Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 365
Filter
Add more filters

Publication year range
1.
J Pain ; 22(4): 440-453, 2021 04.
Article in English | MEDLINE | ID: mdl-33227509

ABSTRACT

Oral amitriptyline hydrochloride (amitriptyline) is ineffective against some forms of chronic pain and is often associated with dose-limiting adverse events. We evaluated the potential effectiveness of high-dose topical amitriptyline in a preliminary case series of chemotherapy-induced peripheral neuropathy patients and investigated whether local or systemic adverse events associated with the use of amitriptyline were present in these patients. We also investigated the mechanism of action of topically administered amitriptyline in mice. Our case series suggested that topical 10% amitriptyline treatment was associated with pain relief in chemotherapy-induced peripheral neuropathy patients, without the side effects associated with systemic absorption. Topical amitriptyline significantly increased mechanical withdrawal thresholds when applied to the hind paw of mice, and inhibited the firing responses of C-, Aß- and Aδ-type peripheral nerve fibers in ex vivo skin-saphenous nerve preparations. Whole-cell patch-clamp recordings on cultured sensory neurons revealed that amitriptyline was a potent inhibitor of the main voltage-gated sodium channels (Nav1.7, Nav1.8, and Nav1.9) found in nociceptors. Calcium imaging showed that amitriptyline activated the transient receptor potential cation channel, TRPA1. Our case series indicated that high-dose 10% topical amitriptyline could alleviate neuropathic pain without adverse local or systemic effects. This analgesic action appeared to be mediated through local inhibition of voltage-gated sodium channels. PERSPECTIVE: Our preliminary case series suggested that topical amitriptyline could provide effective pain relief for chemotherapy-induced peripheral neuropathy patients without any systemic or local adverse events. Investigation of the mechanism of this analgesic action in mice revealed that this activity was mediated through local inhibition of nociceptor Nav channels.


Subject(s)
Amitriptyline/pharmacology , Analgesics, Non-Narcotic/pharmacology , Antineoplastic Agents/adverse effects , Nociceptive Pain/drug therapy , Nociceptors/drug effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , TRPA1 Cation Channel/drug effects , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/drug effects , Administration, Topical , Adolescent , Adult , Aged , Aged, 80 and over , Amitriptyline/administration & dosage , Amitriptyline/adverse effects , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/adverse effects , Animals , Behavior, Animal/drug effects , Child , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , NAV1.7 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel , NAV1.9 Voltage-Gated Sodium Channel , Voltage-Gated Sodium Channel Blockers/administration & dosage , Voltage-Gated Sodium Channel Blockers/adverse effects , Young Adult
2.
Viruses ; 12(11)2020 11 10.
Article in English | MEDLINE | ID: mdl-33182673

ABSTRACT

Dengue is an acute viral disease caused by Dengue virus (DENV) and is considered to be the most common arbovirus worldwide. The clinical characteristics of dengue may vary from asymptomatic to severe complications and severe organ impairment, particularly affecting the liver. Dengue treatment is palliative with acetaminophen (APAP), usually known as Paracetamol, being the most used drug aiming to relieve the mild symptoms of dengue. APAP is a safe and effective drug but, like dengue, can trigger the development of liver disorders. Given this scenario, it is necessary to investigate the effects of combining these two factors on hepatocyte homeostasis. Therefore, this study aimed to evaluate the molecular changes in hepatocytes resulting from the association between DENV infection and treatment with sub-toxic APAP concentrations. Using an in vitro experimental model of DENV-2 infected hepatocytes (AML-12 cells) treated with APAP, we evaluated the influence of the virus and drug association on the transcriptome of these hepatocytes by RNA sequencing (RNAseq). The virus-drug association was able to induce changes in the gene expression profile of AML-12 cells and here we highlight and explore these changes and its putative influence on biological processes for cellular homeostasis.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Dengue Virus/drug effects , Hepatocytes/drug effects , Hepatocytes/virology , Host Microbial Interactions , Transcriptome , Animals , Cell Line , Homeostasis/drug effects , Host Microbial Interactions/drug effects , Host Microbial Interactions/genetics , Liver/cytology , Liver/drug effects , Liver/virology , Mice , Sequence Analysis, RNA , Virus Replication/drug effects
3.
Phytomedicine ; 78: 153307, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32846405

ABSTRACT

BACKGROUND: Current therapies for neuropathic pain are generally symptomatic and possess several side effects, limiting their prolonged usage. HYPOTHESIS/PURPOSE: Thus, it is urgent to develop novel and safe candidates for the management of this chronical condition. For this purpose, we investigated the analgesic effect of a standardized extract from Zingiber officinale Roscoe rhizomes (ZOE) obtained by CO2 supercritical extraction, in a mice model of peripheral neuropathy. We also explored the mechanism of action of ZOE and its main constituents using an in vitro model of neuroinflammation. METHODS: Peripheral mono-neuropathy was induced in mice, by spared nerve injury (SNI). The analgesic effect of ZOE after oral administration was assessed by measuring mechanical and thermal allodynia in SNI mice. The mechanism of action of ZOE and its main constituents were investigated using spinal cords samples and in an in vitro model of neuroinflammation by ELISA, western blotting and immunofluorescence techniques. RESULTS: Oral administration of ZOE 200 mg kg-1 ameliorated mechanical and thermal allodynia in SNI mice, with a rapid and a long-lasting effect. ZOE did not alter locomotor activity. In BV2 cells and spinal cord samples, ZOE, 6-gingerol and 6-shogaol reduced pERK levels, whereas ZOE and terpene fraction reduced HDAC1 protein levels, inhibited NF-κB signalling activation and decreased IL-1ß, TNF-α and IL-6 release. ZOE and each tested constituent had a positive effect on inflammation-impaired SH-SY5Y cell viability. CONCLUSIONS: The oral administration of ZOE attenuated SNI-induced neuropathic pain symptoms by reducing spinal neuroinflammation, suggesting ZOE as a novel and interesting candidate for the management of neuropathic pain.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Neuralgia/drug therapy , Plant Extracts/chemistry , Plant Extracts/pharmacology , Zingiber officinale/chemistry , Administration, Oral , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/chemistry , Animals , Cytokines/metabolism , Disease Models, Animal , Histone Deacetylase 1/metabolism , Hyperalgesia/drug therapy , Inflammation/drug therapy , Locomotion/drug effects , Male , Mice, Inbred Strains , Neuralgia/metabolism , Plant Extracts/administration & dosage , Rhizome/chemistry , Spinal Cord/drug effects
4.
Phytomedicine ; 67: 153166, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31955133

ABSTRACT

BACKGROUND: Peripheral nerve injury can produce chronic and ultimately neuropathic pain. The chronic constriction injury (CCI) model has provided a deeper understanding of nociception and chronic pain. Loganin is a well-known herbal medicine with glucose-lowering action and neuroprotective activity. PURPOSE: This study investigated the molecular mechanisms by which loganin reduced CCI-induced neuropathic pain. METHODS: Sprague-Dawley rats were randomly divided into four groups: sham, sham+loganin, CCI and CCI+loganin. Loganin (1 or 5 mg/kg/day) was injected intraperitoneally once daily for 14 days, starting the day after CCI. For behavioral testing, mechanical and thermal responses were assessed before surgery and on d1, d3, d7 and d14 after surgery. Sciatic nerves (SNs) were collected to measure proinflammatory cytokines. Proximal and distal SNs were collected separately for Western blotting and immunofluorescence studies. RESULTS: Thermal hyperalgesia and mechanical allodynia were reduced in the loganin-treated group as compared to the CCI group. Loganin (5 mg/kg/day) prevented CCI from inducing proinflammatory cytokines (TNF-α, IL-1ß), inflammatory proteins (TNF-α, IL-1ß, pNFκB, pIκB/IκB, iNOS) and receptor (TNFR1, IL-1R), adaptor protein (TRAF2) of TNF-α, and Schwann cell demyelination and axonal damage. Loganin also blocked IκB phosphorylation (p-IκB). Double immunofluorescent staining further demonstrated that pNFκB/pIκB protein was reduced by loganin in Schwann cells on d7 after CCI. In the distal stumps of injured SN, Schwann cell demyelination was correlated with pain behaviors in CCI rats. CONCLUSION: Our findings indicate that loganin improves CCI-induced neuroinflammation and pain behavior by downregulating TNF-α/IL-1ß-dependent NF-κB activation.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Iridoids/pharmacology , NF-kappa B/metabolism , Neuralgia/drug therapy , Schwann Cells/drug effects , Animals , Chronic Pain/drug therapy , Chronic Pain/metabolism , Chronic Pain/pathology , Constriction , Cytokines/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Hyperalgesia/pathology , Interleukin-1beta/metabolism , Male , Neuralgia/metabolism , Neuralgia/pathology , Rats, Sprague-Dawley , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Pak J Pharm Sci ; 33(4(Supplementary)): 1899-1906, 2020 Jul.
Article in English | MEDLINE | ID: mdl-33612475

ABSTRACT

Aim of this study to evaluate the safety profile, hepatoprotective and in-vivo antioxidant activities of Dicliptera bupleuroides Nees. Toxicity studies were conducted in human RBCs and DNA by using standard procedures. Acute hepatoprotective investigation was carried out in albino rats by treated with all six fractions of D. bupleuroides 350 mg/kg/day. ALT, AST, ALP and total bilirubin (TB) were performed. The n-hexane fraction (200 mg/kg/day) exhibited appropriate hepatoprotective activity hence subjected to chronic study (14 days). Paracetamol induced the hepatotoxicity (350mg/kg) and silymarin (50 mg/kg) was standard drug. Liver function tests, liver peroxidation tests and histopathological examination were performed at the end. Hexane fraction showed significant decrease in the level of ALT (88.1±7.8), AST (93.8±7.6), ALP (136.3±8.4) and TB (0.6±0.03) as compared to the standard drug (p>0.05). Rats treated with ethyl acetate fraction showed decrease in MDA (42.8±0.7) while GSH was found to be increased (107.7±1.8) against the toxic group (51.3±2.9), (73.6±4.0) respectively. All the drug extracts decreased the oxidative stress and protect the DNA from free hydroxyl radicals. DNA damage protection activity of these fractions is due to phytochemicals present in these fractions. These results indicate that the plant fractions possess significant hepatoprotective and antioxidant activities with no toxic effects.


Subject(s)
Acanthaceae/chemistry , Acetaminophen/pharmacology , Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Liver/drug effects , Plant Extracts/pharmacology , Protective Agents/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Carbon Tetrachloride/pharmacology , Female , Humans , Liver Function Tests/methods , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Silymarin/pharmacology
6.
Pharmacol Res ; 151: 104549, 2020 01.
Article in English | MEDLINE | ID: mdl-31743775

ABSTRACT

We now appreciate that the mechanism of resolution depends on an active and time-dependent biosynthetic shift from pro-inflammatory to pro-resolution mediators, the so-called specialized pro-resolving lipid mediators (SPMs). These SPMs are biosynthesized from the omega-3 fatty acids arachidonic acid (AA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), or docosahexaenoic acid (DHA). Despite effective for a fraction of patients with rheumatic diseases and neuropathic pain, current analgesic therapies such as biological agents, opioids, corticoids, and gabapentinoids cause unwanted side effects, such as immunosuppression, addiction, or induce analgesic tolerance. A growing body of evidence demonstrates that isolated SPMs show efficacy at very low doses and have been successively used as therapeutic drugs to treat pain and infection in experimental models showing no side effects. Moreover, SPMs work as immunoresolvents and some of them present long-lasting analgesic and anti-inflammatory effects (i.e. block pain without immunosuppressive effects). In this review, we focus on how SPMs block pain, infection and neuro-immune interactions and, therefore, emerge as a new class of non-immunosuppressive and non-opioid analgesic drugs.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Pain/drug therapy , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Fatty Acids, Omega-3/chemistry , Fatty Acids, Omega-3/pharmacology , Humans , Inflammation/drug therapy
7.
Int J Neuropsychopharmacol ; 22(11): 735-745, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31613314

ABSTRACT

BACKGROUND: New treatments for stress-related disorders including depression, anxiety, and substance use disorder are greatly needed. Kappa opioid receptors are expressed in the central nervous system, including areas implicated in analgesia and affective state. Although kappa opioid receptor agonists share the antinociceptive effects of mu opioid receptor agonists, they also tend to produce negative affective states. In contrast, selective kappa opioid receptor antagonists have antidepressant- and anxiolytic-like effects, stimulating interest in their therapeutic potential. The prototypical kappa opioid receptor antagonists (e.g., norBNI, JDTic) have an exceptionally long duration of action that complicates their use in humans, particularly in tests to establish safety. This study was designed to test dose- and time-course effects of novel kappa opioid receptor antagonists with the goal of identifying short-acting lead compounds for future medication development. METHODS: We screened 2 novel, highly selective kappa opioid receptor antagonists (CYM-52220 and CYM-52288) with oral efficacy in the warm water tail flick assay in rats to determine initial dose and time course effects. For comparison, we tested existing kappa opioid receptor antagonists JDTic and LY-2456302 (also known as CERC-501 or JNJ-67953964). RESULTS: In the tail flick assay, the rank order of duration of action for the antagonists was LY-2456302 < CYM-52288 < CYM-52220 << JDTic. Furthermore, LY-2456302 blocked the depressive (anhedonia-producing) effects of the kappa opioid receptor agonist U50,488 in the intracranial self-stimulation paradigm, albeit at a higher dose than that needed for analgesic blockade in the tail flick assay. CONCLUSIONS: These results suggest that structurally diverse kappa opioid receptor antagonists can have short-acting effects and that LY-2456302 reduces anhedonia as measured in the intracranial self-stimulation test.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/pharmacology , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Benzamides/pharmacology , Narcotic Antagonists/pharmacology , Piperidines/pharmacology , Pyrrolidines/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Tetrahydroisoquinolines/pharmacology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage , Analgesics, Non-Narcotic/administration & dosage , Animals , Anti-Anxiety Agents/administration & dosage , Antidepressive Agents/administration & dosage , Benzamides/administration & dosage , Drug Development , Drug Evaluation, Preclinical , Male , Narcotic Antagonists/administration & dosage , Piperidines/administration & dosage , Pyrrolidines/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/agonists , Tetrahydroisoquinolines/administration & dosage
8.
Biomed Res Int ; 2019: 6593125, 2019.
Article in English | MEDLINE | ID: mdl-31467905

ABSTRACT

Methanolic extract of Clinacanthus nutans Lindau leaves (MECN) has been reported to exert antinociceptive activity. The present study aimed to elucidate the possible antinociceptive mechanisms of a lipid-soluble fraction of MECN, which was obtained after sequential extraction in petroleum ether. The petroleum ether fraction of C. nutans (PECN), administered orally to mice, was (i) subjected to capsaicin-, glutamate-, phorbol 12-myristate 13-acetate-, bradykinin-induced nociception model; (ii) prechallenged (intraperitoneal (i.p.)) with 0.15 mg/kg yohimbine, 1 mg/kg pindolol, 3 mg/kg caffeine, 0.2 mg/kg haloperidol, or 10 mg/kg atropine, which were the respective antagonist of α 2-adrenergic, ß-adrenergic, adenosinergic, dopaminergic, or muscarinic receptors; and (iii) prechallenged (i.p.) with 10 mg/kg glibenclamide, 0.04 mg/kg apamin, 0.02 mg/kg charybdotoxin, or 4 mg/kg tetraethylammonium chloride, which were the respective inhibitor of ATP sensitive-, small conductance Ca2+-activated-, large conductance Ca2+-activated-, or nonselective voltage-activated-K+ channel. Results obtained demonstrated that PECN (100, 250, and 500 mg/kg) significantly (P<0.05) inhibited all models of nociception described earlier. The antinociceptive activity of 500 mg/kg PECN was significantly (P<0.05) attenuated when prechallenged with all antagonists or K+ channel blockers. However, only pretreatment with apamin and charybdotoxin caused full inhibition of PECN-induced antinociception. The rest of the K+ channel blockers and all antagonists caused only partial inhibition of PECN antinociception, respectively. Analyses on PECN's phytoconstituents revealed the presence of antinociceptive-bearing bioactive compounds of volatile (i.e., derivatives of γ-tocopherol, α-tocopherol, and lupeol) and nonvolatile (i.e., cinnamic acid) nature. In conclusion, PECN exerts a non-opioid-mediated antinociceptive activity involving mainly activation of adenosinergic and cholinergic receptors or small- and large-conductance Ca2+-activated-K+ channels.


Subject(s)
Acanthaceae/chemistry , Analgesics/pharmacology , Nociceptive Pain/drug therapy , Plant Extracts/pharmacology , Alkanes/chemistry , Analgesics/chemistry , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Animals , Bradykinin/toxicity , Capsaicin/toxicity , Glutamic Acid/toxicity , Humans , Methanol/chemistry , Mice , Nociceptive Pain/chemically induced , Nociceptive Pain/pathology , Plant Extracts/chemistry , Plant Leaves/chemistry , Potassium Channels/genetics , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/toxicity
9.
Psychopharmacology (Berl) ; 236(11): 3169-3182, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31172225

ABSTRACT

RATIONALE: Levo-tetrahydropalmatine (l-THP), an active ingredient of Corydalis yanhusuo, has been reported to be a partial agonist for dopamine D1 receptors (D1R) and an antagonist for D2R. Although it has been safely used clinically in China for decades as an analgesic with sedative/hypnotic properties, there are few studies that address the mechanisms by which l-THP exerts its beneficial effects in chronic pain-induced sleep disturbance. OBJECTIVES: To investigate the effects and mechanisms of l-THP on sleep disturbance in a neuropathic pain-like condition. METHODS: A mouse model of chronic neuropathic pain induced by partial sciatic nerve ligation (PSNL) was employed. The antinociceptive and hypnotic effects of l-THP were evaluated by measurement of mechanical allodynia, thermal hyperalgesia, and electroencephalogram (EEG) recordings in PSNL mice. Pharmacological approaches and c-Fos expression were used to clarify the mechanisms of l-THP. RESULTS: Intraperitoneal injection of l-THP at 5 and 10 mg/kg not only significantly increased the mechanical threshold by 134.4% and 174.8%, and prolonged the thermal latency by 49.4% and 69.2%, but also increased non-rapid eye movement sleep by 17.5% and 29.6%, and decreased sleep fragmentation in PSNL mice, compared with the vehicle control. Moreover, the antinociceptive effect of l-THP was prevented by D1R antagonist SCH23390 or D2R agonist quinpirole; meanwhile, the hypnotic effect of l-THP was blocked by quinpirole rather than by SCH23390. Immunohistochemistry demonstrated that l-THP inhibited c-Fos overexpression induced by PSNL in the cingulate cortex and the periaqueductal gray. CONCLUSIONS: These findings indicated that l-THP exerted analgesic effects by agonism D1R and antagonism D2R, and the antagonism of D2R mediated the hypnotic effect of l-THP in PSNL mice.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Berberine Alkaloids/therapeutic use , Hypnotics and Sedatives/therapeutic use , Neuralgia/drug therapy , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Analgesics, Non-Narcotic/pharmacology , Animals , Berberine Alkaloids/pharmacology , Disease Models, Animal , Dopamine D2 Receptor Antagonists/pharmacology , Dopamine D2 Receptor Antagonists/therapeutic use , Electroencephalography/drug effects , Hypnotics and Sedatives/pharmacology , Male , Mice , Mice, Inbred C57BL , Neuralgia/physiopathology , Receptors, Dopamine D1/agonists
11.
Phytomedicine ; 61: 152836, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31035053

ABSTRACT

BACKGROUND: Oncological pain is one of the most prevalent and difficult-to-treat symptoms in patients with cancer. p-Cymene (PC) is a monoterpene found in more than 100 different plant species, endowed with various pharmacological properties-particularly antinociceptive. HYPOTHESIS/PURPOSE: PC has antinociceptive effect in a model of oncologic pain due to the activation of the descending inhibitory pathway of pain. STUDY DESIGN: A pre-clinical, longitudinal, blind and randomized study. METHODS: Male Swiss mice were induced with S180 cells in the right hind paw, then treated daily with PC (12.5, 25 and 50 mg/kg, s.c.) and screened for mechanical hyperalgesia, spontaneous nociception, nociception induced by non-noxious palpation, tumor growth, changes in the neuromuscular function and existence of bone degradation in the tumor area. The effect of PC on Ca2+ currents (electrophysiological records), histological and neurochemical changes (immunofluorescence for Fos) were also evaluated. RESULTS: PC reduced (p < 0.05) the mechanical hyperalgesia, the spontaneous (p < 0.001) and non-noxious palpation (p < 0.001) nociceptions, not changing the tumor development, neuromuscular function or histopathological aspects of the paw affected. PC reduced Fos expression in the spinal cord (p < 0.001) and increased this expression in the PAG (p < 0.05) and in the NRM (p < 0.01). PC decreased the density of calcium channel currents (p < 0.05). CONCLUSION: These results suggest the antinociceptive effect of PC on oncologic pain, probably acting in both ascending and descending pain pathways, and modulating the calcium channel currents in order to exert its effects.


Subject(s)
Calcium/metabolism , Cancer Pain/drug therapy , Cymenes/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Cancer Pain/metabolism , Hyperalgesia/drug therapy , Male , Mice , Neurons/drug effects , Neurons/metabolism , Nociceptive Pain/drug therapy , Proto-Oncogene Proteins c-fos/metabolism , Random Allocation , Sarcoma 180/complications , Spinal Cord/drug effects , Spinal Cord/metabolism
12.
Future Oncol ; 15(9): 1035-1049, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30720344

ABSTRACT

Despite remaining one of the most widely abused drugs worldwide, Cannabis sativa exhibits remarkable medicinal properties. The phytocannabinoids, cannabidiol and Δ-9-tetrahydrocannabinol, reduce nausea and vomiting, particularly during chemotherapy. This is attributed to their ability to reduce the release of serotonin from enterochromaffin cells in the small intestine, which would otherwise orchestrate the vomiting reflex. Although there are many preclinical and clinical studies on the effects of Δ-9-tetrahydrocannabinol during nausea and vomiting, little is known about the role that cannabidiol plays in this scenario. Since cannabidiol does not induce psychotropic effects, in contrast to other cannabinoids, its use as an anti-emetic is of great interest. This review aims to summarize the available literature on cannabinoid use, with a specific focus on the nonpsychotropic drug cannabidiol, as well as the roles that cannabinoids play in preventing several other adverse side effects of chemotherapy including organ toxicity, pain and loss of appetite.


Subject(s)
Antineoplastic Agents/adverse effects , Cancer Pain/prevention & control , Cannabidiol/therapeutic use , Feeding and Eating Disorders/prevention & control , Nausea/drug therapy , Vomiting/drug therapy , Analgesics, Non-Narcotic/pharmacology , Analgesics, Non-Narcotic/therapeutic use , Antiemetics/pharmacology , Antiemetics/therapeutic use , Appetite/drug effects , Appetite Stimulants/pharmacology , Appetite Stimulants/therapeutic use , Cancer Pain/chemically induced , Cannabidiol/pharmacology , Cannabis/chemistry , Feeding and Eating Disorders/chemically induced , Humans , Nausea/chemically induced , Neoplasms/drug therapy , Vomiting/chemically induced
13.
Int J Hyperthermia ; 35(1): 435-440, 2018.
Article in English | MEDLINE | ID: mdl-30303410

ABSTRACT

OBJECTIVE: Recent evidence suggests the α2-adrenoreceptor agonist dexmedetomidine may promote metastasis of cancer cells. In this study we sought to evaluate the impact of dexmedetomidine administration on the survival of children and adolescents with cancer. DESIGN: Retrospective chart review. SETTING: Comprehensive cancer center. PATIENTS: Children and adolescents who had undergone cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis. INTERVENTION: Intraoperative and/or early postoperative (within 24 hours of surgery) administration of dexmedetomidine. MEASUREMENTS: Multivariable cox proportional hazard models were used to assess the association between dexmedetomidine administration and progression free survival (PFS) or overall survival (OS). MAIN RESULTS: Ninety-three patients were identified. The median age was 12 years, 42% were female, and 35% received dexmedetomidine. There were no significant differences between the baseline and perioperative characteristics of patients who received dexmedetomidine and those who did not. In the multivariable analysis, the administration of dexmedetomidine was not associated with PFS (HR = 1.20, 95% CI [0.60-2.41], p = .606) or OS (HR = 0.81, 95% CI [0.35-1.85], p = .611). CONCLUSION: In this retrospective study of children and adolescents who had undergone a major oncologic surgery, the intraoperative and/or early postoperative administration of dexmedetomidine was not associated with survival.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Cytoreduction Surgical Procedures/adverse effects , Dexmedetomidine/therapeutic use , Hyperthermia, Induced/methods , Adolescent , Analgesics, Non-Narcotic/pharmacology , Child , Cytoreduction Surgical Procedures/methods , Dexmedetomidine/pharmacology , Female , Humans , Hyperthermia, Induced/mortality , Male , Retrospective Studies , Survival Analysis
14.
PLoS One ; 13(10): e0205612, 2018.
Article in English | MEDLINE | ID: mdl-30312340

ABSTRACT

Edible portions of bananas contain high levels of polyphenol oxidase, which catalyzes reactions in the melanin formation pathway. Tyrosine, a physiological substrate of polyphenol oxidase, has an analogous structure to acetaminophen. We investigated whether banana extract causes structural changes in acetaminophen and a decrease in its potency. Acetaminophen concentration in banana extract was measured under different conditions to characterize incompatibility. Reaction products in solution were identified using liquid chromatography/electrospray ionization/mass spectrometry (LC/ESI/MS). Acetaminophen potency decreased with time in the presence of banana extract. The reaction proceeded most efficiently in temperatures 30-37°C and neutral to weakly acidic conditions. Molecular ion peaks derived from the oxidized catechol moiety of acetaminophen were identified in LC/ESI/MS spectra. Our findings suggest that incorporation or simultaneous administration of acetaminophen medication and banana juice may result in decreased efficacy of the clinically important drug. This interaction is likely due to the oxidation of acetaminophen by polyphenol oxidase activity in banana pulp. Therefore, we investigated and characterized a novel interaction between bananas and acetaminophen. To establish a safe and effective antipyretic analgesic regimen using acetaminophen, future studies of this interaction are expected to be performed in humans.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Food-Drug Interactions , Musa , Acetaminophen/chemistry , Analgesics, Non-Narcotic/chemistry , Dose-Response Relationship, Drug , Fruit and Vegetable Juices , Hydrogen-Ion Concentration , Models, Chemical , Musa/chemistry , Oxidation-Reduction , Plant Extracts/chemistry , Plant Extracts/pharmacology , Species Specificity , Temperature
15.
J Med Chem ; 61(16): 7103-7115, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30035538

ABSTRACT

The neurotensin receptors are attractive targets for the development of new analgesic compounds. They represent potential alternatives or adjuvants to opioids. Herein, we report the structural optimization of our recently reported macrocyclic peptide analogues of NT(8-13). The macrocycle was formed via ring-closing metathesis (RCM) between an ortho allylated tyrosine residue in position 11 and the side chain of alkene-functionalized amino acid in position 8 of NT(8-13). Minute modifications led to significant binding affinity improvement ( Ki improved from 5600 to 15 nM) with greatly improved plasma stability compared to NT(8-13). This study also delineates the structural features influencing these parameters. The signaling profiles of the new macrocycles were determined on the NTS1 receptor, and the physiological effects of the two most potent and stable analogues were assessed in vivo using rodent models. Both compounds displayed strong analgesic effects.


Subject(s)
Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Neurotensin/pharmacology , Peptide Fragments/pharmacology , Peptides, Cyclic/chemistry , Receptors, Neurotensin/metabolism , Animals , Binding, Competitive , Blood Pressure/drug effects , Body Temperature/drug effects , CHO Cells , Cricetulus , Cyclization , Drug Evaluation, Preclinical/methods , Drug Stability , Male , Molecular Docking Simulation , Neurotensin/agonists , Neurotensin/chemistry , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptides, Cyclic/blood , Peptides, Cyclic/pharmacology , Rats, Sprague-Dawley , Receptors, Neurotensin/chemistry , Structure-Activity Relationship , Tyrosine/chemistry
16.
Neuropharmacology ; 138: 232-244, 2018 08.
Article in English | MEDLINE | ID: mdl-29913186

ABSTRACT

Since Cav3.2 T-type Ca2+ channels (T-channels) expressed in the primary afferents and CNS contribute to intractable pain, we explored T-channel-blocking components in distinct herbal extracts using a whole-cell patch-clamp technique in HEK293 cells stably expressing Cav3.2 or Cav3.1, and purified and identified sophoraflavanone G (SG) as an active compound from SOPHORAE RADIX (SR). Interestingly, hop-derived SG analogues, (2S)-6-prenylnaringenin (6-PNG) and (2S)-8-PNG, but not naringenin, also blocked T-channels; IC50 (µM) of SG, (2S)-6-PNG and (2S)-8-PNG was 0.68-0.75 for Cav3.2 and 0.99-1.41 for Cav3.1. (2S)-6-PNG and (2S)-8-PNG, but not SG, exhibited reversible inhibition. The racemic (2R/S)-6-PNG as well as (2S)-6-PNG potently blocked Cav3.2, but exhibited minor effect on high-voltage-activated Ca2+ channels and voltage-gated Na+ channels in differentiated NG108-15 cells. In mice, the mechanical allodynia following intraplantar (i.pl.) administration of an H2S donor was abolished by oral or i.p. SR extract and by i.pl. SG, (2S)-6-PNG or (2S)-8-PNG, but not naringenin. Intraperitoneal (2R/S)-6-PNG strongly suppressed visceral pain and spinal ERK phosphorylation following intracolonic administration of an H2S donor in mice. (2R/S)-6-PNG, administered i.pl. or i.p., suppressed the neuropathic allodynia induced by partial sciatic nerve ligation or oxaliplatin, an anti-cancer agent, in mice. (2R/S)-6-PNG had little or no effect on open-field behavior, motor performance or cardiovascular function in mice, and on the contractility of isolated rat aorta. (2R/S)-6-PNG, but not SG, was detectable in the brain after their i.p. administration in mice. Our data suggest that 6-PNG, a hop component, blocks T-channels, and alleviates neuropathic and visceral pain with little side effects.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Calcium Channel Blockers/pharmacology , Flavonoids/pharmacology , Neuralgia/drug therapy , Visceral Pain/drug therapy , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/isolation & purification , Animals , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/isolation & purification , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Disease Models, Animal , Flavonoids/chemistry , Flavonoids/isolation & purification , HEK293 Cells , Humans , Humulus , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neuralgia/metabolism , Phytochemicals/chemistry , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Random Allocation , Rats, Wistar , Visceral Pain/metabolism
17.
Phytomedicine ; 38: 45-56, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29425654

ABSTRACT

BACKGROUND: Blocking the formation and invasive growth of pannus and its secretion of inflammatory cytokines and MMPs is important for treating rheumatoid arthritis. HYPOTHESIS/PURPOSE: Anti-arthritic activity of Aralia continentalis Kitag., an oriental herbal medicine, and the underlying mechanisms involved were investigated. STUDY DESIGN: Anti-inflammatory and anti-nocicpetive activities of the ethanolic extract (50% v/v) of Aralia continentalis Kitag. harvested from Imsil, Korea (ACI) were investigated in IL-1ß-stimulated human fibroblast-like synoviocyte (FLS) cells and rodent models of collagen-induced polyarthritis and carrageenan-induced acute paw pain. METHODS: In IL-1ß-stimulated FLS cells derived from rheumatoid arthritis patients, the anti-inflammatory activity of ACI was examined by analyzing the expression levels of inflammatory mediators such as TNF-α, IL-6, IL-8, MMP-1, MMP-3, MMP-13, PGE2, and COX-2 using ELISA and RT-PCR analysis. The anti-arthritic activity of ACI was investigated by measuring body weight, squeaking score, paw volume, and arthritis index in collagen-induced polyarthritis mice. The anti-nociceptive activity of ACI was examined in the paw-pressure test and Tail-flick latency test in rats. RESULTS: The ethanolic extract (50% v/v) of ACI reduced the levels of TNF-α, IL-6, IL-8, MMP-1, and MMP-13 secreted by IL-1ß-stimulated FLS cells, whereas MMP-3, COX-2, and PGE2 were not significantly affected. ACI inhibited the migration of NF-κB into the nucleus through the inhibition of ERK- and JNK-dependent MAP kinase pathways in IL-1ß-stimulated FLS cells. In collagen-induced polyarthritis mice, oral administration of ACI extract (200 mg/kg) significantly alleviated arthritic behaviors. Histological observations of arthritic mouse knees were consistent with their behaviors. The anti-arthritic and anti-inflammatory activities of 200 mg/kg ACI extract were comparable to those of 10 mg/kg prednisolone when administered to mice. However, ACI administration did not significantly affect carrageenan-induced hyperalgesia or thermal nociception in rats. CONCLUSION: These results suggest that the ethanolic extract of ACI have significant anti-inflammatory and anti-arthritic effects in a rodent arthritis model and in IL-1ß-stimulated FLS cells. Thus, ACI may be a useful candidate for developing pharmaceuticals or dietary supplements for the treatment of inflammatory arthritis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aralia/chemistry , Arthritis/drug therapy , Synoviocytes/drug effects , Analgesics, Non-Narcotic/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Arthritis/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/pharmacology , Male , Mice, Inbred DBA , NF-kappa B/metabolism , Nociception/drug effects , Pain/drug therapy , Rats, Sprague-Dawley , Republic of Korea , Synoviocytes/metabolism , Synoviocytes/pathology
18.
World Neurosurg ; 113: 333-347.e5, 2018 May.
Article in English | MEDLINE | ID: mdl-29421449

ABSTRACT

Neuropathic pain is a subset of chronic pain that is caused by neurons that are damaged or firing aberrantly in the peripheral or central nervous systems. The treatment guidelines for neuropathic pain include antidepressants, calcium channel α2 delta ligands, topical therapy, and opioids as a second-line option. Pharmacotherapy has not been effective in the treatment of neuropathic pain except in the treatment of trigeminal neuralgia with carbamazepine. The inability to properly treat neuropathic pain causes frustration in both the patients and their treating physicians. Venoms, which are classically believed to be causes of pain and death, have peptide components that have been implicated in pain relief. Although some venoms are efficacious and have shown benefits in patients, their side-effect profile precludes their more widespread use. This review identifies and explores the use of venoms in neuropathic pain relief. This treatment can open doors to potential therapeutic targets. We believe that further research into the mechanisms of action of these receptors as well as their functions in nature will provide alternative therapies as well as a window into how they affect neuropathic pain.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Neuralgia/drug therapy , Peptides/therapeutic use , Toxins, Biological/therapeutic use , Venoms/therapeutic use , Analgesics, Non-Narcotic/isolation & purification , Analgesics, Non-Narcotic/pharmacology , Animals , Humans , Neuralgia/diagnosis , Neuralgia/epidemiology , Pain Management/methods , Peptides/isolation & purification , Peptides/pharmacology , Toxins, Biological/isolation & purification , Toxins, Biological/pharmacology , Venoms/isolation & purification , Venoms/pharmacology , omega-Conotoxins/isolation & purification , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use
19.
Neuropsychopharmacology ; 43(1): 52-79, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28857069

ABSTRACT

A great need exists for the development of new medications to treat pain resulting from various disease states and types of injury. Given that the endogenous cannabinoid (that is, endocannabinoid) system modulates neuronal and immune cell function, both of which play key roles in pain, therapeutics targeting this system hold promise as novel analgesics. Potential therapeutic targets include the cannabinoid receptors, type 1 and 2, as well as biosynthetic and catabolic enzymes of the endocannabinoids N-arachidonoylethanolamine and 2-arachidonoylglycerol. Notably, cannabinoid receptor agonists as well as inhibitors of endocannabinoid-regulating enzymes fatty acid amide hydrolase and monoacylglycerol lipase produce reliable antinociceptive effects, and offer opioid-sparing antinociceptive effects in myriad preclinical inflammatory and neuropathic pain models. Emerging clinical studies show that 'medicinal' cannabis or cannabinoid-based medications relieve pain in human diseases such as cancer, multiple sclerosis, and fibromyalgia. However, clinical data have yet to demonstrate the analgesic efficacy of inhibitors of endocannabinoid-regulating enzymes. Likewise, the question of whether pharmacotherapies aimed at the endocannabinoid system promote opioid-sparing effects in the treatment of pain reflects an important area of research. Here we examine the preclinical and clinical evidence of various endocannabinoid system targets as potential therapeutic strategies for inflammatory and neuropathic pain conditions.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Endocannabinoids/metabolism , Inflammation/metabolism , Neuralgia/metabolism , Analgesics, Non-Narcotic/therapeutic use , Animals , Cannabinoid Receptor Modulators/therapeutic use , Drug Discovery , Humans , Inflammation/drug therapy , Neuralgia/drug therapy , Receptors, Cannabinoid/metabolism
20.
Environ Health Prev Med ; 22(1): 54, 2017 Jun 12.
Article in English | MEDLINE | ID: mdl-29165178

ABSTRACT

BACKGROUND: The aim of this study was to investigate the therapeutic effects of a Sasa veitchii leaf extract (SE) on acetaminophen (APAP)-induced hepatotoxicity. METHODS: Seven-week-old male ddY mice were orally administered SE or saline (0.2 mL) once a day for a week. Twenty-four hours after the last pretreatment, the mice were intraperitoneally injected with 550 mg/kg APAP or saline under fasting conditions. The mice from each group were euthanized and bled for plasma analysis 2, 6, 24, and 72 h after the injection. RESULTS: We found that pretreatment with SE significantly decreased hepatic injury markers (i.e., alanine aminotransferase and aspartate aminotransferase), oxidative stress (malondialdehyde and glutathione level), inflammatory cytokines, histological damage, c-jun N-terminal kinase activation, and receptor-interacting protein-1 activation. Further, SE pretreatment decreased Cyp2e1 expression and increased total antioxidant capacity in the liver. CONCLUSION: Our findings demonstrate that prophylactic SE treatment protects mice from APAP-induced hepatotoxicity through modulation of Cyp2e1 expression and antioxidant capacity.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Protective Agents/metabolism , Sasa/metabolism , Alanine Transaminase/drug effects , Animals , Antioxidants/metabolism , Aspartate Aminotransferases , Blotting, Western , Cytochrome P450 Family 2/metabolism , Liver/drug effects , Liver/physiopathology , Male , Mice , Oxidative Stress , Plant Preparations
SELECTION OF CITATIONS
SEARCH DETAIL