Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Alcohol Alcohol ; 52(2): 172-179, 2017 Mar 09.
Article in English | MEDLINE | ID: mdl-28043969

ABSTRACT

AIMS: Nuclear factor kappa light chain enhancer of activated B cells (NFkB) is a ubiquitous transcription factor well known for its role in the innate immune response. As such, NFkB is a transcriptional activator of inflammatory mediators such as cytokines. It has recently been demonstrated that alcohol and other drugs of abuse can induce NFkB activity and cytokine expression in the brain. A number of reviews have been published highlighting this effect of alcohol, and have linked increased NFkB function to neuroimmune-stimulated toxicity. However, in this review we focus on the potentially non-immune functions of NFkB as possible links between NFkB and addiction. METHODS: An extensive review of the literature via Pubmed searches was used to assess the current state of the field. RESULTS: NFkB can induce the expression of a diverse set of gene targets besides inflammatory mediators, some of which are involved in addictive processes, such as opioid receptors and neuropeptides. NFkB mediates complex behaviors including learning and memory, stress responses, anhedonia and drug reward, processes that may lie outside the role of NFkB in the classic neuroimmune response. CONCLUSIONS: Future studies should focus on these non-immune functions of NFkB signaling and their association with addiction-related processes.


Subject(s)
Gene Expression Regulation , NF-kappa B/physiology , Substance-Related Disorders/genetics , Substance-Related Disorders/physiopathology , Anhedonia/physiology , Behavior, Addictive/genetics , Behavior, Addictive/physiopathology , Humans , Inflammation/genetics , Inflammation/immunology , Learning/physiology , Memory/physiology , NF-kappa B/immunology , Neuropeptides/biosynthesis , Receptors, Opioid/biosynthesis
2.
Biopolymers ; 106(4): 460-9, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27271345

ABSTRACT

We previously showed that an antagonist-based peptide ligand, H-Cys(Npys)-Arg-Tyr-Tyr-Arg- Ile-Lys-NH2 , captures the free thiol groups in the ligand-binding site of the nociceptin receptor ORL1. However, the exact receptor sites of this thiol-disulfide exchange reaction have not been uncovered, although such identification would help to clarify the ligand recognition site. Since the Cys→Ala substitution prevents the reaction, we performed the so-called Ala scanning for all the Cys residues in the transmembrane (TM) domains of the ORL1 receptor. Seven different mutant receptors were soundly expressed in the COS-7 cells and examined for their specific affinity labeling by a competitive binding assay using nociceptin and [(3) H]nociceptin. The results of in vitro Ala scanning analyses revealed that the labeled residues were Cys59 in TM1, Cys215 and Cys231 in TM5, and Cys310 in TM7. The present study has provided a novel method of Cys(Npys)-affinity labeling for identification of the ligand-binding sites in the ORL1 receptor. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 460-469, 2016.


Subject(s)
Peptides/chemistry , Receptors, Opioid , Staining and Labeling/methods , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Humans , Ligands , Mutation, Missense , Receptors, Opioid/biosynthesis , Receptors, Opioid/chemistry , Receptors, Opioid/genetics , Nociceptin Receptor
3.
Nat Med ; 13(1): 35-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17159985

ABSTRACT

Abdominal pain is common in the general population and, in patients with irritable bowel syndrome, is attributed to visceral hypersensitivity. We found that oral administration of specific Lactobacillus strains induced the expression of mu-opioid and cannabinoid receptors in intestinal epithelial cells, and mediated analgesic functions in the gut-similar to the effects of morphine. These results suggest that the microbiology of the intestinal tract influences our visceral perception, and suggest new approaches for the treatment of abdominal pain and irritable bowel syndrome.


Subject(s)
Abdominal Pain/physiopathology , Intestines/physiopathology , Lactobacillus acidophilus/physiology , Receptors, Cannabinoid/physiology , Receptors, Opioid/physiology , Abdominal Pain/prevention & control , Administration, Oral , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Cannabinoid Receptor Antagonists , Colon/drug effects , Colon/microbiology , Colon/physiopathology , Dose-Response Relationship, Drug , HT29 Cells , Humans , Indoles/administration & dosage , Indoles/pharmacology , Intestines/drug effects , Intestines/microbiology , Male , Mice , Mice, Inbred BALB C , Morphine/administration & dosage , Morphine/pharmacology , Naloxone/administration & dosage , Naloxone/pharmacology , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Probiotics/administration & dosage , Probiotics/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/biosynthesis , Receptor, Cannabinoid, CB2/physiology , Receptors, Cannabinoid/biosynthesis , Receptors, Opioid/biosynthesis , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid, mu/physiology , Rectum/drug effects , Rectum/microbiology , Rectum/physiopathology
4.
Gynecol Oncol ; 124(2): 319-24, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22037317

ABSTRACT

OBJECTIVE: The opioid growth factor (OGF) and its receptor (OGFr), serve as inhibitory axis regulating cell proliferation in normal cells and cancer. We investigated the presence and relative expression of OGF and OGFr in normal human ovarian surface epithelial (HOSE) cells, benign ovarian cysts, and ovarian cancers. METHODS: Surgical samples of 16 patients with ovarian cancer and 27 patients with ovarian benign cysts were obtained intraoperatively. HOSE were collected by scraping the surface of normal ovaries of 10 post menopausal women undergoing hysterectomy and oophorectomy. Semiquantitative immunohistochemistry was used to assess the presence, distribution, and levels of OGF and OGFr. Receptor binding assays measured binding capacity and affinity of OGFr for radiolabeled OGF. RESULTS: OGF and OGFr were present in HOSE cells, ovarian cysts, and ovarian cancers. Compared to HOSE cells, OGF and OGFr protein levels were reduced 29% and 34% (p<0.001), respectively, in ovarian cysts, and decreased 58% and 48% (p<0.001), respectively, in ovarian cancers. Binding assays revealed 5.4 fold fewer OGFr binding sites in cancers than cysts (p<0.05). Levels of OGF and OGFr were comparable in primary, metastatic, or recurrent ovarian cancers. CONCLUSION: We have shown that a native opioid pathway, the OGF-OGFr axis, is present in human ovarian cancer. Importantly, the expression of OGF and OGFr is diminished in human ovarian cancer. As OGF and OGFr normally function in maintaining cell proliferation, therapy to harness OGF/OGFr function could provide a useful biologic-based treatment for human ovarian cancer.


Subject(s)
Ovarian Neoplasms/metabolism , Receptors, Opioid/biosynthesis , Adult , Aged , Aged, 80 and over , Epithelial Cells/metabolism , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Cysts/metabolism , Ovary/cytology , Ovary/metabolism
5.
Parasite Immunol ; 34(11): 536-46, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22889318

ABSTRACT

Primary exposure of mice to the nematode Heligmosomoides polygyrus infection reduces inflammation in an experimental model of colitis. The aim of the present investigation was to evaluate whether the reduced inflammation provoked by H. polygyrus L4 larvae in BALB/c mice treated with dextran sulphate sodium is associated with changed expression of opioids in the small intestine and colon. Colitis was induced by 5% Dextran sulphate sodium (DSS) oral administration for 3 days before oral infection with 200 infective larvae (L3) H. polygyrus until the end of the experiment, 6 days post-infection. Clinical disease symptoms were monitored daily. The expressions of proopiomelanocortin POMC1, MOR1 (Oprm1) - opioid receptor and ß-endorphin were determined by RT-PCR, Western blot and immunoassay, respectively, in the colon and small intestine of mice. RT-PCR analysis of colon tissues showed up-regulation of the expression of POMC and MOR1 opioid-dependent genes in mice with DSS-induced colitis. H. polygyrus L4 larvae inhibited DSS-induced colitis symptoms that were correlated with increased IL-1ß, TNF-α, IL-6, myeloperoxidase (MPO) concentration, macrophages infiltration and MOR1, POMC and ß-endorphin increased expression in the small intestine and inhibition of those in the colon.


Subject(s)
Colitis/prevention & control , Intestines/physiology , Nematospiroides dubius/immunology , Pro-Opiomelanocortin/biosynthesis , Receptors, Opioid/biosynthesis , beta-Endorphin/biosynthesis , Animals , Blotting, Western , Colitis/chemically induced , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate , Gene Expression Profiling , Larva/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Peroxidase/metabolism , Real-Time Polymerase Chain Reaction , Severity of Illness Index
6.
Vis Neurosci ; 29(3): 203-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22643230

ABSTRACT

The presence of opioid receptors has been confirmed by a variety of techniques in vertebrate retinas including those of mammals; however, in most reports, the location of these receptors has been limited to retinal regions rather than specific cell types. Concurrently, our knowledge of the physiological functions of opioid signaling in the retina is based on only a handful of studies. To date, the best-documented opioid effect is the modulation of retinal dopamine release, which has been shown in a variety of vertebrate species. Nonetheless, it is not known if opioids can affect dopaminergic amacrine cells (DACs) directly, via opioid receptors expressed by DACs. This study, using immunohistochemical methods, sought to determine whether (1) µ- and δ-opioid receptors (MORs and DORs, respectively) are present in the mouse retina, and if present, (2) are they expressed by DACs. We found that MOR and DOR immunolabeling were associated with multiple cell types in the inner retina, suggesting that opioids might influence visual information processing at multiple sites within the mammalian retinal circuitry. Specifically, colabeling studies with the DAC molecular marker anti-tyrosine hydroxylase antibody showed that both MOR and DOR immunolabeling localize to DACs. These findings predict that opioids can affect DACs in the mouse retina directly, via MOR and DOR signaling, and might modulate dopamine release as reported in other mammalian and nonmammalian retinas.


Subject(s)
Amacrine Cells/metabolism , Dopaminergic Neurons/metabolism , Receptors, Opioid/biosynthesis , Retina/metabolism , Animals , Antibodies, Monoclonal/biosynthesis , Data Interpretation, Statistical , Female , Goats/immunology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Receptors, Opioid, delta/immunology , Receptors, Opioid, delta/physiology , Receptors, Opioid, mu/immunology , Receptors, Opioid, mu/physiology , Tyrosine 3-Monooxygenase/metabolism
7.
Methods ; 55(4): 310-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22041719

ABSTRACT

The biophysical characterization of purified membrane proteins typically requires detergent mediated extraction from native lipid membrane environments. In the case of human G protein-coupled receptors (GPCRs), this process has been complicated by their conformational heterogeneity and the general lack of understanding the composition and interactions within the diverse human cellular membrane environment. Several successful GPCR structure determination efforts have shown that the addition of cholesterol analogs is often critical for maintaining protein stability. We have identified sterols that substantially increase the stability of the NOP receptor (ORL-1), a member of the opioid GPCR family, in a mixed micelle environment. Using dynamic light scattering and small-angle X-ray scattering, we have determined that the most thermal stabilizing sterol, cholesteryl hemisuccinate, induces the formation of a bicelle-like micelle architecture when mixed with dodecyl maltoside detergent. Together with mutagenesis studies and recent GPCR structures, our results provide indications that stabilization is attained through a combination of specific sterol binding to GPCRs and modulation of micelle morphology.


Subject(s)
Cholesterol Esters/chemistry , Glucosides/chemistry , Micelles , Receptors, Opioid/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Algorithms , Humans , Light , Models, Molecular , Molecular Conformation , Particle Size , Protein Stability , Receptors, Opioid/chemistry , Recombinant Fusion Proteins/chemistry , Scattering, Radiation , Scattering, Small Angle , Transition Temperature , X-Ray Diffraction , Nociceptin Receptor
8.
JCI Insight ; 7(3)2022 02 08.
Article in English | MEDLINE | ID: mdl-34914639

ABSTRACT

Ca2+/calmodulin-stimulated group I adenylyl cyclase (AC) isoforms AC1 and AC8 have been involved in nociceptive processing and morphine responses. However, whether AC3, another member of group I ACs, is involved in nociceptive transmission and regulates opioid receptor signaling remains elusive. Here, we report that conditional KO of AC3 (AC3 CKO) in L3 and L4 DRGs robustly facilitated the mouse nociceptive responses, decreased voltage-gated potassium (Kv) channel currents, and increased neuronal excitability. Furthermore, we report AC3 CKO eliminated the analgesic effect of κ-opioid receptor (KOR) agonist and its inhibition on Kv channel by classical Gαi/o signaling or nonclassical direct interaction of KOR and AC3 proteins. Interestingly, significantly upregulated AC1 level and cAMP concentration were detected in AC3-deficient DRGs. Inhibition of AC1 completely reversed cAMP upregulation, neuronal excitability enhancement, and nociceptive behavioral hypersensitivity in AC3-CKO mice. Our findings suggest a crucial role of peripheral AC3 in nociceptive modulation and KOR opioid analgesia.


Subject(s)
Ablation Techniques/methods , Adenylyl Cyclases/genetics , Analgesia/methods , Gene Expression Regulation , Hyperalgesia/genetics , Receptors, Opioid/genetics , Adenylyl Cyclases/biosynthesis , Animals , Cells, Cultured , Disease Models, Animal , Hyperalgesia/metabolism , Hyperalgesia/therapy , Mice , RNA/genetics , RNA/metabolism , Receptors, Opioid/biosynthesis , Nociceptin Receptor
9.
Neurochem Res ; 36(11): 2008-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21671107

ABSTRACT

Methadone and buprenorphine are used in maintenance therapy for heroin addicts. In this study, we compared their effects on adenylate cyclase (AC) activity in human embryonic kidney (HEK) 293 cells stably overexpressing human µ-opioid receptor (MOR) and nociceptin/opioid receptor-like 1 receptor (ORL1) simultaneously. After acute exposure, methadone inhibited AC activity; however, buprenorphine induced compromised AC inhibition. When naloxone was introduced after 30 min incubation with methadone, the AC activity was enhanced. This was not observed in the case of buprenorphine. Enhancement of the AC activity was more significant when the incubation lasted for 4 h, and prolonged exposure to buprenorphine elevated the AC activity as well. The removal of methadone and buprenorphine by washing also obtained similar AC superactivation as that revealed by naloxone challenge. The study demonstrated that methadone and buprenorphine exert initially different yet eventually convergent adaptive changes of AC activity in cells coexpressing human MOR and ORL1 receptors.


Subject(s)
Buprenorphine/pharmacology , Methadone/pharmacology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/drug effects , Drug Interactions , Enzyme Activation , HEK293 Cells , Humans , Naloxone/pharmacology , Opioid Peptides/agonists , Receptors, Opioid/biosynthesis , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/biosynthesis , Nociceptin Receptor , Nociceptin
10.
Br J Anaesth ; 106(4): 566-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21324928

ABSTRACT

BACKGROUND: A role of nociceptin and its receptor (NOP) in pain and immune function has been suggested. The hypothesis was that mRNA expression of NOP and the nociceptin precursor pre-pronociceptin (pN/OFQ) in peripheral blood cells differs in end-stage cancer patients suffering from chronic pain and septic intensive care unit (ICU) patients compared with healthy controls. METHODS: Blood samples were drawn from end-stage cancer patients and septic ICU patients. Additionally, postoperative patients representing individuals with surgical stress and healthy controls were enrolled as comparative groups. NOP and pN/OFQ mRNA expression, quantified by real-time polymerase chain reaction (RT-PCR), was compared between study groups, and associated to opioid medication, pain intensities, and the inflammatory markers procalcitonin (PCT) and interleukin-6. RESULTS: NOP expression was significantly higher in cancer patients [normalized ratio, median (inter-quartile range): 10.2 (7.4/17.8)], postoperative patients [8.0 (5.3/10.2)], and ICU patients [6.6 (4.2/9.5)] compared with healthy controls [4.4 (2.7/7.0); P<0.001]. Expression of pN/OFQ was lower in cancer patients [3.8 (1.9/5.9)] and ICU patients [1.9 (1.0/2.7)] but not in postoperative patients compared with healthy controls [7.2 (6.1/9.4); P<0.001]. Increased plasma PCT was associated with decreased pN/OFQ in all patient groups. In cancer patients, no association was seen with pain scores, opioid medication or duration of analgesia, and NOP or pN/OFQ mRNA. CONCLUSIONS: NOP and pN/OFQ expression in peripheral blood cells was modulated in end-stage cancer and septic patients compared with healthy controls, whereas changes in postoperative patients were minor. The involvement of the NOP-pN/OFQ system in inflammation, impaired immune function, and pain has to be further elucidated.


Subject(s)
Neoplasms/blood , Protein Precursors/biosynthesis , Receptors, Opioid/biosynthesis , Sepsis/blood , Adult , Aged , Case-Control Studies , Critical Care , Female , Gene Expression , Humans , Inflammation/blood , Inflammation/etiology , Inflammation Mediators/blood , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/blood , Neoplasm Proteins/genetics , Neoplasms/complications , Pain/blood , Pain/etiology , Protein Precursors/blood , Protein Precursors/genetics , RNA, Messenger/genetics , Receptors, Opioid/blood , Receptors, Opioid/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Sepsis/complications , Nociceptin Receptor
11.
Exp Neurol ; 337: 113572, 2021 03.
Article in English | MEDLINE | ID: mdl-33340498

ABSTRACT

Neuropathic pain is the most common clinical disorder destroying the quality of patient life and leading to a marked economic and social burden. Opioids are still last option for pharmacological treatment of this disorder, but their antinociceptive effects are limited in part due to the downregulation of opioid receptors in the primary afferent neurons after peripheral nerve trauma. How this downregulation occurs is not completely understood, but recent studies have demonstrated that peripheral nerve trauma drives the alterations in epigenetic modifications (including DNA methylation, histone methylation and mciroRNAs), expression of transcription factors, post-transcriptional modifications (e.g., RNA methylation) and protein translation initiation in the neurons of nerve trauma-related dorsal root ganglion (DRG) and that these alternations may be associated with nerve trauma-caused downregulation of DRG opioid receptors. This review presents how opioid receptors are downregulated in the DRG after peripheral nerve trauma, specifically focusing on distinct molecular mechanisms underlying transcriptional and translational processes. This review also discusses how this downregulation contributes to the induction and maintenance of neuropathic pain. A deeper understanding of these molecular mechanisms likely provides a novel avenue for prevention and/or treatment of neuropathic pain.


Subject(s)
Neurons, Afferent/metabolism , Peripheral Nerve Injuries/genetics , Receptors, Opioid/genetics , Animals , Down-Regulation/genetics , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Humans , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Receptors, Opioid/biosynthesis
12.
Br J Anaesth ; 104(6): 698-704, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20413378

ABSTRACT

BACKGROUND: The expression of micro (mu: MOP) and nociceptin/orphanin FQ (NOP) receptors in the human myocardium is controversial. In this polymerase chain reaction (PCR)-based study using human right atrial biopsies, we have (i) probed for mRNA encoding NOP receptor and its endogenous peptide precursor, ppN/OFQ, and mRNA encoding MOP and (ii) attempted to correlate expression with cardiac function. METHODS: mRNA encoding MOP, NOP, and the precursor for NOP (ppN/OFQ) was assessed by quantitative real-time PCR (Q-PCR) using validated TaqMan primers and compared with a housekeeper (glyceraldehyde-3-phosphate dehydrogenase, GAPDH). Q-PCR data are expressed as the difference in cycle threshold (DeltaC(t)=C(tGene of interest)-C(tGAPDH): high value, low expression) and patients were grouped according to left ventricular ejection fraction (LVEF). RESULTS: Forty patients were recruited; NOP, MOP, and ppN/OFQ mRNA were measured in 38, 29, and 10 patients, respectively. DeltaC(t) (median and range) values for NOP and MOP were 10.9 (7.8-13.7) and 16.0 (12.3-18.9), respectively, representing low expression of MOP and approximately 34-fold more NOP. MOP mRNA was not detected in seven samples and with DeltaC(t) values of approximately 20, ppN/OFQ was considered absent. When patients were grouped into normal (>50%) and impaired (<50%) LVEF, there was no difference between the groups for either NOP or MOP. In some patients, intraoperative LVEF was estimated using transoesophageal echocardiography, and there was no correlation with either NOP or MOP. CONCLUSIONS: The human right atrium of patients with coronary artery disease and heart failure expresses mRNA encoding NOP and possibly low levels of MOP. This does not correlate with degree of cardiac dysfunction. In addition, the atrium does not express ppN/OFQ mRNA.


Subject(s)
Coronary Artery Disease/metabolism , Myocardium/metabolism , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid/biosynthesis , Adult , Aged , Aged, 80 and over , Coronary Artery Bypass , Coronary Artery Disease/surgery , Female , Heart Atria/metabolism , Humans , Male , Middle Aged , Polymerase Chain Reaction/methods , RNA, Messenger/genetics , Receptors, Opioid/genetics , Receptors, Opioid, mu/genetics , Stroke Volume/physiology , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left/physiology , Nociceptin Receptor
13.
Article in English | MEDLINE | ID: mdl-31904442

ABSTRACT

Potent opioid-based therapies are often unsuccessful in promoting satisfactory analgesia in neuropathic pain. Moreover, the side effects associated with opioid therapy are still manifested in neuropathy-like diseases, including tolerance, abuse, addiction and hyperalgesia, although the mechanisms underlying these effects remain unclear. Studies in the spinal cord and periphery indicate that neuropathy alters the expression of mu-[MOP], delta-[DOP] or kappa-[KOP] opioid receptors, interfering with their activity. However, there is no consensus as to the supraspinal opioidergic modulation provoked by neuropathy, the structures where the sensory and affective-related pain components are processed. In this study we explored the effect of chronic constriction of the sciatic nerve (CCI) over 7 and 30 days (CCI-7d and CCI-30d, respectively) on MOP, DOP and KOP mRNAs expression, using in situ hybridization, and the efficacy of G-protein stimulation by DAMGO, DPDPE and U-69593 (MOP, DOP and KOP specific agonists, respectively), using [35S]GTPγS binding, within opioid-sensitive brain structures. After CCI-7d, CCI-30d or both, opioid receptor mRNAs expression was altered throughout the brain: MOP - in the paracentral/centrolateral thalamic nuclei, ventral posteromedial thalamic nuclei, superior olivary complex, parabrachial nucleus [PB] and posterodorsal tegmental nucleus; DOP - in the somatosensory cortex [SSC], ventral tegmental area, caudate putamen [CPu], nucleus accumbens [NAcc], raphe magnus [RMg] and PB; and KOP - in the locus coeruleus. Agonist-stimulated [35S]GTPγS binding was altered following CCI: MOP - CPu and RMg; DOP - prefrontal cortex [PFC], SSC, RMg and NAcc; and KOP - PFC and SSC. Thus, this study shows that several opioidergic circuits in the brain are recruited and modified following neuropathy.


Subject(s)
Analgesics, Opioid/therapeutic use , Brain/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid/biosynthesis , Analgesics, Opioid/pharmacology , Animals , Brain/drug effects , Gene Expression , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid/genetics , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/metabolism
14.
Int Immunopharmacol ; 86: 106718, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32585612

ABSTRACT

The metastasis of cervical cancer has always been a clinical challenge. We investigated the effects of low-dose naltrexone (LDN) on the epithelial mesenchymal transition of cervical cancer cells in vitro as well as its influence on macrophage polarization and associated cytokines in vivo. The results suggested that LDN supressed the proliferation, migration and invasion abilities and promote their apoptosis in Hela cells, whereas the opioid growth factor receptor (OGFr) silenced significantly reversed these effects in vitro. Knockdown the expression of OGFr, the inhibitory of LDN on EMT was weakened. LDN could inhibit cervical cancer progression in nude mice. In additon, LDN indirectly reduced the number of tumor-associated macrophages (TAMs), mainly M2 macrophages, and decreased expression of anti-inflammatory factor IL-10 in the serum of nude mice. These findings demonstrate that LDN could be a potential treatment for cervical cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Naltrexone/pharmacology , Uterine Cervical Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytokines/blood , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , Humans , Macrophage Activation/drug effects , Mice, Inbred BALB C , Mice, Nude , Naltrexone/therapeutic use , Receptors, Opioid/biosynthesis , Receptors, Opioid/drug effects , Receptors, Opioid/genetics , Snail Family Transcription Factors/metabolism , Tumor-Associated Macrophages/drug effects , Uterine Cervical Neoplasms/pathology , Vimentin/metabolism , Xenograft Model Antitumor Assays
15.
Front Immunol ; 11: 615327, 2020.
Article in English | MEDLINE | ID: mdl-33408720

ABSTRACT

Clinical management of neuropathic pain is unsatisfactory, mainly due to its resistance to the effects of available analgesics, including opioids. Converging evidence indicates the functional interactions between chemokine and opioid receptors and their influence on nociceptive processes. Recent studies highlight that the CC chemokine receptors type 2 (CCR2) and 5 (CCR5) seem to be of particular interest. Therefore, in this study, we investigated the effects of the dual CCR2/CCR5 antagonist, cenicriviroc, on pain-related behaviors, neuroimmune processes, and the efficacy of opioids in rats after chronic constriction injury (CCI) of the sciatic nerve. To define the mechanisms of action of cenicriviroc, we studied changes in the activation/influx of glial and immune cells and, simultaneously, the expression level of CCR2, CCR5, and important pronociceptive cytokines in the spinal cord and dorsal root ganglia (DRG). We demonstrated that repeated intrathecal injections of cenicriviroc, in a dose-dependent manner, alleviated hypersensitivity to mechanical and thermal stimuli in rats after sciatic nerve injury, as measured by von Frey and cold plate tests. Behavioral effects were associated with the beneficial impact of cenicriviroc on the activation/influx level of C1q/IBA-1-positive cells in the spinal cord and/or DRG and GFAP-positive cells in DRG. In parallel, administration of cenicriviroc decreased the expression of CCR2 in the spinal cord and CCR5 in DRG. Concomitantly, we observed that the level of important pronociceptive factors (e.g., IL-1beta, IL-6, IL-18, and CCL3) were increased in the lumbar spinal cord and/or DRG 7 days following injury, and cenicriviroc was able to prevent these changes. Additionally, repeated administration of this dual CCR2/CCR5 antagonist enhanced the analgesic effects of morphine and buprenorphine in neuropathic rats, which can be associated with the ability of cenicriviroc to prevent nerve injury-induced downregulation of all opioid receptors at the DRG level. Overall, our results suggest that pharmacological modulation based on the simultaneous blockade of CCR2 and CCR5 may serve as an innovative strategy for the treatment of neuropathic pain, as well as in combination with opioids.


Subject(s)
Analgesics/therapeutic use , CCR5 Receptor Antagonists/therapeutic use , Hyperalgesia/drug therapy , Imidazoles/therapeutic use , Neuralgia/drug therapy , Receptors, CCR2/antagonists & inhibitors , Sciatic Neuropathy/complications , Sulfoxides/therapeutic use , Analgesics/administration & dosage , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , CCR5 Receptor Antagonists/administration & dosage , CCR5 Receptor Antagonists/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , Dose-Response Relationship, Drug , Drug Synergism , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Imidazoles/administration & dosage , Imidazoles/pharmacology , Injections, Spinal , Male , Morphine/pharmacology , Morphine/therapeutic use , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuralgia/etiology , Neuralgia/physiopathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, CCR2/biosynthesis , Receptors, CCR2/genetics , Receptors, CCR5/biosynthesis , Receptors, CCR5/genetics , Receptors, Opioid/biosynthesis , Receptors, Opioid/genetics , Spinal Cord/metabolism , Sulfoxides/administration & dosage , Sulfoxides/pharmacology
16.
Biochem Biophys Res Commun ; 385(3): 375-9, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19463790

ABSTRACT

Guanine nucleotide-binding protein (G protein) coupled receptors (GPCRs) are frequently expressed by a baculovirus expression vector system (BEVS). We recently established a novel BEVS using the bacmid system of Bombyx mori nucleopolyhedrovirus (BmNPV), which is directly applicable for protein expression in silkworms. Here, we report the first example of GPCR expression in silkworms by the simple injection of BmNPV bacmid DNA. Human nociceptin receptor, an inhibitory GPCR, and its fusion protein with inhibitory G protein alpha subunit (G(i)alpha) were both successfully expressed in the fat bodies of silkworm larvae as well as in the BmNPV viral fraction. Its yield was much higher than that from Sf9 cells. The microsomal fractions including the nociceptin receptor fusion, which are easily prepared by only centrifugation steps, exhibited [35S]GTPgammaS-binding activity upon specific stimulation by nociceptin. Therefore, this rapid method is easy-to-use and has a high expression level, and thus will be an important tool for human GPCR production.


Subject(s)
Bombyx/metabolism , Protein Biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Opioid/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Animals , Bombyx/genetics , Bombyx/virology , DNA/genetics , Genetic Vectors/genetics , Humans , Nucleopolyhedroviruses/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Opioid/genetics , Recombinant Fusion Proteins/genetics , Nociceptin Receptor
17.
Exp Biol Med (Maywood) ; 234(5): 532-41, 2009 May.
Article in English | MEDLINE | ID: mdl-19244545

ABSTRACT

The opioid growth factor receptor (OGFr) mediates the inhibitory action of OGF on cell replication of normal and neoplastic cells. The spatiotemporal course of OGFr nucleocytoplasmic trafficking was determined with a probe of full-length OGFr fused to enhanced green fluorescent protein (eGFP). Translation of OGFr required 8.5 hours, and transit into the nucleus required 8 hours; OGFr remained in the nucleus for 8 days. OGFr was initially expressed on the outer nuclear envelope, transited to the paranuclear cytoplasm, and into the nucleus. Transport through the nuclear pore was elucidated by mutation of the nuclear localization signal (NLS) sequences in full-length OGFr. Mutation of each NLS reduced nuclear localization by 5%-50%, whereas simultaneous mutation of NLS383-386 and NLS456-460 abolished OGFr-eGFP nuclear localization in 80% of the cells. To determine whether intact NLSs are important for the inhibition of cell proliferation, DNA synthesis was monitored with BrdU. Wild-type OGFr-eGFP-transfected cells had 20% BrdU-positive cells, whereas cells with simultaneous mutation of all three NLS sites had a 70% labeling index. These results indicate that the regulation of cell proliferation by the OGF-OGFr axis is dependent on nucleocytoplasmic translocation and reliant on the integrity of two NLSs in OGFr to interact with transport receptors.


Subject(s)
Cell Proliferation , DNA/biosynthesis , Nuclear Envelope/metabolism , Nuclear Localization Signals/metabolism , Protein Biosynthesis/physiology , Receptors, Opioid/biosynthesis , Active Transport, Cell Nucleus/physiology , Cell Line, Tumor , DNA/genetics , Humans , Mutation , Nuclear Envelope/genetics , Nuclear Localization Signals/genetics , Receptors, Opioid/genetics , Time Factors
18.
J Neuroendocrinol ; 31(12): e12810, 2019 12.
Article in English | MEDLINE | ID: mdl-31715027

ABSTRACT

The onset of puberty is the result of an increase in secretion of hypothalamic gonadotrophin-releasing hormone (GnRH). This action is a result of not only the development of stimulatory inputs to its release, but also the gradual decrease in inhibitory inputs that restrain release of the peptide prior to pubertal onset. Dynorphin (DYN) is one of the inhibitory inputs produced in the medial basal hypothalamus (MBH); however, little is known about what substance(s) control its prepubertal synthesis and release. Because neurokinin B (NKB) increases in the hypothalamus as puberty approaches, we considered it a candidate for such a role. An initial study investigated the acute effects of an NKB agonist, senktide, on the secretion of DYN from MBH tissues incubated in vitro. In other experiments, central injections of senktide were administered to animals for 4 days then MBHs were collected for assessment of DYN synthesis or for the in vitro secretion of both DYN and GnRH. Because insulin-like growth factor (IGF)-1 has been shown to play an important role at puberty, additional animals received central injections of this peptide for 4 days to assess NKB and DYN synthesis or the in vitro secretion of NKB. The results obtained show that senktide administration up-regulates the NKB receptor protein, at the same time as suppressing the DYN and its receptor. Senktide consistently suppressed DYN and elevated GnRH secretion in the same tissue incubates from both the acute and chronic studies. IGF-1 administration caused an increase in NKB protein, at the same time as decreasing DYN protein. Furthermore, the central administration of IGF-1 caused an increase in NKB release, an action blocked by the IGF-1 receptor blocker, JB-1. These results indicate that the IGF-1/NKB pathway contributes to suppressing the DYN inhibitory tone on prepubertal GnRH secretion and thus facilitates the puberty-related increase in the release of GnRH to accelerate the onset of puberty.


Subject(s)
Dynorphins/metabolism , Hypothalamus, Middle/metabolism , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus, Middle/drug effects , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Microinjections , Neurokinin B/metabolism , Peptide Fragments/pharmacology , Peptides/pharmacology , Rats , Receptor, IGF Type 1/antagonists & inhibitors , Receptors, Neurokinin-3/biosynthesis , Receptors, Opioid/biosynthesis , Sexual Maturation , Substance P/analogs & derivatives , Substance P/pharmacology , Up-Regulation
19.
J Neurosci ; 27(49): 13371-5, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-18057194

ABSTRACT

Dysfunction of mesocorticolimbic dopaminergic neurons is considered a common feature of all drugs of abuse, yet few investigations have evaluated the dopamine (DA) system in nonstimulant human abusers. We examined mRNA expression levels of DA transporter (DAT), tyrosine hydroxylase (TH), dopamine D2 receptor, alpha-synuclein, and nuclear receptor-related 1 (Nurr1) in discrete mesocorticolimbic and nigrostriatal subpopulations of heroin users and control subjects. The chronic use of heroin was significantly associated with decreased DAT mRNA expression localized to the paranigral nucleus (PN) and the mesolimbic division of the ventral tegmental area (VTA) with no alterations in nigrostriatal populations. Consistently, the density of DAT immunoreactivity was significantly reduced in the nucleus accumbens but not in dorsal striatum, mesolimbic and nigrostriatal efferent targets, respectively. Significant alteration of the mRNA expression of Nurr1, a transcription factor that regulates DAT expression, was also confined to the PN. Moreover, the results revealed an exaggerated reduction of Nurr1 expression with age in heroin users (r = -0.8268, p < 0.001 vs controls, r = -0.6204, p = 0.0746). TH and alpha-synuclein mRNA levels were, in contrast, elevated in the VTA PN in heroin users with no change of the D2 receptor. Evaluating midbrain mu- and kappa-opioid receptors, relevant for the action of heroin and regulation of DA neurons, revealed dysregulation of G-protein coupling selective to the VTA PN. Altogether the current findings provide direct neurobiological evidence that midbrain reward circuits have the most prominent DA and opioid impairments in human heroin abusers and that abnormal Nurr1 transcription with opiate use may exacerbate limbic dysfunction with age.


Subject(s)
Aging/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , Dopamine/biosynthesis , Heroin Dependence/metabolism , Receptors, Opioid/biosynthesis , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription, Genetic/physiology , Aging/genetics , DNA-Binding Proteins/genetics , Dopamine/genetics , Dopamine Plasma Membrane Transport Proteins/biosynthesis , Dopamine Plasma Membrane Transport Proteins/genetics , Heroin Dependence/genetics , Humans , Limbic System/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2 , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Opioid/genetics , Transcription Factors/genetics
20.
Neuroscience ; 151(1): 129-37, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18082336

ABSTRACT

Zebrafish is a novel experimental model that has been used in developmental studies as well as in the study of pathological processes involved in human diseases. It has been demonstrated that the endogenous opioid system is involved in developmental mechanisms. We have studied the relationship between the different embryonic stages and opioid receptor expression for the four known opioid receptors in zebrafish (mu, delta 1, delta 2 and kappa). The mu opioid receptor is detected at higher levels than the other opioid receptors before the midblastula transition and during the segmentation period. The delta duplicate 2 exhibits only one peak of expression at 21 h postfertilization (hpf), when the motor nervous system is forming. The kappa receptor is expressed at very low levels. In situ hybridization studies at 24 hpf show that the opioid receptors are widely distributed in zebrafish CNS and at 48 hpf their localization is detected in more defined structures. Our results support specific implications of the opioid receptors in developmental processes such as morphogenesis of the CNS, neurogenesis, neuroprotection and development of neuromuscular and digestive system. Pain-related alterations can be a consequence of changes in the endogenous opioid system during development, hence we provide important information that might help to solve pain-related pathological situations.


Subject(s)
Aging/metabolism , Receptors, Opioid/biosynthesis , Receptors, Opioid/metabolism , Actins/biosynthesis , Actins/genetics , Animals , Brain Chemistry/genetics , Brain Chemistry/physiology , Cloning, Molecular , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Embryo, Nonmammalian , In Situ Hybridization , RNA/biosynthesis , RNA/isolation & purification , Receptors, Opioid, delta/biosynthesis , Receptors, Opioid, delta/genetics , Receptors, Opioid, kappa/biosynthesis , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid, mu/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL