Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 486: 116950, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38701902

RESUMO

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.


Assuntos
Quimiocina CCL5 , Cloridrato de Duloxetina , Mucosa Gástrica , Indometacina , Proteínas Proto-Oncogênicas c-akt , Ratos Sprague-Dawley , Serotonina , Transdução de Sinais , Úlcera Gástrica , Fator A de Crescimento do Endotélio Vascular , Animais , Cloridrato de Duloxetina/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Mucosa Gástrica/metabolismo , Masculino , Indometacina/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quimiocina CCL5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ratos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Úlcera Gástrica/induzido quimicamente , Úlcera Gástrica/prevenção & controle , Úlcera Gástrica/patologia , Úlcera Gástrica/metabolismo , Serotonina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo
2.
J Neurosci ; 42(3): 405-415, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-34880120

RESUMO

Duloxetine, a serotonin and norepinephrine reuptake inhibitor, is the best-established treatment for painful chemotherapy-induced peripheral neuropathy (CIPN). While it is only effective in little more than half of patients, our ability to predict patient response remains incompletely understood. Given that stress exacerbates CIPN, and that the therapeutic effect of duloxetine is thought to be mediated, at least in part, via its effects on adrenergic mechanisms, we evaluated the contribution of neuroendocrine stress axes, sympathoadrenal and hypothalamic-pituitary-adrenal, to the effect of duloxetine in preclinical models of oxaliplatin- and paclitaxel-induced CIPN. Systemic administration of duloxetine, which alone had no effect on nociceptive threshold, both prevented and reversed mechanical hyperalgesia associated with oxaliplatin- and paclitaxel-CIPN. It more robustly attenuated oxaliplatin CIPN in male rats, while it was more effective for paclitaxel CIPN in females. Gonadectomy attenuated these sex differences in the effect of duloxetine. To assess the role of neuroendocrine stress axes in the effect of duloxetine on CIPN, rats of both sexes were submitted to adrenalectomy combined with fixed level replacement of corticosterone and epinephrine. While CIPN, in these rats, was of similar magnitude to that observed in adrenal-intact animals, rats of neither sex responded to duloxetine. Furthermore, duloxetine blunted an increase in corticosterone induced by oxaliplatin, and prevented the exacerbation of CIPN by sound stress. Our results demonstrate a role of neuroendocrine stress axes in duloxetine analgesia (anti-hyperalgesia) for the treatment of CIPN.SIGNIFICANCE STATEMENT Painful chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating dose-dependent and therapy-limiting side effect of many of the cytostatic drugs used to treat cancer (Argyriou et al., 2010; Marmiroli et al., 2017). Duloxetine is the only treatment for CIPN currently recommended by the American Society of Clinical Oncology (Hershman et al., 2014). In the present study, focused on elucidating mechanisms mediating the response of oxaliplatin- and paclitaxel-induced painful peripheral neuropathy to duloxetine, we demonstrate a major contribution to its effect of neuroendocrine stress axis function. These findings, which parallel the clinical observation that stress may impact response of CIPN to duloxetine (Taylor et al., 2007), open new approaches to the treatment of CIPN and other stress-associated pain syndromes.


Assuntos
Analgésicos/uso terapêutico , Antineoplásicos/efeitos adversos , Cloridrato de Duloxetina/uso terapêutico , Limiar da Dor/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Analgésicos/farmacologia , Animais , Antineoplásicos/uso terapêutico , Corticosterona/sangue , Cloridrato de Duloxetina/farmacologia , Feminino , Masculino , Oxaliplatina/efeitos adversos , Paclitaxel/efeitos adversos , Manejo da Dor , Doenças do Sistema Nervoso Periférico/sangue , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Ratos , Ratos Sprague-Dawley
3.
Cell Biochem Funct ; 41(7): 814-822, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37814477

RESUMO

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.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Cloridrato de Duloxetina/farmacologia , Fluvoxamina/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Hepáticas/patologia , Norepinefrina , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serotonina/metabolismo , Sertralina/farmacologia , Fator de Crescimento Transformador alfa/farmacologia , Fator de Crescimento Transformador alfa/metabolismo , Regulação para Cima
4.
Int J Mol Sci ; 24(9)2023 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-37176065

RESUMO

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.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Neuralgia , Camundongos , Animais , Paclitaxel/efeitos adversos , Cloridrato de Duloxetina/farmacologia , Cloridrato de Duloxetina/uso terapêutico , Rosuvastatina Cálcica/efeitos adversos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Medição da Dor , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Neuralgia/complicações , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Analgésicos/efeitos adversos
5.
FASEB J ; 35(2): e21329, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33484186

RESUMO

L1 syndrome is a rare developmental disorder characterized by hydrocephalus of varying severity, intellectual deficits, spasticity of the legs, and adducted thumbs. Therapy is limited to symptomatic relief. Numerous gene mutations in the L1 cell adhesion molecule (L1CAM, hereafter abbreviated L1) were identified in L1 syndrome patients, and those affecting the extracellular domain of this transmembrane type 1 glycoprotein show the most severe phenotypes. Previously analyzed rodent models of the L1 syndrome focused on L1-deficient animals or mouse mutants with abrogated cell surface expression of L1, making it difficult to test L1 function-triggering mimetic compounds with potential therapeutic value. To overcome this impasse, we generated a novel L1 syndrome mouse with a mutation of aspartic acid at position 201 in the extracellular part of L1 (p.D201N, hereafter termed L1-201) that displays a cell surface-exposed L1 accessible to the L1 mimetics. Behavioral assessment revealed an increased neurological deficit score and increased locomotor activity in male L1-201 mice carrying the mutation on the X-chromosome. Histological analyses of L1-201 mice showed features of the L1 syndrome, including enlarged ventricles and reduced size of the corpus callosum. Expression levels of L1-201 protein as well as extent of cell surface biotinylation and immunofluorescence labelling of cultured cerebellar neurons were normal. Importantly, treatment of these cultures with the L1 mimetic compounds duloxetine, crotamiton, and trimebutine rescued impaired cell migration and survival as well as neuritogenesis. Altogether, the novel L1 syndrome mouse model provides a first experimental proof-of-principle for the potential therapeutic value of L1 mimetic compounds.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X/tratamento farmacológico , Deficiência Intelectual/tratamento farmacológico , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Peptidomiméticos/uso terapêutico , Paraplegia Espástica Hereditária/tratamento farmacológico , Animais , Células Cultivadas , Cerebelo/citologia , Cerebelo/metabolismo , Cerebelo/patologia , Ventrículos Cerebrais/metabolismo , Ventrículos Cerebrais/patologia , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Cloridrato de Duloxetina/farmacologia , Cloridrato de Duloxetina/uso terapêutico , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Molécula L1 de Adesão de Célula Nervosa/genética , Neurogênese , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Peptidomiméticos/farmacologia , Fenótipo , Paraplegia Espástica Hereditária/genética , Paraplegia Espástica Hereditária/patologia , Toluidinas/farmacologia , Toluidinas/uso terapêutico , Trimebutina/farmacologia , Trimebutina/uso terapêutico
6.
Cell Mol Life Sci ; 79(1): 31, 2021 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-34936033

RESUMO

The benefits of current treatments for depression are limited by low response rates, delayed therapeutic effects, and multiple side effects. Antidepressants affect a variety of neurotransmitter systems in different areas of the brain, and the mechanisms underlying their convergent effects on behavior have been unclear. Here we identify hippocampal bone morphogenetic protein (BMP) signaling as a common downstream pathway that mediates the behavioral effects of five different antidepressant classes (fluoxetine, bupropion, duloxetine, vilazodone, trazodone) and of electroconvulsive therapy. All of these therapies decrease BMP signaling and enhance neurogenesis in the hippocampus. Preventing the decrease in BMP signaling blocks the effect of antidepressant treatment on behavioral phenotypes. Further, inhibition of BMP signaling in hippocampal newborn neurons is sufficient to produce an antidepressant effect, while chemogenetic silencing of newborn neurons prevents the antidepressant effect. Thus, inhibition of hippocampal BMP signaling is both necessary and sufficient to mediate the effects of multiple classes of antidepressants.


Assuntos
Antidepressivos/farmacologia , Proteínas Morfogenéticas Ósseas/metabolismo , Hipocampo/metabolismo , Transdução de Sinais , Envelhecimento/patologia , Animais , Ansiolíticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Giro Denteado/efeitos dos fármacos , Giro Denteado/metabolismo , Cloridrato de Duloxetina/farmacologia , Eletroconvulsoterapia , Fluoxetina/farmacologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/patologia , Hipocampo/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Estresse Psicológico/complicações , Trazodona/farmacologia , Cloridrato de Vilazodona/farmacologia
7.
Pharmacology ; 107(9-10): 472-479, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35584608

RESUMO

INTRODUCTION: The heat and redox-sensitive ion channel TRPM2 was reported to be a causative mechanism for depression in a mouse model and to be upregulated in the hippocampus in patients suffering from depressive disorders. TRPM2 may thus be a novel target for antidepressants, but so far, selective TRPM2-inhibitors have not yet been developed. In this in vitro study, we examined the inhibitory effects of several established antidepressants on heat-evoked inward currents of TRPM2. METHODS: Human (h) TRPM2 expressed in HEK293 cells was examined by means of whole-cell patch clamp recordings. Effects of duloxetine, amitriptyline, sertraline, fluoxetine, paroxetine, citalopram, escitalopram, ketamine, pregabalin, lidocaine, and QX-314 were explored on heat-evoked currents in cells pretreated with ADP-ribose (ADPR). RESULTS: While inward currents induced by 1 mM ADPR in the pipette solution displayed a strong rundown hampering pharmacological experiments, heat-evoked currents in cells loaded with 200 µM APDR remained stable upon repetitive activation. Among all substances examined, only inhibition induced by duloxetine displayed a clear concentration-dependency. Thirty micromolar duloxetine was required for 50% inhibition, the same degree of inhibition was also induced by 30 µM amitriptyline, fluoxetine, and paroxetine. While citalopram, escitalopram, ketamine, and pregabalin failed to robustly modify TRPM2, sertraline and low concentrations of lidocaine even potentiated heat-evoked currents. CONCLUSION: Our data indicate that some, but not all established antidepressants inhibit hTRPM2 when it is activated by heat and ADPR in vitro, e.g., presumably relevant endogenous agonists. However, none of the examined substances exhibited a potent inhibition which is likely to translate into a clinically relevant effect at effective plasma concentrations. Whether or not TRPM2 may be a relevant target for antidepressants cannot be conclusively assessed by a single in vitro study, thus further studies are required along these lines. Nevertheless, future studies may get simplified by the novel approach we developed for in vitro pharmacological analysis of TRPM2.


Assuntos
Ketamina , Canais de Cátion TRPM , Adenosina Difosfato Ribose/farmacologia , Amitriptilina/farmacologia , Animais , Antidepressivos/farmacologia , Citalopram/farmacologia , Cloridrato de Duloxetina/farmacologia , Fluoxetina/farmacologia , Células HEK293 , Temperatura Alta , Humanos , Lidocaína/farmacologia , Camundongos , Paroxetina , Pregabalina , Sertralina
8.
An Acad Bras Cienc ; 94(2): e20211021, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35544847

RESUMO

This study aimed to evaluate the effect of duloxetine hydrochloride (DH) on Cryptococcus neoformans. DH minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC) were 18.5 µg/mL, and the combination with fluconazole (FLZ) reduced the MIC value by 16-and 4-fold for DH and FLZ, respectively. The capsule size decreased by 67% ​​and 16% when treated with DH and DH with FLZ, respectively. Therefore, this study showed that DH is active against C. neoformans alone and in combination with FLZ, leading to the reduction of the capsule size of this yeast.


Assuntos
Cryptococcus neoformans , Fluconazol , Antifúngicos/farmacologia , Cloridrato de Duloxetina/farmacologia , Fluconazol/farmacologia , Testes de Sensibilidade Microbiana
9.
Int J Mol Sci ; 23(5)2022 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-35269729

RESUMO

While cardiovascular disease (CVD) is the leading cause of death, major depressive disorder (MDD) is the primary cause of disability, affecting more than 300 million people worldwide. Interestingly, there is evidence that CVD is more prevalent in people with MDD. It is well established that neurotransmitters, namely serotonin and norepinephrine, are involved in the biochemical mechanisms of MDD, and consequently, drugs targeting serotonin-norepinephrine reuptake, such as duloxetine, are commonly prescribed for MDD. In this connection, serotonin and norepinephrine are also known to play critical roles in primary hemostasis. Based on these considerations, we investigated if duloxetine can be repurposed as an antiplatelet medication. Our results-using human and/or mouse platelets show that duloxetine dose-dependently inhibited agonist-induced platelet aggregation, compared to the vehicle control. Furthermore, it also blocked agonist-induced dense and α-granule secretion, integrin αIIbß3 activation, phosphatidylserine expression, and clot retraction. Moreover duloxetine-treated mice had a significantly prolonged occlusion time. Finally, duloxetine was also found to impair hemostasis. Collectively, our data indicate that the antidepressant duloxetine, which is a serotonin-norepinephrine antagonist, exerts antiplatelet and thromboprotective effects and inhibits hemostasis. Consequently, duloxetine, or a rationally designed derivative, presents potential benefits in the context of CVD, including that associated with MDD.


Assuntos
Transtorno Depressivo Maior , Trombose , Animais , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Transtorno Depressivo Maior/tratamento farmacológico , Cloridrato de Duloxetina/farmacologia , Cloridrato de Duloxetina/uso terapêutico , Hemostasia , Humanos , Camundongos , Norepinefrina/farmacologia , Ativação Plaquetária , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Serotonina/farmacologia , Antagonistas da Serotonina/farmacologia , Trombose/tratamento farmacológico , Trombose/metabolismo , Trombose/prevenção & controle
10.
Molecules ; 27(9)2022 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-35566106

RESUMO

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.


Assuntos
Antidepressivos , Piranos , Animais , Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Cloridrato de Duloxetina/farmacologia , Elevação dos Membros Posteriores/métodos , Camundongos , Natação
11.
Biochem Cell Biol ; 99(5): 578-586, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33646885

RESUMO

Duloxetine, a selective serotonin-norepinephrine reuptake inhibitor, is currently recommended for the treatment of chronic painful disorders such as fibromyalgia, chronic musculoskeletal pain, and diabetic peripheral neuropathy. We previously demonstrated that bone morphogenetic protein-4 (BMP-4) stimulates osteoprotegerin (OPG) production in osteoblast-like MC3T3-E1 cells, and that p70 S6 kinase positively regulates OPG synthesis. The present study aimed to investigate the effect of duloxetine on BMP-4-stimulated OPG synthesis in these cells. Duloxetine dose-dependently suppressed OPG release stimulated by BMP-4. Fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), reduced BMP-4-stimulated OPG release, whereas a selective and specific norepinephrine reuptake inhibitor, reboxetine, failed to affect OPG release. In addition, another SSRI sertraline also inhibited BMP-4-stimulated OPG release. On the other hand, siRNA of SMAD1 reduced the OPG release stimulated by BMP-4, indicating the involvement of the SMAD1/5/8 pathway in OPG release. Rapamycin inhibited BMP-4-stimulated p70 S6 kinase phosphorylation, and compound C suppressed the SMAD1/5/8 phosphorylation stimulated by BMP-4. Duloxetine did not affect BMP-4-induced phosphorylation of p70 S6 kinase but suppressed SMAD1/5/8 phosphorylation. Both fluvoxamine and sertraline also inhibited BMP-4-elicited phosphorylation of SMAD1/5/8. These results strongly suggest that duloxetine suppresses BMP-4-stimulated OPG release via inhibition of the Smad1/5/8 signaling pathway in osteoblasts.


Assuntos
Proteína Morfogenética Óssea 4/antagonistas & inibidores , Cloridrato de Duloxetina/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoprotegerina/antagonistas & inibidores , Células 3T3 , Animais , Proteína Morfogenética Óssea 4/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Camundongos , Osteoblastos/metabolismo , Osteoprotegerina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/metabolismo , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/metabolismo , Proteína Smad8/antagonistas & inibidores , Proteína Smad8/metabolismo
12.
Hum Brain Mapp ; 42(8): 2593-2605, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33638263

RESUMO

Previous neuroimaging studies have revealed abnormal functional connectivity of brain networks in patients with major depressive disorder (MDD), but findings have been inconsistent. A recent big-data study found abnormal intrinsic functional connectivity within the default mode network in patients with recurrent MDD but not in first-episode drug-naïve patients with MDD. This study also provided evidence for reduced default mode network functional connectivity in medicated MDD patients, raising the question of whether previously observed abnormalities may be attributable to antidepressant effects. The present study (ClinicalTrials.gov identifier: NCT03294525) aimed to disentangle the effects of antidepressant treatment from the pathophysiology of MDD and test the medication normalization hypothesis. Forty-one first-episode drug-naïve MDD patients were administrated antidepressant medication (escitalopram or duloxetine) for 8 weeks, with resting-state functional connectivity compared between posttreatment and baseline. To assess the replicability of the big-data finding, we also conducted a cross-sectional comparison of resting-state functional connectivity between the MDD patients and 92 matched healthy controls. Both Network-Based Statistic analyses and large-scale network analyses revealed intrinsic functional connectivity decreases in extensive brain networks after treatment, indicating considerable antidepressant effects. Neither Network-Based Statistic analyses nor large-scale network analyses detected significant functional connectivity differences between treatment-naïve patients and healthy controls. In short, antidepressant effects are widespread across most brain networks and need to be accounted for when considering functional connectivity abnormalities in MDD.


Assuntos
Antidepressivos/farmacologia , Córtex Cerebral , Conectoma , Rede de Modo Padrão , Transtorno Depressivo Maior , Rede Nervosa , Adulto , Antidepressivos/administração & dosagem , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiopatologia , Rede de Modo Padrão/diagnóstico por imagem , Rede de Modo Padrão/efeitos dos fármacos , Rede de Modo Padrão/fisiopatologia , Transtorno Depressivo Maior/diagnóstico por imagem , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/fisiopatologia , Cloridrato de Duloxetina/farmacologia , Escitalopram/farmacologia , Feminino , Seguimentos , Humanos , Imageamento por Ressonância Magnética , Masculino , Rede Nervosa/diagnóstico por imagem , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiopatologia , Resultado do Tratamento , Adulto Jovem
13.
Pharmacol Res ; 174: 105921, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34601079

RESUMO

Neurotoxicity is a frequent side effect of cisplatin (CisPt)-based anticancer therapy whose pathophysiology is largely vague. Here, we exploited C. elegans as a 3R-compliant in vivo model to elucidate molecular mechanisms contributing to CisPt-induced neuronal dysfunction. To this end, we monitored the impact of CisPt on various sensory functions as well as pharyngeal neurotransmission by recording electropharyngeograms (EPGs). CisPt neither affected food and odor sensation nor mechano-sensation, which involve dopaminergic and glutaminergic neurotransmission. However, CisPt reduced serotonin-regulated pharyngeal pumping activity independent of changes in the morphology of related neurons. CisPt-mediated alterations in EPGs were fully rescued by addition of serotonin (5-HT) (≤ 2 mM). Moreover, the CisPt-induced pharyngeal injury was prevented by co-incubation with the clinically approved serotonin re-uptake inhibitory drug duloxetine. A protective effect of 5-HT was also observed with respect to CisPt-mediated impairment of another 5-HT-dependent process, the egg laying activity. Importantly, CisPt-induced apoptosis in the gonad and learning disability were not influenced by 5-HT. Using different C. elegans mutants we found that CisPt-mediated (neuro)toxicity is independent of serotonin biosynthesis and re-uptake and likely involves serotonin-receptor subtype 7 (SER-7)-related functions. In conclusion, by measuring EPGs as a surrogate parameter of neuronal dysfunction, we provide first evidence that CisPt-induced neurotoxicity in C. elegans involves 5-HT-dependent neurotransmission and SER-7-mediated signaling mechanisms and can be prevented by the clinically approved antidepressant duloxetine. The data highlight the particular suitability of C. elegans as a 3R-conform in vivo model in molecular (neuro)toxicology and, moreover, for the pre-clinical identification of neuroprotective candidate drugs.


Assuntos
Antineoplásicos/toxicidade , Caenorhabditis elegans/efeitos dos fármacos , Cisplatino/toxicidade , Modelos Animais de Doenças , Síndromes Neurotóxicas/metabolismo , Serotonina/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Cloridrato de Duloxetina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Síndromes Neurotóxicas/genética , Síndromes Neurotóxicas/fisiopatologia , Faringe/efeitos dos fármacos , Faringe/fisiologia , Inibidores da Recaptação de Serotonina e Norepinefrina/farmacologia , Transmissão Sináptica/efeitos dos fármacos
14.
Eur Arch Psychiatry Clin Neurosci ; 271(3): 521-526, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32715345

RESUMO

It is sometimes clinically believed that major depression with melancholic features is more responsive to antidepressants than non-melancholic depression. Proper analysis and, therefore, valid evidence to support or refute this common clinical lore is lacking. The sample was taken from three placebo-controlled randomized trials of duloxetine, escitalopram and paroxetine (n = 1219). We conducted a two-step individual participant data meta-analysis to combine linear mixed-effects regressions modeling melancholic features as prognostic factor (variable that predicts overall response regardless of the treatments) and as effect modifier (variable that predict differential response to drug over placebo). Melancholic features represented a statistically significant prognostic factor for greater reduction in depression severity both on antidepressants and on placebo, especially after 4 weeks of treatment. However, they were not an effect modifier of the antidepressant treatment through the acute phase treatment: in other words. The superiority of antidepressants over placebo was not influenced by the melancholic features. The treatment decision-making as to the benefits of antidepressant treatment for patients with major depression should not be influenced by the presence or absence of melancholic features.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/fisiopatologia , Avaliação de Resultados em Cuidados de Saúde , Adulto , Citalopram/farmacologia , Cloridrato de Duloxetina/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Avaliação de Resultados em Cuidados de Saúde/estatística & dados numéricos , Paroxetina/farmacologia , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Adulto Jovem
15.
Biol Pharm Bull ; 44(2): 271-274, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33518680

RESUMO

The anticancer agents including oxaliplatin, paclitaxel, and bortezomib cause severe peripheral neuropathy. The Kampo medicine Sokeikakketsuto (SOKT) has been widely used to treat several types of pain. In this study, the analgesic effects of SOKT on oxaliplatin-, paclitaxel-, and bortezomib-induced peripheral neuropathy were investigated in rat models. Rats were treated with oxaliplatin (4 mg/kg, intraperitoneally (i.p.), twice a week for four weeks), paclitaxel (4 mg/kg, i.p., twice a week for two weeks), or bortezomib (0.2 mg/kg, i.p., twice a week for two weeks). SOKT (0.3 or 1.0 g/kg) or duloxetine hydrochloride (30 mg/kg, as a positive control) was administered orally after neuropathy developed. Mechanical allodynia and cold hyperalgesia were assessed using the von Frey test and the acetone test, respectively. These tests were performed immediately before and 30, 60, 90, and 120 min after the administration of the drugs. Repeated treatment of oxaliplatin induced mechanical allodynia and cold hyperalgesia. A single administration of SOKT (1 g/kg, per os (p.o.)), as well as duloxetine, temporarily reversed both the mechanical allodynia and the cold hyperalgesia. Repeated administration of paclitaxel and bortezomib also induced the mechanical allodynia. SOKT and duloxetine reversed the mechanical allodynia caused by bortezomib, but not by paclitaxel. SOKT might have the potential to become a new drug to relieve the symptom of oxaliplatin- or bortezomib-induced peripheral neuropathy.


Assuntos
Analgésicos/farmacologia , Antineoplásicos/efeitos adversos , Temperatura Baixa/efeitos adversos , Medicamentos de Ervas Chinesas/farmacologia , Hiperalgesia/tratamento farmacológico , Analgésicos/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Bortezomib/administração & dosagem , Bortezomib/efeitos adversos , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/uso terapêutico , Cloridrato de Duloxetina/farmacologia , Cloridrato de Duloxetina/uso terapêutico , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/diagnóstico , Masculino , Medicina Kampo/métodos , Oxaliplatina/administração & dosagem , Oxaliplatina/efeitos adversos , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Medição da Dor , Ratos , Ratos Sprague-Dawley
16.
Molecules ; 26(3)2021 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-33503911

RESUMO

BACKGROUND: Wide use of oxaliplatin as an antitumor drug is limited by severe neuropathy with pharmacoresistant cold hypersensitivity as the main symptom. Novel analgesics to attenuate cold hyperalgesia and new methods to detect drug candidates are needed. METHODS: We developed a method to study thermal preference of oxaliplatin-treated mice and assessed analgesic activity of intraperitoneal duloxetine and pregabalin used at 30 mg/kg. A prototype analgesiameter and a broad range of temperatures (0-45 °C) were used. Advanced methods of image analysis (deep learning and machine learning) enabled us to determine the effectiveness of analgesics. The loss or reversal of thermal preference of oxaliplatin-treated mice was a measure of analgesia. RESULTS: Duloxetine selectively attenuated cold-induced pain at temperatures between 0 and 10 °C. Pregabalin-treated mice showed preference towards a colder plate of the two used at temperatures between 0 and 45 °C. CONCLUSION: Unlike duloxetine, pregabalin was not selective for temperatures below thermal preferendum. It influenced pain sensation at a much wider range of temperatures applied. Therefore, for the attenuation of cold hypersensitivity duloxetine seems to be a better than pregabalin therapeutic option. We propose wide-range measurements of thermal preference as a novel method for the assessment of analgesic activity in mice.


Assuntos
Analgésicos/farmacologia , Hiperalgesia/tratamento farmacológico , Dor/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Temperatura Baixa , Modelos Animais de Doenças , Cloridrato de Duloxetina/farmacologia , Temperatura Alta , Masculino , Camundongos , Oxaliplatina/farmacologia , Medição da Dor/métodos , Pregabalina/farmacologia , Temperatura
17.
Molecules ; 26(12)2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-34208184

RESUMO

The antitumor drug, oxaliplatin, induces neuropathic pain, which is resistant to available analgesics, and novel mechanism-based therapies are being evaluated for this debilitating condition. Since activated microglia, impaired serotonergic and noradrenergic neurotransmission and overexpressed sodium channels are implicated in oxaliplatin-induced pain, this in vivo study assessed the effect of minocycline, a microglial activation inhibitor used alone or in combination with ambroxol, a sodium channel blocker, or duloxetine, a serotonin and noradrenaline reuptake inhibitor, on oxaliplatin-induced tactile allodynia and cold hyperalgesia. To induce neuropathic pain, a single dose (10 mg/kg) of intraperitoneal oxaliplatin was used. The mechanical and cold pain thresholds were assessed using mouse von Frey and cold plate tests, respectively. On the day of oxaliplatin administration, only duloxetine (30 mg/kg) and minocycline (100 mg/kg) used alone attenuated both tactile allodynia and cold hyperalgesia 1 h and 6 h after administration. Minocycline (50 mg/kg), duloxetine (10 mg/kg) and combined minocycline + duloxetine influenced only tactile allodynia. Seven days after oxaliplatin, tactile allodynia (but not cold hyperalgesia) was attenuated by minocycline (100 mg/kg), duloxetine (30 mg/kg) and combined minocycline and duloxetine. These results indicate a potential usefulness of minocycline used alone or combination with duloxetine in the treatment of oxaliplatin-induced pain.


Assuntos
Cloridrato de Duloxetina/farmacologia , Microglia/efeitos dos fármacos , Minociclina/farmacologia , Neuralgia/tratamento farmacológico , Oxaliplatina/toxicidade , Limiar da Dor/efeitos dos fármacos , Analgésicos/farmacologia , Animais , Antibacterianos/farmacologia , Antineoplásicos/toxicidade , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Combinação de Medicamentos , Masculino , Camundongos , Microglia/fisiologia , Neuralgia/induzido quimicamente , Neuralgia/patologia
18.
Mol Pain ; 16: 1744806920938237, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32600103

RESUMO

The Merkel disc is a main type of tactile end organs formed by Merkel cells and Aß-afferent endings as first tactile sensory synapses. They are highly abundant in fingertips, touch domes, and whisker hair follicles of mammals and are essential for sensory tasks including social interaction, environmental exploration, and tactile discrimination. We have recently shown that Merkel discs use serotonin to transmit tactile signals from Merkel cells to Aß-afferent endings to drive slowly adapting type 1 impulses on the Aß-afferent nerves. This raises a question as whether the serotoninergic transmission at Merkel discs may be regulated by serotonin transporters and whether serotonin transporter inhibitors may affect the tactile transmission. Here, we made recordings from whisker afferent nerves of mouse whisker hair follicles and tested the effects of monoamine transporter inhibitors on slowly adapting type 1 impulses. We show that methamphetamine, a monoamine releasing facilitator and reuptake inhibitor, elicited spontaneous impulses as well as increased the numbers of slowly adapting type 1 impulses elicited by whisker hair deflections. S-duloxetine, a potent inhibitor of transporters of serotonin and norepinephrine, and fluoxetine, a selective inhibitor of serotonin transporters, both also increased the numbers of slowly adapting type 1 impulses. Prolonged treatment of whisker hair follicles with methamphetamine abolished most of slowly adapting type 1 impulses. Furthermore, the treatment of whisker hair follicles with methamphetamine resulted in serotonin release from whisker hair follicles. Taken together, our results suggest that serotonin transporters play a role in regulating tactile transmission at Merkel discs.


Assuntos
Folículo Piloso/fisiologia , Células de Merkel/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Tato/fisiologia , Vibrissas/fisiologia , Animais , Cloridrato de Duloxetina/farmacologia , Fluoxetina/farmacologia , Folículo Piloso/efeitos dos fármacos , Células de Merkel/efeitos dos fármacos , Metanfetamina/farmacologia , Camundongos Endogâmicos C57BL , Serotonina/metabolismo , Vibrissas/efeitos dos fármacos
19.
J Pharmacol Exp Ther ; 373(3): 453-462, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32238452

RESUMO

Paclitaxel (PTX) is an antineoplastic drug extracted from the Taxus species, and peripheral neuropathy is a common side effect. Paclitaxel-induced peripheral neuropathy (PIPN) seriously affects patient quality of life. Currently, the mechanism of PIPN is still unknown, and few treatments are recognized clinically. Duloxetine is recommended as the only potential treatment of chemotherapy-induced peripheral neuropathy (CIPN) by the American Society of Clinical Oncology. However, this guidance lacks a theoretical basis and experimental evidence. Our study suggested that duloxetine could improve PIPN and provide neuroprotection. We explored the potential mechanisms of duloxetine on PIPN. As a result, duloxetine acts by inhibiting poly ADP-ribose polymerase cleavage (PARP) and tumor suppressor gene p53 activation and regulating apoptosis regulator the Bcl2 family to reverse PTX-induced oxidative stress and apoptosis. Taken together, the present study shows that using duloxetine to attenuate PTX-induced peripheral nerve injury and peripheral pain may provide new clinical therapeutic targets for CIPN. SIGNIFICANCE STATEMENT: This study reported that duloxetine significantly alleviates neuropathic pain induced by paclitaxel and is related to poly ADP-ribose polymerase (PARP), tumor suppressor gene p53, and apoptosis regulator the Bcl2 family. Our findings thus not only provide important guidance to support duloxetine to become the first standard chemotherapy-induced peripheral neuropathy (CIPN) drug but also will find potential new targets and positive control for new CIPN drug development.


Assuntos
Cloridrato de Duloxetina/farmacologia , Paclitaxel/farmacologia , Traumatismos dos Nervos Periféricos/induzido quimicamente , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Traumatismos dos Nervos Periféricos/metabolismo , Ratos , Ratos Sprague-Dawley
20.
Prostaglandins Other Lipid Mediat ; 151: 106481, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33002595

RESUMO

Duloxetine, a serotonin-norepinephrine reuptake inhibitor, is currently recommended as a useful medicine to chronic pain including low back pain. However, as the analogy of classical selective serotonin reuptake inhibitors, there is a concern to deteriorate osteoporosis with remaining to clarify the exact mechanism of duloxetine in bone metabolism. We have previously reported that prostaglandin E1 (PGE1) induces the synthesis of both osteoprotegerin (OPG) and interleukin-6 (IL-6), essential regulators of bone metabolism, in osteoblast-like MC3T3-E1 cells. Based upon them, we herein investigated the mechanism whereby the effect of duloxetine on the synthesis of OPG and IL-6 induced by PGE1 in these cells. Duloxetine enhanced the release from MC3T3-E1 cells of both OPG and IL-6 stimulated by PGE1. However, reboxetine, a selective and specific inhibitor of norepinephrine reuptake, failed to affect the PGE1-induced release of OPG or IL-6. Oppositely, fluvoxamine and sertraline, agents belonging to the class of selective serotonin reuptake inhibitor, upregulated the PGE1-stimulated release of both OPG and IL-6. Duloxetine amplified the expression of OPG mRNA and IL-6 mRNA stimulated by PGE1. Duloxetine strengthened the PGE1-induced p38 MAP kinase phosphorylation, which was amplified by fluvoxamine as well. SB203880, an inhibitor of p38 MAP kinase, suppressed the amplifying effects by duloxetine or fluvoxamine on the PGE1-stimulated release of OPG and IL-6. These results strongly suggest that duloxetine could strengthen osteoblast activation by PGE1 through the upregulation of p38 MAP kinase, leading to increasing the synthesis of OPG and IL-6.


Assuntos
Alprostadil/farmacologia , Cloridrato de Duloxetina/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células 3T3 , Animais , Interações Medicamentosas , Camundongos , Osteoblastos/citologia , Fosforilação/efeitos dos fármacos , Resveratrol/farmacologia , Ativação Transcricional/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA