Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(17): 11985-12004, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37611316

ABSTRACT

We have designed orally bioavailable, non-brain-penetrant antagonists of the cannabinoid-1 receptor (CB1R) with a built-in biguanide sensor to mimic 5'-adenosine monophosphate kinase (AMPK) activation for treating obesity-associated co-morbidities. A series of 3,4-diarylpyrazolines bearing rational pharmacophoric pendants designed to limit brain penetration were synthesized and evaluated in CB1R ligand binding assays and recombinant AMPK assays. The compounds displayed high CB1R binding affinity and potent CB1R antagonist activities and acted as AMPK activators. Select compounds showed good oral exposure, with compounds 36, 38-S, and 39-S showing <5% brain penetrance, attesting to peripheral restriction. In vivo studies of 38-S revealed decreased food intake and body weight reduction in diet-induced obese mice as well as oral in vivo efficacy of 38-S in ameliorating glucose tolerance and insulin resistance. The designed "cannabinoformin" four-arm CB1R antagonists could serve as potential leads for treatment of metabolic syndrome disorders with negligible neuropsychiatric side effects.


Subject(s)
Cannabinoids , Metabolic Diseases , Metabolic Syndrome , Animals , Mice , Metabolic Syndrome/drug therapy , AMP-Activated Protein Kinases , Biguanides/pharmacology , Biguanides/therapeutic use , Cannabinoid Receptor Antagonists , Mice, Obese
2.
Toxicol Appl Pharmacol ; 470: 116548, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37182749

ABSTRACT

Nowadays, more and more new synthetic cannabinoids (SCs) appearing on the illicit market present challenges to analytical, forensic, and toxicology experts. For a better understanding of the physiological effect of SCs, the key issue is studying their metabolomic and psychoactive properties. In this study, our validated targeted reversed phase UHPLC-MS/MS method was used for determination of urinary concentration of 5F-MDMB-PICA, 4F-MDMB-BICA, and their primary metabolites. The liquid-liquid extraction procedure was applied for the enrichment of SCs. The pharmacological characterization of investigated SCs were studied by radioligand competition binding and ligand stimulated [35S]GTPγS binding assays. For 5F-MDMB-PICA and 4F-MDMB-BICA, the median urinary concentrations were 0.076 and 0.312 ng/mL. For primary metabolites, the concentration range was 0.029-881.02* ng/mL for 5F-MDMB-PICA-COOH, and 0.396-4579* ng/mL for 4F-MDMB-BICA-COOH. In the polydrug aspect, the 22 urine samples were verified to be abused with 6 illicit drugs. The affinity of the metabolites to CB1R significantly decreased compared to the parent ligands. In the GTPγS functional assay, both 5F-MDMB-PICA and 4F-MDMB-BICA were acting as full agonists, while the metabolites were found as weak inverse agonists. Additionally, the G-protein stimulatory effects of the full agonist 5F-MDMB-PICA and 4F-MDMB-BICA were reduced by metabolites. These results strongly indicate the dose-dependent CB1R-mediated weak inverse agonist effects of the two butanoic acid metabolites. The obtained high concentration of main urinary metabolites of 5F-MDMB-PICA and 4F-MDMB-BICA confirmed the relevance of their routine analysis in forensic and toxicological practices. Based on in vitro binding assays, the metabolites presumably might cause a lower psychoactive effect than parent compounds.


Subject(s)
Cannabinoids , Tandem Mass Spectrometry , Tandem Mass Spectrometry/methods , Drug Inverse Agonism , Guanosine 5'-O-(3-Thiotriphosphate) , Cannabinoids/pharmacology
3.
ACS Med Chem Lett ; 14(4): 479-486, 2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37077391

ABSTRACT

In this study, 1H-pyrazole-3-carboxylic acids related to the cannabinoid type 1 (CB1) receptor antagonist rimonabant were amidated with valine or tert-leucine, and the resulting acids were further diversified as methyl esters, amides, and N-methyl amides. In vitro receptor binding and functional assays demonstrated a wide series of activities related to the CB1 receptors (CB1Rs). Compound 34 showed a high CB1R binding affinity (K i = 6.9 nM) and agonist activity (EC50 = 46 nM; E max = 135%). Radioligand binding and [35S]GTPγS binding assays also demonstrated its selectivity and specificity to CB1Rs. Moreover, in vivo experiments revealed that 34 was slightly more effective than the CB1 agonist WIN55,212-2 in the early phase of the formalin test, indicating a short duration of the analgesic effect. Interestingly, in a mouse model of zymosan-induced hindlimb edema, 34 was able to maintain the percentage of paw volume below 75% for 24 h following subcutaneous injection. After intraperitoneal administration, 34 increased the food intake of mice, suggesting potential activity on CB1Rs.

4.
Antioxidants (Basel) ; 11(1)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35052666

ABSTRACT

Over the last decades, growing interest has turned to preventive and therapeutic approaches for achieving successful aging. Oxidative stress and inflammation are fundamental features of cardiovascular diseases; therefore, potential targets of them can improve cardiac outcomes. Our study aimed to examine the involvement of the endocannabinoid system, especially the CB1 receptor blockade, on inflammatory and oxidant/antioxidant processes. Twenty-month-old female and male Wistar rats were divided into rimonabant-treated and aging control (untreated) groups. Rimonabant, a selective CB1 receptor antagonist, was administered at the dose of 1 mg/kg/day intraperitoneally for 2 weeks. Cardiac amounts of ROS, the antioxidant glutathione and superoxide dismutase (SOD), and the activity and concentration of the heme oxygenase (HO) enzyme were detected. Among inflammatory parameters, nuclear factor-kappa B (NF-κB), tumor necrosis factor-alpha (TNF-α), and myeloperoxidase (MPO) enzyme activity were measured. Two weeks of low dose rimonabant treatment significantly reduced the cardiac ROS via boosting of the antioxidant defense mechanisms as regards the HO system, and the SOD and glutathione content. Consistently, the age-related inflammatory response was alleviated. Rimonabant-treated animals showed significantly decreased NF-κB, TNF-α, and MPO levels. Our findings prove the beneficial involvement of CB1 receptor blocker rimonabant on inflammatory and oxidative damages to the aging heart.

5.
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
6.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34072767

ABSTRACT

Substance use/abuse is one of the main causes of depressive symptoms. Cannabis and synthetic cannabinoids in particular gained significant popularity in the past years. There is an increasing amount of clinical data associating such compounds with the inflammatory component of depression, indicated by the up-regulation of pro-inflammatory cytokines. Pro-inflammatory cytokines are also well-known to regulate the enzymes of the kynurenine pathway (KP), which is responsible for metabolizing tryptophan, a precursor in serotonin synthesis. Enhanced pro-inflammatory cytokine levels may over-activate the KP, leading to tryptophan depletion and reduced serotonin levels, which can subsequently precipitate depressive symptoms. Therefore, such mechanism might represent a possible link between the endocannabinoid system (ECS) and the KP in depression, via the inflammatory and dysregulated serotonergic component of the disorder. This review will summarize the data regarding those natural and synthetic cannabinoids that increase pro-inflammatory cytokines. Furthermore, the data on such cytokines associated with KP activation will be further reviewed accordingly. The interaction of the ECS and the KP has been postulated and demonstrated in some studies previously. This review will further contribute to this yet less explored connection and propose the KP to be the missing link between cannabinoid-induced inflammation and depressive symptoms.


Subject(s)
Cytokines/metabolism , Depression/etiology , Depression/metabolism , Inflammation Mediators/metabolism , Animals , Biomarkers , Depression/psychology , Disease Susceptibility , Endocannabinoids/metabolism , Humans , Inflammation/complications , Inflammation/etiology , Inflammation/metabolism , Kynurenine/metabolism , Metabolic Networks and Pathways , Serotonin/metabolism , Signal Transduction
7.
Neuropharmacology ; 192: 108612, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34023338

ABSTRACT

Dimethyltryptamine (DMT), an endogenous ligand of sigma-1 receptors (Sig-1Rs), acts against systemic hypoxia, but whether DMT may prevent cerebral ischemic injury is unexplored. Here global forebrain ischemia was created in anesthetized rats and aggravated with the induction of spreading depolarizations (SDs) and subsequent short hypoxia before reperfusion. Drugs (DMT, the selective Sig-1R agonist PRE-084, the Sig-1R antagonist NE-100, or the serotonin receptor antagonist asenapine) were administered intravenously alone or in combination while physiological variables and local field potential from the cerebral cortex was recorded. Neuroprotection and the cellular localization of Sig-1R were evaluated with immunocytochemistry. Plasma and brain DMT content was measured by 2D-LC-HRMS/MS. The affinity of drugs for cerebral Sig-1R was evaluated with a radioligand binding assay. Both DMT and PRE-084 mitigated SDs, counteracted with NE-100. Further, DMT attenuated SD when co-administered with asenapine, compared to asenapine alone. DMT reduced the number of apoptotic and ferroptotic cells and supported astrocyte survival. The binding affinity of DMT to Sig-1R matched previously reported values. Sig-1Rs were associated with the perinuclear cytoplasm of neurons, astrocytes and microglia, and with glial processes. According to these data, DMT may be considered as adjuvant pharmacological therapy in the management of acute cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Cortical Spreading Depression/drug effects , N,N-Dimethyltryptamine/pharmacology , Neurodegenerative Diseases/metabolism , Receptors, sigma/metabolism , Animals , Brain/drug effects , Brain Ischemia/drug therapy , Cortical Spreading Depression/physiology , Dose-Response Relationship, Drug , Male , N,N-Dimethyltryptamine/therapeutic use , Neurodegenerative Diseases/prevention & control , Rats , Rats, Sprague-Dawley , Receptors, sigma/agonists , Sigma-1 Receptor
8.
Int J Mol Sci ; 21(11)2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32516975

ABSTRACT

Inflammatory Bowel Disease (IBD) is an autoimmune ailment of the gastrointestinal (GI) tract, which is characterized by enhanced activation of proinflammatory cytokines. It is suggested that the sigma-1 receptor (σ1R) confers anti-inflammatory effects. As the exact pathogenesis of IBD is still unknown and treatment options are limited, we aimed to investigate the effects of σ1R in 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced experimental colitis. To this end, male Wistar-Harlan rats were used to model colitic inflammation through the administration of TNBS. To investigate the effects of σ1R, Fluvoxamine (FLV, σ1R agonist) and BD1063 (σ1R antagonist) were applied via intracolonic administration to the animals once a day for three days. Our radioligand binding studies indicated the existence of σ1Rs as [3H](+)-pentazocine binding sites, and FLV treatment increased the reduced σ1R maximum binding capacity in TNBS-induced colitis. Furthermore, FLV significantly attenuated the colonic damage, the effect of which was abolished by the administration of BD1063. Additionally, FLV potentially increased the expression of ubiquitin C-terminal hydrolase ligase-1 (UCHL-1) and the levels of endothelial nitric oxide synthase (eNOS), and decreased the levels of interleukin-6 (IL-6) and inducible NOS (iNOS) expression. In summary, our study offers evidence for the anti-inflammatory potential of FLV and σ1R in experimental colitis, and our results present a promising approach to the development of new σ1R-targeted treatment options against IBD.


Subject(s)
Colitis/etiology , Colitis/metabolism , Interleukin-6/metabolism , Receptors, sigma/metabolism , Signal Transduction , Trinitrobenzenesulfonic Acid/adverse effects , Ubiquitin Thiolesterase/metabolism , Animals , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Fluvoxamine/pharmacology , Gene Expression Regulation/drug effects , Heme Oxygenase (Decyclizing)/metabolism , Inflammation Mediators/metabolism , Ligands , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Protein Binding , Rats , Receptors, sigma/agonists , Receptors, sigma/genetics , Severity of Illness Index , Sigma-1 Receptor
9.
Neurochem Int ; 138: 104752, 2020 09.
Article in English | MEDLINE | ID: mdl-32445659

ABSTRACT

Kynurenic acid (KYNA) is one of the most significant metabolite of the kynurenine pathway both in terms of functional and potential therapeutic value. It is an N-methyl-D-aspartate (NMDA) receptor antagonist, but it can also activate the G-protein coupled receptor 35 (GPR35), which shares several structural and functional properties with cannabinoid receptors. Previously our group demonstrated that systemic chronic KYNA treatment altered opioid receptor G-protein activity. Opioid receptors also overlap in many features with cannabinoid receptors. Thus, our aim was to examine the direct in vitro and systemic, chronic in vivo effect of KYNA on type 1 cannabinoid receptor (CB1R) binding and G-protein activity. Based on competition and [35S]GTPγS G-protein binding assays in rat brain, KYNA alone did not show significant binding towards the CB1R, nor did it alter CB1R ligand binding and agonist activity in vitro. When rats were chronically treated with KYNA (single daily, i.p., 128 mg/kg for 9 days), the KYNA plasma and cerebrospinal fluid levels significantly increased compared to vehicle treated group. Furthermore, in G-protein binding assays, in the whole brain the amount of G-proteins in basal and in maximum activity coupled to the CB1R also increased due to the treatment. At the same time, the overall stimulatory properties of the receptor remained unaltered in vehicle and KYNA treated samples. Similar observations were made in rat hippocampus, but not in the cortex and brainstem. In saturation binding assays the density of CB1Rs in rat whole brain and hippocampus were also significantly enhanced after the same treatment, without significantly affecting ligand binding affinity. Thus, KYNA indirectly and brain region specifically increases the abundance of functional CB1Rs, without modifying the overall binding and activity of the receptor. Supposedly, this can be a compensatory mechanism on the part of the endocannabinoid system induced by the long-term KYNA exposure.


Subject(s)
Brain/drug effects , Brain/metabolism , Kynurenic Acid/administration & dosage , Kynurenic Acid/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Benzoxazines/metabolism , Benzoxazines/pharmacology , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Male , Morpholines/metabolism , Morpholines/pharmacology , Naphthalenes/metabolism , Naphthalenes/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
10.
Molecules ; 24(20)2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31619006

ABSTRACT

Schizophrenia, which affects around 1% of the world's population, has been described as a complex set of symptoms triggered by multiple factors. However, the exact background mechanisms remain to be explored, whereas therapeutic agents with excellent effectivity and safety profiles have yet to be developed. Kynurenines and the endocannabinoid system (ECS) play significant roles in both the development and manifestation of schizophrenia, which have been extensively studied and reviewed previously. Accordingly, kynurenines and the ECS share multiple features and mechanisms in schizophrenia, which have yet to be reviewed. Thus, the present study focuses on the main common points and potential interactions between kynurenines and the ECS in schizophrenia, which include (i) the regulation of glutamatergic/dopaminergic/γ-aminobutyric acidergic neurotransmission, (ii) their presence in astrocytes, and (iii) their role in inflammatory mechanisms. Additionally, promising pharmaceutical approaches involving the kynurenine pathway and the ECS will be reviewed herein.


Subject(s)
Endocannabinoids/metabolism , Kynurenine/metabolism , Schizophrenia/etiology , Schizophrenia/metabolism , Animals , Biomarkers , Disease Susceptibility , Humans , Metabolic Networks and Pathways , Molecular Targeted Therapy , Schizophrenia/drug therapy , Synaptic Transmission
11.
Biomolecules ; 9(9)2019 09 16.
Article in English | MEDLINE | ID: mdl-31527522

ABSTRACT

The endocannabinoid system represents an integrated neuronal network involved in the control of several organisms' functions, such as feeding behavior. A series of hybrids of 5-(4-chlorophenyl)-1-(2,4-dichloro-phenyl)-4-methyl-N-(piperidin-1-yl)-1H-pyrazole-3-carboxamide (mimonabant), a well-known inverse agonist of the type-1 cannabinoid receptor (CB1), once used as an antiobesity drug, and the N-(2S)-substitutes of 1-[(4-fluorophenyl)methyl]indazole-3-carboxamide with 1-amino-3-methyl-1-oxobutane (AB-Fubinaca), 1-amino-3,3-dimethyl-1-oxobutane (ADB-Fubinaca), and 3-methylbutanoate (AMB-Fubinaca), endowed with potent agonistic activity towards cannabinoid receptors CB1 and CB2 were in solution as C-terminal amides, acids, methyl esters and N-methyl amides. These compounds have been studied by binding assays to cannabinoid receptors and by functional receptor assays, using rat brain membranes in vitro. The most active among them as an agonist, (S)-1-(2,4-dichlorobenzyl)-N-(3,3-dimethyl-1-(methylamino)-1-oxobutan-2-yl)-1H-indazole-3-carboxamide (LONI11), and an antagonist, (S)-2-(1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxamido)-3-methylbutanoic acid (LONI4), were tested in vivo in mic, to evaluate their ability to stimulate or suppress feeding behavior after intraperitoneal (i.p.) administration. For a LONI11 formalin test and a tail flick test after an administration by the subcutaneous (s.c.) and intracerebroventricular (i.c.v.) routes, respectively, were also carried out in vivo in mice to investigate the antinociceptive property at the central and peripheral levesl. We observed a significant orexant effect for LONI11 and an intense anorexant effect for (S)-methyl 2-(1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxamido)-3,3-dimethylbutanoate (LONI2) and LONI4. In zymosan-induced edema and hyperalgesia, LONI11 reduced the percent of paw volume increase and paw latency after s.c. administration, also suggesting a possible peripheral anti-inflammatory activity.


Subject(s)
Edema/drug therapy , Indazoles/administration & dosage , Indazoles/chemical synthesis , Rimonabant/chemistry , Valine/analogs & derivatives , Animals , Disease Models, Animal , Edema/chemically induced , Feeding Behavior/drug effects , Indazoles/chemistry , Indazoles/pharmacology , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice , Molecular Structure , Rats , Receptors, Cannabinoid/physiology , Valine/chemistry , Zymosan/adverse effects
12.
Eur J Med Chem ; 178: 571-588, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31220675

ABSTRACT

In order to obtain novel pharmacological tools and to investigate a multitargeting analgesic strategy, the CB1 and CB2 cannabinoid receptor agonist JWH-018 was conjugated with the opiate analgesic oxycodone or with an enkephalin related tetrapeptide. The opioid and cannabinoid pharmacophores were coupled via spacers of different length and chemical structure. In vitro radioligand binding experiments confirmed that the resulting bivalent compounds bound both to the opioid and to the cannabinoid receptors with moderate to high affinity. The highest affinity bivalent derivatives 11 and 19 exhibited agonist properties in [35S]GTPγS binding assays. These compounds activated MOR and CB (11 mainly CB2, whereas 19 mainly CB1) receptor-mediated signaling, as it was revealed by experiments using receptor specific antagonists. In rats both 11 and 19 exhibited antiallodynic effect similar to the parent drugs in 20 µg dose at spinal level. These results support the strategy of multitargeting G-protein coupled receptors to develop lead compounds with antinociceptive properties.


Subject(s)
Analgesics, Opioid/pharmacology , Enkephalins/pharmacology , Indoles/pharmacology , Naphthalenes/pharmacology , Oxycodone/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/chemistry , Animals , Dose-Response Relationship, Drug , Enkephalins/chemistry , Indoles/chemistry , Mice , Molecular Structure , Naphthalenes/chemistry , Oxycodone/chemistry , Rats , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
13.
Neuropeptides ; 72: 38-46, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30396596

ABSTRACT

Palatability and variety of foods are major reasons for hedonic eating, and hence for obesity. Hemopressin, a hemoglobin α chain-derived peptide, plays antagonist/inverse agonist role on cannabinoid (CB)1 receptors, while RVD-hemopressin(α)[RVD-hp(α)], a N-terminally extended form of hemopressin, has been reported as an allosteric modulator of CB1 and CB2 receptors. We investigated the effects of 14 daily intraperitoneal injections of RVD-hp(α), in Sprague-Dawley rats fed a highly palatable cafeteria-style (CAF) diet (30% fat, 56% carbohydrate, 14% protein; 4.20 kcal/g) compared to standard laboratory chow (STD) food (3.5% fat, 63% carbohydrate, 14% protein, 19.5% other components without caloric value; 3.20 kcal). Food intake, body weight and locomotor activity were recorded throughout the study. Finally, rats were sacrificed and agouti-related peptide (AgRP), neuropeptide Y (NPY), pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) and fatty acid amide hydrolase (FAAH) gene expression in the hypothalamus was measured by real-time reverse transcription polymerase chain reaction. We found that CAF diet increased food intake as compared to STD diet. In both STD and CAF diet fed rats, RVD-hp(α) treatment inhibited food intake, increased locomotor activity but did not modify body weight. In vehicle injected animals, CAF as compared to STD diet increased AgRP gene expression. RVD-hp(α) treatment decreased POMC mRNA levels in both diet groups and lowered the elevated AgRP levels induced by CAF diet. RVD-hp(α) treatment plays an anorexigenic role paralleled by increased locomotor activity both in STD and CAF diet fed rats. The inhibition of feeding could be partially mediated by lowering of hypothalamic POMC and AgRP gene expression levels.


Subject(s)
Body Weight/drug effects , Diet , Eating/drug effects , Hemoglobins/pharmacology , Hypothalamus/drug effects , Peptide Fragments/pharmacology , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Hypothalamus/metabolism , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley
14.
Amino Acids ; 50(11): 1595-1605, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30145711

ABSTRACT

The discovery of novel modulators of the cannabinoid system is a current topic in medicinal chemistry. In this paper, we report nine novel carboxamides designed as hybrids of Fubinaca family compounds and Rimonabant. These hybrids were obtained by linking the 1-benzyl-2,5-dichloroindazole-3-carboxylic acid to different amino acids bearing a hydrophobic side chain and three different C-terminus. The new chemical entities were tested in vitro to evaluate their bioactivity by means of receptor binding assays and [35S]GTPγS stimulation assays to reveal their affinity and potency. We found that all compounds were able to bind to the cannabinoid receptors in the low nanomolar range with a marked selectivity towards the CB1 cannabinoid receptor. Some of them are full agonists, whereas the others act as partial agonists. These molecules could be potentially used as anti-obesity agents, antiemetic and analgesics.


Subject(s)
Cannabinoid Receptor Antagonists , Piperidines , Pyrazoles , Animals , Cannabinoid Receptor Antagonists/chemical synthesis , Cannabinoid Receptor Antagonists/chemistry , Cannabinoid Receptor Antagonists/pharmacology , Piperidines/chemical synthesis , Piperidines/chemistry , Piperidines/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats , Rats, Wistar , Rimonabant
15.
Pharmacol Rep ; 70(4): 650-657, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29902669

ABSTRACT

BACKGROUND: The endocannabinoid (eCB) system is strongly involved in the regulation of anxiety and feeding behavior. RVD-hemopressin(α) [RVD-hp(α)], a N-terminally extended form of hemopressin, is a negative allosteric modulator of the cannabinoid (CB) 1 receptor and a positive allosteric modulator of CB2 receptor which has been recently reported to exert anxiolytic/antidepressant and anorexigenic effects after peripheral administration in rats. Pharmacological evidences reported a possible link between brain hypocretin/orexin, monoamine and eCB systems, as regards appetite and emotional behavior control. Considering this, the aim of our work was to investigated the effects of RVD-hp(α) on anxiety like behavior and food intake after central administration and related it to monoamine levels and orexin-A gene expression, in the hypothalamus. METHODS: We have studied the effects of central RVD-hp(α) (10nmol) injection on anxiety-like behavior and feeding using different behavioral tests. Hypothalamic levels of norepinephrine (NE), dopamine (DA) and serotonin (5-hydroxytryptamine, 5-HT) and gene expression of orexin-A and proopiomelanocortin (POMC) were measured by high performance liquid chromatography (HPLC) and real-time reverse transcription polymerase chain reaction (RT-PCR) analysis, respectively. RESULTS: Central RVD-hp(α) administration decreased locomotion activity and stereotypies. Moreover, RVD-hp(α) treatment inhibited anxiogenic-like behavior and food intake, NE levels and orexin-A gene expression, in the hypothalamus. CONCLUSION: Concluding, in the present study we demonstrated that central RVD-hp(α) induced anxiolytic and anorexigenic effects possibly related to reduced NE and orexin-A and POMC signaling, in the hypothalamus. These findings further support the central role of the peptide in rat brain thus representing an innovative pharmacological approach for designing new anorexigenic drugs targeting eCB system.


Subject(s)
Exploratory Behavior/drug effects , Feeding Behavior/drug effects , Hemoglobins/pharmacology , Hypothalamus/drug effects , Maze Learning/drug effects , Norepinephrine/metabolism , Orexins/biosynthesis , Peptide Fragments/pharmacology , Animals , Dopamine/metabolism , Hemoglobins/administration & dosage , Hypothalamus/metabolism , Injections, Intraventricular , Male , Motor Activity/drug effects , Peptide Fragments/administration & dosage , Pro-Opiomelanocortin/biosynthesis , Rats , Serotonin/metabolism , Stereotyped Behavior/drug effects
16.
Pharmacol Rep ; 69(6): 1402-1407, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29145068

ABSTRACT

BACKGROUND: Hemopressin, VD-hemopressin(α) and RVD-hemopressin(α) are hemoglobin α chain derived-peptides which have been found in mouse brain, and where they modulate cannabinoid (CB) receptor function. The nonapeptide hemopressin has been reported to inhibit feeding after both central and peripheral administration, possibly playing a role of antagonist/inverse agonist of CB1 receptors, and consequently blocking the orexigenic effects of endogenous cannabinoids. VD-hemopressin(α) and RVD- hemopressin(α), are N-terminal extended forms of hemopressin. VD-hemopressin(α) has CB1 agonist activity, and as such it has been shown to stimulate feeding. RVD-hemopressin(α) is reported to play a negative allosteric modulatory function on CB1 receptors, but there are no data on its possible effects on feeding and metabolic control. METHODS: We have studied, in rats, the effects of 14 daily intraperitoneal (ip) injections of RVD-hemopressin(α) (10nmol). RESULTS: We found that RVD-hemopressin(α) treatment inhibited food intake while total body weight was not affected. The null effect on body weight despite diminished feeding could be related to decreased uncoupling protein 1 (UCP-1) gene expression in brown adipose tissue (BAT). We also investigated the underlying neuromodulatory effects of RVD-hemopressin(α) and found it to down regulate proopiomelanocortin (POMC) gene expression, together with norepinephrine (NE) levels, in the hypothalamus. CONCLUSIONS: In conclusion, RVD-hemopressin(α) administration has an anorectic effect, possibly related to inhibition of POMC and NE levels in the hypothalamus. Despite decreased food intake, body weight is not affected by RVD-hemopressin(α) treatment, possibly due to inhibition of UCP-1 gene expression in BAT.


Subject(s)
Anorexia/chemically induced , Appetite Regulation/drug effects , Eating/drug effects , Hemoglobins/pharmacology , Peptide Fragments/pharmacology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Hemoglobins/administration & dosage , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraperitoneal , Male , Norepinephrine/metabolism , Peptide Fragments/administration & dosage , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Uncoupling Protein 1/genetics
17.
J Pept Sci ; 23(12): 864-870, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29110363

ABSTRACT

Three novel morphiceptin analogs, in which Pro in position 2 and/or 4 was replaced by cis-4-aminoproline connected with the preceding amino acid through the primary amino group, were synthesized. The opioid receptor affinities, functional assay results, enzymatic degradation studies and experimental and in silico structural analysis of such analogs are presented. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Endorphins/chemistry , Peptides/chemical synthesis , Peptides/pharmacology , Receptors, Opioid/metabolism , Animals , Computer Simulation , Humans , Molecular Dynamics Simulation , Peptides/chemistry , Protein Binding , Structure-Activity Relationship
18.
Pharmacol Rep ; 69(6): 1247-1253, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29128806

ABSTRACT

BACKGROUND: The endocannabinoid (eCB) system plays an important role in regulating emotional disorders, and is involved, directly or indirectly, in psychiatric diseases, such as anxiety and depression. Hemopressin, a hemoglobin α chain-derived peptide, and RVD-hemopressin(α), a N-terminally extended form of hemopressin, act as antagonist/inverse agonist and negative allosteric modulator of the cannabinoid 1 (CB1) receptor, respectively. METHODS: Considering the possible involvement of these peptides on emotional behaviour, the aim of our study was to investigate the behavioural effects of a single intraperitoneal (ip) injection of hemopressin (0.05mg/kg) and RVD-hemopressin(α) (0.05mg/kg), using a series of validated behavioural tests (locomotor activity/open field test, light-dark exploration test, forced swim test) in rats. Prefrontal cortex levels of norepinephrine (NE), dopamine (DA) and serotonin (5-hydroxytryptamine, 5-HT) and the gene expression of monoamine oxidase (MAO-B) and catechol-O-methyltransferase (COMT) were measured by high performance liquid chromatography (HPLC) analysis and real-time reverse transcription polymerase chain reaction (RT-PCR), respectively. RESULTS: Hemopressin administration induced anxiogenic and depressive behaviour, decreased monoamine steady state levels in prefrontal cortex, and increased the gene expression of the enzymes involved in their catabolism. By contrast, RVD- hemopressin(α) induced anxiolytic and antidepressive effects, increased monoamines and decreased the enzymes in prefrontal cortex. CONCLUSION: In conclusion, in the present study we demonstrated behavioral effects induced by peripheral hemopressin and RVD-hemopressin(α) injections, that could involve modulatory effects on monoaminergic signaling, in the prefrontal cortex.


Subject(s)
Behavior, Animal/drug effects , Hemoglobins/pharmacology , Peptide Fragments/pharmacology , Prefrontal Cortex/metabolism , Animals , Anxiety/chemically induced , Anxiety/physiopathology , Catechol O-Methyltransferase/metabolism , Chromatography, High Pressure Liquid , Depression/chemically induced , Depression/physiopathology , Dopamine/metabolism , Hemoglobins/administration & dosage , Injections, Intraperitoneal , Male , Norepinephrine/metabolism , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/metabolism
19.
Int J Mol Sci ; 18(8)2017 Jul 30.
Article in English | MEDLINE | ID: mdl-28758944

ABSTRACT

Both the kynurenine and the endocannabinoid systems are involved in several neurological disorders, such as migraine and there are increasing number of reports demonstrating that there are interactions of two systems. Although their cooperation has not yet been implicated in migraine, there are reports suggesting this possibility. Additionally, the individual role of the endocannabinoid and kynurenine system in migraine is reviewed here first, focusing on endocannabinoids, kynurenine metabolites, in particular kynurenic acid. Finally, the function of NMDA and cannabinoid receptors in the trigeminal system-which has a crucial role in the pathomechanisms of migraine-will also be discussed. The interaction of the endocannabinoid and kynurenine system has been demonstrated to be therapeutically relevant in a number of pathological conditions, such as cannabis addiction, psychosis, schizophrenia and epilepsy. Accordingly, the cross-talk of these two systems may imply potential mechanisms related to migraine, and may offer new approaches to manage the treatment of this neurological disorder.


Subject(s)
Endocannabinoids/metabolism , Kynurenine/metabolism , Migraine Disorders/metabolism , Receptors, Cannabinoid/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Humans , Migraine Disorders/therapy
20.
Neurosci Lett ; 633: 87-93, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27639959

ABSTRACT

Schizophrenia is a serious mental health disorder characterized by several behavioral and biochemicel abnormalities. In a previous study we have shown that mu-opioid (MOP) receptor signaling is impaired in specific brain regions of our three-hit animal model of schizophrenia. Since the cannabinoid system is significantly influenced in schizophrenic patients, in the present work we investigated cannabinoid (CB) receptor binding and G-protein activation in cortical, subcortical and cerebellar regions of control and 'schizophrenic' rats. Cannabinoid agonist (WIN-55,212-2 mesylate) mediated G-protein activation was consistently decreased in all areas tested, and the difference was extremely significant in membranes prepared from the cerebellum. Interestingly, the cerebellar activity of WIN-55,212-2 stimulated G-proteins was substantially higher than those of cerebral cortex and subcortical region in control animals, indicating a primordial role of the cannabinoid system in the cerebellum. At the level of radioligand binding, the affinities of the CB receptors were also markedly decreased in the model animals. Capacity of the [3H]WIN-55,212-2 binding was only higher in the cerebellum of 'schizophrenic' model rats. Taken together, in all three brain areas of model rats both cannabinoid receptor binding and cannabinoid agonist-mediated G-protein activation were regularly decreased. Our results revealed that besides the opioids, the endocannabinoid - cannabis receptor system also shows impairment in our rat model, increasing its face validity and translational utility.


Subject(s)
Brain/metabolism , Endocannabinoids/metabolism , Receptors, Cannabinoid/metabolism , Schizophrenia/metabolism , Animals , Benzoxazines/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cerebellum/metabolism , Cerebral Cortex/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Male , Morpholines/pharmacology , Naphthalenes/pharmacology , Radioligand Assay , Rats, Wistar , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...