Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 24(10): 2319-2327, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29511031

ABSTRACT

Purpose: The purpose of this study is to investigate the potential interplay between opioid analgesia and tumor metastasis through modulation of µ-opioid receptor (MOR), Toll-like receptor 4 (TLR4) activation, and matrix degradation potential.Experimental Design: Plasma samples were collected from 60 patients undergoing elective lower limb joint replacement preoperatively and at 3, 6, and 24 hours after surgery; pain scores were documented at the same time points. Opioid administration was recorded and converted into morphine IV equivalents. Plasma samples were also collected from 10 healthy volunteers. Alphascreen cyclic AMP assay and MOR-overexpressing cells were employed to quantify MOR activation. HEK-Blue hTLR4 were utilized to measure TLR4 activation. Circulating matrix metalloprotease and tissue inhibitor of matrix protease activities were assessed by gelatin zymography and reverse zymography, respectively.Results: Postoperative plasma samples displayed the ability to activate MOR and to inhibit lipopolysaccharide (LPS)-induced TLR4 activation. Linear mixed model analysis revealed that MOR activation had a significant effect on inhibition of LPS-induced TLR4 activation. Furthermore, TLR4 had a significant effect to explain pain scores. Postoperative samples also displayed altered circulating matrix-degrading enzymes activity potential, but this was correlated neither to opioid administration nor to MOR activation potential.Conclusions: Our results show for the first time that (i) opioids administered to surgery patients result in modulation of ligand-induced TLR4 activation and (ii) postoperative pain is associated with increased circulating TLR4 activation potential. Our study further promotes the use of MOR activation potential rather than opioid intake in clinical studies measuring opioid exposure at a given time point. Clin Cancer Res; 24(10); 2319-27. ©2018 AACR.


Subject(s)
Analgesics, Opioid/pharmacology , Hemodynamics/drug effects , Neoplasms/metabolism , Receptors, Opioid, mu/agonists , Toll-Like Receptor 4/agonists , Analgesics, Opioid/administration & dosage , Biomarkers , Cancer Pain/diagnosis , Cancer Pain/drug therapy , Cancer Pain/metabolism , Humans , Neoplasm Staging , Neoplasms/complications , Neoplasms/pathology , Neoplasms/surgery , Pain Measurement , Perioperative Care , Proteolysis
2.
FASEB J ; 31(12): 5208-5216, 2017 12.
Article in English | MEDLINE | ID: mdl-28784632

ABSTRACT

Opioids modulate the tumor microenvironment with potential functional consequences for tumor growth and metastasis. We evaluated the effects of morphine administration on the circulating proteolytic profile of tumor-free mice. Serum from morphine-treated (1 or 10 mg/kg, i.p. every 12 h) or saline-treated mice was collected at different time points and tested ex vivo in endothelial, lymphatic endothelial, and breast cancer cell migration assays. Serum from mice that were treated with 10 mg/kg morphine for 3 d displayed reduced chemotactic potential for endothelial and breast cancer cells, and elicited reduced cancer cell invasion through reconstituted basement membrane compared with serum from saline controls. This was associated with decreased circulating matrix metalloproteinase 9 (MMP-9) and increased circulating tissue inhibitor of metalloproteinase 1 (TIMP-1) and TIMP-3/4 as assessed by zymography and reverse zymography. By using quantitative RT-PCR, we confirmed morphine-induced alterations in MMP-9 and TIMP expression and identified organs, including the liver and spleen, in which these changes originated. Pharmacologic inhibition of MMP-9 abrogated the difference in chemotactic attraction between serum from saline-treated and morphine-treated mice, which indicated that reduced proteolytic ability mediated the decreased migration toward serum from morphine-treated mice. This novel mechanism may enable morphine administration to promote an environment that is less conducive to tumor growth, invasion, and metastasis.-Xie, N., Khabbazi, S., Nassar, Z. D., Gregory, K., Vithanage, T., Anand-Apte, B., Cabot, P. J., Sturgess, D., Shaw, P. N., Parat, M.-O. Morphine alters the circulating proteolytic profile in mice: functional consequences on cellular migration and invasion.


Subject(s)
Cell Movement/drug effects , Matrix Metalloproteinase 9/metabolism , Morphine/pharmacology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Analgesics, Opioid/pharmacology , Animals , Cattle , Cell Line , Cell Line, Tumor , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics
3.
Brain Behav Immun ; 61: 244-258, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27939249

ABSTRACT

In this study, we quantified the ability of opioids present in biological samples to activate the µ-opioid receptor and TLR4 using cell-based assays. Each assay was standardised, in the presence of plasma, using morphine, its µ receptor-active metabolite morphine-6 glucuronide (M6G) and its µ receptor-inactive, but TLR4-active metabolite morphine-3 glucuronide (M3G). Specificity was verified using antagonists. Morphine- and M6G-spiked plasma samples exhibited µ receptor activation, which M3G-spiked plasma lacked. In contrast, M3G showed moderate but consistent activation of TLR-4. Plasma samples were collected at a number of time points from mice administered morphine (1 or 10mg/kg every 12h for 3days) or saline. Morphine administration led to intermittent µ receptor activation, reversed by µ receptor antagonists, and to TRL4 activation at time points where M3G is measured in plasma. Interestingly, this protocol of morphine administration also led to TLR4-independent NF-κB activation, at time points where M3G was not detected, presumably via elevation of circulating cytokines including, but not limited to, TNFα. Circulating TNFα was increased after three days of morphine administration, and TNFα mRNA elevated in the spleen of morphine-treated mice.


Subject(s)
Morphine Derivatives/pharmacology , Morphine/pharmacology , Plasma/drug effects , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Female , HEK293 Cells , Humans , Mice
4.
Case Rep Emerg Med ; 2013: 728405, 2013.
Article in English | MEDLINE | ID: mdl-23956890

ABSTRACT

Respiratory failure due to subglottic stenosis is a rare but serious condition. A 22-year-old male presented to the emergency department (ED) with shortness of breath, stridor, and change in tone of voice. The patient did not complain of B-symptoms (fever, weight loss, and night sweats). In the week before this presentation, he was diagnosed with an upper respiratory tract infection with associated bronchospasm and discharged on oral antibiotics and inhaled salbutamol without effect. He developed hypercapnic respiratory failure in the ED after a coughing episode. A normal nasopharyngoscopic examination and a subtle mediastinal abnormality on chest radiograph lead to a working diagnosis of retrosternal subglottic obstruction. The complexities of his airway management and suggestions for multidisciplinary approach are discussed.

SELECTION OF CITATIONS
SEARCH DETAIL
...