Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
1.
Toxicol Appl Pharmacol ; 486: 116950, 2024 May.
Article in English | MEDLINE | ID: mdl-38701902

ABSTRACT

Antidepressant duloxetine has been shown protective effect on indomethacin-induced gastric ulcer, which was escorted by inflammation in the gastric mucosa. Cytokines are the principal mediators of inflammation. Thus, by screening the differential expression of cytokines in the gastric mucosa using cytokine array at 3 h after indomethacin exposure, when the gastric ulcer began to format, we found that indomethacin increased cytokines which promoted inflammation responses, whereas duloxetine decreased pro-inflammatory cytokines increased by indomethacin and increased RANTES expression. RANTES was consistently increased by pretreated with both 5 mg/kg and 20 mg/kg duloxetine at 3 h and 6 h after indomethacin exposure in male rats. Selective blockade of RANTES-CCR5 axis by a functional antagonist Met-RANTES or a CCR5 antagonist maraviroc suppressed the protection of duloxetine. Considering the pharmacologic action of duloxetine on reuptake of monoamine neurotransmitters, we examined the serotonin (5-HT), norepinephrine and dopamine contents in the blood and discovered 20 mg/kg duloxetine increased 5-HT levels in platelet-poor plasma, while treatment with 5-HT promoted expression of RANTES in the gastric mucosa and alleviated the indomethacin-induced gastric injury. Furthermore, duloxetine activated PI3K-AKT-VEGF signaling pathway, which was regulated by RANTES-CCR5, and selective inhibitor of VEGF receptor axitinib blocked the prophylactic effect of duloxetine. Furthermore, duloxetine also protected gastric mucosa from indomethacin in female rats, and RANTES was increased by duloxetine after 6 h after indomethacin exposure too. Together, our results identified the role of cytokines, particularly RANTES, and the underlying mechanisms in gastroprotective effect of duloxetine against indomethacin, which advanced our understanding in inflammatory modulation by monoamine-based antidepressants.


Subject(s)
Chemokine CCL5 , Duloxetine Hydrochloride , Gastric Mucosa , Indomethacin , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Serotonin , Signal Transduction , Stomach Ulcer , Vascular Endothelial Growth Factor A , Animals , Duloxetine Hydrochloride/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Male , Indomethacin/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Chemokine CCL5/metabolism , Signal Transduction/drug effects , Rats , Vascular Endothelial Growth Factor A/metabolism , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Stomach Ulcer/pathology , Stomach Ulcer/metabolism , Serotonin/metabolism , Phosphatidylinositol 3-Kinases/metabolism
2.
Neuroreport ; 35(6): 380-386, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38526956

ABSTRACT

This study aims to investigate the functional connectivity (FC) changes of the habenula (Hb) among patients with major depressive disorder (MDD) after 12 weeks of duloxetine treatment (MDD12). Patients who were diagnosed with MDD for the first time and were drug-naïve were recruited at baseline as cases. Healthy controls (HCs) matched for sex, age, and education level were also recruited at the same time. At baseline, all participants underwent resting-state functional MRI. FC analyses were performed using the Hb seed region of interest, and three groups including HCs, MDD group and MDD12 group were compared using whole-brain voxel-wise comparisons. Compared to the HCs, the MDD group had decreased FC between the Hb and the right anterior cingulate cortex at baseline. Compared to the HCs, the FC between the Hb and the left medial superior frontal gyrus decreased in the MDD12 group. Additionally, the FC between the left precuneus, bilateral cuneus and Hb increased in the MDD12 group than that in the MDD group. No significant correlation was found between HDRS-17 and the FC between the Hb, bilateral cuneus, and the left precuneus in the MDD12 group. Our study suggests that the FC between the post-default mode network and Hb may be the treatment mechanism of duloxetine and the treatment mechanisms and the pathogenesis of depression may be independent of each other.


Subject(s)
Depressive Disorder, Major , Habenula , Humans , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/drug therapy , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , Default Mode Network , Magnetic Resonance Imaging , Rest/physiology
3.
Biomed Pharmacother ; 168: 115810, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37913736

ABSTRACT

BACKGROUND: Several studies have reported the association between osteoporosis and major depressive disorder (MDD) as well as the use of antidepressants. However, it remains to be elucidated whether these associations are related to exposure to antidepressants, a consequence of a disease process, or a combination of both. METHODS: This study investigates the independent effect of the antidepressant duloxetine hydrochloride (DH) on ovariectomy-induced bone loss in mice. One week after ovariectomy, the treated mice received DH. To explore the mechanism underlying the rescue of bone loss, bone marrow cells were isolated from mouse femurs and tibias, and macrophages extracted from them were induced to become osteoclasts in vitro while being treated with DH. Subsequently, the osteoclasts underwent Bulk RNA-Seq to reveal the involved signaling pathways. The results of the bioinformatic analysis were then validated through in vitro experiments. RESULTS: The in vivo experiments demonstrated that DH treatment compromised ovariectomy-induced bone loss after 7 weeks. The in vitro experiments suggested that DH treatment attenuated osteoclast differentiation via the MAPKs/NFATc1 signaling pathway. CONCLUSION: The findings from this study suggest that DH, instead of causing bone mass loss, may assist in alleviating postmenopausal osteoporosis. These results can serve as a reference for the clinical treatment of patients with perimenopausal or postmenopausal depression using antidepressants.


Subject(s)
Depressive Disorder, Major , Osteoclasts , Humans , Female , Animals , Mice , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , Depressive Disorder, Major/metabolism , Cell Differentiation , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Ovariectomy/adverse effects , Osteogenesis , RANK Ligand/metabolism
4.
Cell Biochem Funct ; 41(7): 814-822, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37814477

ABSTRACT

Duloxetine, a selective reuptake inhibitor for serotonin and norepinephrine, is a medication widely used for major depression. Currently, duloxetine is also recommended for pain related to chemotherapy-induced peripheral neuropathy or cancer. Previously, we showed that transforming growth factor-α (TGF-α) induces the migration of human hepatocellular carcinoma (HCC)-derived HuH7 cells through the activation of c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK) and AKT. In the present study, we investigate whether duloxetine affects cell migration and its mechanism. Duloxetine significantly enhanced the TGF-α-induced migration of HuH7 cells. Fluvoxamine and sertraline, specific inhibitors of serotonin reuptake, also upregulated the TGF-α-induced cell migration. On the contrary, reboxetine, a specific norepinephrine reuptake inhibitor, failed to affect cell migration. Duloxetine significantly amplified the TGF-α-stimulated phosphorylation of JNK, but not p38 MAPK and AKT. In addition, fluvoxamine and sertraline, but not reboxetine, enhanced the phosphorylation of JNK. SP600125, a JNK inhibitor, suppressed the enhancement by duloxetine, fluvoxamine, or sertraline of TGF-α-induced migration of HuH7 cells. Taken together, our results strongly suggest that duloxetine strengthens the TGF-α-induced activation of JNK via inhibition of serotonin reuptake in HCC cells, leading to the enhancement of cell migration.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Duloxetine Hydrochloride/pharmacology , Fluvoxamine/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/pathology , Norepinephrine , p38 Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Serotonin/metabolism , Sertraline/pharmacology , Transforming Growth Factor alpha/pharmacology , Transforming Growth Factor alpha/metabolism , Up-Regulation
5.
Biomed Pharmacother ; 166: 115360, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37657261

ABSTRACT

Osteoarthritis represents a leading cause of disability with limited treatment options. Furthermore, it is frequently accompanied by cardiovascular and cognitive disorders, which can be exacerbated by osteoarthritis or drugs used for its treatment. Here, we examined the behavioral and cardiac effects of the novel antidepressant vortioxetine in an osteoarthritis model, and compared them to duloxetine (an established osteoarthritis treatment). Osteoarthritis was induced in male and female rats with an intraarticular sodium-monoiodoacetate injection. Antidepressants were orally administered for 28 days following induction. During this period the acetone, burrowing and novel-object-recognition tests (NORT) were used to assess the effects of antidepressants on pain hypersensitivity (cold allodynia), animal well-being and cognitive performance, respectively. Following behavioral experiments, heart muscles were collected for assessment of redox status/histology. Antidepressant treatment dose-dependently reduced cold allodynia in rats with osteoarthritis. Duloxetine (but not vortioxetine) depressed burrowing behavior in osteoarthritic rats in a dose-related manner. Osteoarthritis induction reduced cognitive performance in NORT, which was dose-dependently alleviated by vortioxetine (duloxetine improved performance only in female rats). Furthermore, duloxetine (but not vortioxetine) increased oxidative stress parameters in the heart muscles of female (but not male) rats and induced histological changes in cardiomyocytes indicative of oxidative damage. Vortioxetine displayed comparable efficacy to duloxetine in reducing pain hypersensitivity. Furthermore, vortioxetine (unlike duloxetine) dose-dependently improved cognitive performance and had no adverse effect on burrowing behavior (animal surrogate of well-being) and cardiac redox status/histology. Our results indicate that vortioxetine could be a potential osteoarthritis treatment (with better characteristics compared to duloxetine).


Subject(s)
Hypersensitivity , Osteoarthritis , Female , Animals , Rats , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , Vortioxetine , Hyperalgesia , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Myocytes, Cardiac , Osteoarthritis/drug therapy , Cognition
6.
Inflammation ; 46(6): 2449-2469, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37644164

ABSTRACT

Asthma is an inflammatory disease characterized by airway hyperresponsiveness, airway remodeling, and airway inflammation. In recent years, the prevalence of asthma has been increasing steadily and the pathogenesis of asthma varies from person to person. Due to poor compliance or resistance, existing drugs cannot achieve the desired therapeutic effect. Therefore, developing or screening asthma therapeutic drugs with high curative effects, low toxicity, and strong specificity is very urgent. Duloxetine HCl (DUX) is a selective serotonin and norepinephrine reuptake inhibitor, and it was mainly used to treat depression, osteoarthritis, and neuropathic pain. It was also reported that DUX has potential anti-infection, anti-inflammation, analgesic, antioxidative, and other pharmacological effects. However, whether DUX has some effects on asthma remains unknown. In order to investigate it, a series of ex vivo and in vivo experiments, including biological tension tests, patch clamp, histopathological analysis, lung function detection, oxidative stress enzyme activity detection, and molecular biology experiments, were designed in this study. We found that DUX can not only relax high potassium or ACh precontracted tracheal smooth muscle by regulating L-type voltage-dependent Ca2+ channel (L-VDCC) and nonselective cation channel (NSCC) ion channels but also alleviate asthma symptoms through anti-inflammatory and antioxidative response regulated by PI3K/AKT/mTOR and Nrf2/HO-1 signaling pathways. Our data suggests that DUX is expected to become a potential new drug for relieving or treating asthma.


Subject(s)
Asthma , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , NF-E2-Related Factor 2/metabolism , Asthma/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Anti-Inflammatory Agents/pharmacology
7.
Steroids ; 198: 109272, 2023 10.
Article in English | MEDLINE | ID: mdl-37468115

ABSTRACT

Allopregnanolone (AP) is a neurosteroid synthesized in the brain and a positive allosteric modulator of γ-aminobutyric acid (GABA) type A receptors. Some drugs possessing the aryloxypropanamine (AOPA) pharmacophore, such as fluoxetine, exert their central nervous system (CNS) effects by increasing the brain AP. Although duloxetine (DLX), dapoxetine (DPX), atomoxetine (ATX) and propranolol (PRL) also possess the AOPA pharmacophore and are used to treat some psychiatric disorders, the capabilities of these drugs to increase the brain AP and the possible involvement of AP in their CNS effects remain to be fully elucidated. To clarify these points, we first developed a method for quantifying AP in the rat brain by liquid chromatography/electrospray ionization-tandem mass spectrometry. Analysis of the changes in the brain AP levels using this method revealed that the intraperitoneal administration of DLX (10 mg/kg), DPX (10 mg/kg) and PRL (20 mg/kg) significantly increased the brain AP (DLX: < 0.40-2.74 ng/g tissue, DPX: 1.48-3.83 ng/g tissue and PRL: < 0.40-2.09 ng/g tissue) compared to the saline administration (<0.40 ng/g tissue). These results suggested the possible involvement of the GABAergic neurosteroid, AP, in the central actions of DLX, DPX and PRL. In contrast, ATX (10 mg/kg) did not affect the AP levels in the brain. In addition, the brain and serum AP levels had a remarkably high positive correlation after the administration of DLX, DPX and PRL. Thus, this study proposed the AP-related novel mechanism of actions of DLX, DPX and PRL in the CNS.


Subject(s)
Neurosteroids , Pregnanolone , Animals , Rats , Brain , Duloxetine Hydrochloride/pharmacology , Pharmaceutical Preparations , Pharmacophore , Pregnanolone/pharmacology , Propranolol/pharmacology , Propylamines/chemistry , Propylamines/pharmacology
8.
Int J Mol Sci ; 24(9)2023 May 06.
Article in English | MEDLINE | ID: mdl-37176065

ABSTRACT

Paclitaxel, a widely used cancer chemotherapeutic agent, has high incidence of neurotoxicity associated with the production of neuropathic pain, for which only duloxetine has shown significant but moderate analgesic effect. Since statins, classically used to reduce hypercholesterolemia, have shown antinociceptive effect in preclinical studies on neuropathic pain, we studied whether the antinociceptive efficacy of duloxetine could be synergistically potentiated by rosuvastatin in a model of paclitaxel-induced neuropathy in mice. The astrocytic and microglial responses in the spinal cord of paclitaxel-treated mice were also assessed by measuring GFAP and CD11b proteins, respectively. Paclitaxel treatment did not impair motor coordination and balance in rotarod testing. Rosuvastatin, duloxetine, and the rosuvastatin/duloxetine combination (combined at equieffective doses) dose-dependently decreased mechanical allodynia (ED30, von Frey testing) and thermal hyperalgesia (ED50, hot plate testing) in paclitaxel-treated mice. Isobolographic analysis showed a superadditive interaction for rosuvastatin and duloxetine, as both the ED30 and ED50 for the rosuvastatin/duloxetine combination contained only a quarter of each drug compared to the individual drugs. The rosuvastatin/duloxetine combination reversed paclitaxel-induced GFAP overexpression, indicating that such effects might depend in part on astrocyte inactivation. Results suggest that statins could be useful in synergistically enhancing the efficacy of duloxetine in some chemotherapy-induced neuropathic conditions.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Neuralgia , Mice , Animals , Paclitaxel/adverse effects , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , Rosuvastatin Calcium/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Pain Measurement , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/complications , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Analgesics/adverse effects
9.
ACS Chem Neurosci ; 14(7): 1261-1277, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36930958

ABSTRACT

Antidepressants, such as duloxetine and amitriptyline, are effective for treating patients with chronic neuropathic pain. Inhibiting norepinephrine and serotonin transporters at presynaptic terminals raises extracellular concentrations of norepinephrine. The α1- and α2-adrenergic receptor agonists inhibit glutamatergic input from primary afferent nerves to the spinal dorsal horn. However, the contribution of spinal α1- and α2-adrenergic receptors to the analgesic effect of antidepressants and associated synaptic plasticity remains uncertain. In this study, we showed that systemic administration of duloxetine or amitriptyline acutely reduced tactile allodynia and mechanical and thermal hyperalgesia caused by spinal nerve ligation in rats. In contrast, duloxetine or amitriptyline had no effect on nociception in sham rats. Blocking α1-adrenergic receptors with WB-4101 or α2-adrenergic receptors with yohimbine at the spinal level diminished the analgesic effect of systemically administered duloxetine and amitriptyline. Furthermore, intrathecal injection of duloxetine or amitriptyline similarly attenuated pain hypersensitivity in nerve-injured rats; the analgesic effect was abolished by intrathecal pretreatment with both WB-4101 and yohimbine. In addition, whole-cell patch-clamp recordings in spinal cord slices showed that duloxetine or amitriptyline rapidly inhibited dorsal root-evoked excitatory postsynaptic currents in dorsal horn neurons in nerve-injured rats but had no such effect in sham rats. The inhibitory effect of duloxetine and amitriptyline was abolished by the WB-4101 and yohimbine combination. Therefore, antidepressants attenuate neuropathic pain predominantly by inhibiting primary afferent input to the spinal cord via activating both α1- and α2-adrenergic receptors. This information helps the design of new strategies to improve the treatment of neuropathic pain.


Subject(s)
Amitriptyline , Neuralgia , Rats , Animals , Duloxetine Hydrochloride/pharmacology , Amitriptyline/pharmacology , Rats, Sprague-Dawley , Antidepressive Agents , Spinal Cord Dorsal Horn , Norepinephrine , Posterior Horn Cells , Hyperalgesia/drug therapy , Yohimbine/pharmacology , Neuralgia/drug therapy , Analgesics/pharmacology , Receptors, Adrenergic
10.
PLoS One ; 18(1): e0279011, 2023.
Article in English | MEDLINE | ID: mdl-36638092

ABSTRACT

Tramadol is a useful analgesic which acts as a serotonin and noradrenaline reuptake inhibitor in addition to µ-opioid receptor agonist. Cytoplasmic serotonin modulates the small GTPase activity through serotonylation, which is closely related to the human platelet activation. We recently reported that the combination of subthreshold collagen and CXCL12 synergistically activates human platelets. We herein investigated the effect and the mechanism of tramadol on the synergistic effect. Tramadol attenuated the synergistically stimulated platelet aggregation (300 µM of tramadol, 64.3% decrease, p<0.05). Not morphine or reboxetine, but duloxetine, fluvoxamine and sertraline attenuated the synergistic effect of the combination on the platelet aggregation (30 µM of fluvoxamine, 67.3% decrease, p<0.05; 30 µM of sertraline, 67.8% decrease, p<0.05). The geranylgeranyltransferase inhibitor GGTI-286 attenuated the aggregation of synergistically stimulated platelet (50 µM of GGTI-286, 80.8% decrease, p<0.05), in which GTP-binding Rac was increased. The Rac1-GEF interaction inhibitor NSC23766 suppressed the platelet activation and the phosphorylation of p38 MAPK and HSP27 induced by the combination of collagen and CXCL12. Tramadol and fluvoxamine almost completely attenuated the levels of GTP-binding Rac and the phosphorylation of both p38 MAPK and HSP27 stimulated by the combination. Suppression of the platelet aggregation after the duloxetine administration was observed in 2 of 5 patients in pain clinic. These results suggest that tramadol negatively regulates the combination of subthreshold collagen and CXCL12-induced platelet activation via Rac upstream of p38 MAPK.


Subject(s)
Tramadol , Humans , Tramadol/pharmacology , HSP27 Heat-Shock Proteins/metabolism , rho-Associated Kinases , Duloxetine Hydrochloride/pharmacology , Fluvoxamine , Serotonin/pharmacology , Sertraline/pharmacology , Blood Platelets/metabolism , Platelet Aggregation , Collagen/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Guanosine Triphosphate , Phosphorylation
11.
Eur J Pharmacol ; 941: 175499, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36627097

ABSTRACT

Duloxetine has been shown to produce gastroprotective effect against gastric ulcer induced by water immersion restraint stress (WIRS) via modulation of NADPH oxidases in the gastric mucosa and neurometabolites of central nucleus of amygdala. However, the underlying mechanism based on the basic pharmacological function of duloxetine-regulation on serotonin (5-HT) and norepinephrine (NE) remains unclear. Here, we found that 5-HT level in platelet-poor plasma (PPP) was decreased but NE level in plasma was increased in rats exposed to WIRS, while pretreatment with duloxetine increased 5-HT in PPP dose-dependently and decreased NE in plasma of rats after WIRS. We further showed that depletion of 5-HT by 4-chloro-DL-phenylalanine (PCPA) aggravated gastric mucosa damage and supplement of 5-HT alleviated gastric ulcers induced by WIRS. Blockade of NE receptors also mitigated the stress gastric ulcers. Using adrenalectomy and chemical blocking, we identified that it was NE from adrenal medulla rather than sympathetic nerve that was more critical in the gastroprotection of duloxetine, and intriguingly, glucocorticoid did not make a difference in WIRS-provoked gastric ulcers as a classic stress hormone. Together, our work demonstrated prophylactic protection of duloxetine from the stress gastric ulcer depended on enhancing peripheral 5-HT content and reducing NE from adrenal medulla, which provided insight into treatments of WIRS-induced gastric ulcer.


Subject(s)
Norepinephrine , Stomach Ulcer , Rats , Animals , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , Serotonin , Stomach Ulcer/etiology , Stomach Ulcer/prevention & control , Stomach Ulcer/drug therapy , Gastric Mucosa
12.
J Pharm Sci ; 112(2): 544-561, 2023 02.
Article in English | MEDLINE | ID: mdl-36063878

ABSTRACT

Duloxetine HCl (DXH) is a psychiatric medicine employed for treating major depressive disorder. Nonetheless, its low water solubility, high first-pass metabolism, and acid instability diminish the absolute oral bioavailability to 40%, thus necessitating frequent administration. Therefore, the aim of the current study was to formulate DXH as nasal chitosan-grafted polymeric nanoparticles to improve its pharmacokinetic and pharmacodynamic properties. Applying the Box-Behnken design, DXH loaded PLGA-Chitosan nanoparticles (DXH-PLGA-CS-NPs) were fabricated and optimized using polylactide-co-glycolic acid (PLGA), chitosan (CS), and polyvinyl alcohol (PVA) as the independent factors. Particle size, entrapment efficiency, release percent, and cumulative amount permeated after 24 h of DXH-PLGA-CS-NPs (dependent variables) were evaluated. The in-vivo biodistribution and pharmacodynamic studies were done in male Wistar rats. The optimized DXH-PLGA-CS-NPs had a vesicle size of 122.11 nm and EE% of 66.95 with 77.65% release and Q24 of 555.34 (µg/cm2). Ex-vivo permeation study revealed 4-folds increase in DXH permeation from DXH-PLGA-CS-NPs after 24 h compared to DXH solution. Intranasal administration of optimized DXH-PLGA-CS-NPs resulted in significantly higher (p < 0.05) Cmax, AUCtotal, t1/2, and MRT in rat brain and plasma than oral DXH solution. Pharmacodynamics investigation revealed that intranasally exploited optimal DXH-PLGA-CS-NPs could be deemed a fruitful horizon for DXH as a treatment for depression.


Subject(s)
Chitosan , Depressive Disorder, Major , Nanoparticles , Rats , Animals , Male , Polylactic Acid-Polyglycolic Acid Copolymer , Chitosan/metabolism , Duloxetine Hydrochloride/pharmacology , Rats, Wistar , Drug Carriers/metabolism , Tissue Distribution , Particle Size
13.
Eur J Pain ; 27(1): 129-147, 2023 01.
Article in English | MEDLINE | ID: mdl-36198034

ABSTRACT

BACKGROUND: Exercise is a known trigger of the inhibitory pain modulation system and its analgesic effect is termed exercise-induced hypoalgesia (EIH). Previous studies have demonstrated that rats with deficient analgesic response following exercise develop more significant hypersensitivity following nerve injury compared to rats with substantial analgesic response following exercise. OBJECTIVES: A rat model of EIH as an indicator of the pain inhibitory system's efficiency was used to explore the association between EIH profiles and the effect of pharmacotherapy on rat's neuropathic pain. METHODS: EIH profiles were assessed by evaluating paw responses to mechanical stimuli before and after exercise on a rotating rod. Rats with a reduction of ≤33% in responses were classified as low EIH and those with ≥67% as high EIH. Low and high EIH rats underwent sciatic nerve chronic constriction injury (CCI). Paw responses to mechanical stimuli were measured at baseline, following CCI, and after treatment with diclofenac, duloxetine or pregabalin. In a different group of low and high EIH rats, EIH was measured before and following treatment with the same medications. RESULTS: Low EIH rats developed more significant hypersensitivity following CCI. Duloxetine and pregabalin successfully reduced hypersensitivity, although significantly more so in low EIH rats. Diclofenac had limited effects, and only on low EIH rats. Four days of duloxetine administration transformed low EIH rats' profiles to high EIH. CONCLUSIONS: The findings of this study suggest that EIH profiles in rats can not only predict the development of hypersensitivity following injury but may also support targeted pharmacological treatment. SIGNIFICANCE: Exercise is a known trigger of the inhibitory pain modulation. Rats with deficient analgesic response following exercise develop more significant hypersensitivity following nerve injury. Pain modulation profiles in rats can also support targeted pharmacological treatment; rats with deficient analgesic response following exercise benefit more from treatment with duloxetine and gabapentin. Treatment with duloxetine can improve pain modulation profile.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , Rats , Animals , Pregabalin/pharmacology , Pregabalin/therapeutic use , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , Diclofenac/adverse effects , Neuralgia/drug therapy , Neuralgia/chemically induced , Analgesics/adverse effects , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/drug therapy , Constriction, Pathologic/drug therapy , Pain Threshold/physiology
14.
Cells ; 11(24)2022 12 14.
Article in English | MEDLINE | ID: mdl-36552814

ABSTRACT

Neuropathic pain is a chronic pain condition that occurs after nerve damage; allodynia, which refers to pain caused by generally innocuous stimuli, is a hallmark symptom. Although allodynia is often resistant to analgesics, the antidepressant duloxetine has been used as an effective therapeutic option. Duloxetine increases spinal noradrenaline (NA) levels by inhibiting its transporter at NAergic terminals in the spinal dorsal horn (SDH), which has been proposed to contribute to its pain-relieving effect. However, the mechanism through which duloxetine suppresses neuropathic allodynia remains unclear. Here, we identified an SDH inhibitory interneuron subset (captured by adeno-associated viral (AAV) vectors incorporating a rat neuropeptide Y promoter; AAV-NpyP+ neurons) that is mostly depolarized by NA. Furthermore, this excitatory effect was suppressed by pharmacological blockade or genetic knockdown of α1B-adrenoceptors (ARs) in AAV-NpyP+ SDH neurons. We found that duloxetine suppressed Aß fiber-mediated allodynia-like behavioral responses after nerve injury and that this effect was not observed in AAV-NpyP+ SDH neuron-selective α1B-AR-knockdown. These results indicate that α1B-AR and AAV-NpyP+ neurons are critical targets for spinal NA and are necessary for the therapeutic effect of duloxetine on neuropathic pain, which can support the development of novel analgesics.


Subject(s)
Hyperalgesia , Neuralgia , Rats , Animals , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , Hyperalgesia/complications , Neuralgia/drug therapy , Neuralgia/etiology , Interneurons , Analgesics/pharmacology , Analgesics/therapeutic use
15.
Cancer Res Commun ; 2(11): 1334-1343, 2022 11.
Article in English | MEDLINE | ID: mdl-36506732

ABSTRACT

Oxaliplatin-induced peripheral neurotoxicity (OIPN) is a debilitating side effect that afflicts ~90% of patients that is initiated by OCT2-dependent uptake of oxaliplatin in DRG neurons. The antidepressant drug duloxetine has been used to treat OIPN, although its usefulness in preventing this side effect remains unclear. We hypothesized that duloxetine has OCT2-inhibitory properties and can be used as an adjunct to oxaliplatin-based regimens to prevent OIPN. Transport studies were performed in cells stably transfected with mouse or human OCT2 and in isolated mouse DRG neurons ex vivo. Wild-type and OCT2-deficient mice were used to assess effects of duloxetine on hallmarks of OIPN, endogenous OCT2 biomarkers, and the pharmacokinetics of oxaliplatin, and the translational feasibility of a duloxetine-oxaliplatin combination was evaluated in various models of colorectal cancer. We found that duloxetine potently inhibited the OCT2-mediated transport of several xenobiotic substrates, including oxaliplatin, in a reversible, concentration-dependent manner, and independent of species and cell context. Furthermore, duloxetine restricted access of these substrates to DRG neurons ex vivo and prevented OIPN in wild-type mice to a degree similar to the complete protection observed in OCT2-deficient mice, without affecting the plasma levels of oxaliplatin. Importantly, the uptake and cytotoxicity of oxaliplatin in tumor cell lines in vitro and in vivo were not negatively influenced by duloxetine. The observed OCT2-targeting properties of duloxetine, combined with the potential for clinical translation, provide support for its further exploration as a therapeutic candidate for studies aimed at preventing OIPN in cancer patients requiring treatment with oxaliplatin. Significance: We found that duloxetine has potent OCT2-inhibitory properties and can diminish excessive accumulation of oxaliplatin into DRG neurons. In addition, pre-treatment of mice with duloxetine prevented OIPN without significantly altering the plasma pharmacokinetics and antitumor properties of oxaliplatin. These results suggest that intentional inhibition of OCT2-mediated transport by duloxetine can be employed as a prevention strategy to ameliorate OIPN without compromising the effectiveness of oxaliplatin-based treatment.


Subject(s)
Antineoplastic Agents , Neurotoxicity Syndromes , Peripheral Nervous System Diseases , Humans , Mice , Animals , Oxaliplatin/adverse effects , Antineoplastic Agents/toxicity , Duloxetine Hydrochloride/pharmacology , Peripheral Nervous System Diseases/chemically induced , Neurotoxicity Syndromes/drug therapy
16.
Sci Rep ; 12(1): 16359, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36175479

ABSTRACT

Traumatic brain injury (TBI) is a significant public health concern, with the majority of injuries being mild. Many TBI victims experience chronic pain. Unfortunately, the mechanisms underlying pain after TBI are poorly understood. Here we examined the contribution of spinal monoamine signaling to dysfunctional descending pain modulation after TBI. For these studies we used a well-characterized concussive model of mild TBI. Measurements included mechanical allodynia, the efficacy of diffuse noxious inhibitory control (DNIC) endogenous pain control pathways and lumber norepinephrine and serotonin levels. We observed that DNIC is strongly reduced in both male and female mice after mild TBI for at least 12 weeks. In naïve mice, DNIC was mediated through α2 adrenoceptors, but sensitivity to α2 adrenoceptor agonists was reduced after TBI, and reboxetine failed to restore DNIC in these mice. The intrathecal injection of ondansetron showed that loss of DNIC was not due to excess serotonergic signaling through 5-HT3 receptors. On the other hand, the serotonin-norepinephrine reuptake inhibitor, duloxetine and the serotonin selective reuptake inhibitor escitalopram both effectively restored DNIC after TBI in both male and female mice. Therefore, enhancing serotonergic signaling as opposed to noradrenergic signaling alone may be an effective pain treatment strategy after TBI.


Subject(s)
Brain Concussion , Brain Injuries, Traumatic , Chronic Pain , Amines , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Duloxetine Hydrochloride/pharmacology , Female , Male , Mice , Norepinephrine , Ondansetron , Reboxetine , Receptors, Adrenergic , Serotonin , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use
17.
Cell Death Dis ; 13(8): 669, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35915074

ABSTRACT

Duloxetine (DLX) has been approved for the successful treatment of psychiatric diseases, including major depressive disorder, diabetic neuropathy, fibromyalgia and generalized anxiety disorder. However, since the usage of DLX carries a manufacturer warning of hepatotoxicity given its implication in numerous cases of drug-induced liver injuries (DILI), it is not recommended for patients with chronic liver diseases. In our previous study, we developed an enhanced human-simulated hepatic spheroid (EHS) imaging model system for performing drug hepatotoxicity evaluation using the human hepatoma cell line HepaRG and the support of a pulverized liver biomatrix scaffold, which demonstrated much improved hepatic-specific functions. In the current study, we were able to use this robust model to demonstrate that the DLX-DILI is a human CYP450 specific, metabolism-dependent, oxidative stress triggered complex hepatic injury. High-content imaging analysis (HCA) of organoids exposed to DLX showed that the potential toxicophore, naphthyl ring in DLX initiated oxidative stress which ultimately led to mitochondrial dysfunction in the hepatic organoids, and vice versa. Furthermore, DLX-induced hepatic steatosis and cholestasis was also detected in the exposed EHSs. We also discovered that a novel compound S-071031B, which replaced DLX's naphthyl ring with benzodioxole, showed dramatically lower hepatotoxicities through reducing oxidative stress. Thus, we conclusively present the human-relevant EHS model as an ideal, highly competent system for evaluating DLX induced hepatotoxicity and exploring related mechanisms in vitro. Moreover, HCA use on functional hepatic organoids has promising application prospects for guiding compound structural modifications and optimization in order to improve drug development by reducing hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury , Depressive Disorder, Major , Fatty Liver , Duloxetine Hydrochloride/pharmacology , Humans
18.
Psychopharmacology (Berl) ; 239(10): 3133-3143, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35882635

ABSTRACT

RATIONALE: It is known that both selective serotonin and serotonin noradrenaline reuptake inhibitors (SSRI, SNRI) are first-line drugs for the treatment of major depressive disorder. It has also been considered that both SSRI and SNRI can improve the symptoms of major depressive disorder by increasing the concentration of monoamine in the synaptic cleft based on the monoamine hypothesis. However, accumulating evidence has indicated that inflammation in the brain may be a key factor in the pathophysiological mechanisms that underlie the development of major depressive disorder. OBJECTIVES: It has been advocated that microglial cells may regulate the inflammatory response under pathological conditions such as major depressive disorder. In this study, we focused on whether duloxetine can ameliorate the inflammatory response induced by lipopolysaccharide (LPS) in BV-2 microglial cells. RESULTS: Our results indicated that duloxetine significantly decreased the NO production induced by LPS. The increase in the protein expression level of iNOS induced by LPS was significantly decreased by treatment with duloxetine. Moreover, the increases in the protein expression levels of phosphorylated-IκBα, phosphorylated-Akt and Akt induced by LPS were also significantly decreased. Unexpectedly, the protein expression levels of other pro-inflammatory factors such as COX-2 and the phosphorylation ratios for various molecules including IκBα and Akt were not changed by treatment with duloxetine. CONCLUSIONS: These findings suggest that duloxetine may have an anti-inflammatory effect, which could contribute to its therapeutic effectiveness for major depressive disorder.


Subject(s)
Depressive Disorder, Major , Serotonin and Noradrenaline Reuptake Inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Cyclooxygenase 2/metabolism , Depressive Disorder, Major/metabolism , Duloxetine Hydrochloride/pharmacology , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/pharmacology , Norepinephrine/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serotonin/metabolism , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology
19.
Biomed Pharmacother ; 152: 113262, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35691156

ABSTRACT

Transient receptor potential canonical 5 (TRPC5) is a polymodal, calcium-permeable, nonselective ion channel that is expressed in the brain and 75 % of human sensory neurons. Its pharmacological or genetic inhibition leads to the relief of neuropathic and inflammatory pain. The clinically approved drug duloxetine is superior to other serotonin and norepinephrine reuptake inhibitors at managing painful neuropathies, but it is not known why. Here we ask whether the TRPC5 receptor is modulated by duloxetine and may contribute to its analgesic effect. Electrophysiological measurements of heterologously expressed human TRPC5 in HEK293T cells were performed to evaluate the effect of duloxetine. The interaction site was identified by molecular docking and molecular dynamics simulations in combination with point mutagenesis. We found that duloxetine inhibits TRPC5 in a concentration-dependent manner with a high potency (IC50 = 0.54 ± 0.03 µM). Our data suggest that duloxetine binds into a voltage sensor-like domain. For the interaction, Glu418 exhibited particular importance due to putative hydrogen bond formation. Duloxetine effectively inhibits TRPC5 currents induced by cooling, voltage, direct agonists and by the stimulation of the PLC pathway. The finding that this TRPC5 inhibitor is widely used and well tolerated provides a scaffold for new pain treatment strategies.


Subject(s)
Pain , TRPC Cation Channels , Duloxetine Hydrochloride/pharmacology , HEK293 Cells , Humans , Molecular Docking Simulation , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
20.
Molecules ; 27(9)2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35566106

ABSTRACT

Here, we explored the possible interaction between duloxetine and SEP-363856 (SEP-856) in depression-related reactions. The results showed that oral administration of duloxetine showed powerful antidepressant-like effects in both the forced swimming test (FST) and the suspension tail test (TST). SEP-856 orally administered alone also exerted an antidepressant-like effect in FST and TST, especially at doses of 0.3, 1, and 10 mg/kg. In addition, duloxetine (15 mg/kg) and SEP-856 (15 mg/kg) both showed antidepressant-like effects in the sucrose preference test (SPT). Most importantly, in the above experiments, compared with duloxetine alone, the simultaneous use of duloxetine and SEP-856 caused a more significant antidepressant-like effect. It is worth noting that doses of drug combination in FST and TST did not change the motor activities of mice in the open-field test (OFT). Thus, duloxetine and SEP-856 seem to play a synergistic role in regulating depression-related behaviors and might be beneficial for refractory depression.


Subject(s)
Antidepressive Agents , Pyrans , Animals , Antidepressive Agents/pharmacology , Depression/drug therapy , Duloxetine Hydrochloride/pharmacology , Hindlimb Suspension/methods , Mice , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL
...