Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-39192554

RESUMEN

BACKGROUND: Peripheral nerve blocks effectively alleviate postoperative pain. Animal studies and human research suggest that opioid tolerance may reduce the effectiveness of local analgesics. The reduced effectiveness has been observed in opioid-tolerant humans and animals undergoing spinal and infiltration anaesthesia with both lidocaine and bupivacaine. However, the impact on peripheral nerve blocks in humans has not been evaluated. This study aims to assess the onset time and duration of a radial nerve block in opioid-tolerant individuals compared to opioid-naive individuals. We hypothesise that peripheral nerve blocks may be less effective in producing sensory and motor blockades in opioid-tolerant individuals compared to their opioid-naive counterparts. METHODS: Twenty opioid-tolerant individuals will be matched by sex and age with opioid-naïve counterparts. Participants will receive an ultrasound-guided radial nerve block. The primary outcome is the difference in the duration of sensory nerve blockade between the two groups. The secondary outcomes include the onset time of sensory blockade, onset time of motor blockade, and difference in duration of motor nerve blockade. CONCLUSION: This study will compare the effectiveness of a peripheral nerve block between opioid-tolerant and opioid-naïve individuals. Any found differences could support a specific postoperative protocol for opioid-tolerant individuals regarding the use of peripheral nerve blocks.

2.
IBRO Neurosci Rep ; 17: 38-51, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38933596

RESUMEN

First-line pharmacotherapy for peripheral neuropathic pain (NP) of diverse pathophysiology consists of antidepressants and gabapentinoids, but only a minority achieve sufficient analgesia with these drugs. Opioids are considered third-line analgesics in NP due to potential severe and unpredictable adverse effects in long-term use. Also, opioid tolerance and NP may have shared mechanisms, raising further concerns about opioid use in NP. We set out to further elucidate possible shared and separate mechanisms after chronic morphine treatment and oxaliplatin-induced and diabetic polyneuropathies, and to identify potential diagnostic markers and therapeutic targets. We analysed thermal nociceptive behaviour, the transcriptome of dorsal root ganglia (DRG) and the metabolome of cerebrospinal fluid (CSF) in these three conditions, in rats. Several genes were differentially expressed, most following oxaliplatin and least after chronic morphine treatment, compared with saline-treated rats. A few genes were differentially expressed in the DRGs in all three models (e.g. Csf3r and Fkbp5). Some, e.g. Alox15 and Slc12a5, were differentially expressed in both diabetic and oxaliplatin models. Other differentially expressed genes were associated with nociception, inflammation, and glial cells. The CSF metabolome was most significantly affected in the diabetic rats. Interestingly, we saw changes in nicotinamide metabolism, which has been associated with opioid addiction and withdrawal, in the CSF of morphine-tolerant rats. Our results offer new hypotheses for the pathophysiology and treatment of NP and opioid tolerance. In particular, the role of nicotinamide metabolism in opioid addiction deserves further study.

3.
Biomed Pharmacother ; 176: 116879, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38850666

RESUMEN

Cannabinoid CB2 agonists show therapeutic efficacy without unwanted CB1-mediated side effects. The G protein-biased CB2 receptor agonist LY2828360 attenuates the maintenance of chemotherapy-induced neuropathic nociception in male mice and blocks development of morphine tolerance in this model. However, the cell types involved in this phenomenon are unknown and whether this therapeutic profile is observed in female mice has never been investigated. We used conditional deletion of CB2 receptors to determine the cell population(s) mediating the anti-allodynic and morphine-sparing effects of CB2 agonists. Anti-allodynic effects of structurally distinct CB2 agonists (LY2828360 and AM1710) were present in paclitaxel-treated CB2f/f mice and in mice lacking CB2 receptors in CX3CR1 expressing microglia/macrophages (CX3CR1CRE/+; CB2f/f), but were absent in mice lacking CB2 receptors in peripheral sensory neurons (AdvillinCRE/+; CB2f/f). The morphine-sparing effect of LY28282360 occurred in a sexually-dimorphic manner, being present in male, but not female, mice. LY2828360 treatment (3 mg/kg per day i.p. x 12 days) blocked the development of morphine tolerance in male CB2f/f and CX3CR1CRE/+; CB2f/f mice with established paclitaxel-induced neuropathy but was absent in male (or female) AdvillinCRE/+; CB2f/f mice. Co-administration of morphine with a low dose of LY2828360 (0.1 mg/kg per day i.p. x 6 days) reversed morphine tolerance in paclitaxel-treated male CB2f/f mice, but not AdvillinCRE/+; CB2f/f mice of either sex. LY2828360 (3 mg/kg per day i.p. x 8 days) delayed, but did not prevent, the development of paclitaxel-induced mechanical or cold allodynia in either CB2f/f or CX3CR1CRE/+; CB2f/f mice of either sex. Our findings have potential clinical implications.


Asunto(s)
Tolerancia a Medicamentos , Morfina , Neuralgia , Paclitaxel , Receptor Cannabinoide CB2 , Células Receptoras Sensoriales , Animales , Masculino , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/metabolismo , Receptor Cannabinoide CB2/genética , Femenino , Morfina/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Tolerancia a Medicamentos/fisiología , Ratones , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Nocicepción/efectos de los fármacos , Ratones Endogámicos C57BL , Caracteres Sexuales , Ratones Noqueados , Agonistas de Receptores de Cannabinoides/farmacología
4.
J Pain Symptom Manage ; 68(2): e146-e151, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38729533

RESUMEN

In the cancer pain setting, ketamine has been typically employed as a co-analgesic for opioid refractory and neuropathic pain. One controversial topic is whether subanesthetic ketamine be considered when managing opioid refractory cancer pain. In this "Controversies in Palliative Care" article, three clinicians independently answer this question. Specifically, each clinician provides a synopsis of the key studies that inform their thought processes, share practical advice on their clinical approach, and highlight the opportunities for future research. Three independent clinicians reported a divergence of opinion regarding the usefulness of subanesthetic ketamine for managing opioid refractory cancer pain. All investigators acknowledged the lack of high-quality trials. All agreed on the need for adequately powered trials, the development of standardized methodology, and the exploration of any patient sub-populations that may benefit from ketamine for cancer related pain.


Asunto(s)
Analgésicos Opioides , Analgésicos , Dolor en Cáncer , Ketamina , Dolor Intratable , Humanos , Analgésicos/uso terapéutico , Analgésicos Opioides/uso terapéutico , Dolor en Cáncer/tratamiento farmacológico , Ketamina/uso terapéutico , Dolor Intratable/tratamiento farmacológico , Cuidados Paliativos/métodos
5.
Anesth Pain Med ; 14(2): e141975, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38741899

RESUMEN

Background: Total knee arthroplasty (TKA) is a standard surgical procedure for individuals with debilitating knee arthritis. Effective postoperative pain management is essential for successful recovery, although traditional opioid-based methods have limitations. Objectives: This study aimed to compare the efficacy of Apotel and remifentanil patient-controlled analgesia in managing postoperative pain after TKA. Methods: This double-blind, randomized, controlled clinical trial took place at Amir-al-Momenin and Qods Hospitals in Arak, Iran, spanning from June 2022 to September 2023. Sixty-two eligible patients scheduled for knee joint replacement were randomly assigned to receive either Apotel (Group A) or remifentanil (Group R) as part of multimodal analgesia administered via a pain pump for postoperative pain relief in TKA. The study assessed hemodynamic parameters, pain levels (measured using the Visual Analog Scale), analgesic duration, and narcotic consumption. Statistical analyses were performed using SPSS v.27 and Plotly. Results: Subjects exhibited no statistically significant differences in age, gender distribution, duration of surgery, or anesthesia. The hemodynamic status assessment in the recovery room showed no significant differences in SPO2, PR, or MAP between the groups. However, remifentanil demonstrated superior effectiveness in reducing pain over 24 hours post TKA surgery compared to Apotel, as evidenced by lower average Visual Analog Scale (VAS) scores (P < 0.001), longer duration without the need for narcotic painkillers (P < 0.001), and lower cumulative opioid analgesic consumption in Group R (P < 0.001). Conclusions: Remifentanil demonstrates superior pain control in a multimodal pain management approach compared to Apotel, providing sustained pain reduction over 24 hours post-surgery. Moreover, remifentanil offers longer-lasting pain relief and results in lower cumulative narcotic painkiller consumption compared to Apotel.

6.
Brain ; 147(7): 2507-2521, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38577773

RESUMEN

Opioid pain medications, such as morphine, remain the mainstay for treating severe and chronic pain. Prolonged morphine use, however, triggers analgesic tolerance and hyperalgesia (OIH), which can last for a long period after morphine withdrawal. How morphine induces these detrimental side effects remains unclear. Here, we show that morphine tolerance and OIH are mediated by Tiam1-coordinated synaptic structural and functional plasticity in the spinal nociceptive network. Tiam1 is a Rac1 GTPase guanine nucleotide exchange factor that promotes excitatory synaptogenesis by modulating actin cytoskeletal dynamics. We found that prolonged morphine treatment activated Tiam1 in the spinal dorsal horn and Tiam1 ablation from spinal neurons eliminated morphine antinociceptive tolerance and OIH. At the same time, the pharmacological blockade of Tiam1-Rac1 signalling prevented the development and reserved the established tolerance and OIH. Prolonged morphine treatment increased dendritic spine density and synaptic NMDA receptor activity in spinal dorsal horn neurons, both of which required Tiam1. Furthermore, co-administration of the Tiam1 signalling inhibitor NSC23766 was sufficient to abrogate morphine tolerance in chronic pain management. These findings identify Tiam1-mediated maladaptive plasticity in the spinal nociceptive network as an underlying cause for the development and maintenance of morphine tolerance and OIH and provide a promising therapeutic target to reduce tolerance and prolong morphine use in chronic pain management.


Asunto(s)
Analgésicos Opioides , Tolerancia a Medicamentos , Hiperalgesia , Morfina , Plasticidad Neuronal , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Animales , Morfina/farmacología , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Tolerancia a Medicamentos/fisiología , Ratones , Analgésicos Opioides/farmacología , Masculino , Ratones Endogámicos C57BL , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/metabolismo , Proteína de Unión al GTP rac1/metabolismo
7.
Pain Physician ; 27(1): 69-77, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38285037

RESUMEN

BACKGROUND: Outcome optimization after the placement of a spinal cord stimulator (SCS) is critical. The objective of this study was to determine if an association existed between pre-procedural opioid use (compared to patients who were opioid-naïve) and postoperative long-term outcomes following SCS placement. OBJECTIVE: To examine the impact of preprocedural opioid use on long-term outcomes after SCS therapy. STUDY DESIGN: Cohort study utilizing a nationwide database. SETTING: Retrospective. METHODS: With the use of data from HCA Healthcare's national database, a retrospective cohort study was performed to analyze differences in outcomes between opioid-naïve patients and preoperative opioid users who underwent SCS placements. The primary outcome of interest was device explantation at 6 months and 12 months. Secondary outcome measurements included reoperations and readmissions at 6 months and 12 months, as well as operative complications. Multivariable logistic regression models were performed to analyze the association of preoperative opioid use with those outcomes. The odds ratio (OR), 95% confidence intervals (CI), and P values were reported for the independent variables. RESULTS: The final study population consisted of 13,893 patients who underwent SCS placements. In univariate analyses, patients who used opioids preoperatively had higher 6-month (3.6% vs. 2.6%) and one-year removal rates (3.6% vs. 2.8%) (all P < 0.009). On multivariable logistic regression, those using opioids preoperatively had higher odds of removal at 6 months (OR = 1.290, 95% CI 1.05-1.58, P = 0.01) and at one year (OR = 1.23, 95% CI 1.01-1.50, P = 0.04). There was no difference between patients requiring preoperative opioids and patients who were opioid-naive as far as the odds of 6- or 12-month readmissions were concerned. Compared to the opioid-naive group, patients requiring preoperative opioids had increased odds of reoperation at 6 months (OR = 1.2, 95% CI 1.02-1.40, P = 0.03). There were no differences in the odds of complications between both cohorts. LIMITATIONS: Opioid use in this study was defined as using opioids preoperatively in the 30 days leading up to surgery. CONCLUSION: Patients requiring preoperative opioids before SCS placements had increased odds of SCS explantation at 6 months and 12 months, as well as increased odds of reoperation at 6 months.


Asunto(s)
Analgésicos Opioides , Trastornos Relacionados con Opioides , Humanos , Analgésicos Opioides/uso terapéutico , Estudios Retrospectivos , Estudios de Cohortes , Médula Espinal
8.
Acta Anaesthesiol Scand ; 68(3): 410-416, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37877497

RESUMEN

BACKGROUND: Patients with a daily use of opioids have a higher risk of insufficient pain treatment during hospitalization than other patients. This study aimed to examine whether as-needed opioid doses (PRN) were adequately adjusted when patients were admitted to the emergency department (ED) with pain. METHODS: Patients, with a daily use of opioids, who received PRN opioid within the first 3 h after admission at the ED at Aarhus University Hospital, Denmark, were prospectively included from February 2021 to June 2021. The primary outcome was the proportion of patients receiving an inadequate initial dose of PRN opioid, here defined as <15% of daily dose of opioids (DDO) based on sparse evidence, but aligning with the prevailing clinical practice. Secondary outcomes included risk of an inadequate PRN dose in relation to DDO (patients were dichotomized into two groups (DDO <60 or ≥60 mg/day). RESULTS: Among 252 patients admitted to the ED with a daily use of opioids, 149 were admitted with pain and 82 received a PRN opioid dose within 3 h. Twenty-seven out of 82 (33%) patients received a PRN dose of <15% of DDO (95% CI: 23.7-43). When dichotomised; 10 out of 50 (20%) patients with a DDO <60 mg/day (95% CI: 10.0-33.7) versus 17 out of 32 (53.1%) patients with a DDO ≥60 mg/day (95% CI: 34.7-70.9) received an inadequate PRN dose (relative risk, RR: 2.65 [95% CI: 1.4-5.1]). CONCLUSIONS: Patients with daily use of opioids presenting in the ED with acute pain had a high risk of inadequate PRN opioid dose, especially if the DDO was high. Awareness about and education focusing on sufficient PRN doses for patients with a daily use of opioids is (still) called for.


Asunto(s)
Dolor Agudo , Analgésicos Opioides , Humanos , Analgésicos Opioides/uso terapéutico , Manejo del Dolor , Dolor Agudo/tratamiento farmacológico , Servicio de Urgencia en Hospital , Pacientes
9.
J Perianesth Nurs ; 39(1): 87-92, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37855765

RESUMEN

PURPOSE: Perioperative pain management of opioid-tolerant patients can be challenging. Although regional anesthesia and multimodal analgesics may be beneficial, these modalities are often underused. It is unclear whether practice patterns for perioperative pain management are determined by the knowledge, attitudes, and beliefs of surgeons and anesthesiologists. DESIGN: Descriptive survey. METHODS: Using a Qualtrics survey, we polled a randomly selected group of 25 surgeons and 25 anesthesiologists regarding their knowledge, attitudes, beliefs, and practices for pain management in an opioid-tolerant patient. FINDINGS: Of 25, 23 anesthesiologists and 18/25 surgeons responded to the survey. Demographics were similar between the 2 groups. Most of the participant surgeons and anesthesiologists believed that pain management may be challenging in an opioid-tolerant patient. However, only 56% of surgeons would recommend a preoperative pain consultation. Most surgeons and anesthesiologists believed in the efficacy of regional anesthetics. However, 43% of surgeons would not advocate for a regional block, perhaps due to their perception of the added perioperative time. Multimodal analgesics were widely accepted by both surgeons and anesthesiologists. CONCLUSIONS: There is an urgent need to reinforce the importance of patient-centered care, with a specific focus on addressing knowledge gaps and improving perceptions for all the members of the team, including surgeons, anesthesiologists, and perioperative nursing teams, if optimal outcomes are to be achieved for our patients.


Asunto(s)
Analgesia , Analgésicos Opioides , Humanos , Analgesia/métodos , Analgésicos Opioides/farmacología , Anestesiólogos , Manejo del Dolor , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/etiología , Cirujanos , Encuestas y Cuestionarios
10.
J Neurosci ; 44(6)2024 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-38124021

RESUMEN

Prolonged exposure to opioids causes an enhanced sensitivity to painful stimuli (opioid-induced hyperalgesia, OIH) and a need for increased opioid doses to maintain analgesia (opioid-induced tolerance, OIT), but the mechanisms underlying both processes remain obscure. We found that pharmacological block or genetic deletion of HCN2 ion channels in primary nociceptive neurons of male mice completely abolished OIH but had no effect on OIT. Conversely, pharmacological inhibition of central HCN channels alleviated OIT but had no effect on OIH. Expression of C-FOS, a marker of neuronal activity, was increased in second-order neurons of the dorsal spinal cord by induction of OIH, and the increase was prevented by peripheral block or genetic deletion of HCN2, but block of OIT by spinal block of HCN channels had no impact on C-FOS expression in dorsal horn neurons. Collectively, these observations show that OIH is driven by HCN2 ion channels in peripheral nociceptors, while OIT is driven by a member of the HCN family located in the CNS. Induction of OIH increased cAMP in nociceptive neurons, and a consequent shift in the activation curve of HCN2 caused an increase in nociceptor firing. The shift in HCN2 was caused by expression of a constitutively active µ-opioid receptor (MOR) and was reversed by MOR antagonists. We identified the opioid-induced MOR as a six-transmembrane splice variant, and we show that it increases cAMP by coupling constitutively to Gs HCN2 ion channels therefore drive OIH, and likely OIT, and may be a novel therapeutic target for the treatment of addiction.


Asunto(s)
Analgésicos Opioides , Hiperalgesia , Ratones , Masculino , Animales , Analgésicos Opioides/efectos adversos , Hiperalgesia/metabolismo , Canales Iónicos , Nociceptores , Médula Espinal/metabolismo , Dolor/metabolismo
11.
Int Immunopharmacol ; 125(Pt A): 111142, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37918085

RESUMEN

Opioids are widely used in treating patients with acute and chronic pain; however, this class of drugs is also commonly abused. Opioid use disorder and associated overdoses are becoming more prevalent as the opioid crisis continues. Chronic opioid use is associated with tolerance, which decreases the efficacy of opioids over time, but also puts individuals at risk of fatal overdoses. Therefore, it is essential to identify strategies to reduce opioid tolerance in those that use these agents. The gut microbiome has been found to play a critical role in opioid tolerance, with opioids causing dysbiosis of the gut, and changes in the gut microbiome impacting opioid tolerance. These changes in turn have a detrimental effect on the gut microbiome, creating a positive feedback cycle. We review the bidirectional relationship between the gut microbiome and opioid tolerance, discuss the role of modulation of the gut microbiome as a potential therapeutic option in opioid-induced gut dysbiosis, and suggest opportunities for further research and clinical interventions.


Asunto(s)
Microbioma Gastrointestinal , Trastornos Relacionados con Opioides , Humanos , Analgésicos Opioides/efectos adversos , Disbiosis/tratamiento farmacológico , Disbiosis/inducido químicamente , Tolerancia a Medicamentos , Trastornos Relacionados con Opioides/tratamiento farmacológico
12.
Ophthalmologie ; 120(12): 1204-1215, 2023 Dec.
Artículo en Alemán | MEDLINE | ID: mdl-37930369

RESUMEN

The quality of postoperative pain management is still considered insufficient in many cases, also in surgical ophthalmology. Complex constellations and comorbidities, such as pre-existing chronic pain, opioid consumption and opioid use disorders represent a special challenge due to psychosocial influencing factors and sometimes psychological and psychiatric comorbidities but also due to pharmacological effects, such as the development of opioid tolerance, the opioid-induced hyperalgesia. This review article aims to impart knowledge on aspects of these comorbidities and the perioperative management to improve the treatment skills of ophthalmologists in the management of pain in these complex patients.


Asunto(s)
Dolor Crónico , Oftalmología , Trastornos Relacionados con Opioides , Humanos , Analgésicos Opioides/uso terapéutico , Dolor Crónico/tratamiento farmacológico , Dolor Postoperatorio/tratamiento farmacológico , Tolerancia a Medicamentos , Trastornos Relacionados con Opioides/tratamiento farmacológico
14.
Int J Mol Sci ; 24(9)2023 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-37175678

RESUMEN

Despite the large arsenal of analgesic medications, neuropathic pain (NP) management is not solved yet. Angiotensin II receptor type 1 (AT1) has been identified as a potential target in NP therapy. Here, we investigate the antiallodynic effect of AT1 blockers telmisartan and losartan, and particularly their combination with morphine on rat mononeuropathic pain following acute or chronic oral administration. The impact of telmisartan on morphine analgesic tolerance was also assessed using the rat tail-flick assay. Morphine potency and efficacy in spinal cord samples of treated neuropathic animals were assessed by [35S]GTPγS-binding assay. Finally, the glutamate content of the cerebrospinal fluid (CSF) was measured by capillary electrophoresis. Oral telmisartan or losartan in higher doses showed an acute antiallodynic effect. In the chronic treatment study, the combination of subanalgesic doses of telmisartan and morphine ameliorated allodynia and resulted in a leftward shift in the dose-response curve of morphine in the [35S]GTPγS binding assay and increased CSF glutamate content. Telmisartan delayed morphine analgesic-tolerance development. Our study has identified a promising combination therapy composed of telmisartan and morphine for NP and opioid tolerance. Since telmisartan is an inhibitor of AT1 and activator of PPAR-γ, future studies are needed to analyze the effect of each component.


Asunto(s)
Analgésicos Opioides , Neuralgia , Ratas , Animales , Analgésicos Opioides/uso terapéutico , Telmisartán/farmacología , Telmisartán/uso terapéutico , Losartán/uso terapéutico , Guanosina 5'-O-(3-Tiotrifosfato) , Tolerancia a Medicamentos , Analgésicos/farmacología , Analgésicos/uso terapéutico , Morfina/farmacología , Morfina/uso terapéutico , Neuralgia/tratamiento farmacológico , Glutamatos/uso terapéutico
15.
Front Neurosci ; 17: 1134842, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37090798

RESUMEN

Opioids provide analgesia, as well as modulate sleep and respiration, all by possibly acting on the µ-opioid receptors (MOR). MOR's are ubiquitously present throughout the brain, posing a challenge for understanding the precise anatomical substrates that mediate opioid induced respiratory depression (OIRD) that ultimately kills most users. Sleep is a major modulator not only of pain perception, but also for changing the efficacy of opioids as analgesics. Therefore, sleep disturbances are major risk factors for developing opioid overuse, withdrawal, poor treatment response for pain, and addiction relapse. Despite challenges to resolve the neural substrates of respiratory malfunctions during opioid overdose, two main areas, the pre-Bötzinger complex (preBötC) in the medulla and the parabrachial (PB) complex have been implicated in regulating respiratory depression. More recent studies suggest that it is mediation by the PB that causes OIRD. The PB also act as a major node in the upper brain stem that not only receives input from the chemosensory areas in medulla, but also receives nociceptive information from spinal cord. We have previously shown that the PB neurons play an important role in mediating arousal from sleep in response to hypercapnia by its projections to the forebrain arousal centers, and it may also act as a major relay for the pain stimuli. However, due to heterogeneity of cells in the PB, their precise roles in regulating, sleep, analgesia, and respiratory depression, needs addressing. This review sheds light on interactions between sleep and pain, along with dissecting the elements that adversely affects respiration.

16.
Neuroscience ; 519: 147-161, 2023 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-36997020

RESUMEN

The analgesic effect of opioids decreases over time due to the development of analgesic tolerance. We have shown that inhibition of the platelet-derived growth factor beta (PDGFR-ß) signaling eliminates morphine analgesic tolerance in rats. Although the PDGFR-ß and its ligand, the platelet-derived growth factor type B (PDGF-B), are expressed in the substantia gelatinosa of the spinal cord (SG) and in the dorsal root ganglia (DRG), their precise distribution within different cell types of these structures is unknown. Additionally, the impact of a tolerance-mediating chronic morphine treatment, on the expression and distribution of PDGF-B and PDGFR-ß has not yet been studied. Using immunohistochemistry (IHC), we found that in the spinal cord, PDGFR-ß and PDGF-B were expressed in neurons and oligodendrocytes and co-localized with the mu-opioid receptor (MOPr) in opioid naïve rats. PDGF-B was also found in microglia and astrocytes. Both PDGFR-ß and PDGF-B were detected in DRG neurons but not in spinal primary afferent terminals. Chronic morphine exposure did not change the cellular distribution of PDGFR-ß or PDGF-B. However, PDGFR-ß expression was downregulated in the SG and upregulated in the DRG. Consistent with our previous finding that morphine caused tolerance by inducing PDGF-B release, PDGF-B was upregulated in the spinal cord. We also found that chronic morphine exposure caused a spinal proliferation of oligodendrocytes. The changes in PDGFR-ß and PDGF-B expression induced by chronic morphine treatment suggest potential mechanistic substrates underlying opioid tolerance.


Asunto(s)
Analgésicos Opioides , Morfina , Ratas , Masculino , Animales , Morfina/farmacología , Analgésicos Opioides/farmacología , Analgésicos Opioides/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Ratas Sprague-Dawley , Ganglios Espinales/metabolismo , Tolerancia a Medicamentos/fisiología , Médula Espinal/metabolismo
17.
Int J Mol Sci ; 24(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36675147

RESUMEN

Endometriosis (EM), defined as the presence of endometrial-like tissue with surrounding smooth muscle cells outside the uterus, is a disregarded gynecological disease reported to affect 6-10% of women of reproductive age, with 30-50% of them suffering from chronic pelvic pain and infertility. Since the exact pathogenic mechanisms of EM are still unclear, no curative therapy is available. As pain is an important factor in EM, optimal analgesia should be sought, which to date has been treated primarily with non-steroidal anti-inflammatory drugs (NSAIDs), metamizole or, in extreme cases, opioids. Here, we review the pain therapy options, the mechanisms of pain development in EM, the endogenous opioid system and pain, as well as the opioid receptors and EM-associated pain. We also explore the drug abuse and addiction to opioids and the possible use of NOP receptors in terms of analgesia and improved tolerability as a target for EM-associated pain treatment. Emerging evidence has shown a promising functional profile of bifunctional NOP/MOP partial agonists as safe and nonaddictive analgesics. However, until now, the role of NOP receptors in EM has not been investigated. This review offers a thought which still needs further investigation but may provide potential options for relieving EM-associated pain.


Asunto(s)
Endometriosis , Receptores Opioides , Femenino , Humanos , Receptores Opioides/agonistas , Analgésicos Opioides/efectos adversos , Endometriosis/tratamiento farmacológico , Dolor/tratamiento farmacológico , Analgésicos/uso terapéutico , Receptores Opioides mu/agonistas
18.
Br J Pharmacol ; 180(7): 894-909, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-34877650

RESUMEN

The use of opioids in pain management is hampered by the emergence of analgesic tolerance, which leads to increased dosing and side effects, both of which have contributed to the opioid epidemic. One promising potential approach to limit opioid analgesic tolerance is activating the endocannabinoid system in the CNS, via activation of CB1 receptors in the descending pain inhibitory pathway. In this review, we first discuss preclinical and clinical evidence revealing the potential of pharmacological activation of CB1 receptors in modulating opioid tolerance, including activation by phytocannabinoids, synthetic CB1 receptor agonists, endocannabinoid degradation enzyme inhibitors, and recently discovered positive allosteric modulators of CB1 receptors. On the other hand, as non-pharmacological pain relief is advocated by the US-NIH to combat the opioid epidemic, we also discuss contributions of peripheral neuromodulation, involving the electrostimulation of peripheral nerves, in addressing chronic pain and opioid tolerance. The involvement of supraspinal endocannabinoid systems in peripheral neuromodulation-induced analgesia is also discussed. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.


Asunto(s)
Analgésicos Opioides , Endocannabinoides , Humanos , Endocannabinoides/metabolismo , Analgésicos Opioides/efectos adversos , Manejo del Dolor , Epidemia de Opioides , Tolerancia a Medicamentos , Dolor/tratamiento farmacológico , Dolor/metabolismo , Analgésicos/farmacología , Receptor Cannabinoide CB1
19.
Pharmacol Res ; 187: 106560, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36417942

RESUMEN

Painful peripheral neuropathy is a common neurological complication associated with human immunodeficiency virus (HIV) infection and anti-retroviral therapy. We characterized the impact of two CB2 cannabinoid agonists (AM1710 and LY2828360 - ligands differing in signaling bias and CNS penetration) on neuropathic nociception induced by the antiretroviral agent Zalcitabine (2',3'-dideoxycytidine; ddC). We also used a conditional knockout approach to identify cell types mediating CB2 agonist-induced antinociceptive efficacy and sparing of morphine tolerance. AM1710 and LY2828360 alleviated ddC-induced neuropathic nociception in mice of both sexes. These benefits were absent in global CB2 knockout mice, which exhibited robust morphine antinociception. Like morphine, AM1710 blunted ddC-induced increases in proinflammatory cytokine (IL-1ß, TNF-α) and chemokine (CCL2) mRNA expression levels. We generated advillinCre/+;CB2f/f conditional knockout mice to ascertain the role of CB2 localized to primary sensory neurons in CB2-mediated therapeutic effects. Antinociceptive efficacy of both AM1710 and LY2828360, but not reference analgesics, were absent in advillinCre/+;CB2f/f mice, which exhibited robust ddC-induced neuropathy. In ddC-treated CB2f/f mice, LY2828360 suppressed development of morphine tolerance and reversed established morphine tolerance, albeit with greater efficacy in male compared to female mice. LY2828360 failed to block or reverse morphine tolerance in advillinCre/+;CB2f/f mice. The present studies indicate that CB2 activation may alleviate HIV-associated antiretroviral neuropathy and identify a previously unreported mechanism through which CB2 activation produces antinociceptive efficacy. Our results also provide the first evidence that a CB2 agonist can reverse established morphine tolerance and demonstrate that CB2 localized to peripheral sensory neurons mediates the opioid tolerance sparing efficacy of CB2 agonists.


Asunto(s)
Analgésicos Opioides , Terapia Antirretroviral Altamente Activa , Tolerancia a Medicamentos , Hiperalgesia , Morfina , Enfermedades del Sistema Nervioso Periférico , Receptor Cannabinoide CB2 , Animales , Femenino , Humanos , Masculino , Ratones , Analgésicos Opioides/uso terapéutico , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Ratones Noqueados , Morfina/uso terapéutico , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/metabolismo , Células Receptoras Sensoriales/metabolismo , Terapia Antirretroviral Altamente Activa/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico
20.
Neurosci Biobehav Rev ; 145: 104992, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36509207

RESUMEN

Placentophagia, ingestion of placenta and amniotic fluid, usually during parturition, is a behavioral feature of nearly all nonaquatic, placental mammals, and is a nexus for several interlocking behavioral phenomena. Placentophagia has not been typical of human cultures, but in recent years, some women in affluent societies have engaged in it, thereby bringing publicity to the behavior. First, we summarized benefits of placentophagia for nonhuman mammals, which include increased attractiveness of neonates, enhanced onset of maternal behavior, suppression of pseudopregnancy, and enhancement of opioid hypoalgesia by Placental Opioid-Enhancing Factor (POEF), a benefit that may extend well outside the context of parturition. The research on POEF in animals was discussed in detail. Then we discussed placentophagia (placentophagy) in humans, and whether there is validity to the claims of various benefits reported primarily in the pro-placentophagy literature, and, although human afterbirth shows POEF activity, the POEF effect has not yet been tested in humans. Finally, we discussed the general possible implications, for the management of pain and addiction, of isolating and characterizing POEF.


Asunto(s)
Analgésicos Opioides , Placenta , Animales , Recién Nacido , Femenino , Embarazo , Humanos , Periodo Posparto , Dolor , Conducta Materna , Mamíferos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...