Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Pharmacol Rep ; 76(1): 112-126, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38236555

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the leading causes of death globally. Multiple factors may contribute to the pathogenesis of CRC, including the abnormalities in the functioning of the endogenous opioid system (EOS) or adiponectin-related signaling. The aim of our study was to evaluate if differences in the expression of opioid receptors (ORs) influence the development of CRC and if modulation of adiponectin receptors using AdipoRon, a selective AdipoR1 receptor agonist, affects colorectal carcinogenesis. METHODS: Naltrexone, an opioid receptor antagonist, was injected intraperitoneally every second day for 2 weeks, at the dose of 1 mg/kg in healthy Balb/C mice to induce changes in ORs expression. CRC was induced by a single intraperitoneal injection of azoxymethane (AOM) and the addition of dextran sodium sulfate (DSS) into drinking water in three-week cycles. The development of CRC was assessed using macro- and microscopic scoring and molecular analysis (RT qPCR, ELISA) after 14 weeks. RESULTS: Naltrexone significantly increased the mRNA expression of Oprm1, Oprd1, and Oprk1 in the mouse colon and in the brain (non-significantly). The pretreatment of mice with naltrexone aggravated the course of CRC (as indicated by tumor area, colon thickness, and spleen weight). The level of circulatory adiponectin was lowered in mice with CRC and increased in the colon as compared with healthy mice. The ß-endorphin level was increased in the plasma of mice with CRC and decreased in the colon as compared to healthy mice. AdipoRon, AdipoR1 agonist, worsened the CRC development, and pretreatment with naltrexone enhanced this negative effect in mice. CRC did not affect the expression of the Adipor1 gene, but the Adipor1 level was increased in mice pretreated with naltrexone (AOM/DSS and healthy mice). AdipoRon did not influence the expression of opioid receptors at the mRNA level in the colon of mice with CRC. The mRNA expression of Ptgs2, Il6, Nos2, Il1b, Il18, Gsdmd, and Rela was increased in mice with CRC as compared to the healthy colon. AdipoRon significantly decreased mRNA expression of Ptgs2, Il6, Il1b, and Il18 as compared to CRC mice. CONCLUSION: EOS and adiponectin-related signaling may play a role in the pathogenesis of CRC and these systems may present some additivity during carcinogenesis.


Subject(s)
Colitis-Associated Neoplasms , Colitis , Colorectal Neoplasms , Mice , Animals , Interleukin-18 , Analgesics, Opioid/adverse effects , Interleukin-6 , Adipokines , Naltrexone/pharmacology , Adiponectin/adverse effects , Cyclooxygenase 2 , Carcinogenesis , Azoxymethane/toxicity , Disease Models, Animal , Receptors, Opioid/genetics , RNA, Messenger , Dextran Sulfate , Colorectal Neoplasms/genetics , Mice, Inbred C57BL , Colitis/chemically induced
2.
Molecules ; 27(19)2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36235308

ABSTRACT

The enteric nervous system (ENS) is a part of the autonomic nervous system that intrinsically innervates the gastrointestinal (GI) tract. Whereas enteric neurons have been deeply studied, the enteric glial cells (EGCs) have received less attention. However, these are immune-competent cells that contribute to the maintenance of the GI tract homeostasis through supporting epithelial integrity, providing neuroprotection, and influencing the GI motor function and sensation. The endogenous cannabinoid system (ECS) includes endogenous classical cannabinoids (anandamide, 2-arachidonoylglycerol), cannabinoid-like ligands (oleoylethanolamide (OEA) and palmitoylethanolamide (PEA)), enzymes involved in their metabolism (FAAH, MAGL, COX-2) and classical (CB1 and CB2) and non-classical (TRPV1, GPR55, PPAR) receptors. The ECS participates in many processes crucial for the proper functioning of the GI tract, in which the EGCs are involved. Thus, the modulation of the EGCs through the ECS might be beneficial to treat some dysfunctions of the GI tract. This review explores the role of EGCs and ECS on the GI tract functions and dysfunctions, and the current knowledge about how EGCs may be modulated by the ECS components, as possible new targets for cannabinoids and cannabinoid-like molecules, particularly those with potential nutraceutical use.


Subject(s)
Cannabinoids , Endocannabinoids , Cannabinoids/metabolism , Cannabinoids/pharmacology , Cyclooxygenase 2 , Dietary Supplements , Endocannabinoids/metabolism , Neuroglia/metabolism , Peroxisome Proliferator-Activated Receptors
3.
Int J Mol Sci ; 23(7)2022 Mar 24.
Article in English | MEDLINE | ID: mdl-35408926

ABSTRACT

Nearly 20% of elderly patients suffer from constipation, but the age-related changes in the gastrointestinal (GI) tract remain insufficiently elucidated. In this study, the alterations within the endogenous opioid system (EOS) as a potential cause of constipation in the elderly were evaluated. The GI functions were assessed in vitro and in vivo and compared between 6-, 12- and 18-month old mice. Moreover, the effect of opioid receptor (MOP, DOP, KOP) agonists on the mouse GI tract functions and the EOS components expression in mouse tissues and colonic biopsies from patients with functional constipation were determined. In the oldest mice, the GI peristalsis was significantly impaired as compared to the younger groups. The tissue response to MOP and DOP, but not KOP, agonists weakened with age in vitro; for DOP, it was confirmed in vivo. In the mouse upper GI tract, Oprm1, Oprd1, Oprk1 expression decreased with age; in the colon, Oprm1 expression increased. There were no differences in the expression of these genes in the colonic biopsies from patients >50 years old as compared to the younger group. In conclusion, the age-related impairment of the GI peristalsis may result from reduced MOP and DOP response to the activation with opioid agonists or the alterations in the EOS expression.


Subject(s)
Analgesics, Opioid , Receptors, Opioid , Aged , Aging/genetics , Analgesics, Opioid/pharmacology , Animals , Constipation , Gastrointestinal Tract/metabolism , Humans , Mice , Opioid Peptides , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , Receptors, Opioid, mu/metabolism
4.
J Integr Neurosci ; 21(1): 1, 2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35164437

ABSTRACT

A nutraceutical is a food-derived molecule that provides medical or health benefits beyond its basic nutritional role, including the prevention and treatment of disease and its symptoms. In the peripheral nervous system, satellite glial cells are found in close relationship with neurons, mainly in peripheral sensory ganglia, but, compared with other glial cells, the relationship between these cells and nutraceuticals has received little attention. After describing satellite glial cells and their role and changes in physiology and pathology, we review the studies on the effects of nutraceuticals as modulators of their functions. Maybe due to the difficulties in selectively labeling these cells, only a few studies, performed mainly in rodent models, have analyzed nutraceutical effects, showing that N-acetylcysteine, curcumin, quercetin, osthole and resveratrol may palliate neuropathic pain through satellite glial cells-dependent pathways, namely antioxidant mechanisms and/or interference with purinergic signaling. Neither other conditions in which satellite glial cells are involved (visceral pain, nerve regeneration) nor other nutraceuticals or mechanisms of action have been studied. Although more preclinical and clinical research is needed, the available reports support the general notion that nutraceuticals may become interesting alternatives in the prevention and/or treatment of peripheral gliopathies and their associated conditions, including those affecting the satellite glial cells.


Subject(s)
Curcumin/therapeutic use , Dietary Supplements , Neuroglia/drug effects , Peripheral Nervous System Diseases/therapy , Quercetin/therapeutic use , Resveratrol/therapeutic use , Animals , Humans
5.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G66-G78, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34755545

ABSTRACT

Allosteric modulators (AMs) are molecules that can fine-tune signaling by G protein-coupled receptors (GPCRs). Although they are a promising therapeutic approach for treating a range of disorders, allosteric modulation of GPCRs in the context of the enteric nervous system (ENS) and digestive dysfunction remains largely unexplored. This study examined allosteric modulation of the delta opioid receptor (DOR) in the ENS and assessed the suitability of DOR AMs for the treatment of irritable bowel syndrome (IBS) symptoms using mouse models. The effects of the positive allosteric modulator (PAM) of DOR, BMS-986187, on neurogenic contractions of the mouse colon and on DOR internalization in enteric neurons were quantified. The ability of BMS-986187 to influence colonic motility was assessed both in vitro and in vivo. BMS-986187 displayed DOR-selective PAM-agonist activity and orthosteric agonist probe dependence in the mouse colon. BMS-986187 augmented the inhibitory effects of DOR agonists on neurogenic contractions and enhanced reflex-evoked DOR internalization in myenteric neurons. BMS-986187 significantly increased DOR endocytosis in myenteric neurons in response to the weakly internalizing agonist ARM390. BMS-986187 reduced the generation of complex motor patterns in the isolated intact colon. BMS-986187 reduced fecal output and diarrhea onset in the novel environment stress and castor oil models of IBS symptoms, respectively. DOR PAMs enhance DOR-mediated signaling in the ENS and have potential benefit for the treatment of dysmotility. This study provides proof of concept to support the use of GPCR AMs for the treatment of gastrointestinal motility disorders.NEW & NOTEWORTHY This study assesses the use of positive allosteric modulation as a pharmacological approach to enhance opioid receptor signaling in the enteric nervous system. We demonstrate that selective modulation of endogenous delta opioid receptor signaling can suppress colonic motility without causing constipation. We propose that allosteric modulation of opioid receptor signaling may be a therapeutic strategy to normalize gastrointestinal motility in conditions such as irritable bowel syndrome.


Subject(s)
Enteric Nervous System/drug effects , Gastrointestinal Motility/drug effects , Receptors, Opioid, delta/drug effects , Xanthones/pharmacology , Analgesics, Opioid/pharmacology , Benzamides/pharmacology , Colon/drug effects , Enteric Nervous System/physiopathology , Gastrointestinal Motility/physiology , Humans , Receptors, Opioid/drug effects , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/drug effects , Signal Transduction/drug effects
6.
Postepy Biochem ; 67(1): 34-43, 2021 03 31.
Article in English | MEDLINE | ID: mdl-34378903

ABSTRACT

Constipation occurs more often in old patients, because the intestinal peristalsis decreases with aging. Constipation is caused due to impaired motility of the intestines, intestinal barrier damage and the imbalance between the absorption and secretion of water and electrolytes, as well as disturbed production and release of intestinal hormones, infiltration of the gastrointestinal tract with immune cells, excessive production of pro-inflammatory cytokines and the alterations in the functions of enteric nervous system. In this review we will discuss the most important issues associated with the process of aging of the digestive tract, focusing on the enteric nervous system.


Subject(s)
Enteric Nervous System , Peristalsis , Aging , Gastrointestinal Tract , Humans
7.
Molecules ; 26(12)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205534

ABSTRACT

Until recently, glia were considered to be a structural support for neurons, however further investigations showed that glial cells are equally as important as neurons. Among many different types of glia, enteric glial cells (EGCs) found in the gastrointestinal tract, have been significantly underestimated, but proved to play an essential role in neuroprotection, immune system modulation and many other functions. They are also said to be remarkably altered in different physiopathological conditions. A nutraceutical is defined as any food substance or part of a food that provides medical or health benefits, including prevention and treatment of the disease. Following the description of these interesting peripheral glial cells and highlighting their role in physiological and pathological changes, this article reviews all the studies on the effects of nutraceuticals as modulators of their functions. Currently there are only a few studies available concerning the effects of nutraceuticals on EGCs. Most of them evaluated molecules with antioxidant properties in systemic conditions, whereas only a few studies have been performed using models of gastrointestinal disorders. Despite the scarcity of studies on the topic, all agree that nutraceuticals have the potential to be an interesting alternative in the prevention and/or treatment of enteric gliopathies (of systemic or local etiology) and their associated gastrointestinal conditions.


Subject(s)
Enteric Nervous System/drug effects , Neuroglia/drug effects , Animals , Antioxidants/pharmacology , Dietary Supplements , Gastrointestinal Diseases/drug therapy , Gastrointestinal Tract/drug effects , Humans , Neurons/drug effects
8.
Pharmacol Rep ; 73(4): 1147-1154, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34133018

ABSTRACT

INTRODUCTION: Tolerance (TOL) and physical dependence (PD) constitute important limitations of opioid therapy. The aim of our study was to validate research tools to investigate TOL and PD and to characterize the interactions between opioid (OR) and cannabinoid (CB) receptors in these processes in the GI tract. METHODS: TOL was assessed through the comparison of morphine ability to inhibit electrically evoked smooth muscles contractility in the mouse ileum that was previously incubated with/without morphine for 1 h. To evaluate the PD, the ileum was incubated with morphine for 10 min, then challenged with naloxone to induce withdrawal response (WR). The OR/CB interactions were evaluated using mixed agonist (PR-38) and AM-251 (CB1 antagonist). RESULTS: The inhibitory effect of morphine on ileal contractions was weaker in tissue incubated with this opioid than in tissue incubated without opioid. The opposite was noted for PR-38. In tissues exposed to morphine, but not to PR-38, naloxone induced a WR. The blockage of CB1 receptors with AM-251 before the addition of PR-38 resulted in a naloxone-induced WR. CONCLUSION: The co-activation of OR and CB reduced development of TOL and PD to opioids in the mouse GI tract and mixed OR/CB agonists are promising alternative to currently used opioid drugs.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance/physiology , Ileum/drug effects , Ileum/metabolism , Receptors, Cannabinoid/metabolism , Animals , Cannabinoids/metabolism , Diterpenes, Clerodane/pharmacology , Male , Mice , Mice, Inbred BALB C , Morphine/pharmacology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Naloxone/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Substance Withdrawal Syndrome/metabolism
9.
Biochim Biophys Acta Rev Cancer ; 1875(1): 188460, 2021 01.
Article in English | MEDLINE | ID: mdl-33184028

ABSTRACT

Colorectal cancer (CRC) is one of the most common cause of death among neoplasms around the world. The environmental factors, like diet and obesity, are crucial in CRC pathogenesis by creating cancer-favorable microenvironment and hormonal changes. Adiponectin, the adipose tissue-specific hormone, is generally considered to negatively correlate with CRC development. The interleukin 6 (IL-6) is one of the most important pro-inflammatory cytokine connected with CRC, which is strongly inflammation-associated. The opioids are variable group substantially correlated with cancers - the endogenous opioids affect immune system and cell cycle including proliferation and cell death whereas exogenous opioids are leading clinically used analgesics in terminal cancer patients. In this review we discuss the involvement of adiponectin, IL-6 and opioids in CRC pathogenesis, their link with obesity, possible cross-talk and potential novel therapeutic approach in CRC treatment.


Subject(s)
Adiponectin/genetics , Analgesics, Opioid/metabolism , Colorectal Neoplasms/genetics , Interleukin-6/genetics , Carcinogenesis/genetics , Colorectal Neoplasms/pathology , Humans , Signal Transduction/genetics
10.
Postepy Biochem ; 66(1): 42-48, 2020 03 31.
Article in Polish | MEDLINE | ID: mdl-33320478

ABSTRACT

Sphingosine-1-phosphate (S1P) belongs to the group of biologically active sphingolipids. Because of its ability to regulate the migration of lymphocytes, S1P constitutes an important element of pathophysiology of several diseases, such as: lupus erythematosus, multiple sclerosis or inflammatory bowel diseases. Inflammatory bowel diseases (IBD) are the group of chronic and recurrent diseases of the gastrointestinal tract. The most common among IBD are: Crohn's disease and ulcerative colitis. Drugs that are currently used in the therapy of IBD alleviate symptoms, improve patients' quality of life and induce remission but their efficacy is not satisfactory. Modulators of S1P receptors constitu­te an emerging option in the therapy of IBD. In this review we will discuss the role of S1P, its receptor and enzymes that participate in the metabolism of S1P under physiological conditions and in the course of IBD. Moreover, we will sum up the results of preclinical and clinical studies on S1P receptors modulators in IBD.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Crohn Disease/drug therapy , Crohn Disease/metabolism , Humans , Quality of Life , Sphingosine/metabolism
11.
Eur J Pharmacol ; 885: 173463, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32835668

ABSTRACT

Endogenous opioid system is involved in the maintenance of the intestinal homeostasis. Recently, we proved that stimulation of opioid receptors using P-317, a cyclic morphiceptin analog, resulted in the alleviation of acute colitis in mice. The aim of the current study was to assess the effect of P-317 during colitis and colitis-associated colorectal cancer in mice. Colitis was induced by addition of dextran sodium sulfate (DSS) into drinking water. Colitis-associated colorectal cancer was induced by a single intraperitoneal injection of azoxymethane (AOM) and subsequent addition of DSS into drinking water (week 2, 5, 8). During macroscopic damage evaluation the samples were collected and used for biochemical (MPO activity assay), molecular (qPCR and western blot) and histological studies. In experimental colitis, P-317 induced an anti-inflammatory response as indicated by macroscopic and microscopic scores. In the colitis-associated colorectal cancer model, a significant difference in colorectal tumor development was observed between vehicle- and P-317-treated mice. P-317 decreased the total number of colonic tumors and inhibited MPO activity. Hematoxylin and eosin staining confirmed anti-tumor activity of P-317. The expression of TNF-α was decreased in P-317-treated mice as compared to the vehicle-treated group. P-317 decreased proliferation as well as ß-catenin expression in tumors. P-317, a mixed MOP and KOP receptor agonist, induced an anti-inflammatory response in experimental colitis and decreased tumor development in colitis-associated colorectal cancer in mice.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Colitis/drug therapy , Colorectal Neoplasms/drug therapy , Endorphins/pharmacology , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/therapeutic use , Azoxymethane , Carcinogens , Cell Proliferation/drug effects , Colitis/chemically induced , Colitis/complications , Colorectal Neoplasms/etiology , Dextran Sulfate , Endorphins/therapeutic use , Male , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/antagonists & inhibitors
12.
Pharmacol Rep ; 72(2): 314-321, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32125684

ABSTRACT

BACKGROUND: Irritable bowel syndrome (IBS) is a chronic condition with recurring gastrointestinal (GI) symptoms: altered motility and abdominal pain. As endogenous opioid system participates in pain perception and in the control of GI peristalsis, opioids have been proposed as a promising therapy in IBS. In a previous study, we observed that morphiceptin derivative, P-317 (Dmt-cyclo-(D-Lys-Phe-D-Pro-Asp)-NH2), presents promising features to be applied in IBS. In this project, we tested whether modifications in cyclic morphiceptin-based structure: fluorination (compound 1) or peptide bond reduction (compound 2) improve pharmacological effect. METHODS: We evaluated tested derivatives in the mouse GI system under physiological (GI transit) and pathophysiological (castor oil diarrhea, stress-induced hypermotility, visceral pain) conditions. RESULTS: Both compounds prolonged GI transit. Compound 1 and P-317 inhibited upper GI transit and motility of the colon; compound 2 remained inactive. Compound 1 and P-317 inhibited hypermotility in stressed mice and delayed the acute diarrhea in comparison to control. Only P-317 exerted antinociceptive effect. None of tested derivatives, similar to P-317, affected locomotor activity. CONCLUSIONS: Compound 1 is equally effective as P-317 in the mouse GI tract. The peptide bond reduction decreased the activity of compound 2. Fluorination appears to be an efficient way to increase the effects of morphiceptin analogs in the GI tract.


Subject(s)
Abdominal Pain/drug therapy , Endorphins/pharmacology , Gastrointestinal Tract/drug effects , Gastrointestinal Transit/drug effects , Peptides, Cyclic/pharmacology , Abdominal Pain/physiopathology , Animals , Endorphins/chemistry , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Peptides, Cyclic/chemistry , Structure-Activity Relationship
13.
Naunyn Schmiedebergs Arch Pharmacol ; 393(8): 1357-1364, 2020 08.
Article in English | MEDLINE | ID: mdl-32002574

ABSTRACT

TRPV1 are involved in the control of the gastrointestinal (GI) functions and pain sensation. Their activation induces pain but it is followed by desensitization, which in turn causes analgesia. The studies from the last two decades indicate that TRPV1 are involved in visceral hypersensitivity in the GI tract and pathogenesis of irritable bowel syndrome (IBS). Therefore, the aim of this study is to assess the action of fast desensitizing agonist of TRPV1, palvanil (N-palmitoyl-vanillamine), in the murine GI tract and on nociception to evaluate its potential application in the therapy of IBS. The effect of palvanil on smooth muscle contractility was evaluated using organ baths. The impact of palvanil on intestinal secretion was assessed in Ussing chambers. In vivo, the action of palvanil (0.1-1 mg/kg) was assessed in whole GI transit, fecal pellet output, and colonic bead expulsion tests. The antinociceptive potency of palvanil was tested in the mustard oil-induced pain test. Palvanil inhibited colonic contractions (evoked by electrical field stimulation, EFS) and decreased the ion transport in the colon stimulated with forskolin. It did not affect secretion in experiments with veratridine. In vivo, palvanil prolonged whole GI transit at all doses tested. At the lower dose tested, it accelerated colonic motility during first 60 min following injection. By contrast, at the dose of 1 mg/kg, colonic motility was inhibited. Palvanil induced antinociceptive action at all tested doses in mustard oil-induced pain test. TRPV1 fast-desensitizing compounds, i.e., palvanil, may be promising agents in the therapy of IBS since it modulates intestinal motility and reduces visceral pain.


Subject(s)
Abdominal Pain/prevention & control , Analgesics/pharmacology , Capsaicin/analogs & derivatives , Colon/drug effects , Gastrointestinal Motility/drug effects , Irritable Bowel Syndrome/drug therapy , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/drug effects , Abdominal Pain/chemically induced , Abdominal Pain/physiopathology , Animals , Behavior, Animal/drug effects , Capsaicin/pharmacology , Colon/metabolism , Colon/physiopathology , Disease Models, Animal , In Vitro Techniques , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/physiopathology , Male , Mice, Inbred BALB C , Mustard Plant , Plant Oils , Time Factors
14.
Neurogastroenterol Motil ; 31(4): e13526, 2019 04.
Article in English | MEDLINE | ID: mdl-30549162

ABSTRACT

The endogenous opioid system (EOS) is considered being a crucial element involved in the pathophysiology of irritable bowel syndrome (IBS) as it regulates gastrointestinal (GI) homeostasis through modulation of motility and water and ion secretion/absorption. Along with opioid receptors (ORs), the following components of EOS can be distinguished: 1. endogenous opioid peptides (EOPs), namely enkephalins, endorphins, endomorphins and dynorphins, and 2. peptidases, which regulate the metabolism (synthesis and degradation) of EOPs. Enkephalins, which are δ-opioid receptors agonists, induce significant effects in the GI tract as they act as potent pro-absorptive neurotransmitters. The action of enkephalins and other EOPs is limited, since EOPs are easily and rapidly inactivated by a natural metalloendopeptidase (enkephalinase/neprilysin) and aminopeptidase N. Studies show that the activity of EOPs can be enhanced by inhibition of these enzymes. In this review, we discuss the antidiarrheal and antinociceptive potential of enkephalinase inhibitors. Furthermore, our review is to answer the question whether enkephalinase inhibitors may be helpful in the future treatment of diarrhea predominant functional GI disorders.


Subject(s)
Diarrhea/drug therapy , Enzyme Inhibitors/therapeutic use , Gastrointestinal Diseases/drug therapy , Neprilysin/antagonists & inhibitors , Diarrhea/metabolism , Gastrointestinal Diseases/metabolism , Humans , Opioid Peptides/metabolism
15.
Peptides ; 105: 51-57, 2018 07.
Article in English | MEDLINE | ID: mdl-29684591

ABSTRACT

Morphine and related drugs, which are the most effective analgesics for the relief of severe pain, act through activating opioid receptors. The endogenous ligands of these receptors are opioid peptides which cannot be used as antinociceptive agents due to their low bioactivity and stability in biological fluids. The major goal of opioid research is to understand the mechanism of action of opioid receptor agonists in order to improve therapeutic utility of opioids. Analgesic effects of morphine are mediated mostly through activation of the mu opioid receptor. However, in the search for safer and more effective drug candidates, analogs with mixed opioid receptor profile gained a lot of interest. Recently, the concept of biased agonists able to differentially activate GPCR downstream pathways, became a new approach in the design of novel drug candidates. It is hypothesized that compounds promoting G-protein signaling may produce analgesia while ß-arrestin recruitment may be responsible for opioid side effects. In this report we showed that replacement of the tyrosine residue in the mu-selective ligand Tyr-c[d-Lys-Phe-Asp]NH2 with 2',6'-dimethyltyrosine (Dmt) produced a cyclopeptide Dmt-c[d-Lys-Phe-Asp]NH2 with mu/delta opioid receptor agonist profile. This analog showed improved antinociception in the hot-plate test, probably due to the simultaneous activation of mu and delta receptors but also significantly inhibited the gastrointestinal transit. Using the bioluminescence resonance energy transfer (BRET) assay it was shown that this analog was a mu receptor agonist biased toward ß-arrestin. ß-Arrestin-dependent signaling is most likely responsible for the observed inhibition of gastrointestinal motility exerted by the novel cyclopeptide.


Subject(s)
Pain/drug therapy , Peptides, Cyclic/chemistry , Receptors, Opioid, mu/metabolism , beta-Arrestins/metabolism , Analgesics/administration & dosage , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics, Opioid/administration & dosage , Animals , Ligands , Mice , Morphine/administration & dosage , Pain/metabolism , Pain/pathology , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemical synthesis , Receptors, Opioid, mu/agonists , Structure-Activity Relationship , beta-Arrestins/genetics
16.
J Enzyme Inhib Med Chem ; 33(1): 560-566, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29513114

ABSTRACT

Opioid peptides and opiate drugs such as morphine, mediate their analgesic effects, but also undesired side effects, mostly through activation of the mu opioid receptor. However, delta- and kappa-opioid receptors can also contribute to the analgesic effects of opioids. Recent findings showed that simultaneous activation of multiple opioid receptors may result in additional analgesia with fewer side effects. Here, we evaluated the pharmacological profile of our formerly developed mixed mu/kappa-opioid receptor ligands, Dmt-c[D-Lys-Phe-Phe-Asp]NH2 (C-36) and Dmt-c[D-Lys-Phe-p-CF3-Phe-Asp]NH2 (F-81). The ability of these peptides to cross the blood-brain barrier was tested in the parallel artificial membrane permeability (PAMPA) assay. On the basis of the hot-plate test in mice after central and peripheral administration, analog F-81 was selected for the anti-nociceptive and anti-inflammatory activity assessment after peripheral administration.


Subject(s)
Analgesics, Opioid/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Pain/drug therapy , Peptides, Cyclic/pharmacology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Colitis/drug therapy , Colitis/pathology , Dose-Response Relationship, Drug , Halogenation , Male , Mice , Molecular Structure , Mustard Plant , Pain/chemically induced , Pain/pathology , Pain Measurement , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , Plant Oils , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
17.
Naunyn Schmiedebergs Arch Pharmacol ; 391(4): 435-444, 2018 04.
Article in English | MEDLINE | ID: mdl-29404698

ABSTRACT

Cannabinoid-1 (CB1) receptors are broadly distributed in the central and peripheral nervous systems; among others, they are located in the enteric nervous system. In the gastrointestinal (GI) system, they participate in regulation of intestinal motility or ion transport. The aim of our study was to assess the effect of 1,2,3-triazole derivatives (compound 1: 2-[4,5-bis(2,4-dichlorophenyl)-2H-1,2,3-triazol-2-yl]-N-(2-fluorobenzyl)acetamide, compound 2: 2-[4,5-bis(2,4-dichlorophenyl)-2H-1,2,3-triazol-2-yl]-N-(4-fluorobenzyl)acetamide, compound 3: N-benzyl-2-[4-(4-chlorophenyl)-5-(2,4-dichlorophenyl)-2H-1,2,3-triazol-2-yl]acetamide]), characterized in vitro as CB1 antagonists with high CB1 over CB2 selectivity, in the mouse GI tract. The action of compounds 1-3 was assessed in vitro (electrical field stimulated smooth muscle contractility of the mouse ileum and colon) and in vivo (whole GI transit time). Compound 1 decreased ileal (10-6 M) and colonic (10-7-10-6 M) smooth muscles contractility. Moreover, it prolonged whole GI transit. Compound 2 (10-10-10-8 M) slightly increased the amplitude of muscle contractions in the ileum, but at a higher concentration (10-6 M), the amplitude was decreased. Compound 2 reduced colonic contractility but accelerated GI transit. Compound 3 decreased the amplitude of intestinal muscle contractions in the ileum (10-6 M) and colon (10-10-10-6 M). Moreover, it increased the GI transit time in vivo. Triazole derivatives possess easily modifiable structure and interesting pharmacological action in the GI tract; further, alterations may enhance their efficacy at CB receptors and provide low side effect profile in clinical conditions.


Subject(s)
Cannabinoid Receptor Antagonists/pharmacology , Gastrointestinal Motility/drug effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Triazoles/pharmacology , Animals , Colon/drug effects , Colon/physiology , Ileum/drug effects , Ileum/physiology , In Vitro Techniques , Ligands , Male , Mice , Muscle, Smooth/drug effects , Muscle, Smooth/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...