Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
Medicine (Baltimore) ; 100(34): e27064, 2021 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-34449500

RESUMO

BACKGROUND: Dexmedetomidine (Dexm), a selective alpha-2 adrenoceptor agonist, and dexamethasone (Dexa), a very potent and highly selective glucocorticoid, have both been proven effectively to prolong the duration of local anesthetics (LA) in regional anesthesia. However, data comparing the efficacy of Dexm and Dexa as perineural adjuvants are inconsistent. Therefore, this systematic review and meta-analysis of randomized and quasi-randomized controlled trials (RCTs) was conducted to compare the effects of Dexm and Dexa when used as LA adjuvants on peripheral nerve block (PNB). METHODS: We systematically searched PubMed, Cochrane Library, EMBASE, Web of Science, and ScienceDirect databases up to October, 2020. The primary outcome was the duration of analgesia. Secondary outcomes included incidence of rescue analgesia, cumulative opioid consumption, time required for onset of sensory and motor blockades, duration of sensory and motor blockades, incidence of postoperative nausea and vomiting (PONV), and side effect-associated outcomes (e.g., bradycardia, sedation, hypotension, rates of infection, and neurological complications). The study was registered on PROSPERO, number CRD42020188796. RESULTS: After screening of full-text relevant articles, 13 RCTs that met the inclusion criteria were retrieved for this systematic review. It was revealed that perineural Dexm provided equivalent analgesic duration to perineural Dexa. Besides, the intake of Dexm increased the incidence of rescue analgesia in limbs surgery, as well as the cumulative opioid consumption, and decreased the time required for onset of sensory and motor blockades for long-acting LA (all P < .05). Other analysis revealed insignificant difference between the 2 groups in terms of the incidence of PONV (P > .05). Additionally, 2 studies demonstrated that Dexm possesses more sedative properties than Dexa (P < .05). CONCLUSIONS: This meta-analysis indicated that the analgesic duration of Dexm and Dexa as LA adjuvants in PNB is the same. Meanwhile, the effects of perineural Dexm and Dexa on some secondary outcomes, including the incidence of rescue analgesia, cumulative opioid consumption, and time required for onset of sensory and motor blockades, are associated with the surgical site and type of LA.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Anestesia Local/métodos , Dexametasona/uso terapêutico , Dexmedetomidina/uso terapêutico , Glucocorticoides/uso terapêutico , Bloqueio Nervoso/métodos , Adjuvantes Anestésicos , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Dexametasona/administração & dosagem , Dexmedetomidina/administração & dosagem , Glucocorticoides/administração & dosagem , Humanos , Nervos Periféricos , Ensaios Clínicos Controlados Aleatórios como Assunto
2.
Int J Mol Sci ; 22(8)2021 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-33923533

RESUMO

It has been established that the selective α2A adrenoceptor agonist guanfacine reduces hyperactivity and improves cognitive impairment in patients with attention-deficit/hyperactivity disorder (ADHD). The major mechanisms of guanfacine are considered to involve the activation of the postsynaptic α2A adrenoceptor of glutamatergic pyramidal neurons in the frontal cortex, but the effects of chronic guanfacine administration on catecholaminergic and glutamatergic transmissions associated with the orbitofrontal cortex (OFC) are yet to be clarified. The actions of guanfacine on catecholaminergic transmission, the effects of acutely local and systemically chronic (for 7 days) administrations of guanfacine on catecholamine release in pathways from the locus coeruleus (LC) to OFC, the ventral tegmental area (VTA) and reticular thalamic-nucleus (RTN), from VTA to OFC, from RTN to the mediodorsal thalamic-nucleus (MDTN), and from MDTN to OFC were determined using multi-probe microdialysis with ultra-high performance liquid chromatography. Additionally, the effects of chronic guanfacine administration on the expression of the α2A adrenoceptor in the plasma membrane fraction of OFC, VTA and LC were examined using a capillary immunoblotting system. The acute local administration of therapeutically relevant concentrations of guanfacine into the LC decreased norepinephrine release in the OFC, VTA and RTN without affecting dopamine release in the OFC. Systemically, chronic administration of therapeutically relevant doses of guanfacine for 14 days increased the basal release of norepinephrine in the OFC, VTA, RTN, and dopamine release in the OFC via the downregulation of the α2A adrenoceptor in the LC, OFC and VTA. Furthermore, systemically, chronic guanfacine administration did not affect intrathalamic GABAergic transmission, but it phasically enhanced thalamocortical glutamatergic transmission. The present study demonstrated the dual actions of guanfacine on catecholaminergic transmission-acute attenuation of noradrenergic transmission and chronic enhancement of noradrenergic transmission and thalamocortical glutamatergic transmission. These dual actions of guanfacine probably contribute to the clinical effects of guanfacine against ADHD.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Guanfacina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Animais , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Guanfacina/administração & dosagem , Guanfacina/uso terapêutico , Masculino , Norepinefrina/metabolismo , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Tálamo/metabolismo , Tálamo/fisiopatologia , Ácido gama-Aminobutírico/metabolismo
3.
Int J Med Sci ; 17(16): 2454-2467, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33029088

RESUMO

Aims: Dexmedetomidine (Dex) as a highly selective α2-adrenoceptor agonist, was widely used anesthetic in perioperative settings, whether Dex induces cardiac hypertrophy during perioperative administration is unknown. Methods: The effects of Dex on cardiac hypertrophy were explored using the transverse aortic constriction model and neonatal rat cardiomyocytes. Results: We reported that Dex induces cardiomyocyte hypertrophy with activated ERK, AKT, PKC and inactivated AMPK in both wild-type mice and primary cultured rat cardiomyocytes. Additionally, pre-administration of Dex protects against transverse aortic constriction induced-heart failure in mice. We found that Dex up-regulates the activation of ERK, AKT, and PKC via suppression of AMPK activation in rat cardiomyocytes. However, suppression of mitochondrial coupling efficiency and membrane potential by FCCP blocks Dex induced AMPK inactivation as well as ERK, AKT, and PKC activation. All of these effects are blocked by the α2-adrenoceptor antagonist atipamezole. Conclusion: The present study demonstrates Dex preconditioning induces cardiac hypertrophy that protects against heart failure through mitochondria-AMPK pathway in perioperative settings.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Cardiomegalia/induzido quimicamente , Dexmedetomidina/farmacologia , Insuficiência Cardíaca/prevenção & controle , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Animais , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/administração & dosagem , Células Cultivadas , Dexmedetomidina/uso terapêutico , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/patologia , Humanos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Cultura Primária de Células , Ratos , Transdução de Sinais/efeitos dos fármacos
4.
J Glaucoma ; 29(4): 280-286, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32053556

RESUMO

PRéCIS:: Adjuvant diclofenac and apraclonidine eye drop given in conjunction with selective laser trabeculoplasty (SLT) do not significantly impact medium-term intraocular pressure (IOP) reduction compared with placebo, but apraclonidine can be used to blunt immediate postlaser pressure spikes. PURPOSE: There is limited high-grade evidence guiding the choice of eye drops given before and after SLT. The authors chose to measure IOP during the first 24 hours, at 1 week, 6 weeks, and 6 months after SLT, and compare the effect of apraclonidine before SLT and diclofenac after SLT, with placebo. MATERIALS AND METHODS: In this double-blind, randomized, placebo-controlled trial, patients with open-angle glaucoma or ocular hypertension referred for SLT were recruited between 2016 and 2018. Patients were randomized to receive either apraclonidine pre-SLT with placebo post-SLT, placebo pre-SLT with diclofenac post-SLT, or placebo before and after SLT. RESULTS: Sixty eyes from 35 patients were treated with 360-degree SLT. Twenty-four-hour IOP measurements with patient self-monitoring after SLT demonstrated a moderate IOP spike at 1 hour and 2 hours post-SLT in the placebo and diclofenac study arms (mean=+4.05±0.58 mm Hg and +4.47±0.73, respectively, P<0.001 vs. pre-SLT IOP), which was prevented by apraclonidine (mean=-2.41±0.88 mm Hg, P<0.0001 vs. other study arms post-SLT). There were no significant differences between the 3 arms of the study on the long-term IOP reduction achieved by SLT (6 wk: P=0.51, 6 mo: P=0.42). CONCLUSIONS: Neither the use of apraclonidine before SLT nor diclofenac after SLT significantly influenced the IOP reduction induced by SLT. Except for a slight and transient reduction in intraocular inflammation, there was no beneficial effect of diclofenac on early IOP changes or the degree of patient discomfort relative to placebo.


Assuntos
Clonidina/análogos & derivados , Diclofenaco/uso terapêutico , Glaucoma de Ângulo Aberto/cirurgia , Pressão Intraocular/efeitos dos fármacos , Hipertensão Ocular/cirurgia , Trabeculectomia , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Idoso , Anti-Inflamatórios não Esteroides/uso terapêutico , Clonidina/uso terapêutico , Método Duplo-Cego , Feminino , Glaucoma de Ângulo Aberto/fisiopatologia , Humanos , Terapia a Laser/efeitos adversos , Lasers Semicondutores/uso terapêutico , Masculino , Pessoa de Meia-Idade , Hipertensão Ocular/fisiopatologia , Soluções Oftálmicas/uso terapêutico , Tonometria Ocular
6.
Am J Drug Alcohol Abuse ; 46(1): 44-48, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31339797

RESUMO

Background: Currently, there are no established pharmacotherapies for cannabis use disorders (CUDs). As a long-acting alpha-2-adrenergic receptor agonist, guanfacine extended-release (G-XR) could be useful in the treatment of CUDs by mitigating withdrawal and improving behavioral control.Objectives: To evaluate the feasibility and tolerability of G-XR as a treatment for CUDs.Methods: In an eight-week open-label outpatient pilot trial, we evaluated the safety and tolerability of G-XR in 22 cannabis dependent individuals. Using 2 different titration schedules, G-XR was gradually titrated to a dose of 4 mg or the highest dose tolerated. All participants received standard medication management.Results: Retention at week eight was 41%. Average daily amount of cannabis use (in grams: F1,86 = 8.74, p = .004; in dollars: F1,86 = 16.67, p < .0001) and cannabis using days (F1,86 = 7.67, p = .007) significantly reduced over the course of study participation. There were no significant differences between the titration schedules on emergence of side effects (Fisher exact test, p = .378) or retention (Log-Rank Test X21 = 0.021, p = .886). A total of 3 participants achieved 3 weeks or greater of total abstinence.Conclusions: G-XR is a feasible treatment for CUDs, and should be evaluated further in an efficacy trial.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Guanfacina/uso terapêutico , Abuso de Maconha/tratamento farmacológico , Adulto , Preparações de Ação Retardada/uso terapêutico , Estudos de Viabilidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Estados Unidos/epidemiologia
7.
Aging (Albany NY) ; 11(21): 9556-9568, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31682592

RESUMO

Dexmedetomidine is known to alleviate cerebral ischemia-reperfusion injury (CIRI). We established a rat model of CIRI, which exhibited higher neurological deficit scores and a greater number of apoptotic cells in the cerebral ischemic penumbra than controls. Dexmedetomidine reversed the neuronal apoptosis and improved neurological function in this model. We then examined Sigma-1 receptor (Sig-1R) expression on the endoplasmic reticulum (ER) in brain tissues at different reperfusion time points. Sig-1R expression increased with CIRI and decreased with increasing reperfusion times. After 24 hours of reperfusion, dexmedetomidine upregulated Sig-1R expression, and ER stress proteins (GRP78, CHOP, JNK and Caspase-3) were detected in brain tissues with Western blotting. Moreover, GRP78 expression followed a pattern similar to Sig-1R. Dexmedetomidine induced GRP78 expression but inhibited CHOP, Caspase-3 and phosphorylated-JNK expression in brain tissues. A Sig-1R-specific inhibitor reduced GRP78 expression and partially inhibited the upregulation of GRP78 by dexmedetomidine. The inhibitor also increased CHOP and Caspase-3 expression and partially reversed the inhibitory effects of dexmedetomidine on these pro-apoptotic ER stress proteins. These results suggest that dexmedetomidine at least partially inhibits ER stress-induced apoptosis by activating Sig-1R, thereby attenuating brain damage after 24 hours of ischemia-reperfusion.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Lesões Encefálicas/prevenção & controle , Dexmedetomidina/uso terapêutico , Receptores sigma/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Apoptose/efeitos dos fármacos , Dexmedetomidina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Masculino , Distribuição Aleatória , Ratos Sprague-Dawley , Receptor Sigma-1
8.
Anesth Analg ; 129(6): 1529-1535, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31743172

RESUMO

BACKGROUND: Dexmedetomidine (Dex) is an attractive agent for procedural sedation due to its unique pharmacodynamic profile, specifically affording predictable sedation without concurrent respiratory depression. However, Dex has previously been reported to prevent or terminate arrhythmias. The purpose of this study was to investigate paroxysmal supraventricular tachycardia (PSVT) inducibility and homeostatic stability during electrophysiology studies (EPSs) and ablation when a standardized Dex protocol was used as the primary sedation agent. METHODS: We performed a retrospective review of 163 consecutive procedures for PSVT ablation that received Dex as the primary sedative with adjunct fentanyl and midazolam boluses (DEX-FENT-MIDAZ). This cohort was compared to 163 consecutive control procedures wherein strictly fentanyl and midazolam were used for sedation. The primary outcome reviewed was PSVT inducibility assessed before ablation. Reviewed secondary outcomes included level of sedation and intraprocedure hemodynamics and oxygenation. RESULTS: The arrhythmia profiles of the DEX-FENT-MIDAZ and control cohorts were very similar. The overall incidence of a "negative" EPSs in which arrhythmia was not induced was 24% in the DEX-FENT-MIDAZ group and 26% in the control group (P = .7). Unintended deep sedation was significantly less with DEX-FENT-MIDAZ (4.3% vs 27%; P ≤ .0001). However, DEX-FENT-MIDAZ use was associated with a higher incidence of intraprocedure hypotension. CONCLUSIONS: Dex sedation during EPSs is not associated with a reduction in PSVT inducibility. The therapeutic utility of Dex during EPS arises from the predictable sedation Dex affords but is associated with an increased incidence of intraprocedure hypotension.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Ablação por Cateter , Dexmedetomidina/uso terapêutico , Técnicas Eletrofisiológicas Cardíacas , Frequência Cardíaca , Hipnóticos e Sedativos/uso terapêutico , Taquicardia Supraventricular/cirurgia , Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Adulto , Idoso , Pressão Sanguínea/efeitos dos fármacos , Estimulação Cardíaca Artificial , Dexmedetomidina/efeitos adversos , Feminino , Humanos , Hipnóticos e Sedativos/efeitos adversos , Hipotensão/induzido quimicamente , Hipotensão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Retrospectivos , Taquicardia Supraventricular/diagnóstico , Taquicardia Supraventricular/fisiopatologia , Resultado do Tratamento
9.
Plast Reconstr Surg ; 144(6): 1080e-1094e, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31764672

RESUMO

LEARNING OBJECTIVES: After reading this article, the participant should be able to: 1. Describe the fundamental concepts of multimodal analgesia techniques and how they target pain pathophysiology. 2. Effectively educate patients on postoperative pain and safe opioid use. 3. Develop and implement a multimodal postoperative analgesia regimen. SUMMARY: For many years, opioids were the cornerstone of postoperative pain control, contributing to what has become a significant public health concern. This article discusses contemporary approaches to multimodal, opioid-sparing postoperative pain management in the plastic surgical patient.


Assuntos
Recuperação Pós-Cirúrgica Melhorada , Dor Pós-Operatória/prevenção & controle , Acetaminofen/uso terapêutico , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Analgésicos Opioides/uso terapêutico , Anestesia por Condução/métodos , Anestesia Local/métodos , Anestésicos Combinados , Anestésicos Locais/farmacocinética , Anti-Inflamatórios não Esteroides/uso terapêutico , Antidepressivos/uso terapêutico , Bloqueio do Plexo Braquial/métodos , Aconselhamento , Gabapentina/uso terapêutico , Humanos , Educação de Pacientes como Assunto , Modalidades de Fisioterapia , Guias de Prática Clínica como Assunto , Cuidados Pré-Operatórios , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
10.
Kidney Int ; 96(5): 1150-1161, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31530477

RESUMO

Norepinephrine exacerbates renal medullary hypoxia in experimental septic acute kidney injury. Here we examined whether dexmedetomidine, an α2-adrenergic agonist, can restore vasopressor responsiveness, decrease the requirement for norepinephrine and attenuate medullary hypoxia in ovine gram-negative sepsis. Sheep were instrumented with pulmonary and renal artery flow probes, and laser Doppler and oxygen-sensing probes in the renal cortex and medulla. Conscious sheep received an infusion of live Escherichia coli for 30 hours. Eight sheep in each group were randomized to receive norepinephrine, norepinephrine with dexmedetomidine, dexmedetomidine alone or saline vehicle, from 24-30 hours of sepsis. Sepsis significantly reduced the average mean arterial pressure (84 to 67 mmHg), average renal medullary perfusion (1250 to 730 perfusion units), average medullary tissue pO2 (40 to 21 mmHg) and creatinine clearance (2.50 to 0.78 mL/Kg/min). Norepinephrine restored baseline mean arterial pressure (to 83 mmHg) but worsened medullary hypoperfusion (to 330 perfusion units) and medullary hypoxia (to 9 mmHg). Dexmedetomidine (0.5 µg/kg/h) co-administration significantly reduced the norepinephrine dose (0.8 to 0.4 µg/kg/min) required to restore baseline mean arterial pressure, attenuated medullary hypoperfusion (to 606 perfusion units), decreased medullary tissue hypoxia (to 29 mmHg), and progressively increased creatinine clearance (to 1.8 mL/Kg/min). Compared with vehicle time-control, dexmedetomidine given alone significantly prevented the temporal reduction in mean arterial pressure, but had no significant effects on medullary perfusion and oxygenation or creatinine clearance. Thus, in experimental septic acute kidney injury, dexmedetomidine reduced norepinephrine requirements, attenuated its adverse effects on the renal medulla, and maintained renal function.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Agonistas alfa-Adrenérgicos/uso terapêutico , Dexmedetomidina/uso terapêutico , Norepinefrina/uso terapêutico , Injúria Renal Aguda/sangue , Injúria Renal Aguda/etiologia , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Citocinas/sangue , Dexmedetomidina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Escherichia coli , Hemodinâmica/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/metabolismo , Norepinefrina/farmacologia , Oxigênio/metabolismo , Sepse/complicações , Ovinos
11.
Am J Addict ; 28(6): 455-464, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31483544

RESUMO

BACKGROUND AND OBJECTIVES: No medication has Food and Drug Administration approval for cannabis use disorder (CUD), and most medication development focuses on the withdrawal syndrome. We evaluated the effects of short-term treatment using the α-2A-adrenergic receptor agonist, guanfacine, on withdrawal symptoms in volunteers with CUD and a history of early onset of cannabis use. METHODS: Non-treatment-seeking healthy volunteers (n = 7) who met the Diagnostic and Statistical Manual of Mental Disorders, 4th Edition criteria for CUD participated in a two-phase, within-subjects study. Volunteers received placebo or guanfacine (3 mg/day) for the first 8-day inpatient study and the alternative medication for the second 8-day inpatient study. On day 1 of both treatment periods, participants received 30 mg of synthetic Δ9 -tetrahydrocannabinol for standardization of abstinence onset. On days 2 to 7, participants received study medication. Cannabis withdrawal symptoms, sleep, craving, and physiology were assessed on all inpatient days. RESULTS: Compared with placebo, guanfacine did not show significant effects on withdrawal, craving, or sleep, although there were trends for guanfacine to increase positive mood symptoms and decrease craving-associated compulsivity. DISCUSSION AND CONCLUSIONS: Compared with former studies, we could not prove significant improvement in sleep or decrease of negative symptoms, but we found trends for increased positive mood symptoms. Our data did not show significant effects of guanfacine on withdrawal symptoms or craving. Due to early and longer cannabis use, our subjects indicate a great severity of illness increasing the likelihood of treatment resistance. SCIENTIFIC SIGNIFICANCE: On the basis of trends demonstrated here and other lines of evidence, further investigation is warranted regarding the utility of guanfacine as a potential treatment for CUD. (Am J Addict 2019;00:1-10).


Assuntos
Dronabinol/efeitos adversos , Guanfacina/uso terapêutico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Adulto , Afeto/efeitos dos fármacos , Fissura/efeitos dos fármacos , Feminino , Humanos , Masculino , Método Simples-Cego , Sono/efeitos dos fármacos , Adulto Jovem
12.
Am J Vet Res ; 80(9): 868-877, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31449445

RESUMO

OBJECTIVE: To determine the effects of 3 α2-adrenergic receptor agonists (α2-ARAs), alone or in combination with butorphanol tartrate, on objective measurements of lameness in horses. ANIMALS: 17 adult polo horses with naturally occurring forelimb or hind limb lameness (or both). PROCEDURES: In a crossover design, each horse received each protocol (saline [0.09% NaCl] solution [2 mL, IV] or xylazine hydrochloride [0.33 mg/kg, IV], detomidine hydrochloride [0.007 mg/kg, IV], or romifidine hydrochloride [0.033 mg/kg, IV] alone or in combination with butorphanol [0.007 mg/kg, IV]) in random order, with a washout period (≥ 7 days) between protocols. Horses were assessed immediately prior to (baseline) and 10, 15, 20, 30, and 40 minutes after administration of each protocol for degree of sedation, mechanical nociceptive threshold (MNT), and objective lameness measurements. RESULTS: Compared with baseline values, sedation scores and MNTs were significantly higher at all evaluated time points following administration of all sedation protocols except xylazine alone; following administration of xylazine alone, sedation scores and MNTs were significantly higher at ≤ 30 minutes and ≤ 20 minutes, respectively. Significant differences in objective forelimb lameness measurements were noted after administration of the 3 α2-ARA-butorphanol combinations. Most significant differences in objective measurements of hind limb lameness were detected after administration of detomidine or romifidine, alone or in combination with butorphanol. CONCLUSIONS AND CLINICAL RELEVANCE: In the study horses, xylazine alone had the least impact on objective lameness measurements. The administration of α2-ARAs, particularly detomidine or romifidine, alone or in combination with butorphanol, resulted in small but significant effects on objective lameness measurements.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Butorfanol/uso terapêutico , Imidazóis/uso terapêutico , Coxeadura Animal/tratamento farmacológico , Xilazina/uso terapêutico , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Animais , Estudos Cross-Over , Quimioterapia Combinada , Feminino , Membro Anterior/efeitos dos fármacos , Marcha/efeitos dos fármacos , Cavalos , Imidazóis/administração & dosagem , Masculino , Distribuição Aleatória
13.
CNS Drugs ; 33(5): 417-430, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30977108

RESUMO

Novel pharmacological treatments are needed for Tourette syndrome. Our goal was to examine the current evidence base and biological rationale for the use of cannabis-derived medications or medications that act on the cannabinoid system in Tourette syndrome. We conducted a comprehensive literature search of PubMed for randomized controlled trials or clinical trials of cannabis-derived medications in Tourette syndrome. Data regarding the population, intervention, safety profile, and outcomes for each trial were extracted and reported and the evidence supporting use of individual cannabis-derived medications was critiqued. There is a strong biological rationale regarding how cannabis-derived medications could affect tic severity. Anecdotal case reports and series have noted that many patients report that their tics improve after using cannabis. However, only two small randomized, placebo-controlled trials of Δ9-tetrahydrocannabinol have been published; these suggested possible benefits of cannabis-derived agents for the treatment of tics. Trials examining other agents active on the cannabinoid system for tic disorders are currently ongoing. Cannabinoid-based treatments are a promising avenue of new research for medications that may help the Tourette syndrome population. However, given the limited research available, the overall efficacy and safety of cannabinoid-based treatments is largely unknown. Further trials are needed to examine dosing, active ingredients, and optimal mode of administration of cannabis-derived compounds, assuming initial trials suggest efficacy. Clinical use for refractory patients should at the very least be restricted to adult populations, given the uncertain efficacy and risk of developmental adverse effects that cannabinoids may have in children. Even in adult populations, cannabis-derived medications are associated with significant issues such as the effects they have on driving safety and the fact that they cause positive urine drug screens that can affect employment.


Assuntos
Canabinoides/uso terapêutico , Síndrome de Tourette/tratamento farmacológico , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Antipsicóticos/uso terapêutico , Terapia Comportamental , Estimulação Encefálica Profunda , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
14.
J Pharm Biomed Anal ; 172: 67-77, 2019 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-31029802

RESUMO

A potent synthetic α2-adrenergic agonist called PT-31, (3-(2-chloro-6-fluorobenzyl)-imidazolidine-2,4-dione), was recently detected as a potential drug to be used as an adjuvant drug to treat chronic pain. The excellent pharmacological property of PT-31 highlights the importance in elucidating its metabolism, which could provide valuable information about its metabolite profile for further pharmacokinetics studies and additionally to estimate the impact of its metabolites on the efficacy, safety and elimination of PT-31. In this work, the study of the in vitro metabolism of PT-31 was initially carried out by using a liquid chromatography coupled to ion trap multiple-stage mass spectrometer (LC-IT-MSn) and a hybrid triple quadrupole/linear ion trap mass spectrometer (LC-QTrap). The production of at least three unknown oxidative metabolites was observed. Structural identification of the unknown metabolites was carried out by combination of LC-MS experiments, including selected reaction monitoring (SRM) and multi-stage full scan experiments. Further analysis of 1H-NMR led to the structural confirmation of the major metabolite. The results indicated that PT-31 was metabolized by a hydroxylation reaction in the imidazolidine-2,4-dione ring in rat and human liver microsomes, producing the metabolite 3-(2-chloro-6-fluorobenzyl)-5-hydroxyimidazolidine-2,4-dione in rat liver microsomes. A carbon hydroxylation onto the benzyl ring, produced two other minor metabolites of the PT-31 in rat liver microsomes.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/metabolismo , Analgésicos/metabolismo , Microssomos Hepáticos/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Analgésicos/farmacocinética , Analgésicos/uso terapêutico , Animais , Dor Crônica/tratamento farmacológico , Avaliação Pré-Clínica de Medicamentos , Humanos , Imidazolidinas/metabolismo , Imidazolidinas/farmacocinética , Imidazolidinas/uso terapêutico , Espectroscopia de Ressonância Magnética , Oxirredução , Ratos , Espectrometria de Massas em Tandem
15.
Addict Biol ; 24(4): 707-716, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-29659126

RESUMO

The α2a-adrenergic agonist, lofexidine, reduced cannabis withdrawal-related sleep disruption in the laboratory, but side effects (e.g. fatigue, hypotension) limit its utility as a treatment for cannabis use disorder. This study tested the potential efficacy and tolerability of a daily bedtime administration of the FDA-approved α2a-adrenergic agonist, guanfacine, in a human laboratory model of cannabis use disorder. Daily, nontreatment-seeking cannabis smokers (13M, 2F) completed a within-subject study comprising two 9-day inpatient study phases. Each phase tested the effects of daily placebo or immediate-release guanfacine (2 mg) on cannabis intoxication (5.6 percent THC; 2 days), withdrawal (4 days of abstinence) and subsequent 'relapse' (3 days of cannabis self-administration). Ratings of mood, sleep, cardiovascular effects, food intake, psychomotor performance and cannabis self-administration were assessed. An outpatient phase preceded each inpatient phase for medication clearance or dose induction. Under placebo medication conditions, cannabis abstinence produced significant withdrawal, including irritability, sleep disruption and anorexia. Guanfacine reduced ratings of irritability and improved objective measures of sleep during cannabis withdrawal relative to placebo but did not reduce cannabis self-administration. Guanfacine was well tolerated with little evidence of fatigue and only small decreases in blood pressure: no dose was held due to hypotension. Thus, a single daily administration of guanfacine at bedtime improved sleep and mood during cannabis withdrawal relative to placebo. This positive signal supports further studies varying the guanfacine dose, formulation or frequency of administration, or combining it with other medications to increase the likelihood of having an impact on cannabis use.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Guanfacina/uso terapêutico , Abuso de Maconha , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Adulto , Afeto , Anorexia/etiologia , Anorexia/fisiopatologia , Pressão Sanguínea , Cannabis/efeitos adversos , Comportamento Alimentar , Feminino , Humanos , Humor Irritável , Masculino , Desempenho Psicomotor , Autoadministração , Sono , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Síndrome de Abstinência a Substâncias/psicologia , Adulto Jovem
17.
Clin Obstet Gynecol ; 61(2): 260-268, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29419532

RESUMO

For women at elevated risk of thrombosis, clinicians are challenged to relieve menopausal symptoms without increasing the risk of thrombosis. Oral menopausal hormone therapy increases the risk of venous thromboembolism by 2-fold to 3-fold. Observational studies suggest less thrombotic risk with transdermal therapies and with progesterone over synthetic progestogens (progestins), but the data are limited. Beneficial nonpharmacologic therapies include cognitive behavioral therapy and clinical hypnosis, whereas beneficial nonhormonal pharmacologic therapies include selective serotonin reuptake inhibitors and serotonin-norepinephrine reuptake inhibitors. For treatment of the genitourinary syndrome of menopause, vaginal lubricants and moisturizers, low-dose vaginal estrogen, and intravaginal dehydroepiandrosterone are options.


Assuntos
Fogachos/prevenção & controle , Menopausa/fisiologia , Trombose/prevenção & controle , Doenças Vaginais/terapia , Doenças da Vulva/terapia , Administração Intravaginal , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Atrofia/fisiopatologia , Atrofia/terapia , Dispareunia/fisiopatologia , Dispareunia/terapia , Estrogênios/administração & dosagem , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Feminino , Terapia de Reposição Hormonal/efeitos adversos , Fogachos/fisiopatologia , Humanos , Terapia a Laser , Lubrificantes/uso terapêutico , Fitoterapia , Prurido/fisiopatologia , Prurido/terapia , Risco , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Sudorese/fisiologia , Trombose/etiologia , Doenças Vaginais/fisiopatologia , Doenças da Vulva/fisiopatologia
19.
J Neuropsychiatry Clin Neurosci ; 30(1): 66-76, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28641496

RESUMO

The cannabinoid-1 receptor (CB1R) agonist Δ9-tetrahydrocannabinol (THC), the main psychoactive constituent of cannabis, adversely effects working memory performance in humans. The α2A-adrenoceptor (AR) agonist guanfacine improves working memory performance in humans. The authors aimed to determine the effects of short-term (6 days) treatment with guanfacine on adverse cognitive effects produced by THC. Employing a double-blind, placebo-controlled crossover design, the cognitive, subjective, and cardiovascular effects produced by oral THC (20 mg) administration were determined twice in the same cannabis users: once after treatment with placebo and once after treatment with guanfacine (3 mg/day). Compared with performance at baseline, THC negatively affected accuracy on spatial working memory trials while participants were maintained on placebo (p=0.012) but not guanfacine (p=0.497); compared with placebo, accuracy was significantly (p=0.003, Cohen's d=-0.640) improved while individuals were treated with guanfacine. Similarly, compared with baseline, THC increased omission errors on an attentional task while participants were maintained on placebo (p=0.017) but not on guanfacine (p=0.709); compared with placebo, there were significantly (p=0.034, Cohen's d=0.838) fewer omissions while individuals were maintained on guanfacine. Although THC increased visual analog scores of subjective effects and heart rate, these increases were similar during treatment with placebo and guanfacine. THC did not significantly affect performance of a recognition memory task or blood pressure while individuals were maintained on either treatment. Although preliminary, these results suggest that guanfacine warrants further testing as a potential treatment for cannabis-induced cognitive deficits.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Agonistas de Receptores de Canabinoides/efeitos adversos , Dronabinol/efeitos adversos , Guanfacina/uso terapêutico , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Memória de Curto Prazo/efeitos dos fármacos , Adolescente , Adulto , Análise de Variância , Pressão Sanguínea/efeitos dos fármacos , Estudos Cross-Over , Método Duplo-Cego , Feminino , Frequência Cardíaca/efeitos dos fármacos , Humanos , Masculino , Fumar Maconha/efeitos adversos , Pessoa de Meia-Idade , Testes Neuropsicológicos , Projetos Piloto , Adulto Jovem
20.
J Minim Invasive Gynecol ; 25(7): 1165-1178, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28964926

RESUMO

In this review, we examine the evidence behind nonopioid medication alternatives, peripheral nerve blocks, surgical techniques, and postoperative recovery protocols that can help minimize and effectively treat postoperative pain after minimally invasive gynecologic surgery (MIGS). Because of the depth and heterogeneity of the data, a narrative review was performed of reported interventions. A comprehensive review was performed of PubMed, Embase, and the Cochrane Database with a focus on randomized controlled trials. In the absence of literature specific to benign gynecology, similar specialty or procedural data were reviewed. A variety of nonopioid medications, surgical techniques, and postoperative recovery protocols have shown significant improvements in postoperative pain after gynecologic surgery. Nonopioid medication options that are beneficial include acetaminophen, nonsteroidal anti-inflammatories, and antiepileptics. Incision infiltration with local anesthesia also significantly reduces pain. Surgically, minimally invasive approaches, reducing the laparoscopic trocar size to <10 mm, and evacuating the pneumoperitoneum at the end of the case all have significant benefits. Lastly, enhanced recovery pathways show promise in reducing pain after MIGS. By using a multimodal approach, minimally invasive gynecologic surgeons can help to minimize and manage postoperative pain with less reliance on opioid pain medications.


Assuntos
Analgésicos/uso terapêutico , Procedimentos Cirúrgicos em Ginecologia/efeitos adversos , Dor Pós-Operatória/prevenção & controle , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Anestésicos Locais/uso terapêutico , Anticonvulsivantes/uso terapêutico , Dióxido de Carbono/administração & dosagem , Procedimentos Clínicos , Dexametasona/uso terapêutico , Desenho de Equipamento , Feminino , Glucocorticoides/uso terapêutico , Humanos , Cuidados Intraoperatórios/métodos , Laparoscopia/instrumentação , Laparoscopia/métodos , Procedimentos Cirúrgicos Minimamente Invasivos/métodos , Bloqueio Nervoso/métodos , Uso Off-Label , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Dor Pós-Operatória/reabilitação , Pneumoperitônio Artificial/métodos , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Técnicas de Sutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA