Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Bioorg Chem ; 148: 107489, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38797065

RESUMO

The number of opioid-related overdose deaths and individuals that have suffered from opioid use disorders have significantly increased over the last 30 years. FDA approved maintenance therapies to treat opioid use disorder may successfully curb drug craving and prevent relapse but harbor adverse effects that reduce patient compliance. This has created a need for new chemical entities with improved patient experience. Previously our group reported a novel lead compound, NAT, a mu-opioid receptor antagonist that potently antagonized the antinociception of morphine and showed significant blood-brain barrier permeability. However, NAT belongs to thiophene containing compounds which are known structural alerts for potential oxidative metabolism. To overcome this, 15 NAT derivatives with various substituents at the 5'-position of the thiophene ring were designed and their structure-activity relationships were studied. These derivatives were characterized for their binding affinity, selectivity, and functional activity at the mu opioid receptor and assessed for their ability to antagonize the antinociceptive effects of morphine in vivo. Compound 12 showed retention of the basic pharmacological attributes of NAT while improving the withdrawal effects that were experienced in opioid-dependent mice. Further studies will be conducted to fully characterize compound 12 to examine whether it would serve as a new lead for opioid use disorder treatment and management.


Assuntos
Receptores Opioides mu , Animais , Relação Estrutura-Atividade , Camundongos , Receptores Opioides mu/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Humanos , Estrutura Molecular , Tiofenos/química , Tiofenos/farmacologia , Tiofenos/síntese química , Tiofenos/uso terapêutico , Masculino , Relação Dose-Resposta a Droga , Analgésicos Opioides/farmacologia , Analgésicos Opioides/química , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/química , Morfina/farmacologia
2.
J Med Chem ; 67(11): 9552-9574, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38814086

RESUMO

Despite the availability of numerous pain medications, the current array of Food and Drug Administration-approved options falls short in adequately addressing pain states for numerous patients and consequently worsens the opioid crisis. Thus, it is imperative for basic research to develop novel and nonaddictive pain medications. Toward addressing this clinical goal, nalfurafine (NLF) was chosen as a lead and its structure-activity relationship (SAR) systematically studied through design, syntheses, and in vivo characterization of 24 analogues. Two analogues, 21 and 23, showed longer durations of action than NLF in a warm-water tail immersion assay, produced in vivo effects primarily mediated by KOR and DOR, penetrated the blood-brain barrier, and did not function as reinforcers. Additionally, 21 produced fewer sedative effects than NLF. Taken together, these results aid the understanding of NLF SAR and provide insights for future endeavors in developing novel nonaddictive therapeutics to treat pain.


Assuntos
Morfinanos , Compostos de Espiro , Relação Estrutura-Atividade , Compostos de Espiro/química , Compostos de Espiro/farmacologia , Compostos de Espiro/síntese química , Animais , Morfinanos/farmacologia , Morfinanos/química , Morfinanos/síntese química , Morfinanos/uso terapêutico , Camundongos , Masculino , Humanos , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Manejo da Dor/métodos , Dor/tratamento farmacológico , Analgésicos/farmacologia , Analgésicos/química , Analgésicos/síntese química , Analgésicos/uso terapêutico
3.
FASEB J ; 38(8): e23603, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38648368

RESUMO

Recent evidence suggests that chronic exposure to opioid analgesics such as morphine disrupts the intestinal epithelial layer and causes intestinal dysbiosis. Depleting gut bacteria can preclude the development of tolerance to opioid-induced antinociception, suggesting an important role of the gut-brain axis in mediating opioid effects. The mechanism underlying opioid-induced dysbiosis, however, remains unclear. Host-produced antimicrobial peptides (AMPs) are critical for the integrity of the intestinal epithelial barrier as they prevent the pathogenesis of the enteric microbiota. Here, we report that chronic morphine or fentanyl exposure reduces the antimicrobial activity in the ileum, resulting in changes in the composition of bacteria. Fecal samples from morphine-treated mice had increased levels of Akkermansia muciniphila with a shift in the abundance ratio of Firmicutes and Bacteroidetes. Fecal microbial transplant (FMT) from morphine-naïve mice or oral supplementation with butyrate restored (a) the antimicrobial activity, (b) the expression of the antimicrobial peptide, Reg3γ, (c) prevented the increase in intestinal permeability and (d) prevented the development of antinociceptive tolerance in morphine-dependent mice. Improved epithelial barrier function with FMT or butyrate prevented the enrichment of the mucin-degrading A. muciniphila in morphine-dependent mice. These data implicate impairment of the antimicrobial activity of the intestinal epithelium as a mechanism by which opioids disrupt the microbiota-gut-brain axis.


Assuntos
Analgésicos Opioides , Disbiose , Fentanila , Microbioma Gastrointestinal , Mucosa Intestinal , Camundongos Endogâmicos C57BL , Morfina , Animais , Morfina/farmacologia , Camundongos , Disbiose/induzido quimicamente , Disbiose/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Masculino , Fentanila/farmacologia , Analgésicos Opioides/farmacologia , Eixo Encéfalo-Intestino/efeitos dos fármacos , Transplante de Microbiota Fecal , Proteínas Associadas a Pancreatite/metabolismo , Akkermansia/efeitos dos fármacos , Peptídeos Antimicrobianos/farmacologia , Bacteroidetes/efeitos dos fármacos
4.
J Med Chem ; 67(1): 603-619, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38156970

RESUMO

While there are approved therapeutics to treat opioid overdoses, the need for treatments to reverse overdoses due to ultrapotent fentanyls remains unmet. This may be due in part to an adrenergic mechanism of fentanyls in addition to their stereotypical mu-opioid receptor (MOR) effects. Herein, we report our efforts to further understanding of the functions these distinct mechanisms impart. Employing the known MOR neutral antagonist phenylfentanil as a lead, 17 analogues were designed based on the concept of isosteric replacement. To probe mechanisms of action, these analogues were pharmacologically evaluated in vitro and in vivo, while in silico modeling studies were also conducted on phenylfentanil. While it did not indicate MOR involvement in vivo, phenylfentanil yielded respiratory minute volumes similar to those caused by fentanyl. Taken together with molecular modeling studies, these results indicated that respiratory effects of fentanyls may also correlate to inhibition of both α1A- and α1B-adrenergic receptors.


Assuntos
Adrenérgicos , Fentanila , Fentanila/farmacologia , Receptores Opioides mu , Antagonistas de Entorpecentes , Analgésicos Opioides/farmacologia
5.
Res Sq ; 2023 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-37503065

RESUMO

Recent evidence suggests that chronic exposure to opioid analgesics such as morphine disrupt the intestinal epithelial layer and cause intestinal dysbiosis. Inhibiting opioid-induced dysbiosis can preclude the development of tolerance to opioid-induced antinociception, suggesting an important role of the gut-brain axis in mediating opioid effects. However, the mechanism underlying opioid-induced dysbiosis remains unclear. Host-produced antimicrobial peptides (AMPs) are critical for the integrity of the intestinal epithelial barrier as they prevent the pathogenesis of the enteric microbiota. Here, we report that chronic morphine exposure reduces expression of the antimicrobial peptide, Regenerating islet-derived 3 gamma (Reg3γ), in the ileum resulting in reduced intestinal antimicrobial activity against Gram-positive bacteria, L. reuteri. Fecal samples from morphine-treated mice had reduced levels of the phylum, Firmicutes, concomitant with reduced levels of short-chain fatty acid, butyrate. Fecal microbial transplant (FMT) from morphine-naïve mice restored the antimicrobial activity, the expression of Reg3γ, and prevented the increase in intestinal permeability and the development of antinociceptive tolerance in morphine-dependent mice. Similarly, oral gavage with sodium butyrate dose-dependently reduced the development of antinociceptive tolerance, and prevented the downregulation of Reg3γ and the reduction in antimicrobial activity. The alpha diversity of the microbiome was also restored by oral butyrate in morphine-dependent mice. These data implicate impairment of the antimicrobial activity of the intestinal epithelium as a mechanism by which morphine disrupts the microbiota-gut-brain axis.

6.
Surg Clin North Am ; 103(3): 483-494, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37149384

RESUMO

Despite the fact that modern burn care has significantly reduced the mortality associated with severe burn injuries, the rehabilitation and community reintegration of survivors continues to be a challenge. An interprofessional team approach is essential for optimal outcomes. This includes early occupational and physical therapy, beginning in the intensive care unit (ICU). Burn-specific techniques (edema management, wound healing, and contracture prevention) are successfully integrated into the burn ICU. Research demonstrates that early intensive rehabilitation of critically ill burn patients is safe and effective. Further work on the physiologic, functional, and long-term impact of this care is needed.


Assuntos
Cuidados Críticos , Unidades de Terapia Intensiva , Humanos , Cuidados Críticos/métodos , Cicatrização/fisiologia , Estado Terminal/terapia
7.
J Med Chem ; 66(1): 577-595, 2023 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-36538027

RESUMO

The search for selective opioid ligands with desired pharmacological potency and improved safety profile has always been an area of interest. Our previous effort yielded a potent opioid modulator, NAN, a 6α-N-7'-indolyl-substituted naltrexamine derivative, which exhibited promising pharmacological activities both in vitro and in vivo. However, significant human ether-a-go-go-related gene (hERG) liability limited its further development. Therefore, a systematic structural modification on NAN was conducted in order to alleviate hERG toxicity while preserving pharmacological properties, which led to the discovery of 2'-methylindolyl derivative compound 21. Compared to NAN, compound 21 manifested overall improved pharmacological profiles. Follow-up hERG channel inhibition evaluation revealed a seven-fold decreased potency of compound 21 compared to NAN. Furthermore, several fundamental drug-like property evaluations suggested a reasonable ADME profile of 21. Collectively, compound 21 appeared to be a promising opioid modulator for further development as a novel therapeutic agent toward opioid use disorder treatments.


Assuntos
Analgésicos Opioides , Receptores Opioides , Humanos , Analgésicos Opioides/farmacologia , Canais de Potássio Éter-A-Go-Go , Ligantes
8.
ACS Chem Neurosci ; 13(24): 3608-3628, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36449691

RESUMO

Discovery of analgesics void of abuse liability is critical to battle the opioid crisis in the United States. Among many strategies to achieve this goal, targeting more than one opioid receptor seems promising to minimize this unwanted side effect while achieving a reasonable therapeutic profile. In the process of understanding the structure-activity relationship of nalfurafine, we identified a potential analgesic agent, NMF, as a dual kappa opioid receptor/delta opioid receptor agonist with minimum abuse liability. Further characterizations, including primary in vitro ADMET studies (hERG toxicity, plasma protein binding, permeability, and hepatic metabolism), and in vivo pharmacodynamic and toxicity profiling (time course, abuse liability, tolerance, withdrawal, respiratory depression, body weight, and locomotor activity) further confirmed NMF as a promising drug candidate for future development.


Assuntos
Analgésicos Opioides , Morfinanos , Humanos , Analgésicos Opioides/química , Receptores Opioides kappa/agonistas , Morfinanos/farmacologia , Analgésicos/farmacologia , Relação Estrutura-Atividade , Receptores Opioides mu/agonistas
9.
Adv Drug Alcohol Res ; 12022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-35909497

RESUMO

Background: Opioid users regularly consume other drugs such as alcohol (ethanol). Acute administration of ethanol rapidly reverses tolerance to morphine-induced respiratory depression. However, recent research has suggested that the primary metabolite of ethanol, acetaldehyde, may play a key role in mediating the CNS effects seen after ethanol consumption. This research investigated the role of acetaldehyde in ethanol reversal of tolerance to morphine-induced respiratory depression. Methods: Tolerance was induced in mice by 6-days implantation of a 75 mg morphine pellet with control mice implanted with a placebo pellet. Tolerance was assessed by acute morphine administration on day 6 and respiration measured by plethysmography. Levels of acetaldehyde were inhibited or enhanced by pre-treatments with the acetaldehyde chelator D-penicillamine and the inhibitor of acetaldehyde dehydrogenase disulfiram respectively. Results: Morphine pellet implanted mice displayed tolerance to an acute dose of morphine compared to placebo pellet implanted controls. Acute acetaldehyde administration dose-dependently reversed tolerance to morphine respiratory depression. As previously demonstrated, ethanol reversed morphine tolerance, and this was inhibited by D-penicillamine pre-treatment. An acute, low dose of ethanol that did not significantly reverse morphine tolerance was able to do so following disulfiram pre-treatment. Conclusion: These data suggest that acetaldehyde, the primary metabolite of ethanol, is responsible for the reversal of morphine tolerance observed following ethanol administration.

10.
J Med Chem ; 65(6): 4991-5003, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35255683

RESUMO

Opioid-induced constipation (OIC) is a common adverse effect of opioid analgesics. Peripherally acting µ opioid receptor antagonists (PAMORAs) can be applied in the treatment of OIC without compromising the analgesic effects. NAP, a 6ß-N-4-pyridyl-substituted naltrexamine derivative, was previously identified as a potent and selective MOR antagonist mainly acting peripherally but with some CNS effects. Herein, we introduced a highly polar aromatic moiety, for example, a pyrazolyl or imidazolyl ring to decrease CNS MPO scores in order to reduce passive BBB permeability. Four compounds 2, 5, 17, and 19, when administered orally, were able to increase intestinal motility during morphine-induced constipation in the carmine red dye assays. Among them, compound 19 (p.o.) improved GI tract motility by 75% while orally administered NAP and methylnaltrexone showed no significant effects at the same dose. Thus, this compound seemed a promising agent to be further developed as an oral treatment for OIC.


Assuntos
Constipação Induzida por Opioides , Analgésicos Opioides/efeitos adversos , Constipação Intestinal/induzido quimicamente , Constipação Intestinal/tratamento farmacológico , Humanos , Ligantes , Naltrexona/farmacologia , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Receptores Opioides mu
11.
J Med Chem ; 65(6): 5095-5112, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35255685

RESUMO

The µ opioid receptor (MOR) has been an intrinsic target to develop treatment of opioid use disorders (OUD). Herein, we report our efforts on developing centrally acting MOR antagonists by structural modifications of 17-cyclopropylmethyl-3,14-dihydroxy-4,5α-epoxy-6ß-[(4'-pyridyl) carboxamido] morphinan (NAP), a peripherally acting MOR-selective antagonist. An isosteric replacement concept was applied and incorporated with physiochemical property predictions in the molecular design. Three analogs, namely, 25, 26, and 31, were identified as potent MOR antagonists in vivo with significantly fewer withdrawal symptoms than naloxone observed at similar doses. Furthermore, brain and plasma drug distribution studies supported the outcomes of our design strategy on these compounds. Taken together, our isosteric replacement of pyridine with pyrrole, furan, and thiophene provided insights into the structure-activity relationships of NAP and aided the understanding of physicochemical requirements of potential CNS acting opioids. These efforts resulted in potent, centrally efficacious MOR antagonists that may be pursued as leads to treat OUD.


Assuntos
Morfinanos , Transtornos Relacionados ao Uso de Opioides , Analgésicos Opioides/química , Sistema Nervoso Central , Humanos , Morfinanos/química , Naloxona , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Receptores Opioides mu
12.
J Neuroimmune Pharmacol ; 17(1-2): 111-130, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35106734

RESUMO

The gastrointestinal epithelium is critical for maintaining a symbiotic relationship with commensal microbiota. Chronic morphine exposure can compromise the gut epithelial barrier in mice and lead to dysbiosis. Recently, studies have implicated morphine-induced dysbiosis in the mechanism of antinociceptive tolerance and reward, suggesting the presence of a gut-brain axis in the pharmacological effects of morphine. However, the mechanism(s) underlying morphine-induced changes in the gut microbiome remains unclear. The pro-inflammatory cytokine, Interleukin-18 (IL-18), released by enteric neurons can modulate gut barrier function. Therefore, in the present study we investigated the effect of morphine on IL-18 expression in the mouse ileum. We observed that chronic morphine exposure in vivo induces IL-18 expression in the ileum myenteric plexus that is attenuated by naloxone. Given that mu-opioid receptors (MORs) are mainly expressed in enteric neurons, we also characterized morphine effects on the excitability of cholinergic (excitatory) and vasoactive intestinal peptide (VIP)-expressing (inhibitory) myenteric neurons. We found fundamental differences in the electrical properties of cholinergic and VIP neurons such that VIP neurons are more excitable than cholinergic neurons. Furthermore, MORs were primarily expressed in cholinergic neurons, although a subset of VIP neurons also expressed MORs and responded to morphine in electrophysiology experiments. In conclusion, these data show that morphine increases IL-18 in ileum myenteric plexus neurons via activation of MORs in a subset of cholinergic and VIP neurons. Thus, understanding the neurochemistry and electrophysiology of MOR-expressing enteric neurons can help to delineate mechanisms by which morphine perturbs the gut barrier.


Assuntos
Morfina , Plexo Mientérico , Camundongos , Animais , Morfina/farmacologia , Interleucina-18 , Colinérgicos , Receptores Opioides
13.
Neuropharmacology ; 209: 108988, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35183539

RESUMO

Opioids are among the most effective analgesics and the mainstay of pain management. However, concerns about safety and abuse liability have challenged their widespread use by the medical community. Opioid-sparing therapies include drugs that in combination with opioids have the ability to enhance analgesia while decreasing opioid requirement as well as their side effects. Sex differences in antinociceptive responses to opioids have received increasing attention in recent years. However, the molecular mechanisms underlying sex differences related to opioid-sparing adjuncts remain largely unexplored. Using warm water tail-withdrawal as a mouse model of acute thermal nociception, our data suggest that adjunctive administration of the serotonin 5-HT2A receptor (5-HT2AR) antagonist volinanserin dose-dependently enhanced potency of the opioid analgesic oxycodone in male, but not female, mice. This antinociceptive-like response induced by oxycodone was also augmented in 5-HT2AR knockout (5-HT2AR-/-) male, but not female mice; an effect that was reversed by Cre-loxP-mediated selective expression of 5-HT2AR in dorsal root ganglion (DRG) neurons of 5-HT2AR-/- littermates. Pharmacological inhibition with volinanserin or genetic deletion in 5-HT2AR-/- animals potentiated the ability of oxycodone to reduce DRG excitability in male mice. Adjunctive volinanserin did not affect oxycodone-induced conditioned place preference (CPP), whereas it reduced oxycodone-induced locomotor sensitization in male and female mice. Together, these results suggest that adjunctive volinanserin augments opioid-induced antinociception, but not abuse-related behavior, through a sex-specific signaling crosstalk mechanism that requires 5-HT2AR expression in mouse DRG neurons. Ultimately, our results may pave the way for the clinical evaluation of volinanserin as a potential sex-specific opioid adjuvant.


Assuntos
Analgésicos Opioides , Oxicodona , Analgésicos Opioides/farmacologia , Animais , Feminino , Masculino , Camundongos , Oxicodona/farmacologia , Receptor 5-HT2A de Serotonina , Recompensa , Serotonina
14.
Physiology (Bethesda) ; 36(5): 315-323, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34431418

RESUMO

Bidirectional interactions of the gut epithelium with commensal bacteria are critical for maintaining homeostasis within the gut. Chronic opioid exposure perturbs gut homeostasis through a multitude of neuro-immune-epithelial mechanisms, resulting in the development of analgesic tolerance, a major underpinning of the current opioid crisis. Differences in molecular mechanisms of opioid tolerance between the enteric and central pain pathways pose a significant challenge for managing chronic pain without untoward gastrointestinal effects.


Assuntos
Microbioma Gastrointestinal , Epidemia de Opioides , Analgésicos Opioides/efeitos adversos , Tolerância a Medicamentos , Humanos , Mucosa Intestinal
15.
Prev Vet Med ; 194: 105419, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34274864

RESUMO

To keep pace with rising opportunities for disease emergence and spread, surveillance in aquaculture must enable the early detection of both known and new pathogens. Conventional surveillance systems (designed to provide proof of disease freedom) may not support detection outside of periodic sampling windows, leaving substantial blind spots to pathogens that emerge in other times and places. To address this problem, we organized an expert panel to envision optimal systems for early disease detection, focusing on Ostreid herpesvirus 1 (OsHV-1), a pathogen of panzootic consequence to oyster industries. The panel followed an integrative group process to identify and weight surveillance system traits perceived as critical to the early detection of OsHV-1. Results offer a road map with fourteen factors to consider when building surveillance systems geared to early detection; factor weights can be used by planners and analysts to compare the relative value of different designs or enhancements. The results were also used to build a simple, but replicable, model estimating the system sensitivity (SSe) of observational surveillance and, in turn, the confidence in disease freedom that negative reporting can provide. Findings suggest that optimally designed observational systems can contribute substantially to both early detection and disease freedom confidence. In contrast, active surveillance as a singular system is likely insufficient for early detection. The strongest systems combined active with observational surveillance and engaged joint industry and government involvement: results suggest that effective partnerships can generate highly sensitive systems, whereas ineffective partnerships may seriously erode early detection capability. Given the costs of routine testing, and the value (via averted losses) of early detection, we conclude that observational surveillance is an important and potentially very effective tool for health management and disease prevention on oyster farms, but one that demands careful planning and participation. This evaluation centered on OsHV-1 detection in farmed oyster populations. However, many of the features likely generalize to other pathogens and settings, with the important caveat that the pathogens need to manifest via morbidity or mortality events in the species, life stages and environments under observation.


Assuntos
Crassostrea , Infecções por Herpesviridae/veterinária , Herpesviridae , Animais , Aquicultura , Crassostrea/virologia , Infecções por Herpesviridae/diagnóstico
16.
Eur J Pharmacol ; 899: 174007, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33705801

RESUMO

G-protein-biased agonists with reduced ß-arrestin-2 activation are being investigated as safer alternatives to clinically-used opioids. ß-arrestin-2 has been implicated in the mechanism of opioid-induced antinociceptive tolerance. Opioid-induced analgesic tolerance is classically considered as centrally-mediated, but recent reports implicate nociceptive dorsal root ganglia neurons as critical mediators in this process. Here, we investigated the role of ß-arrestin-2 in the mechanism of opioid tolerance in dorsal root ganglia nociceptive neurons using ß-arrestin-2 knockout mice and the G-protein-biased µ-opioid receptor agonist, TRV130. Whole-cell current-clamp electrophysiology experiments revealed that 15-18-h overnight exposure to 10 µM morphine in vitro induced acute tolerance in ß-arrestin-2 wild-type but not knockout neurons. Furthermore, in wild-type neurons circumventing ß-arrestin-2 activation by overnight treatment with 200 nM TRV130 attenuated tolerance. Similarly, acute morphine tolerance in vivo in ß-arrestin-2 knockout mice was prevented in the warm-water tail-withdrawal assay. Treatment with 30 mg/kg TRV130 s.c. also inhibited acute antinociceptive tolerance in vivo in wild-type mice. Alternately, in ß-arrestin-2 knockout neurons tolerance induced by 7-day in vivo exposure to 50 mg morphine pellet was conserved. Likewise, ß-arrestin-2 deletion did not mitigate in vivo antinociceptive tolerance induced by 7-day exposure to 25 mg or 50 mg morphine pellet in both female or male mice, respectively. Consequently, these results indicated that ß-arrestin-2 mediates acute but not chronic opioid tolerance in dorsal root ganglia neurons and to antinociception in vivo. This suggests that opioid-induced antinociceptive tolerance may develop even in the absence of ß-arrestin-2 activation, and thus significantly affect the clinical utility of biased agonists.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos , Gânglios Espinais/efeitos dos fármacos , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Dor Nociceptiva/prevenção & controle , Receptores Opioides mu/agonistas , Compostos de Espiro/farmacologia , Tiofenos/farmacologia , beta-Arrestina 2/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Masculino , Camundongos Knockout , Neurônios/metabolismo , Dor Nociceptiva/genética , Dor Nociceptiva/metabolismo , Dor Nociceptiva/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Fatores de Tempo , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
17.
J Burn Care Res ; 41(4): 809-813, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32386300

RESUMO

Patients who suffer hand burns are at a high contracture risk, partly due to numerous cutaneous functional units, or contracture risk areas, located within the hand. Patients who undergo split-thickness skin grafting are often immobilized postoperatively for graft protection. Recent practice at our burn center includes an early range of motion (EROM) following hand grafting to limit unnecessary immobilization. The purpose of this study was to determine whether EROM is safe to perform after hand grafting and if there is any clinical benefit. This retrospective, matched case-control study of adults compared patients who received EROM to subjects who received the standard 3 to 5 days of postoperative immobilization. Patients were evaluated for graft loss and range of motion. Seventy-one patients were included in this study: 37 EROM patients and 34 matched controls. Six patients experienced minor graft loss, three of these were not attributable to EROM. All graft loss was less than 1 cm and none required additional surgery. Significantly more patients who received EROM achieved full-digital flexion by the first outpatient visit (25/27 = 92.6% vs 15/22 = 68.2%; P = .028). Performing EROM does not cause an increase in graft loss. All areas of graft loss from the EROM group healed without intervention. There appears to be a benefit to EROM since there was a significant improvement in the patients' ability to make a full fist at initial outpatient follow-up. Additional prospective analysis is needed to examine the true clinical utility of EROM in the hand and other contracture-prone areas.


Assuntos
Queimaduras/reabilitação , Contratura/reabilitação , Terapia por Exercício , Traumatismos da Mão/reabilitação , Amplitude de Movimento Articular/fisiologia , Transplante de Pele , Adulto , Autoenxertos , Queimaduras/fisiopatologia , Queimaduras/cirurgia , Estudos de Casos e Controles , Contratura/fisiopatologia , Contratura/cirurgia , Feminino , Sobrevivência de Enxerto , Traumatismos da Mão/fisiopatologia , Traumatismos da Mão/cirurgia , Humanos , Imobilização , Masculino , Cuidados Pós-Operatórios , Estudos Retrospectivos , Contenções
18.
Neuropharmacology ; 167: 107949, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31987863

RESUMO

Opioid-sparing adjuncts are treatments that aim to reduce the overall dose of opioids needed to achieve analgesia, hence decreasing the burden of side effects through alternative mechanisms of action. Lorcaserin is a serotonin 5-HT2C receptor (5-HT2CR) agonist that has recently been reported to reduce abuse-related effects of the opioid analgesic oxycodone. The goal of our studies was to evaluate the effects of adjunctive lorcaserin on opioid-induced analgesic-like behavior using the tail-flick reflex (TFR) test as a mouse model of acute thermal nociception. We show that whereas subcutaneous (s.c.) administration of lorcaserin alone was inactive on the TFR test, adjunctive lorcaserin (s.c.) significantly increased the potency of oxycodone as an antinociceptive drug. This effect was prevented by the 5-HT2CR antagonist SB242084. A similar lorcaserin (s.c.)-induced adjunctive phenotype was observed upon administration of the opioid analgesics morphine and fentanyl. Remarkably, we also show that, opposite to the effects observed via s.c. administration, intrathecal (i.t.) administration of lorcaserin alone induced antinociceptive TFR behavior, an effect that was not prevented by the opioid receptor antagonist naloxone. This route of administration (i.t.) also led to a significant augmentation of oxycodone-induced antinociception. Lorcaserin (s.c.) did not alter the brain or blood concentrations of oxycodone, which suggests that its adjunctive effects on opioid-induced antinociception do not depend upon changes in opioid metabolism. Together, these data indicate that lorcaserin-mediated activation of the 5-HT2CR may represent a new pharmacological approach to augment opioid-induced antinociception. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.


Assuntos
Analgésicos Opioides/administração & dosagem , Analgésicos/administração & dosagem , Benzazepinas/administração & dosagem , Medição da Dor/efeitos dos fármacos , Receptor 5-HT2C de Serotonina , Agonistas do Receptor 5-HT2 de Serotonina/administração & dosagem , Aminopiridinas/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Indóis/administração & dosagem , Injeções Espinhais , Masculino , Camundongos , Medição da Dor/métodos , Receptor 5-HT2C de Serotonina/metabolismo , Antagonistas do Receptor 5-HT2 de Serotonina/administração & dosagem
19.
J Pain ; 21(7-8): 751-762, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31841668

RESUMO

It is increasingly recognized that chronic opioid use leads to maladaptive changes in the composition and localization of gut bacteria. Recently, this "opioid-induced dysbiosis" (OID) has been linked to antinociceptive tolerance development in preclinical models and may therefore identify promising targets for new opioid-sparing strategies. Such developments are critical to curb dose escalations in the clinical setting and combat the ongoing opioid epidemic. In this article, we review the existing literature that pertains to OID, including the current evidence regarding its qualitative nature, influence on antinociceptive tolerance, and future prospects. PERSPECTIVE: This article reviews the current literature on OID of gut bacteria, including its qualitative nature, influence on antinociceptive tolerance, and future prospects. This work may help identify targets for new opioid-sparing strategies.


Assuntos
Analgésicos Opioides/efeitos adversos , Analgésicos/efeitos adversos , Tolerância a Medicamentos , Disbiose/induzido quimicamente , Microbioma Gastrointestinal/efeitos dos fármacos , Dor/tratamento farmacológico , Animais , Humanos
20.
Br J Pharmacol ; 177(2): 254-266, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31499594

RESUMO

BACKGROUND AND PURPOSE: Fentanyl overdose deaths have reached "epidemic" levels in North America. Death in opioid overdose invariably results from respiratory depression. In the present work, we have characterized how fentanyl depresses respiration, and by comparing fentanyl with heroin and morphine, the active breakdown product of heroin, we have sought to determine the factors, in addition to high potency, that contribute to the lethality of fentanyl. EXPERIMENTAL APPROACH: Respiration (rate and tidal volume) was measured in awake, freely moving mice by whole body plethysmography. KEY RESULTS: Intravenously administered fentanyl produced more rapid depression of respiration than equipotent doses of heroin or morphine. Fentanyl depressed both respiratory rate and tidal volume. Fentanyl did not depress respiration in µ-opioid receptor knockout mice. Naloxone, the opioid antagonist widely used to treat opioid overdose, reversed the depression of respiration by morphine more readily than that by fentanyl, whereas diprenorphine, a more lipophilic antagonist, was equipotent in reversing fentanyl and morphine depression of respiration. Prolonged treatment with morphine induced tolerance to respiratory depression, but the degree of cross tolerance to fentanyl was less than the tolerance to morphine itself. CONCLUSION AND IMPLICATIONS: We propose that several factors (potency, rate of onset, lowered sensitivity to naloxone, and lowered cross tolerance to heroin) combine to make fentanyl more likely to cause opioid overdose deaths than other commonly abused opioids. Lipophilic antagonists such as diprenorphine may be better antidotes than naloxone to treat fentanyl overdose.


Assuntos
Analgésicos Opioides/toxicidade , Fentanila/toxicidade , Heroína/toxicidade , Pulmão/efeitos dos fármacos , Morfina/toxicidade , Respiração/efeitos dos fármacos , Insuficiência Respiratória/induzido quimicamente , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Overdose de Drogas , Tolerância a Medicamentos , Feminino , Fentanila/administração & dosagem , Heroína/administração & dosagem , Injeções Intraperitoneais , Injeções Intravenosas , Pulmão/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Insuficiência Respiratória/tratamento farmacológico , Insuficiência Respiratória/metabolismo , Insuficiência Respiratória/fisiopatologia , Taxa Respiratória/efeitos dos fármacos , Medição de Risco , Volume de Ventilação Pulmonar/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA