLINE | ID: mdl-35961067

ABSTRACT

To develop safer and potent analgesics, we designed, synthesized, and evaluated a new series of benzylaminofentanyl derivates as bifunctional µ opioid receptor (MOR) and σ1 receptor (σ1R) ligands. Compound 68 (Tao-191) showed desirable MOR agonism (Ki = 6.5 nΜ; EC50 = 48.5 nΜ, Emax = 66.3%) and σ1R antagonism (Ki = 35.7 nM) in vitro, and exerted powerful analgesic effects in the abdominal constriction test (ED50 = 0.32 mg/kg, in mice), formalin-induced pain test (phase II, ED50 = 2.26 mg/kg, in rats), and paclitaxel-induced neuropathic pain model (ED50 = 0.30 mg/kg, in mice). The contributions of MOR and σ1R to its antinociceptive effect were verified by combined administration with the MOR antagonist naloxone and the σ1R agonist PRE-084, respectively. At equianalgesic doses, compound 68 induced fewer MOR-related side effects-including physical and psychological dependence, respiratory depression, constipation, and acute hyperlocomotion-than fentanyl. The results provide a rationale for further exploration of the action and safety of dual MOR/σ1R ligands as a promising avenue for the development of potent and safe analgesics.


Subject(s)
Analgesics , Neuralgia , Receptors, Opioid, mu , Receptors, sigma , Analgesics/pharmacology , Animals , Ligands , Mice , Naloxone/pharmacology , Rats , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Sigma-1 Receptor
6.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35054797

ABSTRACT

Neuropathic pain is a significant problem with few effective treatments lacking adverse effects. The sigma-1 receptor (S1R) is a potential therapeutic target for neuropathic pain, as antagonists for this receptor effectively ameliorate pain in both preclinical and clinical studies. The current research examines the antinociceptive and anti-allodynic efficacy of SI 1/28, a recently reported benzylpiperazine derivative and analog of the S1R antagonist SI 1/13, that was 423-fold more selective for S1R over the sigma-2 receptor (S2R). In addition, possible liabilities of respiration, sedation, and drug reinforcement caused by SI 1/28 have been evaluated. Inflammatory and chemical nociception, chronic nerve constriction injury (CCI) induced mechanical allodynia, and adverse effects of sedation in a rotarod assay, conditioned place preference (CPP), and changes in breath rate and locomotor activity were assessed after i.p. administration of SI 1/28. Pretreatment with SI 1/28 produced dose-dependent antinociception in the formalin test, with an ED50 (and 95% C.I.) value of 13.2 (7.42-28.3) mg/kg, i.p. Likewise, SI 1/28 produced dose-dependent antinociception against visceral nociception and anti-allodynia against CCI-induced neuropathic pain. SI 1/28 demonstrated no impairment of locomotor activity, conditioned place preference, or respiratory depression. In summary, SI 1/28 proved efficacious in the treatment of acute inflammatory pain and chronic neuropathy without liabilities at therapeutic doses, supporting the development of S1R antagonists as therapeutics for chronic pain.


Subject(s)
Analgesics/therapeutic use , Hyperalgesia/drug therapy , Nociception , Receptors, sigma/antagonists & inhibitors , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Disease Models, Animal , Hyperalgesia/complications , Inflammation/complications , Inflammation/pathology , Male , Mice, Inbred C57BL , Receptors, sigma/metabolism , Time Factors , Viscera/pathology , Sigma-1 Receptor
7.
Eur J Med Chem ; 230: 114091, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35016113

ABSTRACT

The development of σ1 receptor antagonists hybridized with a H2S-donor is here reported. We aimed to obtain improved analgesic effects when compared to σ1 receptor antagonists or H2S-donors alone. In an in vivo model of sensory hypersensitivity, thioamide 1a induced analgesia which was synergistically enhanced when associated with the σ1 receptor antagonist BD-1063. The selective σ1 receptor agonist PRE-084 completely reversed this effect. Four thioamide H2S-σ1 receptor hybrids (5a-8a) and their amide derivatives (5b-8b) were synthesized. Compound 7a (AD164) robustly released H2S and showed selectivity for σ1 receptor over σ2 and opioid receptors. This compound induced marked analgesia that was reversed by PRE-084. The amide analogue 7b (AD163) showed only minimal analgesia. Further studies showed that 7a exhibited negligible acute toxicity, together with a favorable pharmacokinetic profile. To the best of our knowledge, compound 7a is the first dual-acting ligand with simultaneous H2S-release and σ1 antagonistic activities.


Subject(s)
Hydrogen Sulfide , Morpholines/pharmacology , Pain/drug therapy , Piperazines/pharmacology , Receptors, sigma , Animals , Guinea Pigs , Hydrogen , Ligands , Male , Rats, Sprague-Dawley , Receptors, sigma/antagonists & inhibitors , Sigma-1 Receptor
8.
Br J Clin Pharmacol ; 88(4): 1644-1654, 2022 02.
Article in English | MEDLINE | ID: mdl-34156715

ABSTRACT

AIMS: Preclinical studies of MR309, a selective sigma-1 receptor (σ1R) antagonist, support a potential role in treating neuropathic pain. We report 2 studies that provide insight into the pharmacokinetics (PK) and brain σ1R binding of MR309. METHODS: Steady-state PK of MR309 (400 mg once daily and 200 mg twice-daily [BID] for 10 days; EudraCT 2015-001818-99 [PK study]) and the relationship between MR309 plasma exposure and brain σ1R occupancy (EudraCT 2017-000670-11 [positron emission tomography study]) were investigated in healthy volunteers. Positron emission tomography using the σ1R ligand [11 C]SA4503 was conducted at baseline, and 2 and 8 hours after a single dose of MR309 (200-800 mg). The relationship between brain σ1R occupancy and MR309 exposure was explored using data-driven model fitting. RESULTS: MR309 was well tolerated, brain σ1R occupancy ranged between 30.5 and 74.9% following single-dose MR309 (n = 7). MR309 BID provided a plasma PK profile with less fluctuation than once daily dosing (n = 16). MR309 200 mg BID yielded average steady state plasma concentrations between 2000 and 4000 ng/mL in the PK study, which corresponded to an estimated brain σ1R occupancy of 59-74%. CONCLUSION: MR309 200 mg BID dose was below the 75% σ1R occupancy threshold expected to elicit maximal antinociceptive effect as observed in neuropathic pain models. Further investigations of MR309 for neuropathic pain will require higher brain σ1R occupancy, and establish the optimal dose by elucidating the clinical impact of a broad range of brain σ1R occupancy across different neuropathic pain indications.


Subject(s)
Neuralgia , Receptors, sigma , Brain/diagnostic imaging , Brain/metabolism , Humans , Neuralgia/drug therapy , Positron-Emission Tomography/methods , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism , Sigma-1 Receptor
9.
Brain Res Bull ; 178: 69-81, 2022 01.
Article in English | MEDLINE | ID: mdl-34813897

ABSTRACT

Recent studies have revealed that glial sigma-1 receptor (Sig-1R) in the spinal cord may be a critical factor to mediate sensory function. However, the functional role of Sig-1R in astrocyte has not been clearly elucidated. Here, we determined whether Sig-1Rs modulate calcium responses in primary cultured astrocytes and pathological changes in spinal astrocytes, and whether they contribute to pain hypersensitivity in naïve mice and neuropathic pain following chronic constriction injury (CCI) of the sciatic nerve in mice. Sig-1R was expressed in glial fibrillary acidic protein (GFAP)-positive cultured astrocytes. Treatment with the Sig-1R agonist, PRE-084 or neurosteroid dehydroepiandrosterone (DHEA) increased intracellular calcium responses in cultured astrocytes, and this increase was blocked by the pretreatment with the Sig-1R antagonist, BD-1047 or neurosteroid progesterone. Intrathecal administration of PRE-084 or DHEA for 10 days induced mechanical and thermal hypersensitivity and increased the number of Sig-1R-immunostained GFAP-positive cells in the superficial dorsal horn (SDH) region of the spinal cord in naïve mice, and these changes were inhibited by administration with BD-1047 or progesterone. In CCI mice, intrathecal administration of BD-1047 or progesterone at post-operative day 14 suppressed the developed mechanical allodynia and the number of Sig-1R-immunostained GFAP-positive cells that were increased in the SDH region of the spinal cord following CCI of the sciatic nerve. These results demonstrate that Sig-1Rs play an important role in the modulation of intracellular calcium responses in cultured astrocytes and pathological changes in spinal astrocytes and that administration of BD-1047 or progesterone alleviates the Sig-1R-induced pain hypersensitivity and the peripheral nerve injury-induced mechanical allodynia.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Hyperalgesia/metabolism , Neuralgia/metabolism , Neurosteroids/metabolism , Peripheral Nerve Injuries/metabolism , Receptors, sigma/metabolism , Spinal Cord/metabolism , Animals , Astrocytes/drug effects , Cells, Cultured , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Mice , Neuralgia/drug therapy , Neuralgia/physiopathology , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/physiopathology , Progesterone/pharmacology , Receptors, sigma/antagonists & inhibitors , Spinal Cord/drug effects , Spinal Cord/physiopathology , Sigma-1 Receptor
10.
Eur J Med Chem ; 228: 114038, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34902734

ABSTRACT

The sigma-1 (σ1) receptor plays a significant role in many normal physiological functions and pathological disease states, and as such represents an attractive therapeutic target for both agonists and antagonists. Here, we describe a novel series of phenoxyalkylpiperidines based on the lead compound 1-[ω-(4-chlorophenoxy)ethyl]-4-methylpiperidine (1a) in which the degree of methylation at the carbon atoms alpha to the piperidine nitrogen was systematically varied. The affinity at σ1 and σ2 receptors and at Δ8-Δ7 sterol isomerase (SI) ranged from subnanomolar to micromolar Ki values. While the highest-affinity was displayed at the σ1, the increase of the degree of methylation in the piperidine ring progressively decreased the affinity. The subnanomolar affinity 1a and 1-[ω-(4-methoxyphenoxy)ethyl]-4-methylpiperidine (1b) displayed potent anti-amnesic effects associated with σ1 receptor agonism, in two memory tests. Automated receptor-small-molecule ligand docking provided a molecular structure-based rationale for the agonistic effects of 1a and 1b. Overall, the class of the phenoxyalkylpiperidines holds potential for the development of high affinity σ1 receptor agonists, and compound 1a, that appears as the best in class (exceeding by far the activity of the reference compound PRE-084) deserves further investigation.


Subject(s)
Amnesia/drug therapy , Drug Development , Piperidines/pharmacology , Receptors, sigma/antagonists & inhibitors , Amnesia/metabolism , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Guinea Pigs , Humans , Ligands , Male , Mice , Models, Molecular , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Rats , Rats, Wistar , Receptors, sigma/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured , Sigma-1 Receptor
11.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769165

ABSTRACT

Our recent study demonstrated that the CC-chemokine ligand 2 (CCL2) present in primary afferent fibers (PAFs) plays an important role in the microglia-dependent neuronal activation associated with zymosan-induced inflammatory pain. The present study was aimed to evaluate whether BD1047 (a prototypical sigma-1 receptor (Sig-1R) antagonist) is capable of modifying elevated levels of inflammation-evoked CCL2 as a peripheral antinociceptive mechanism. In DRG primary culture, zymosan dose-dependently increased CCL2 release from isolectin B4 (IB4)-positive DRG neurons, a process that was inhibited by co-culture with BD1047. Single treatment of BD1047 before intraplantar injection of zymosan in rats significantly reduced thermal hyperalgesia and mechanical hyperalgesia, as well as CCL2 expression in DRG neurons and microglia activation in the spinal dorsal horn. In the Complete Freund's adjuvant (CFA)-induced inflammation model, repeated administration of BD1047 dramatically attenuated thermal hyperalgesia and mechanical hyperalgesia, and significantly diminished CCL2 immunoreactivity and microglia activation. Notably, CFA-induced inflammation significantly increased Sig-1R immunoreactivity in DRG neurons, which was co-localized with CCL2 and IB4, respectively. Taken together, our results suggest that BD1047's anti-nociceptive property was substantially mediated by the inhibition of CCL2 release in unmyelinated PAFs and that this may, in turn, have attenuated the spinal microglia activation that is associated with inflammatory pain.


Subject(s)
Analgesics/therapeutic use , Chemokine CCL2/metabolism , Ethylenediamines/therapeutic use , Hyperalgesia/drug therapy , Receptors, sigma/antagonists & inhibitors , Animals , Cells, Cultured , Hyperalgesia/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, sigma/metabolism , Sigma-1 Receptor
12.
Eur J Med Chem ; 226: 113879, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34628236

ABSTRACT

Here, we describe the optimization, synthesis, and associated pharmacological analgesic activities of a new series of bifunctional piperidinamide derivatives as sigma-1 receptor (σ1R) antagonists and mu opioid receptor (MOR) agonists. The new compounds were evaluated in vitro in σ1R and MOR binding assays. The most promising compound 114 (also called HKC-126), showed superior affinities for σ1R and MOR and good selectivity to additional receptors related to pain. Compound 114 showed powerful dose-dependent analgesic effects in the acetic acid writhing test, formalin test, hot plate test, and chronic constriction injury (CCI) neuropathic pain model. In contrast to an equianalgesic dose of fentanyl, compound 114 produced fewer opioid-like side effects, such as reward liability, respiratory depression, physical dependence, and sedation. Lastly, the pharmacokinetic properties of this drug were also acceptable, and these results suggest that compound 114, as a mixed σ1R/MOR ligand, has potential for treating neuropathic pain.


Subject(s)
Amides/pharmacology , Neuralgia/drug therapy , Piperidines/pharmacology , Receptors, Opioid, mu/agonists , Receptors, sigma/antagonists & inhibitors , Acetic Acid , Amides/chemical synthesis , Amides/chemistry , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Formaldehyde , Guinea Pigs , Mice , Mice, Inbred ICR , Molecular Dynamics Simulation , Molecular Structure , Neuralgia/chemically induced , Neuralgia/metabolism , Pain Measurement , Piperidines/chemical synthesis , Piperidines/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Receptors, sigma/metabolism , Structure-Activity Relationship , Sigma-1 Receptor
13.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34360878

ABSTRACT

Sigma-1 receptor (S1R) is an intracellular, multi-functional, ligand operated protein that also acts as a chaperone. It is considered as a pluripotent drug target in several pathologies. The publication of agonist and antagonist bound receptor structures has paved the way for receptor-based in silico drug design. However, recent studies on this subject payed no attention to the structural differences of agonist and antagonist binding. In this work, we have developed a new ensemble docking-based virtual screening protocol utilizing both agonist and antagonist bound S1R structures. This protocol was used to screen our in-house compound library. The S1R binding affinities of the 40 highest ranked compounds were measured in competitive radioligand binding assays and the sigma-2 receptor (S2R) affinities of the best S1R binders were also determined. This way three novel high affinity S1R ligands were identified and one of them exhibited a notable S1R/S2R selectivity.


Subject(s)
Isoxazoles/chemistry , Molecular Docking Simulation/methods , Pentazocine/chemistry , Pyridines/chemistry , Receptors, sigma/chemistry , Binding Sites , Hydrophobic and Hydrophilic Interactions , Isoxazoles/analysis , Isoxazoles/pharmacology , Ligands , Molecular Structure , Pentazocine/analysis , Pentazocine/pharmacology , Protein Binding , Pyridines/analysis , Pyridines/pharmacology , Radioligand Assay/methods , Receptors, sigma/agonists , Receptors, sigma/analysis , Receptors, sigma/antagonists & inhibitors , Sigma-1 Receptor
14.
J Med Chem ; 64(14): 10139-10154, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34236190

ABSTRACT

A new series of propionamide derivatives was developed as dual µ-opioid receptor agonists and σ1 receptor antagonists. Modification of a high-throughput screening hit originated a series of piperazinylcycloalkylmethyl propionamides, which were explored to overcome the challenge of achieving balanced dual activity and convenient drug-like properties. The lead compound identified, 18g, showed good analgesic effects in several animal models of both acute (paw pressure) and chronic (partial sciatic nerve ligation) pain, with reduced gastrointestinal effects in comparison with oxycodone.


Subject(s)
Amides/pharmacology , Analgesics, Opioid/pharmacology , Narcotic Antagonists/pharmacology , Pain/drug therapy , Receptors, Opioid, mu/agonists , Receptors, sigma/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/chemistry , Animals , Dose-Response Relationship, Drug , Humans , Mice , Molecular Structure , Narcotic Antagonists/chemical synthesis , Narcotic Antagonists/chemistry , Structure-Activity Relationship , Sigma-1 Receptor
15.
Molecules ; 26(13)2021 Jun 24.
Article in English | MEDLINE | ID: mdl-34202711

ABSTRACT

Cancer is a multifactorial disease that may be tackled by targeting different signaling pathways. Heme oxygenase-1 (HO-1) and sigma receptors (σRs) are both overexpressed in different human cancers, including prostate and brain, contributing to the cancer spreading. In the present study, we investigated whether HO-1 inhibitors and σR ligands, as well a combination of the two, may influence DU145 human prostate and U87MG human glioblastoma cancer cells proliferation. In addition, we synthesized, characterized, and tested a small series of novel hybrid compounds (HO-1/σRs) 1-4 containing the chemical features needed for HO-1 inhibition and σR modulation. Herein, we report for the first time that targeting simultaneously HO-1 and σR proteins may be a good strategy to achieve increased antiproliferative activity against DU145 and U87MG cells, with respect to the mono administration of the parent compounds. The obtained outcomes provide an initial proof of concept useful to further optimize the structure of HO-1/σRs hybrids to develop novel potential anticancer agents.


Subject(s)
Antineoplastic Agents , Enzyme Inhibitors , Heme Oxygenase-1/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasms , Receptors, sigma/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Heme Oxygenase-1/metabolism , Humans , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Rats , Receptors, sigma/metabolism
16.
Eur J Med Chem ; 223: 113658, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34175542

ABSTRACT

Opioid analgesics are highly effective painkillers for the treatment of moderate or severe pain, but they are associated with a number of undesirable adverse effects, including the development of tolerance, addiction, constipation and life-threatening respiratory depression. The development of new and safer analgesics with innovative mechanisms of action, which can enhance the efficacy in comparison to available treatments and reduce their side effects, is urgently needed. The sigma-1 receptor (σ1R), a unique Ca2+-sensing chaperone protein, is expressed throughout pain-modulating tissues and affects neurotransmission by interacting with different protein partners, including molecular targets that participate in nociceptive signalling, such as the µ-opioid receptor (MOR), N-methyl-d-aspartate receptor (NMDAR) and cannabinoid 1 receptor (CB1R). Overwhelming pharmacological and genetic evidence indicates that σ1R antagonists induce anti-hypersensitive effects in sensitising pain conditions (e.g. chemically induced, inflammatory and neuropathic pain) and enhance opioid analgesia but not opioid-mediated detrimental effects. It has been suggested that balanced modulation of MORs and σ1Rs may improve both the therapeutic efficacy and safety of opioids. This review summarises the functional profiles of ligands with mixed MOR agonist and σ1R antagonist activities and highlights their therapeutic potentials for pain management. Dual MOR agonism/σ1R antagonism represents a promising avenue for the development of potent and safer analgesics.


Subject(s)
Analgesics, Opioid/chemistry , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Analgesics, Opioid/adverse effects , Analgesics, Opioid/metabolism , Analgesics, Opioid/therapeutic use , Benzopyrans/chemistry , Benzopyrans/metabolism , Humans , Ligands , Pain/drug therapy , Piperazines/chemistry , Piperazines/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism , Sigma-1 Receptor
17.
J Med Chem ; 64(12): 7926-7962, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34076441

ABSTRACT

Since their discovery as distinct receptor proteins, the specific physiopathological role of sigma receptors (σRs) has been deeply investigated. It has been reported that these proteins, classified into two subtypes indicated as σ1 and σ2, might play a pivotal role in cancer growth, cell proliferation, and tumor aggressiveness. As a result, the development of selective σR ligands with potential antitumor properties attracted significant attention as an emerging theme in cancer research. This perspective deals with the recent advances of σR ligands as novel cytotoxic agents, covering articles published between 2010 and 2020. An up-to-date description of the medicinal chemistry of selective σ1R and σ2R ligands with antiproliferative and cytotoxic activities has been provided, including major pharmacophore models and comprehensive structure-activity relationships for each main class of σR ligands.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds/pharmacology , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/therapeutic use , Humans , Ligands , Molecular Structure , Structure-Activity Relationship
18.
Eur J Pharmacol ; 906: 174263, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34144027

ABSTRACT

Sigma-2 receptor/transmembrane protein 97 (TMEM97) is upregulated in cancer cells compared to normal cells. Traditional sigma-2 receptor agonists induce apoptosis and autophagy, making them of interest in cancer therapy. Recently, we reported a novel metabolically stimulative function of the sigma-2 receptor, showing increased 3-(4,5 dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction and stimulation of glycolytic hallmarks. 6-Substituted analogs of the canonical sigma-2 receptor antagonist, 6-acetyl-3-(4-(4-(4-fluorophenyl)piperazin-1-yl)butyl)benzo[d]oxazol-2(3H)-one (SN79), produce both metabolically stimulative and cytotoxic effects. Here, we compare the activities of two related compounds: 6-amino-3-(4-(4-(4-fluorophenyl)piperazin-1-yl)butyl)benzo[d]oxazol-2(3H)-one (CM571), the 6-amino derivative of SN79, which binds with high affinity to both sigma-1 and sigma-2 receptors, and 1,3-bis(3-(4-(4-(4-fluorophenyl)piperazin-1-yl)butyl)-2-oxo-2,3-dihydrobenzo[d]oxazol-6-yl)thiourea (MAM03055A), a homo-bivalent dimer of CM571. MAM03055A resulted from the degradation of 3-(4-(4-(4-fluorophenyl)piperazin-1-yl)butyl)-6-isothiocyanatobenzo[d]oxazol-2(3H)-one (CM572), the cytotoxic 6-isothiocyanato SN79 derivative. MAM03055A exhibited high affinity and strong preference for sigma-2 receptors (sigma-1 Ki = 3371 nM; sigma-2 receptor Ki = 55.9 nM). Functionally, MAM03055A treatment potently induced cell death in SK-N-SH neuroblastoma, MDA-MB-231 breast, and both SW48 and SW480 colorectal cancer cell lines, causing proapoptotic BH3 interacting-domain death agonist (BID) cleavage in SK-N-SH cells. Conversely, CM571 induced metabolic stimulation. CM571 bound reversibly to both receptors, while MAM03055A bound pseudo-irreversibly to sigma-2 receptors and caused residual cytotoxic activity after acute exposure and removal of the compound from the media. Interestingly, MAM03055A induced a time-dependent loss of sigma-2 receptor/TMEM97 protein from cells, whereas monomer CM571 had no effect on receptor levels. These results suggest that monovalent and bivalent sigma-2 receptor ligands in this series interact differently with the receptor, thus resulting in divergent effects.


Subject(s)
Antineoplastic Agents/pharmacology , Membrane Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, sigma/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Isocyanates/pharmacology , Isocyanates/therapeutic use , Membrane Proteins/metabolism , Neoplasms/pathology , Receptors, sigma/metabolism
19.
Alcohol Clin Exp Res ; 45(7): 1398-1407, 2021 07.
Article in English | MEDLINE | ID: mdl-34060104

ABSTRACT

BACKGROUND: Alcohol use disorder (AUD) is a complex psychiatric disease characterized by high alcohol intake as well as hyperkatifeia and hyperalgesia during withdrawal. A role for Sigma-1 receptors (Sig-1Rs) in the rewarding and reinforcing effects of alcohol has started to emerge in recent years, as rat studies have indicated that Sig-1R hyperactivity may result in excessive alcohol drinking. Sig-1R studies in mice are very scarce, and its potential role in alcohol-induced hyperalgesia is also unknown. METHODS: In this study, we investigated the role of Sig-1R in alcohol drinking and associated hyperalgesia in male mice, using an intermittent access 2-bottle choice model of heavy drinking. RESULTS: The Sig-1R antagonist BD-1063 was found dose dependently to reduce both alcohol intake and preference, without affecting either water or sucrose intake, suggesting that the effects are specific for alcohol. Notably, the ability of BD-1063 to suppress ethanol intake correlated with the individual baseline levels of alcohol drinking, suggesting that the treatment was more efficacious in heavy drinking animals. In addition, BD-1063 reversed alcohol-induced hyperalgesia during withdrawal, assessed using an automatic Hargreaves test, without affecting thermal sensitivity in alcohol-naïve animals or locomotor activity in either group. CONCLUSIONS: These data show that Sig-1R antagonism dose-dependently reduced ethanol consumption in heavy drinking mice as well as its efficacy in reducing alcohol-induced hyperalgesia. These findings provide a foundation for the development of novel treatments for AUD and associated pain states.


Subject(s)
Alcohol Drinking/prevention & control , Ethanol/administration & dosage , Hyperalgesia/prevention & control , Piperazines/administration & dosage , Receptors, sigma/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Head , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Pain Measurement , Piperazines/therapeutic use , Receptors, sigma/physiology , Substance Withdrawal Syndrome/drug therapy , Sucrose/administration & dosage , Sigma-1 Receptor
20.
Biochemistry (Mosc) ; 86(4): 471-479, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33941067

ABSTRACT

Huntington's disease (HD) is a neurodegenerative, dominantly inherited genetic disease caused by expansion of the polyglutamine tract in the huntingtin gene. At the cellular level, HD is characterized by the accumulation of mutant huntingtin protein in brain cells, resulting in the development of the HD phenotype, which includes mental disorders, decreased cognitive abilities, and progressive motor impairments in the form of chorea. Despite numerous studies, no unambigous connection between the accumulation of mutant protein and selective death of striatal neurons has yet been established. Recent studies have shown impairments in the calcium homeostasis in striatal neurons in HD. These cells are extremely sensitive to changes in the cytoplasmic concentration of calcium and its excessive increase leads to their death. One of the possible ways to normalize the balance of calcium in striatal neurons is through the sigma 1 receptor (S1R), which act as a calcium sensor that also exhibits modulating chaperone activity upon the cell stress observed during the development of many neurodegenerative diseases. The fact that S1R is a ligand-operated protein makes it a new promising molecular target for the development of drug therapy of HD based on the agonists of this receptor.


Subject(s)
Calcium/metabolism , Corpus Striatum/metabolism , Huntington Disease/metabolism , Neurons/metabolism , Receptors, sigma/antagonists & inhibitors , Animals , Corpus Striatum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Huntington Disease/drug therapy , Neurons/drug effects , Receptors, sigma/metabolism , Sigma-1 Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...