Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38.868
Filtrar
1.
Med Sci Monit ; 30: e945595, 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39267297

RESUMO

BACKGROUND Robot-assisted laparoscopic partial nephrectomy (RAPN) has been increasingly used for treating renal tumors due to its advantages over other approaches. However, RAPN can induce acute incisional, peritoneal, visceral, and referred pain. Therefore, acute pain control in robotic surgery is a concern. This retrospective study aimed to evaluate the efficacy of intrathecal morphine (ITM) for postoperative analgesia and recovery after RAPN. MATERIAL AND METHODS We retrospectively investigated consecutive patients who underwent RAPN at our institute between 2020 and 2021. Among the 272 patients who met the inclusion criteria, 135 patients were administered 200 µg of ITM preoperatively (ITM group), while 137 patients were not (control group). Postoperative pain assessments using the numeric rating scale (NRS), opioid requirements, and recovery profiles during the first postoperative 24 h were compared between the 2 groups. RESULTS As the primary endpoint, the incidence of moderate-to-severe pain (24-h average NRS pain score ≥4) was significantly lower in the ITM group than in the control group (36.3% vs 61.3%, P<0.001). Pain scores and cumulative opioid requirements were also significantly lower in the ITM group for all assessments (P<0.001). Moreover, the ITM group had a higher score on the Quality of Recovery-15 questionnaire on the first postoperative day (129 vs 120, P=0.003) despite an increased rate of postoperative nausea/vomiting (27.4% vs 13.1%, P=0.003). CONCLUSIONS Our findings indicate that ITM provided superior pain control during the early period following RAPN, with reduced postoperative opioid requirements. Moreover, ITM improved patient satisfaction with recovery.


Assuntos
Analgésicos Opioides , Injeções Espinhais , Laparoscopia , Morfina , Nefrectomia , Dor Pós-Operatória , Procedimentos Cirúrgicos Robóticos , Humanos , Masculino , Feminino , Morfina/administração & dosagem , Morfina/uso terapêutico , Nefrectomia/métodos , Dor Pós-Operatória/tratamento farmacológico , Estudos Retrospectivos , Pessoa de Meia-Idade , Laparoscopia/métodos , Procedimentos Cirúrgicos Robóticos/métodos , Injeções Espinhais/métodos , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/uso terapêutico , Idoso , Manejo da Dor/métodos , Medição da Dor/métodos , Adulto , Neoplasias Renais/cirurgia
2.
Sci Rep ; 14(1): 21040, 2024 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-39251806

RESUMO

Long-term use of opioid drugs such as morphine can induce addiction in the central nervous system through dysregulation of the reward system of the brain. Deep brain stimulation (DBS) is a non-pharmacological technique capable of attenuating behavioral responses associated with opioid drug consumption and possesses the capability to selectively activate and target localized brain regions with a high spatial resolution. However, long-term implantation of electrodes in brain tissue may limit the effectiveness of DBS due to changes in impedance, position, and shape of the tip of the stimulation electrode and the risk of infection of nerve tissue around the implanted electrode. The main objective of the current study is to evaluate the effect of temporal interference (TI) brain stimulation on addictive behaviors of morphine-induced conditioned place preference (CPP) in rats. TI stimulation is a non-invasive technique used transcranially to modulate neural activity within targeted brain regions. It involves applying two high-frequency currents with slightly different frequencies, resulting in interference and targeted stimulation of different brain areas with the desired spatial resolution. The results indicated that TI stimulation with the amplitude of I 1 = I 2 = 0.5 mA, carrier frequency of 2 kHz, frequency difference of 25 Hz, ON-OFF stimulation frequency of 0.25 Hz, and total duration of 10 min in three consecutive days resulted in a significant reduction of morphine preference in the morphine-stimulation group in comparison with the morphine group (p < 0.001). These findings highlight the potential of TI stimulation as a modulatory intervention in mitigating the addictive properties of morphine and provide valuable insights into the therapeutic implications of this stimulation paradigm for treatment of opioid drugs in human subjects.


Assuntos
Estimulação Encefálica Profunda , Morfina , Animais , Morfina/farmacologia , Ratos , Estimulação Encefálica Profunda/métodos , Masculino , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Analgésicos Opioides/farmacologia , Ratos Sprague-Dawley
3.
Sci Rep ; 14(1): 21130, 2024 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-39256509

RESUMO

Morphine has been suggested to affect cancer cell dynamics and decrease survival rates in lung cancer patients at specific doses, but the precise mechanisms poorly understood. In this study, we aimed to investigate the molecular mechanisms by which morphine modulates the malignant characteristics of non-small cell lung cancer. Cell proliferation was assessed via the Cell Counting Kit-8 assay, and cell migration and invasion were examined via wound healing and Transwell assays. We employed immunofluorescence staining to evaluate E-cadherin expression in A549 and Lewis lung cancer (LLC) cell lines and immunohistochemistry to evaluate E-cadherin expression in nude mice tumours. Additionally, the in vivo effects of morphine on lung cancer progression were explored in a xenograft tumour experiments, in which naloxone was used as a morphine antagonist. Western blot analysis was performed to detect E-cadherin, phosphorylated mTOR (p-mTOR), mTOR, phosphorylated AKT (p-AKT), AKT, phosphorylated PI3K (p-PI3K), and PI3K protein levels in A549 and LLC cells as well as in tumour samples. Morphine (10 µM) significantly increased the proliferation of A549 and LLC cells in vitro (p < 0.05). It also enhanced the migratory and invasive capacities of these cell lines (p < 0.01). Mechanistically, morphine treatment (10 µM) led to a reduction in the expression of E-cadherin, and an increase in the phosphorylation of PI3K, AKT, and mTOR in A549 and LLC cells (p < 0.01). Morphine treatment (1.5 mg/kg) also reduced E-cadherin expression in xenograft tumours and promoted tumour growth in vivo (p < 0.05). This effect was reversed by naloxone (0.1 mg/kg). The results demonstrated that morphine stimulates the malignant proliferation of A549 and LLC cell lines and promotes xenograft tumour growth. Perhaps by specifically targeting MOR, morphine triggers a signalling cascade that activates the PI3K/AKT/mTOR pathway while inhibiting the EMT marker E-cadherin, which may consequently promote the progression of lung cancer.


Assuntos
Caderinas , Carcinoma Pulmonar de Células não Pequenas , Movimento Celular , Proliferação de Células , Neoplasias Pulmonares , Morfina , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Serina-Treonina Quinases TOR , Caderinas/metabolismo , Humanos , Serina-Treonina Quinases TOR/metabolismo , Animais , Morfina/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Transdução de Sinais/efeitos dos fármacos , Camundongos , Proliferação de Células/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Camundongos Nus , Células A549 , Progressão da Doença , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Regulação para Baixo/efeitos dos fármacos , Naloxona/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Masculino
4.
BMC Anesthesiol ; 24(1): 322, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39256675

RESUMO

BACKGROUND: The study aimed to compare the pain-relieving effectiveness of anterior quadratus lumborum block (QLB3) and erector spinae plane block (ESPB), both of which have been documented to provide relief during abdominal surgery. METHODS: This prospective observational study, conducted between February and July 2023, included 96 patients who had undergone percutaneous nephrolithotomy (PCNL). Patients were divided into three groups: QLB3, ESPB, and control (no block) and received the corresponding nerve block in the preanesthetic room for regional block. Cumulative morphine consumption during the initial 24 h after PCNL, numerical rating scale resting/movement scores, intraoperative remifentanil usage, rescue analgesic requirements, time when the first analgesic was requested, and postoperative nausea and vomiting scores were documented and compared between the groups. RESULTS: Total median morphine consumption in the first 24 h postoperatively was similar in the QLB3 and ESPB groups but higher in the control group (QLB3, 7 mg [(Q1-Q3) 7-8.5]; ESPB, 8 mg [6.5-9]; control, 12.5 [10-17]; P < 0.001). Similarly, median intraoperative remifentanil consumption did not differ between the block groups but was higher in the control group (QLB3, 1082 µg [IQR 805.5-1292.7]; ESPB, 1278 µg [940.2-1297.5]; control, 1561 µg [1315-2068]; P < 0.001). The number of patients receiving rescue analgesic medication was similar in the block groups but higher in the control group (QLB3, n = 9 [30%]; ESPB, n = 14 [46.7%]; control, n = 21 [70%]; P = 0.008). CONCLUSIONS: QLB3 and ESPB were adequate and comparable in providing postoperative analgesia as part of multimodal analgesia after PCNL. TRIAL REGISTRATION: The study was registered on ClinicalTrials.gov (Identifier: NCT05822492).


Assuntos
Analgésicos Opioides , Nefrolitotomia Percutânea , Bloqueio Nervoso , Dor Pós-Operatória , Humanos , Estudos Prospectivos , Dor Pós-Operatória/prevenção & controle , Bloqueio Nervoso/métodos , Masculino , Feminino , Pessoa de Meia-Idade , Nefrolitotomia Percutânea/métodos , Analgésicos Opioides/administração & dosagem , Adulto , Músculos Paraespinais , Morfina/administração & dosagem , Dor Aguda/prevenção & controle , Músculos Abdominais/inervação , Idoso
5.
Sci Prog ; 107(3): 368504241264994, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39228316

RESUMO

Millions of people suffer from opioid use disorder, because of the ongoing opioid epidemic. The aversive symptoms of withdrawal are a leading factor for drug relapses, yet there are limited therapeutic options to minimize or prevent withdrawal symptoms. The mechanism behind opioid withdrawal is still not fully understood, thus preventing the development of new therapeutics. This study is an extension of our previously proposed mechanism of a toll-like receptor 2 (TLR2) mediated withdrawal response as a result of morphine induced microbial change that occurs during morphine withdrawal. Transcriptome analysis of the pre-frontal cortex indicated that there was increased expression of genes related to TLR2 signaling in morphine withdrawal treated animals compared to placebo controls. Antibiotic treatment further altered TLR2 related genes, recovering some of the morphine induced effect and leading to additional suppression of some genes related to the TLR2 pathway. Morphine withdrawal induced gene expression was attenuated in a whole body TLR2 knockout model. These results provide more support that TLR2 plays an integral role in morphine withdrawal mechanisms and could be a potential therapeutic target to minimize opioid withdrawal associated co-morbidities.


Assuntos
Morfina , Córtex Pré-Frontal , Transdução de Sinais , Síndrome de Abstinência a Substâncias , Receptor 2 Toll-Like , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Síndrome de Abstinência a Substâncias/genética , Síndrome de Abstinência a Substâncias/metabolismo , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Animais , Transdução de Sinais/efeitos dos fármacos , Camundongos , Masculino , Camundongos Knockout , Camundongos Endogâmicos C57BL , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Dependência de Morfina/genética , Dependência de Morfina/metabolismo
6.
BMC Anesthesiol ; 24(1): 305, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223470

RESUMO

BACKGROUND: Open liver resection necessitates a substantial upper abdominal inverted-L incision, resulting in severe pain and compromising patient recovery. Despite the efficacy of epidural analgesia in providing adequate postoperative analgesia, the potential epidural-related adverse effects should be carefully considered. This study aims to compare the efficacy and safety of continuous epidural analgesia and intravenous analgesia in open liver resection. METHODS: A retrospective study was conducted, collecting data from patients who underwent open liver resection between 2007 and 2017. Propensity score matching was implemented to mitigate confounding variables, with patients being matched in a 1:1 ratio based on propensity scores. The primary outcome was the comparison of postoperative morphine consumption at 24, 48, and 72 hours between the two groups. Secondary outcomes included pain scores, postoperative outcomes, and epidural-related adverse effects. RESULTS: A total of 612 patients were included, and after matching, there were 204 patients in each group. Opioid consumption at 24, 48, and 72 hours postoperatively was statistically lower in the epidural analgesia group compared to the intravenous analgesia group (p < 0.001). However, there was no significant difference in pain scores (p = 0.422). Additionally, perioperative hypotension requiring treatment, as well as nausea and vomiting, were significantly higher in the epidural analgesia group compared to the intravenous analgesia group (p < 0.001). CONCLUSIONS: Epidural analgesia is superior to intravenous morphine in terms of reducing postoperative opioid consumption within the initial 72 h after open liver resection. Nevertheless, perioperative hypotension, which necessitates management, should be approached with consideration and vigilance. TRIAL REGISTRATION: The study was registered in the Clinical Trials Registry at www. CLINICALTRIALS: gov/ , NCT number: NCT06301932.


Assuntos
Analgesia Epidural , Analgésicos Opioides , Hepatectomia , Morfina , Dor Pós-Operatória , Pontuação de Propensão , Humanos , Analgesia Epidural/métodos , Feminino , Masculino , Estudos Retrospectivos , Dor Pós-Operatória/prevenção & controle , Dor Pós-Operatória/tratamento farmacológico , Pessoa de Meia-Idade , Hepatectomia/métodos , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/uso terapêutico , Morfina/administração & dosagem , Idoso , Resultado do Tratamento , Adulto
7.
J Neurophysiol ; 132(3): 968-978, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39110512

RESUMO

How cellular adaptations give rise to opioid analgesic tolerance to opioids like morphine is not well understood. For one, pain is a complex phenomenon comprising both sensory and affective components, largely mediated through separate circuits. Glutamatergic projections from the medial thalamus (MThal) to the anterior cingulate cortex (ACC) are implicated in processing of affective pain, a relatively understudied component of the pain experience. The goal of this study was to determine the effects of chronic morphine exposure on mu-opioid receptor (MOR) signaling on MThal-ACC synaptic transmission within the excitatory and feedforward inhibitory pathways. Using whole cell patch-clamp electrophysiology and optogenetics to selectively target these projections, we measured morphine-mediated inhibition of optically evoked postsynaptic currents in ACC layer V pyramidal neurons in drug-naïve and chronically morphine-treated mice. We found that morphine perfusion inhibited the excitatory and feedforward inhibitory pathways similarly in females but caused greater inhibition of the inhibitory pathway in males. Chronic morphine treatment robustly attenuated morphine presynaptic inhibition within the inhibitory pathway in males, but not females, and mildly attenuated presynaptic inhibition within the excitatory pathway in both sexes. These effects were not observed in MOR phosphorylation-deficient mice. This study indicates that chronic morphine treatment induces cellular tolerance to morphine within a thalamo-cortical circuit relevant to pain and opioid analgesia. Furthermore, it suggests this tolerance may be driven by MOR phosphorylation. Overall, these findings improve our understanding of how chronic opioid exposure alters cellular signaling in ways that may contribute to opioid analgesic tolerance.NEW & NOTEWORTHY Opioid signaling within the anterior cingulate cortex (ACC) is important for opioid modulation of affective pain. Glutamatergic medial thalamus (MThal) neurons synapse in the ACC and opioids, acting through mu opioid receptors (MORs), acutely inhibit synaptic transmission from MThal synapses. However, the effect of chronic opioid exposure on MThal-ACC synaptic transmission is not known. Here, we demonstrate that chronic morphine treatment induces cellular tolerance at these synapses in a sex-specific and phosphorylation-dependent manner.


Assuntos
Analgésicos Opioides , Morfina , Receptores Opioides mu , Tálamo , Animais , Receptores Opioides mu/metabolismo , Morfina/farmacologia , Morfina/administração & dosagem , Masculino , Feminino , Camundongos , Analgésicos Opioides/farmacologia , Analgésicos Opioides/administração & dosagem , Tálamo/efeitos dos fármacos , Tálamo/fisiologia , Tálamo/metabolismo , Giro do Cíngulo/efeitos dos fármacos , Giro do Cíngulo/fisiologia , Giro do Cíngulo/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Tolerância a Medicamentos/fisiologia , Camundongos Endogâmicos C57BL , Caracteres Sexuais , Transdução de Sinais/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia
9.
Addict Biol ; 29(8): e13429, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39109814

RESUMO

The endocannabinoid system interacts with the reward system to modulate responsiveness to natural reinforcers, as well as drugs of abuse. Previous preclinical studies suggested that direct blockade of CB1 cannabinoid receptors (CB1R) could be leveraged as a potential pharmacological approach to treat substance use disorder, but this strategy failed during clinical trials due to severe psychiatric side effects. Alternative strategies have emerged to circumvent the side effects of direct CB1 binding through the development of allosteric modulators. We hypothesized that negative allosteric modulation of CB1R signalling would reduce the reinforcing properties of morphine and decrease behaviours associated with opioid misuse. By employing intravenous self-administration in mice, we studied the effects of GAT358, a functionally-biased CB1R negative allosteric modulator (NAM), on morphine intake, relapse-like behaviour and motivation to work for morphine infusions. GAT358 reduced morphine infusion intake during the maintenance phase of morphine self-administration under a fixed ratio 1 schedule of reinforcement. GAT358 also decreased morphine-seeking behaviour after forced abstinence. Moreover, GAT358 dose dependently decreased the motivation to obtain morphine infusions under a progressive ratio schedule of reinforcement. Strikingly, GAT358 did not affect the motivation to work for food rewards in an identical progressive ratio task, suggesting that the effect of GAT358 in decreasing opioid self-administration was reward specific. Furthermore, GAT58 did not produce motor ataxia in the rotarod test. Our results suggest that CB1R NAMs reduced the reinforcing properties of morphine and could represent a viable therapeutic route to safely decrease misuse of opioids.


Assuntos
Morfina , Receptor CB1 de Canabinoide , Autoadministração , Animais , Morfina/farmacologia , Morfina/administração & dosagem , Receptor CB1 de Canabinoide/efeitos dos fármacos , Camundongos , Regulação Alostérica/efeitos dos fármacos , Masculino , Comportamento de Procura de Droga/efeitos dos fármacos , Recidiva , Reforço Psicológico , Motivação/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Analgésicos Opioides/administração & dosagem , Administração Intravenosa , Condicionamento Operante/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
10.
Clin Transl Sci ; 17(8): e13888, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39118392

RESUMO

The OPRM1 gene codes for the mu opioid receptor (MOR) and polymorphisms are associated with complex patient clinical responses. The most studied single nucleotide polymorphism (SNP) in OPRM1 is adenine (A) substituted by guanine (G) at position 118 (118A>G, rs1799971) leading to a substitution of asparagine (Asn) for aspartic acid (Asp) at position 40 in the N terminus of the resulting protein. To date, no structural explanation for the associated clinical responses resulting from the 118A>G polymorphism has been proposed. We utilized computational modeling paired with functional cellular assays to predict unstructured N- and C-terminal regions of MOR-1. Using molecular docking and post-docking energy minimizations with morphine, we show that the extracellular substitution of Asn at position 40 alters the cytoplasmic C-terminal conformation, while leaving the G-protein binding interface unaffected. A real-time BRET assay measuring G-protein and ß-arrestin association with MOR r generated data that tested this prediction. Consistent with this in silico prediction, we show changes in morphine-mediated ß-arrestin association with receptor variants with little change in morphine-mediated G-protein association comparing MOR-1 wild type (WT) to MOR-1118A>G. We tested the system with different opioid agonists, the OPRM1 118A>G SNP, and different MOR splice variants (MOR-1 and MOR-1O). These results are consistent with the observation that patients with the 118A>G OPRM1 allele respond more readily to fentanyl than to morphine. In conclusion, the 118A>G substitution alters receptor responses to opioids through variable C-terminal domain movements that are agonist and splice variant dependent.


Assuntos
Simulação de Acoplamento Molecular , Morfina , Polimorfismo de Nucleotídeo Único , Receptores Opioides mu , beta-Arrestinas , Receptores Opioides mu/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Humanos , beta-Arrestinas/metabolismo , Morfina/farmacologia , Células HEK293 , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Ligação Proteica
11.
Turk J Gastroenterol ; 35(6): 453-464, 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-39114901

RESUMO

The pathogenesis mechanism of acute gastric mucosal lesions (AGML) is still unclear; further exploration is urgently needed to find a new therapeutic target. This study aimed to investigate whether morphine might regulate the expression and function of transient receptor potential ankyrin 1 (TRPA1) through a cyclic adenosine monophosphate/protein kinase A (cAMP/PKA)-dependent pathway, thereby alleviating gastric mucosal lesions caused by water-immersion restraint stress (WIRS). Rats were administered with intrathecal morphine, TRPA1 antagonist (HC-030031), µ-opioid receptor antagonist, or protein kinase A inhibitor (H-89), respectively, before WIRS. After 6 hours of WIRS, microscopic lesions, hematoxylin and eosin staining, and transmission electron microscopy were applied to assess the damage of the gastric mucosa. Real-time polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay were conducted to detect the levels of TRPA1 and substance P (SP) in the dorsal root ganglia (DRG) and gastric tissues. In addition, immunofluorescence was used to explore the possible co-expression of TRPA1 and µ-opioid receptors in the DRG. The results indicated that WIRS upregulated TRPA1 and SP in gastric mucosa, and HC-030031 or H-89 could alleviate gastric mucosal lesions caused by WIRS (P < .0001). Morphine was found to suppress both WIRS-induced gastric mucosal lesions (P < .0001) and the upregulation of TRPA1 (P = .0086) and SP (P = .0013). Both TRPA1 and SP play important roles in the pathogenesis of WIRS-induced AGML. Exogenous gastroprotective strategies reduce elevated levels of TRPA1 via the cAMP/PKA-dependent pathway. Inhibition of TRPA1 upregulation in the DRG is critical for intrathecal morphine preconditioning-induced gastric protection.


Assuntos
Gânglios Espinais , Mucosa Gástrica , Isoquinolinas , Morfina , Ratos Sprague-Dawley , Restrição Física , Canal de Cátion TRPA1 , Regulação para Cima , Animais , Morfina/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Regulação para Cima/efeitos dos fármacos , Canal de Cátion TRPA1/metabolismo , Masculino , Gânglios Espinais/metabolismo , Gânglios Espinais/efeitos dos fármacos , Restrição Física/efeitos adversos , Ratos , Isoquinolinas/farmacologia , Acetanilidas/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Purinas/farmacologia , Estresse Psicológico/complicações , Imersão , Receptores Opioides mu/metabolismo , AMP Cíclico/metabolismo , Sulfonamidas
16.
Nutrients ; 16(16)2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39203757

RESUMO

Morphine is an important pain reliever employed in pain management, its extended utilize is hindered by the onset of analgesic tolerance and oxidative stress. Long-term morphine administration causes elevated production of reactive oxygen species (ROS), disrupting mitochondrial function and inducing oxidation. Sirtuin 3 (SIRT3), a mitochondrial protein, is essential in modulating ROS levels by regulating mitochondrial antioxidant enzymes as manganese superoxide dismutase (MnSOD). Our investigation focused on the impact of SIRT3 on hyperalgesia and morphine tolerance in mice, as evaluating the antioxidant effect of the polyphenolic fraction of bergamot (BPF). Mice were administered morphine twice daily for four consecutive days (20 mg/kg). On the fifth day, mice received an acute dose of morphine (3 mg/kg), either alone or in conjunction with BPF or Mn (III)tetrakis (4-benzoic acid) porphyrin (MnTBAP). We evaluated levels of malondialdehyde (MDA), nitration, and the activity of SIRT3, MnSOD, glutamine synthetase (GS), and glutamate 1 transporter (GLT1) in the spinal cord. Our findings demonstrate that administering repeated doses of morphine led to the development of antinociceptive tolerance in mice, accompanied by increased superoxide production, nitration, and inactivation of mitochondrial SIRT3, MnSOD, GS, and GLT1. The combined administration of morphine with either BPF or MnTBAP prevented these effects.


Assuntos
Tolerância a Medicamentos , Hiperalgesia , Mitocôndrias , Morfina , Estresse Oxidativo , Polifenóis , Sirtuína 3 , Animais , Morfina/farmacologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Masculino , Hiperalgesia/tratamento farmacológico , Hiperalgesia/induzido quimicamente , Polifenóis/farmacologia , Sirtuína 3/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Antioxidantes/farmacologia , Analgésicos Opioides/farmacologia , Malondialdeído/metabolismo , Glutamato-Amônia Ligase/metabolismo , Metaloporfirinas/farmacologia
17.
Hosp Pediatr ; 14(9): 758-765, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39193635

RESUMO

BACKGROUND AND OBJECTIVES: Individualized, weight-based opioid dosing poses safety risks and contributes to inefficient medication delivery processes. Dose banding is a patient safety strategy to reduce dosing errors through standardized doses based on weight ranges. Study objectives were (1) determine the frequency of dosing deviation from reference ranges of common intravenous (IV) and oral opioid medications, (2) evaluate the differences in dosing deviations by age, and (3) determine the potential reduction in dose variation that could be achieved by dose banding. METHODS: We conducted a cross-sectional analysis of hospitalized children ≥2 months to ≤24 months old who received IV morphine, oral methadone, or oral oxycodone at a single center. Dosing was categorized as no dosing deviation (within ±5% of the reference range), negative dosing deviation (>5% below the reference range), or positive dosing deviation (>5% above the reference range). Descriptive and bivariate analyses were conducted. RESULTS: A total of 3361 opioid doses met the inclusion criteria. A total of 2663 (79.2%) had no dosing deviation, 214 (6.3%) demonstrated negative deviations, and 484 (14.4%) demonstrated positive deviations. Dosing deviations were more frequent among subjects ≥2 months to ≤6 months old for oral methadone and oxycodone (P < .0001) and more frequent among older age group for IV morphine (P < .0001). Dose banding has the potential to reduce the number of unique doses prescribed for all medications by 75% while eliminating unintended dosing deviations. CONCLUSIONS: A total of 20% of opioid doses prescribed to children ≤24 months of age are outside the recommended ranges. Dose banding represents a promising method for simplifying opioid prescribing in the pediatric inpatient setting.


Assuntos
Analgésicos Opioides , Morfina , Oxicodona , Humanos , Analgésicos Opioides/administração & dosagem , Lactente , Masculino , Estudos Transversais , Feminino , Morfina/administração & dosagem , Oxicodona/administração & dosagem , Metadona/administração & dosagem , Erros de Medicação/prevenção & controle , Pré-Escolar , Administração Oral , Criança Hospitalizada , Relação Dose-Resposta a Droga
18.
J Psychiatr Res ; 178: 225-227, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39163660

RESUMO

The onset of the disease as a morphine addiction is associated with the appearance in the patient's body of antibodies directed against opiate receptors (ORs). Once anti-opiate receptor antibodies (anti-OR antibodies) appear in the blood they will tend to bind to ORs. Such binding will cause blocking of physiological functions of OR. The blockage is felt by a morphine addict as withdrawal syndrome. To get rid of this harmful condition, the addict increases the dose of morphine taken. This is where tolerance manifests itself. The drug addict is forced to increase the dose of morphine from time to time because of the body responds by producing the more and more anti-OR antibodies. The immunological nature of morphine addiction is the reason for lifelong changes in the body's reactivity to the drug. An addict can be cured if he gets rid of B- and T-memory cells, which specifically react to ORs.


Assuntos
Dependência de Morfina , Humanos , Dependência de Morfina/imunologia , Receptores Opioides/metabolismo , Morfina/administração & dosagem , Morfina/farmacologia
19.
Chem Biol Interact ; 402: 111189, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39121896

RESUMO

In the pursuit of new lead compounds with fewer side effects than opioids, the novel synthetic phytochemical core, 3,3-dibromoflavanone (3,3-DBF), has emerged as a promising candidate for pain management. Acute assays demonstrated dose-dependent central and peripheral antinociceptive activity of 3,3-DBF through the µ-opioid receptor. This study aimed to explore repeated administration effects of 3,3-DBF in mice and compare them with morphine. Mice were treated with 3,3-DBF (30 mg/kg), morphine (6 mg/kg), or vehicle for 10 days, alongside single-treatment groups. Unlike morphine, 3,3-DBF demonstrated antinociceptive effects in the hot plate test without inducing tolerance. Locomotor activity and motor coordination tests (evaluated through the inverted screen and rotarod tests) revealed no significant differences between the 3,3-DBF-treated and control groups. The gastrointestinal transit assay indicated that 3,3-DBF did not induce constipation, in contrast to morphine. Furthermore, withdrawal signs assessed with the Gellert-Holtzman scale were not comparable to morphine. Additionally, 3,3-DBF exhibited antidepressant-like activity, reducing immobility time in the forced swimming and tail suspension tests, akin to imipramine. In summary, 3,3-DBF demonstrated antinociceptive effects without inducing tolerance or dependence and exhibited antidepressant properties. These findings highlight the potential of 3,3-DBF as a promising therapeutic agent for pain management and its comorbidities, offering advantages over morphine by minimizing side effects.


Assuntos
Analgésicos , Antidepressivos , Flavonoides , Morfina , Animais , Morfina/farmacologia , Morfina/uso terapêutico , Camundongos , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Antidepressivos/química , Masculino , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Analgésicos/química , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Flavonoides/química , Dor/tratamento farmacológico , Manejo da Dor/métodos , Atividade Motora/efeitos dos fármacos
20.
Neurosci Lett ; 841: 137944, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39154843

RESUMO

Depression and anxiety are prominent symptoms of withdrawal syndrome, often caused by the abuse of addictive drugs like morphine. N-palmitoylethanolamide (PEA), a biologically active lipid, is utilized as an anti-inflammatory and analgesic medication. Recent studies have highlighted PEA's role in mitigating cognitive decline and easing depression resulting from chronic pain. However, it remains unknown whether PEA can influence negative emotions triggered by morphine withdrawal. This study seeks to explore the impact of PEA on such emotions and investigate the underlying mechanisms. Mice subjected to morphine treatment underwent a 10-day withdrawal period, followed by assessments of the effect of PEA on anxiety- and depression-like behaviors using various tests. Enzyme-linked immunosorbent assay was conducted to measure levels of monoamine neurotransmitters in specific brain regions. The findings indicate that PEA mitigated anxiety and depression symptoms and reduced 5-hydroxytryptamine, noradrenaline, and dopamine levels in the hippocampus and prefrontal cortex. In summary, PEA demonstrates a significant positive effect on negative emotions associated with morphine withdrawal, accompanied with the reduction in levels of monoamine neurotransmitters in key brain regions. These insights could be valuable for managing negative emotions arising from morphine withdrawal.


Assuntos
Amidas , Ansiedade , Depressão , Etanolaminas , Morfina , Ácidos Palmíticos , Síndrome de Abstinência a Substâncias , Animais , Síndrome de Abstinência a Substâncias/psicologia , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Etanolaminas/farmacologia , Ácidos Palmíticos/farmacologia , Camundongos , Masculino , Morfina/farmacologia , Depressão/metabolismo , Depressão/tratamento farmacológico , Depressão/psicologia , Depressão/etiologia , Amidas/farmacologia , Ansiedade/tratamento farmacológico , Ansiedade/psicologia , Ansiedade/metabolismo , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Emoções/efeitos dos fármacos , Serotonina/metabolismo , Dependência de Morfina/metabolismo , Dependência de Morfina/psicologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Norepinefrina/metabolismo , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA