Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Endocrinology ; 162(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34270714

RESUMO

Energetic status often affects reproductive function, glucose homeostasis, and feeding in mammals. Malnutrition suppresses pulsatile release of the gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) and increases gluconeogenesis and feeding. The present study aims to examine whether ß-endorphin-µ-opioid receptor (MOR) signaling mediates the suppression of pulsatile GnRH/LH release and an increase in gluconeogenesis/feeding induced by malnutrition. Ovariectomized female rats treated with a negative feedback level of estradiol-17ß (OVX + low E2) receiving 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, intravenously (iv) were used as a malnutrition model. An administration of D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP), a selective MOR antagonist, into the third ventricle blocked the suppression of the LH pulse and increase in gluconeogenesis/feeding induced by iv 2DG administration. Histological analysis revealed that arcuate Kiss1 (kisspeptin gene)-expressing cells and preoptic Gnrh1 (GnRH gene)-expressing cells co-expressed little Oprm1 (MOR gene), while around 10% of arcuate Slc17a6 (glutamatergic marker gene)-expressing cells co-expressed Oprm1. Further, the CTOP treatment decreased the number of fos-positive cells in the paraventricular nucleus (PVN) in OVX + low E2 rats treated with iv 2DG but failed to affect the number of arcuate fos-expressing Slc17a6-positive cells. Taken together, these results suggest that the central ß-endorphin-MOR signaling mediates the suppression of pulsatile LH release and that the ß-endorphin may indirectly suppress the arcuate kisspeptin neurons, a master regulator for GnRH/LH pulses during malnutrition. Furthermore, the current study suggests that central ß-endorphin-MOR signaling is also involved in gluconeogenesis and an increase in food intake by directly or indirectly acting on the PVN neurons during malnutrition in female rats.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/metabolismo , Antagonistas de Entorpecentes/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Opioides mu/metabolismo , beta-Endorfina/metabolismo , Animais , Glicemia/análise , Feminino , Gluconeogênese , Hipotálamo , Kisspeptinas/metabolismo , Ratos , Ratos Wistar , Receptores Opioides mu/biossíntese , Transdução de Sinais , Proteína Vesicular 2 de Transporte de Glutamato/biossíntese
2.
Neurosci Lett ; 746: 135661, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33482308

RESUMO

Pain is a common and devastating symptom among cancer patients. It can be caused by the cancer itself or by certain therapies like surgery, radiation or chemotherapy. Opioids are the first line of treatment for moderate to severe cancer-related pain. Opioids alone or in combination with non-opioid analgesics and adjuvant medications are important components for pain management during the perioperative period for cancer patients. Opioids act on the µ-opioid receptor (MOR), which is expressed in cancer cells and non-malignant cells of the tumor microenvironment. Retrospective studies suggest an association between the expression of MOR in cancers and shorter survival. In addition, recent evidence suggests that opium use and prescription opioids can influence clinical oncological outcomes. In this review, we will summarize the clinical evidence regarding the effect of opioid administration and survival in patients with cancer as well as the current evidence involving MOR expression and cancer progression.


Assuntos
Analgésicos Opioides/efeitos adversos , Dor do Câncer/metabolismo , Medicina Baseada em Evidências/métodos , Neoplasias/metabolismo , Receptores Opioides mu/biossíntese , Microambiente Tumoral/fisiologia , Analgésicos Opioides/administração & dosagem , Animais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Dor do Câncer/diagnóstico , Dor do Câncer/tratamento farmacológico , Humanos , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Manejo da Dor/efeitos adversos , Manejo da Dor/métodos , Prognóstico , Receptores Opioides mu/genética , Estudos Retrospectivos , Microambiente Tumoral/efeitos dos fármacos
3.
J Cancer Res Clin Oncol ; 147(3): 779-792, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33315125

RESUMO

PURPOSE: D,L-methadone (MET), an analgesic drug used for pain treatment and opiate addiction, has achieved attention from oncologists and social media as possible chemoensitizing agent in cancer therapy, notably brain cancer (glioblastoma multiforme, GBM). MET has been reported to enhance doxorubicin-induced cytotoxicity in GBM cells via activation of the µ-opioid receptor (MOR). Here, we extended this work and quantified the toxic effect of MET in comparison to other opioids alone and in combination with doxorubicin and the clinically more relevant alkylating drug temozolomide (TMZ), using a set of GBM cell lines and primary GBM cells. METHODS: MOR expression in GBM cells was investigated by immunofluorescence and immunoblotting. Resistance to drugs alone and in combination with anticancer drugs was assessed by MTT assays. Concentration effect curves were fitted by nonlinear regression analysis and IC50 values were calculated. Apoptosis and necrosis rates were determined by annexin V/propidium iodide (PI)-flow cytometry. RESULTS: MET alone was cytotoxic in all GBM cell lines and primary GBM cells at high micromolar concentrations (IC50 ~ 60-130 µM), observed both in the metabolic MTT assay and by quantifying apoptosis and necrosis, while morphine and oxycodone were not cytotoxic in this concentration range. Naloxone was not able to block MET-induced cytotoxicity, indicating that cell death-inducing effects of MET are not MOR-dependent. We recorded doxorubicin and TMZ concentration- response curves in combination with fixed MET concentrations. MET enhanced doxorubicin-induced cytotoxicity in only one cell line, and in primary cells it was observed only in a particular MET concentration range. In all assays, MET was not effective in sensitizing cells to TMZ. In two cell lines, MET even decreased the cell's sensitivity to TMZ. CONCLUSION: MET was found to be cytotoxic in GBM cells in vitro only at high, clinically not relevant concentrations, where it was effective in inducing apoptosis and necrosis. Sensitizing effects were only observed in combination with doxorubicin, but not with TMZ, and are dependent on cell line and the applied drug concentration. Therefore, our findings do not support the use of MET in the treatment of GBM in combination with TMZ, as no sensitizing effect of MET was observed.


Assuntos
Analgésicos Opioides/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Doxorrubicina/farmacologia , Glioblastoma/tratamento farmacológico , Metadona/farmacologia , Analgésicos Opioides/administração & dosagem , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Metadona/administração & dosagem , Morfina/farmacologia , Naloxona/farmacologia , Oxicodona/farmacologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/biossíntese , Células Tumorais Cultivadas
4.
Br J Anaesth ; 125(5): 722-729, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32900505

RESUMO

BACKGROUND: Expression of the mu-opioid receptor (MOR) is associated with poor long-term outcomes in various types of cancer. The association between MOR expression and clinical outcomes in laryngeal squamous cell carcinoma (LSCC) is not clear. METHODS: This retrospective study included patients who underwent laryngectomy for LSCC. The expression pattern of the MOR protein and OPRM1 gene in tumours and corresponding adjacent non-carcinoma specimens was measured. Propensity score matching was used to minimise bias. The primary endpoints were overall survival (OS) and disease-free survival (DFS). The secondary endpoints were intraoperative sufentanil consumption, grade of surgical complications according to the Clavien-Dindo classification, and hospital length of stay. RESULTS: A total of 207 LSCC patients were enrolled. After propensity score matching, there was a significant difference in DFS between groups at 1, 3, and 5 yr (60.2% vs 81.2%, P=0.019; 39.4% vs 50.2%, P=0.026; 37.5% vs 42.5%, P=0.023, respectively) in patients with high MOR expression. The OS rates at 1, 3, and 5 yr were significantly lower in the high MOR expression group (81.2% vs 93.2%, P=0.027; 57.7% vs 78.3%, P<0.001; 42.5% vs 60.3%, P<0.001, respectively). The multivariate analysis indicated that high MOR expression was associated with worse DFS and OS (hazard ratio: 1.52, 95% confidence interval: 1.07, 2.25, P=0.034; hazard ratio: 1.42, 95% confidence interval: 1.17, 2.34, P=0.032). CONCLUSION: High MOR expression may be associated with poor prognosis in patients with LSCC, suggesting that MOR could be used as a valuable molecular biomarker to predict prognosis of LSCC patients.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidade , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/mortalidade , Receptores Opioides mu/biossíntese , Adulto , Idoso , Anestesia , Carcinoma de Células Escamosas/cirurgia , Intervalo Livre de Doença , Determinação de Ponto Final , Feminino , Humanos , Estimativa de Kaplan-Meier , Neoplasias Laríngeas/cirurgia , Laringectomia , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Pontuação de Propensão , Receptores Opioides mu/genética , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento
5.
Am J Clin Oncol ; 43(8): 591-597, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32482952

RESUMO

OBJECTIVES: Pancreatic adenocarcinoma is frequently associated with pain requiring opioid therapy. Opioids, however, have been implicated in causing tumor progression, ultimately shortening survival. We examined the impact of pain, opioid use, and the mu-opioid receptor (MOP-R) expression in tumor tissue on progression-free survival and overall survival of patients with metastatic pancreatic cancer. METHODS: We identified 103 patients with metastatic pancreatic adenocarcinoma receiving chemotherapy and abstracted data from Tumor Registry, in addition to pain, opioid exposure, carbohydrate antigen 19-9 values, survival, and imaging response. MOP-R expression was evaluated using an immunohistochemistry assay. The association of variables with progression-free survival and overall survival was analyzed in univariate and multivariate models. RESULTS: Patients with low opioid use (<5 mg oral morphine equivalent/d) survived longer than patients with high opioid (HO) use (≥5 mg oral morphine equivalent/d) (median overall survival of 315 vs. 150 d; hazard ratio [HR]=1.79; 95% confidence interval [CI]: 1.13, 2.84). This effect persisted on multivariate models (adjusted HR=2.76; 95% CI: 1.39, 5.48). Low opioid patients tended to respond better to treatment than HO patients, based on carbohydrate antigen 19-9. Patients with low MOP-R expression had longer median survival (230 vs. 193 d), though the HR was not significant (1.15; 95% CI: 0.71, 1.88). Baseline pain was not associated with outcomes. CONCLUSION: In patients with metastatic pancreatic adenocarcinoma, HO use is associated with decreased survival, but the severity of baseline pain and MOP-R expression score in tumor tissue does not correlate with clinical outcomes.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Dor do Câncer/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Receptores Opioides mu/biossíntese , Receptores Opioides mu/efeitos dos fármacos , Adenocarcinoma/mortalidade , Adenocarcinoma/secundário , Adulto , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Pancreáticas/mortalidade , Intervalo Livre de Progressão , Estudos Retrospectivos , Taxa de Sobrevida
6.
Cancer Lett ; 453: 1-9, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30928385

RESUMO

Hepatocellular carcinoma (HCC) is the most prevalent subtype of liver cancer. Anesthetic regimens possibly influence cancer development. Exploration of novel, effective targets for liver cancer is the current hotspot in cancer treatment. A previous study conducted by us has demonstrated that enhanced expression of the µ-opioid receptor (MOR) promotes cell proliferation, adhesion, migration, and tumorigenesis. The current study investigates whether MOR regulates self-renewal of hepatocellular carcinoma stem cells (HCSCs). We utilize cell function assays, siRNA, shRNA, flow cytometry sorting, and other molecular biology techniques for this purpose. The results indicate that MOR expression is positively related to hepatocarcinoma progression. Silencing MOR greatly reduce HCC-related tumorigenesis both in vitro and in vivo and significantly extend the survival of tumor-bearing mice. Moreover, MOR silencing will greatly reduce colony formation by HCC cells, indicating down-regulation of cancer initiation. In conclusion, these results establish that MOR can be a novel and reliable HCSC marker and a potential therapeutic target against HCC via MOR-NFAT signaling.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Receptores Opioides mu/biossíntese , Animais , Carcinogênese , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Células Hep G2 , Xenoenxertos , Humanos , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Receptores Opioides mu/genética , Transcrição Gênica
7.
Br J Anaesth ; 122(6): e157-e167, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30915986

RESUMO

BACKGROUND: Opioid receptors are implicated in cancer progression and long-term patient outcomes. However, the prognostic significance, underlying mechanisms, and therapeutic value of mu-opioid receptor (MOP) in hepatocellular carcinoma (HCC) remain unclear. METHODS: MOP expression in human biopsy HCC samples was evaluated using RNA microarrays, quantitative real-time polymerase chain reaction (qRT-PCR), and immunochemical analyses. Molecular and cellular techniques, including siRNA-mediated depletion and lentiviral vector-mediated overexpression, were used to elucidate the functions and mechanisms of MOP. The effect of the MOP agonist morphine in HCC was evaluated both in vitro and in vivo. The therapeutic value of MOP inhibitors in HCC progression and metastasis was investigated with in vitro experiments and subcutaneous and orthotopic HCC mouse models in vivo. RESULTS: Through microarray analysis and qRT-PCR, we identified that MOP is highly expressed in human HCC tumours. High MOP expression in HCC tumours was confirmed by immunocytochemistry and correlated with aggressive clinicopathological features and a worse prognosis. Depletion of MOP suppressed cell proliferation, migration, and invasion, whereas overexpression of MOP promoted cell growth and metastasis in human HCC cell lines. Both clinical and biological evidence revealed that MOP-mediated epithelial-mesenchymal transition promotes HCC metastasis and poor prognosis. Morphine promotes cell proliferation, migration, and invasion in vitro and in vivo in mouse models. More importantly, MOP inhibitors suppressed cell growth, invasion, and metastasis in vitro and in the subcutaneous and orthotopic xenograft models. CONCLUSIONS: MOP plays a key oncogenic function in hepatocarcinogenesis. Its overexpression is associated with poor prognosis in patients with HCC. Furthermore, MOP inhibitors may be a promising strategy for HCC therapy.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores Opioides mu/biossíntese , Adolescente , Adulto , Idoso , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Animais , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/secundário , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Progressão da Doença , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Terapia de Alvo Molecular/métodos , Morfina/efeitos adversos , Morfina/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Invasividade Neoplásica , Prognóstico , RNA Mensageiro/genética , RNA Neoplásico/genética , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Receptores Opioides mu/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adulto Jovem
8.
Anesth Analg ; 122(4): 1031-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26720619

RESUMO

BACKGROUND: Morphine is widely used in patients with moderate and severe cancer pain, whereas the development of drug tolerance remains a major problem associated with opioid use. Previous studies have shown that cannabinoid type 2 (CB2) receptor agonists induce morphine analgesia, attenuate morphine tolerance in normal and neuropathic pain animals, induce transcription of the µ-opioid receptor (MOR) gene in Jurkat T cells, and increase morphine analgesia in cancer pain animals. However, no studies of the effects of CB2 receptor agonists on morphine tolerance in cancer pain have been performed. Therefore, we investigated the effect of repeated intrathecal (IT) injection of the low-dose CB2 receptor agonist AM1241 on the development of morphine tolerance in walker 256 tumor-bearing rats. We also tested the influence of the CB2 receptor agonist AM1241 on MOR protein and messenger ribonucleic acid (mRNA) expression in the rat spinal cord and dorsal root ganglia (DRG). METHODS: Walker 256 cells were implanted into the plantar region of each rat's right hindpaw. Tumor-bearing rats received IT injection of the CB2 receptor agonist AM1241 or antagonist AM630 with or without morphine subcutaneously twice daily for 8 days. Rats receiving drug vehicle only served as the control group. Mechanical paw withdrawal threshold and thermal paw withdrawal latency were assessed by a von Frey test and hot plate test 30 minutes after drug administration every day. MOR protein and mRNA expression in the spinal cord and DRG were detected after the last day (day 8) of drug administration via Western blot and real-time reverse transcription polymerase chain reaction. The data were analyzed via analysis of variance followed by Student t test with Bonferroni correction for multiple comparisons. RESULTS: Repeated morphine treatments reduced the mechanical withdrawal threshold and thermal latency. Coadministration of a nonanalgetic dose of the CB2 receptor agonist AM1241 with morphine significantly inhibited the development of morphine tolerance and increased the MOR protein expression in the spinal cord and DRG and mRNA expression in the spinal cord in tumor-bearing rats. CONCLUSIONS: Our findings indicate that IT injection of a nonanalgetic dose of a CB2 receptor agonist increased the analgesia effect and alleviated tolerance to morphine in tumor-bearing rats, potentially by regulating MOR expression in the spinal cord and DRG. This receptor may be a new target for prevention of the development of opioid tolerance in cancer pain.


Assuntos
Carcinoma 256 de Walker/metabolismo , Regulação Neoplásica da Expressão Gênica , Morfina/administração & dosagem , Receptor CB2 de Canabinoide/agonistas , Receptores Opioides mu/biossíntese , Animais , Canabinoides/administração & dosagem , Tolerância a Medicamentos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Masculino , Ratos , Ratos Wistar , Receptores Opioides mu/genética
9.
Psychopharmacology (Berl) ; 233(4): 661-72, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26554386

RESUMO

RATIONALE: Repeated administration of methamphetamine (Meth) induces behavioral sensitization which is characterized by a progressive increase in locomotor response after each injection. Previous studies have shown that Mu opioid receptors (MORs) can regulate Meth-mediated behavioral sensitization. However, the reported interactions are controversial; systemic activation of MORs either enhanced or suppressed Meth sensitization. It is possible that alteration of Meth sensitization after systemic administration of MOR ligands reflects the sum of distinct MOR reactions in multiple brain regions. OBJECTIVES: The purpose of the present study was to examine the actions of MORs on Meth sensitization after regionally selective overexpression of human MOR through an AAV6-based gene delivery system. METHOD: We demonstrated that adeno-associated virus (AAV)-MOR increased MOR immunoreactivity and binding in vitro. AAV-MOR or AAV-green fluorescent protein (GFP) was injected into the nucleus accumbens (NAc) or ventral tegmental area (VTA) of adult mice. Two weeks after viral infection, animals received Meth or saline for five consecutive days. Locomotor behavior and striatal dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC) level were determined. RESULTS: Repeated administration of Meth progressively increased locomotor activity; this sensitization reaction was attenuated by intra-NAc AAV-MOR microinjections. Infusion of AAV-MOR to VTA enhanced Meth sensitization. AAV-MOR significantly enhanced DA levels in VTA after VTA infection but reduced DOPAC/DA turnover in the NAc after NAc injection. CONCLUSION: Our data suggest a differential modulation of Meth sensitization by overexpression of MOR in NAc and VTA. Regional manipulation of MOR expression through AAV may be a novel approach to control Meth abuse and psychomimetic activity.


Assuntos
Metanfetamina/administração & dosagem , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptores Opioides mu/biossíntese , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microinjeções , Atividade Motora/fisiologia
10.
Peptides ; 62: 144-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25281794

RESUMO

Atopic dermatitis (AD) is a chronic inflammatory skin disease with heterogeneous clinical phenotypes reflecting genetic predisposition and exposure to environmental factors. Reactions to food may play a significant role especially in young children. Milk proteins are particularly strong allergens and are additional source of bioactive peptides including ß-casomorphin-7 (BCM7, Tyr-Pro-Phe-Pro-Gly-Pro-Ile). BCM7 exerts its influence on nervous, digestive, and immune functions via the µ-opioid receptor (MOR). Proline dipeptidyl peptidase IV (DPPIV; EC 3.4.14.5) appears to be the primary degrading enzyme of BCM7. Moreover, DPPIV is known to restrict activity of proinflammatory peptides. BCM7 is considered to modulate an immune response by affecting MOR and DPPIV genes expression. In this study, we determined the MOR and DPPIV genes expression in children diagnosed with a severe form of AD. 40 healthy children and 62 children diagnosed with severe AD (AD score ≥60) were included in the study. Peripheral blood mononuclear cells (PBMCs) from the studied subjects were incubated with the peptide extracts of raw and hydrolysed cow milk with defined ß-casein genotypes (A1A1, A2A2 and A1A2) and MOR and DPPIV genes expression was determined with real-time PCR. Incubation PBMCs with peptide extracts from cow milk caused an increase of the MOR gene expression (p<0.05; p<0.001) in AD children with a simultaneous decrease in the DPPIV gene expression (p<0.001). The obtained results supplement the knowledge on the BCM7 participation in AD etiology and provide an important diagnostic tool.


Assuntos
Dermatite Atópica/tratamento farmacológico , Endorfinas/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Hipersensibilidade a Leite/tratamento farmacológico , Fragmentos de Peptídeos/administração & dosagem , Adolescente , Alérgenos/efeitos dos fármacos , Animais , Bovinos , Criança , Dermatite Atópica/genética , Dermatite Atópica/patologia , Dipeptidil Peptidase 4/biossíntese , Endorfinas/metabolismo , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Hipersensibilidade a Leite/genética , Hipersensibilidade a Leite/patologia , Proteínas do Leite/efeitos adversos , Fragmentos de Peptídeos/metabolismo , Receptores Opioides mu/biossíntese
11.
Peptides ; 55: 79-84, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24582609

RESUMO

The zwitterionic detergent CHAPS was used to solubilize the human mu-opioid receptor (hMOR) from SH-SY5Y neuroblastoma cells and recombinant hMOR-CHO (CHO-T7-hMOR) and hMOR-SH-SY5Y (SH-SY5Y-T7-hMOR) cell membranes. Agonist stimulation and G-protein activation by the mu-selective opioid agonist DAMGO ([D-Ala2, N-MePhe4, Gly-ol]-enkephalin) were recovered after removing of CHAPS after polyethylene glycol (PEG) precipitation. Binding assays show that hMOR solubilized and reconstituted this way was functional and able to interact with both agonist peptides and with G-protein. The effective solubilization and reconstitution of hMOR from mammalian cells, without truncation and extensive modification, represent an essential step toward the purification of a receptor bearing important post-translational modifications.


Assuntos
Receptores Opioides mu/química , Animais , Células CHO , Linhagem Celular Tumoral , Ácidos Cólicos/química , Cricetulus , Detergentes/química , Diprenorfina/química , Ala(2)-MePhe(4)-Gly(5)-Encefalina/química , Guanosina 5'-O-(3-Tiotrifosfato)/química , Humanos , Ligantes , Antagonistas de Entorpecentes/química , Polietilenoglicóis/química , Ligação Proteica , Redobramento de Proteína , Receptores Opioides mu/agonistas , Receptores Opioides mu/biossíntese , Solubilidade
12.
Cancer Lett ; 344(2): 212-22, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24291668

RESUMO

The aim of this study was to investigate the immunological mechanisms by which synthetic methionine enkephalin (MENK) exerts therapeutic effects on tumor growth. Our findings in vivo or in vitro show that MENK treatment either in vivo or in vitro could up-regulate the percentages of CD8+T cells, induce markers of activated T cells, increased cytotoxic activity against mouse S180 tumor cells and increase secretion of IFNγ. In addition, the adoptively transferred CD8+T cells, after either in vitro or in vivo treatment with MENK, result in significantly increased survival of S180 tumor-bearing mice and significant shrinkage in tumor growth. Opioid receptors are detected on normal CD8+T cells and exposure to MENK leads to increased expression of opioid receptors. Interaction between MENK and the opioid receptors on CD8+T cells appears to be essential for the activation of CTL, since the addition of naltrexone (NTX), an opioid receptor antagonist, significantly inhibits all of the effects of MENK. The evidence obtained indicates that the MENK-induced T cell signaling is associated with a significant up-regulation of Ca2+ influx into the cytoplasm and the translocation of NFAT2 into nucleus, and these signaling effects are also inhibited by naltrexone.


Assuntos
Encefalina Metionina/farmacologia , Imunoterapia Adotiva/métodos , Sarcoma 180/imunologia , Sarcoma 180/terapia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Cálcio/metabolismo , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , Encefalina Metionina/imunologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Receptores Opioides delta/biossíntese , Receptores Opioides delta/imunologia , Receptores Opioides mu/biossíntese , Receptores Opioides mu/imunologia , Sarcoma 180/tratamento farmacológico
13.
PLoS One ; 8(3): e55748, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555556

RESUMO

Morphine has been reported to accelerate the progression of chronic kidney disease. However, whether morphine affects slit diaphragm (SD), the major constituent of glomerular filtration barrier, is still unclear. In the present study, we examined the effect of morphine on glomerular filtration barrier in general and podocyte integrity in particular. Mice were administered either normal saline or morphine for 72 h, then urine samples were collected and kidneys were subsequently isolated for immunohistochemical studies and Western blot. For in vitro studies, human podocytes were treated with morphine and then probed for the molecular markers of slit diaphragm. Morphine-receiving mice displayed a significant increase in albuminuria and showed effacement of podocyte foot processes. In both in vivo and in vitro studies, the expression of synaptopodin, a molecular marker for podocyte integrity, and the slit diaphragm constituting molecules (SDCM), such as nephrin, podocin, and CD2-associated protein (CD2AP), were decreased in morphine-treated podocytes. In vitro studies indicated that morphine modulated podocyte expression of SDCM through opiate mu (MOR) and kappa (KOR) receptors. Since morphine also enhanced podocyte oxidative stress, the latter seems to contribute to decreased SDCM expression. In addition, AKT, p38, and JNK pathways were involved in morphine-induced down regulation of SDCM in human podocytes. These findings demonstrate that morphine has the potential to alter the glomerular filtration barrier by compromising the integrity of podocytes.


Assuntos
Albuminúria/metabolismo , Morfina/efeitos adversos , Entorpecentes/efeitos adversos , Podócitos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Albuminúria/induzido quimicamente , Albuminúria/patologia , Animais , Linhagem Celular Transformada , Proteínas do Citoesqueleto/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Membrana/biossíntese , Camundongos , Morfina/farmacologia , Entorpecentes/farmacologia , Podócitos/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Opioides kappa/biossíntese , Receptores Opioides mu/biossíntese , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Anesthesiology ; 117(4): 847-56, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22913923

RESUMO

BACKGROUND: Patients with peritoneal carcinomatosis often report abdominal pain, which is relatively refractory to morphine. It has been considered that a new animal model is required to investigate the mechanism of abdominal pain for the development of optimal treatments for this type of pain. METHODS: To prepare a peritoneal carcinomatosis model, highly peritoneal-seeding gastric cancer cells, 60As6, were implanted into the abdominal cavity. The nociceptive modality for pain-related behavior was assessed in terms of withdrawal behavior in response to mechanical stimuli and hunching behavior. Tissue samples from mouse dorsal root ganglia and spinal cord were subject to immunohistochemistry and real-time reverse transcription polymerase chain reaction. RESULTS: Mice with peritoneal dissemination showed significant hypersensitivity of the abdomen to mechanical stimulation and spontaneous visceral pain-related behavior. There was a significant increase in c-Fos-positive cells in the spinal cord in tumor-bearing mice. Those mice exhibited a remarkable increase in substance P-positive neurons in the dorsal root ganglia (control vs. tumor, 15.4 ± 1.1 vs. 24.2 ± 3.6, P < 0.05, n = 3). A significant decreases in µ-opioid receptor expression mainly in substance P-positive neurons was observed in tumor-bearing mice (69.3 ± 4.9 vs. 38.7 ± 0.9, P < 0.05, n = 3), and a relatively higher dose of morphine was required to significantly reverse the abdominal hypersensitivity. CONCLUSION: Both the up-regulation of substance P and down-regulation of µ-opioid receptor seen in the dorsal root ganglia may be, at least in part, responsible for the abdominal pain-like state associated with peritoneal carcinomatosis.


Assuntos
Dor Abdominal/etiologia , Dor Abdominal/metabolismo , Carcinoma/complicações , Carcinoma/metabolismo , Receptores Opioides mu/biossíntese , Medula Espinal/metabolismo , Substância P/biossíntese , Dor Abdominal/psicologia , Animais , Comportamento Animal , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Carcinoma/psicologia , Linhagem Celular , Linhagem Celular Tumoral , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Imuno-Histoquímica , Inflamação/complicações , Inflamação/psicologia , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Transplante de Neoplasias , Pancreatite/complicações , Pancreatite/psicologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Opioides mu/genética , Substância P/genética
15.
J Neuroimmune Pharmacol ; 7(4): 835-42, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22926418

RESUMO

We have previously reported that functionally active µ-opioid receptors (MOR) are constitutively expressed at relatively low levels by developing T cells in the thymus. However, very little is known about the regulation of MOR expression by immature T cells. In this report, we first attempted to determine the effect of T cell receptor-induced T cell activation on the expression of MOR. We activated T cells with either the combination of anti-CD3 and CD28, or with superantigen, and observed a substantial increase in MOR transcript expression. We also chose to examine the effect of cytokine-mediated T cell activation on the expression of this opioid receptor. We selected certain cytokines that play a role in T cell development and are known to be present at functional levels in the thymus gland. Our results show that interferon γ (IFNγ), IL-1ß, and IL-2, and in particular transforming growth factor-ß (TGFß), all induced significant increases in MOR transcript expression. On the other hand, both TNFα and IL-7 exhibited much weaker effects on MOR expression. These results show that MOR expression by developing T cells is strongly regulated by several cytokines involved in T cell development in the thymus gland.


Assuntos
Ativação Linfocitária/fisiologia , Receptores Opioides mu/biossíntese , Receptores Opioides mu/efeitos dos fármacos , Linfócitos T/fisiologia , Animais , Southern Blotting , Diferenciação Celular/fisiologia , AMP Cíclico/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Interleucina-2/farmacologia , Interleucina-7/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , RNA/biossíntese , RNA/isolamento & purificação , Linfócitos T/efeitos dos fármacos , Timo/citologia , Fator de Crescimento Transformador beta/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
16.
Anesthesiology ; 116(4): 857-67, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22343475

RESUMO

BACKGROUND: Recent epidemiologic studies suggesting that there were differences in cancer recurrence contingent on anesthetic regimens have raised the possibility that µ-opioid agonists can influence cancer progression. Based on our previous studies indicating the µ-opioid receptor (MOR) is up-regulated in several types of non-small cell lung cancer, this study examined the functional significance of MOR overexpression to elucidate a possible mechanism for the epidemiologic findings. METHODS: Stable vector control and MOR1 overexpressing human bronchioloalveolar carcinoma cells were evaluated using immunoblot analysis, proliferation and transendothelial extravasation assays with or without Akt inhibitor, mTOR inhibitor (temsirolimus), or the peripheral MOR antagonist, methylnaltrexone. In human lung cancer xenograft models, primary tumor growth rates and lung metastasis were analyzed using consecutive tumor volume measurements and nestin immunoreactivity in lungs of the nude mouse model. RESULTS: The authors provide evidence that MOR is an important regulator of lung cancer progression. MOR overexpression increased Akt and mTOR activation, proliferation, and extravasation in human bronchioloalveolar carcinoma cells. In vivo, overexpression of MOR in human bronchoalveolar carcinoma cells increased primary tumor growth rates in nude mice by approximately 2.5-fold and lung metastasis by approximately 20-fold compared with vector control cells (n = 4 per condition). CONCLUSIONS: The overexpression data suggest a possible direct effect of MOR on Akt and mTOR activation and lung cancer progression. Such an effect provides a plausible explanation for the epidemiologic findings. The authors' observations further suggest that exploration of MOR in non-small cell lung carcinoma merits further study both as a diagnostic and therapeutic option.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/secundário , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Opioides mu/biossíntese , Serina-Treonina Quinases TOR/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Opioides mu/genética , Receptores Opioides mu/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
17.
Exp Physiol ; 97(5): 651-62, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22337865

RESUMO

Both low-frequency electro-acupuncture (EA) and manual acupuncture improve menstrual frequency and decrease circulating androgens in women with polycystic ovary syndrome (PCOS). We sought to determine whether low-frequency EA is more effective than manual stimulation in regulating disturbed oestrous cyclicity in rats with PCOS induced by 5α-dihydrotestosterone. To identify the central mechanisms of the effects of stimulation, we assessed hypothalamic mRNA expression of molecules that regulate reproductive and neuroendocrine function. From age 70 days, rats received 2 Hz EA or manual stimulation with the needles five times per week for 4-5 weeks; untreated rats served as control animals. Specific hypothalamic nuclei were obtained by laser microdissection, and mRNA expression was measured with TaqMan low-density arrays. Untreated rats were acyclic. During the last 2 weeks of treatment, seven of eight (88%) rats in the EA group had epithelial keratinocytes, demonstrating oestrous cycle change (P = 0.034 versus control rats). In the manual group, five of eight (62%) rats had oestrous cycle changes (n.s. versus control animals). The mRNA expression of the opioid receptors Oprk1 and Oprm1 in the hypothalamic arcuate nucleus was lower in the EA group than in untreated control rats. The mRNA expression of the steroid hormone receptors Esr2, Pgr and Kiss1r was lower in the manual group than in the control animals. In rats with 5α-dihydrotestosterone-induced PCOS, low-frequency EA restored disturbed oestrous cyclicity but did not differ from the manual stimulation group, although electrical stimulation lowered serum testosterone in responders, those with restored oestrus cyclicity, and differed from both control animals and the manual stimulation group. Thus, EA cannot in all aspects be considered superior to manual stimulation. The effects of low-frequency EA may be mediated by central opioid receptors, while manual stimulation may involve regulation of steroid hormone/peptide receptors.


Assuntos
Terapia por Acupuntura , Eletroacupuntura , Ciclo Estral/fisiologia , Síndrome do Ovário Policístico/fisiopatologia , Animais , Núcleo Arqueado do Hipotálamo/fisiopatologia , Di-Hidrotestosterona , Modelos Animais de Doenças , Feminino , Síndrome do Ovário Policístico/induzido quimicamente , Progesterona/sangue , Ratos , Receptores Opioides kappa/biossíntese , Receptores Opioides mu/biossíntese , Testosterona/sangue
18.
Fundam Clin Pharmacol ; 26(3): 363-72, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21306432

RESUMO

Agonists of µ-opioid receptors are currently used in the management of cancer pain. However, several data suggest that the analgesic effect of morphine can diminish during the development of experimental tumors. By using a thermal test, we have studied whether the analgesic effect evoked by morphine is altered in mice bearing two painful bone tumors. The analgesic effect evoked by systemic morphine remained unaltered after the intratibial inoculation of B16-F10 melanoma cells and was potentiated after the inoculation of NCTC 2472 osteosarcoma cells. Although the number of spinal µ-opioid receptors measured by western blot studies was not augmented in osteosarcoma-bearing mice, the analgesia evoked by intrathecal (i.t.) morphine was also enhanced. The analgesic response produced by the spinal administration of the Gi/o protein activator mastoparan was amplified, whereas the analgesic response evoked by the i.t. administration of the N-type calcium channel blocker ω-conotoxin remained unaltered. The efficacy of the GIRK channel blocker tertiapin-Q to antagonize the analgesic effect produced by a maximal dose of morphine was also increased in osteosarcoma-bearing mice. Our results seem to indicate that the analgesic effect of morphine on thermal nociception can be enhanced in response to the development of particular bone tumors in mice, being this potentiation probably related to a greater efficacy of the transduction system driven by Gi/o proteins and GIRK channels.


Assuntos
Analgesia/métodos , Neoplasias Ósseas/tratamento farmacológico , Temperatura Alta , Morfina/administração & dosagem , Medição da Dor/métodos , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Temperatura Alta/efeitos adversos , Masculino , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Dor/tratamento farmacológico , Dor/metabolismo , Dor/patologia , Receptores Opioides mu/biossíntese
19.
J Neuroimmunol ; 236(1-2): 57-64, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21684020

RESUMO

This study examined the mechanism by which exposure to lipopolysaccharide (LPS) alters mu-opioid receptor (MOR) expression in immune and neuronal cells using an in vitro conditioned medium model system. We found that LPS stimulated the intracellular accumulation of reactive oxygen species (ROS) and MOR expression in macrophage-like TPA-HL-60 cells. Conditioned medium from the LPS-stimulated TPA-HL-60 cells increased MOR expression in SH-SY5Y cells, a neuronal cell model, through actions mediated by TNF-α and GM-CSF. These data suggest that the endotoxin, LPS, modulates MOR expression in nervous and immune cells via ROS signaling, and demonstrates the crosstalk that exists within the neuroimmune axis.


Assuntos
Endotoxinas/toxicidade , Regulação da Expressão Gênica , Neurônios/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Opioides mu/biossíntese , Células HL-60 , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Receptores Opioides mu/genética
20.
Mol Pharmacol ; 78(5): 837-45, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20702761

RESUMO

The µ-opioid receptor (MOR) plays an important role in modulating analgesia, feeding behavior, and a range of autonomic functions. In the current study, we investigated the degree to which 13 naturally occurring missense mutations affect the pharmacological properties of the human MOR. After expression of each receptor in human embryonic kidney 293 cells, signaling (Gα(i/o)-mediated) induced by peptide agonists was assessed using luciferase reporter gene assays. Multiple mutants (S66F, S147C, R260H, R265C, R265H, and S268P) show a significant reduction in agonist potency. At the N190K variant, agonist-mediated signaling was essentially absent. Enzyme-linked immunosorbent assay, microscopic analysis, and radioligand binding assays revealed that this mutant shows markedly reduced cell-surface expression, whereas all other receptor variants were expressed at normal levels. Surface expression of the N190K variant could be increased by incubation with the alkaloid agonist buprenorphine or with either naltrexone or naloxone, structurally related MOR antagonists. We were surprised to find that both putative antagonists, despite being inactive at the wild-type MOR, triggered a concentration-dependent increase in N190K receptor-mediated signaling. In contrast, peptidic ligands failed to promote expression or rescue function of the N190K mutant. Subsequent analysis of the N190K variant in an ethnically diverse cohort identified this isoform in a subgroup of African Americans. Taken together, our studies reveal that the N190K mutation leads to severe functional alterations and, in parallel, changes the response to established MOR ligands. The extent to which this mutation results in physiological abnormalities or affects drug sensitivity in selected populations (e.g., those with chronic pain or addiction) remains to be investigated.


Assuntos
Peptídeos/farmacologia , Receptores Opioides mu/agonistas , Negro ou Afro-Americano , Substituição de Aminoácidos , Linhagem Celular , HDL-Colesterol/sangue , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Feminino , Genes Reporter , Genótipo , Humanos , Luciferases/biossíntese , Luciferases/genética , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Naloxona/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Peptídeos Opioides/farmacologia , Polimorfismo de Nucleotídeo Único , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Transporte Proteico , Ensaio Radioligante , Receptores Opioides mu/biossíntese , Receptores Opioides mu/genética , Transdução de Sinais , População Branca
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA