Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Int Immunopharmacol ; 125(Pt B): 111226, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37976597

RESUMEN

OBJECTIVE: This study aimed to investigate the underlying regulatory effects of methionine enkephalin (MENK) on osteosarcoma. METHODS: The Cell Counting Kit-8 assay, clone formation, wound healing, transwell assay, and flow cytometry were performed to measure the effects of MENK on the proliferation, migration, invasion, and apoptosis of MG-63 and Saos-2 cells. Opiate growth factor receptor expression (OGFr) in cells was stably knocked down using siRNA. A tumor model was established by inoculating MG-63 cells into mice. Flow cytometry was performed to identify alterations in mice bone marrow, spleen, and tumor tissue immune cells. The phenotype of tumor-associated macrophages was determined using immunohistochemistry. After OGFr knockdown or/and treatment with MENK, Bax, Bcl-2, caspase 3, caspase 9, and PARP expression levels were characterized using qRT-PCR, western blot, and WES, respectively. RESULTS: MENK could significantly inhibit the proliferation, invasion, and migration of MG-63 and Saos-2, arrest the cell cycle in the G0/G1 phase, upregulate Bax, caspase 3, caspase 9, and PARP expression, and downregulate Bcl-2 expression. Tumor size and weight were lower in the MENK group than those in the control group. MENK-treated mice exhibited a reduced ratio of CD11b + Gr-1 + myeloid-derived suppressor cells. MENK increased the ratio of M1-type macrophages and decreased the proportion of M2-type macrophages in tumor tissue. Furthermore, the level of TNF-α significantly increased while that of IL-10 decreased in MENK-treated mice. The effect of MENK could be partly reversed by OGFr knockdown. CONCLUSION: MENK reduces the abundance of myeloid-derived suppressor cells, induces M1 polarization of macrophages, and exhibits an inhibitory effect on osteosarcoma.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Animales , Ratones , Caspasa 3 , Caspasa 9 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteína X Asociada a bcl-2 , Osteosarcoma/tratamiento farmacológico , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico
2.
Int Immunopharmacol ; 118: 110064, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36989897

RESUMEN

Metastasis is one of the most difficult challenges for clinical lung cancer treatment. Epithelial-mesenchymal transition (EMT) is the crucial step of tumor metastasis. Immune cells in the tumor microenvironment (TME), such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), promote cancer cell EMT. In this study, we explored the effect of methionine enkephalin (MENK) on the EMT process in vitro and in vivo, and its influence on TAMs, MDSCs, and associated cytokines in vivo. The results showed that MENK suppressed growth, migration, and invasion of lung cancer cells and inhibited the EMT process by interacting with opioid growth factor receptor. MENK reduced the number of M2 macrophages and MDSC infiltration, and downregulated the expression of interleukin-10 and transforming growth factor-ß1 in both primary and metastatic tumors of nude mice. The present findings suggest that MENK is a potential target for suppressing metastasis in lung cancer treatment.


Asunto(s)
Neoplasias Pulmonares , Células Supresoras de Origen Mieloide , Animales , Ratones , Transición Epitelial-Mesenquimal , Macrófagos Asociados a Tumores/metabolismo , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Encefalina Metionina/metabolismo , Microambiente Tumoral , Ratones Desnudos , Línea Celular Tumoral , Movimiento Celular
3.
J Pain ; 24(5): 840-859, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36586660

RESUMEN

Venom-derived NaV1.7 channel blockers have promising prospects in pain management. The 34-residue tarantula peptide GpTx-1 is a potent NaV1.7 channel blocker. Its powerful analog [Ala5, Phe6, Leu26, Arg28]GpTx-1 (GpTx-1-71) displayed excellent NaV1.7 selectivity and analgesic properties in mice. The current study aimed to elucidate the anti-hyperalgesic activities of GpTx-1-71 in inflammatory pain and reveal the underlying mechanisms. Our results demonstrated that intrathecal and intraplantar injections of GpTx-1-71 dose-dependently attenuated CFA-induced inflammatory hypersensitivity in rats. Moreover, GpTx-1-71-induced anti-hyperalgesia was significantly reduced by opioid receptor antagonists and the enkephalin antibody and diminished in proenkephalin (Penk) gene knockout animals. Consistently, GpTx-1-71 treatment increased the enkephalin level in the spinal dorsal horn and promoted the Penk transcription and enkephalin release in primary dorsal root ganglion (DRG) neurons, wherein sodium played a crucial role in these processes. Mass spectrometry analysis revealed that GpTx-1-71 mainly promoted the secretion of Met-enkephalin but not Leu-enkephalin from DRG neurons. In addition, the combination of subtherapeutic Met-enkephalin and GpTx-1-71 produced synergistic anti-hyperalgesia in CFA-induced inflammatory hypersensitivity. These findings suggest that the endogenous enkephalin pathway is essential for GpTx-1-71-induced spinal and peripheral analgesia in inflammatory pain. PERSPECTIVE: This article presents a possible pharmacological mechanism underlying NaV1.7 blocker-induced analgesia in inflammatory pain, which helps us to better understand and develop venom-based painkillers for incurable pain.


Asunto(s)
Hiperalgesia , Dolor , Ratas , Ratones , Animales , Dolor/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Analgésicos/farmacología , Analgésicos/uso terapéutico , Encefalinas/metabolismo , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Ganglios Espinales/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo
4.
Int Immunopharmacol ; 111: 109125, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35988519

RESUMEN

There is evidence that methionine enkephalin (MENK), an opioid peptide, promotes anti-tumor immune responses. In this study, the effect of MENK on colorectal cancer (CRC) and its mechanisms of action were examined in vivo. The intraperitoneal administration of 20 mg/kg MENK effectively inhibited MC38 subcutaneous colorectal tumor growth in mice. MENK inhibited tumor progression by increasing the immunogenicity and recognition of MC38 cells. MENK down-regulated the oncogene Kras and anti-apoptotic Bclxl and Bcl2, suppressed Il1b, Il6, iNOS, and Arg1 (encoding inflammatory cytokines), and increased Il17a and Il10 levels. MENK promoted a tumor suppressive state by decreasing the immune checkpoints Pd-1, Pd-l1, Lag3, Flgl1, and 2b4 in CRC. MENK also altered the immune status of the tumor immune microenvironment (TIME). It increased the infiltration of M1-type macrophages, CD8+T cells, and CD4+T cells and decreased the proportions of G-MDSCs, M-MDSCs, and M2-type macrophages. MENK accelerated CD4+TEM and CD8+TEM cell activation in the TIME and up-regulated IFN-γ, TNF-α, and IL-17A in CD4+T cells and Granzyme B in CD8+T cells. In addition, analyses of PD-1 and PD-L1 expression indicated that MENK promoted the anti-tumor immune response mediated by effector T cells. Finally, OGFr was up-regulated at the protein and mRNA levels by MENK, and the inhibitory effects of MENK on tumor growth were blocked by NTX, a specific blocker of OGFr. These finding indicate that MENK remodels the TIME in CRC to inhibit tumor progression by binding to OGFr. MENK is a potential therapeutic agent for CRC, especially for improving the efficacy of immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Encefalina Metionina , Animales , Antígeno B7-H1 , Neoplasias Colorrectales/tratamiento farmacológico , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Factores Inmunológicos , Ratones , Receptor de Muerte Celular Programada 1 , Microambiente Tumoral
5.
Int Immunopharmacol ; 110: 108933, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35738090

RESUMEN

Immunotherapy for cervical carcinoma is becoming increasingly important recently. In these studies methionine enkephalin (menk) is shown to inhibit cervical tumor cell proliferation in vitro in association with an increase in the expression of apoptosis markers and mediators, including an increase in fas, caspase 8, and caspase 3 expression and intrinsic expression of the signaling pathway mediator bax. In vivo, tumor growth was restrained in mice xenotransplant model with typical pathological features of apoptosis. Furthermore, myeloid derived suppressor cells (MDSCs) had a significant decrease in circulation and in tumor site. In brief, these findings showed menk could inhibit tumor growth in vitro and in vivo, providing direction of further research and clinical application prospect.


Asunto(s)
Carcinoma , Células Supresoras de Origen Mieloide , Neoplasias del Cuello Uterino , Animales , Apoptosis , Línea Celular Tumoral , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Femenino , Humanos , Factores Inmunológicos/metabolismo , Ratones , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo
6.
Eur J Pharmacol ; 882: 173253, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32535097

RESUMEN

Methionine enkephalin (MENK) is an opioid peptide composed of five amino acids with multiple biological activities. Since its discovery, MENK has become prominent in neuroregulation and immunoregulation. Tumors have increasingly been a spotlight because of their terrible trends and refractory characteristic. The therapeutic potential of MENK was investigated on a large scale, and there are numerous evidences that MENK exerts anti-tumor effects via two mechanisms. The first mechanism explains the enhanced anti-tumor immune effects of MENK. The second mechanism shows that MENK directly inhibits tumor cell proliferation. However, numerous reports have clarified the pro-tumor role of MENK by inhibiting T and B cell proliferation, promoting tumor cell growth by binding to opioid receptors, leading to desensitization of lymphocytes, and inducing tolerance. It is particularly intriguing that dual reactions are triggered when MENK combines with its opioid receptors; thus, anti-tumor response of the whole body is influenced. This review will expound the dual roles of MENK in tumor responses based on immune cells, cytokines, and tumor cells to provide better suggestions for its application in tumor treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Encefalina Metionina/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Encefalina Metionina/farmacología , Humanos , Neoplasias/metabolismo
7.
J Leukoc Biol ; 108(1): 215-228, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31994797

RESUMEN

Methionine enkephalin (MENK), an endogenous opioid peptide, has a role in nervous system, immune system, and anticancer therapy. Inflammation, metabolism and cancer are closely intertwined with each other. This study is to identify the correlation of the antitumor effects of MENK with systemic inflammation, liver metabolism, and immune cells as myeloid-derived suppressor cells (MDSCs). We established a subcutaneous CT26 colon carcinoma model and a cyclophosphamide-induced immunosuppressive model subjected to MENK. AML12 and MDSCs were used as in vitro models. The results showed that MENK treatment degraded tumor growth and inhibited proinflammatory cytokines both in tumor tissues and serum. The MENK-treated tumor mice showed normalized liver function with glycolipid metabolic homeostasis. No inhibitory effect on CT26 tumor cell in vitro, but only reduced lipid synthesis in AML12 were presented by MENK. Meanwhile, MENK invigorated immune response in both two animal models by markedly suppressing MDSCs and enhancing T cells response. In vitro MENK-treated MDSCs showed reduced glycolysis and less ROS production, which was mediated by PI3K/AKT/mTOR pathway. Opioid receptor antagonist naltrexone reversed most of the regulation. These results illustrate that MENK preventing development of colon carcinoma might be correlated with the suppression of inflammation, improving metabolism in liver as well as in MDSCs partly through opioid receptor, which brings new elements supporting the adjuvant therapy for tumor by MENK.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/metabolismo , Encefalina Metionina/farmacología , Animales , Antiinflamatorios/uso terapéutico , Antineoplásicos/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Ciclofosfamida/farmacología , Citocinas/metabolismo , Encefalina Metionina/uso terapéutico , Glucolípidos/metabolismo , Glucólisis/efectos de los fármacos , Terapia de Inmunosupresión , Inflamación/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
8.
Int Immunopharmacol ; 78: 106032, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31835089

RESUMEN

MENK, as an immune adjuvant, has potential immune-regulatory activity on innate and adaptive immune cells. The aim of this work was to investigate the antiviral effect of MENK on influenza virus-infected murine macrophage cells (RAW264.7) and its underlying mechanisms. The results showed that MENK markedly inhibited influenza A virus (H1N1) replication in pre- and post-MENK treatment, especially in pre-MENK treatment. The mechanisms exploration revealed that MENK (10 mg/mL) significantly inhibited the nucleoprotein (NP) of influenza virus and up-regulated levels of IL-6, TNF-α and IFN-ß compared with those in H1N1 control group. Further experiments confirmed that antiviral effects of MENK was associated with promotion of opioid receptor (MOR) as well as activation of NF-κB p65 inducing cellular antiviral status. The data suggest that MENK should be potential candidate for prophylactic or therapeutic treatment against H1N1 influenza virus.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Encefalina Metionina/farmacología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Adyuvantes Inmunológicos/uso terapéutico , Animales , Encefalina Metionina/uso terapéutico , Humanos , Gripe Humana/inmunología , Gripe Humana/virología , Interferón beta/metabolismo , Interleucina-6/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Receptores Opioides mu/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Replicación Viral/inmunología
9.
Int Immunopharmacol ; 75: 105785, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31404891

RESUMEN

Endogenous opioids are neuro-peptides with multifunctional properties. Historically, opioids are used to mediate pain; however, excess opiate consumption can lead to addiction. One endogenous opioid, methionine enkephalin (MENK), was reported to modulate cell growth, MENK was identified as an opioid growth factor (OGF) that interacts with the OGF receptor (OGFr) and regulates cell proliferation. Further, opioid antagonists, including naltrexone and naloxone are widely used to reverse drug and alcohol overdoses. Naltrexone (NTX) acts on all opioid receptors, blocking the interaction between OGF and OGFr, and thus influencing cell growth. During the last decades, insights have been made concerning the interaction between OGF and OGFr, confirming that both opioids and opioid antagonists have an important role in balancing host homeostasis, host immunity and mediating cancer therapy. This review provides insight into the interactions between OGF and OGFr in the treatment of cancers.


Asunto(s)
Encefalina Metionina/uso terapéutico , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Encefalina Metionina/farmacología , Humanos , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Neoplasias/inmunología , Neoplasias/metabolismo
10.
Int Immunopharmacol ; 65: 312-322, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30343258

RESUMEN

This study was to explore the effect and mechanisms of anti- human gastric cancer by MENK in vitro and in vivo. The results showed in MENK-treated xenograft tissue, the percentage of M2-type macrophages decreased while M1-type macrophages increased. MENK increased the expression of M1-related cytokine TNF-α and attenuated the expression of M2-related cytokine IL-10 expression. MENK upregulated the expression of opioid receptor (OGFr), while it inhibited HGC27 and SGC7901 cells through blocking PI3K/AKT/mTOR signal pathway in vitro and in vivo. These effects of MENK could be cancelled when OGFr was knockdown. This indicates that binding to OGFr by MENK appears to be essential for the anti- GC cells. Therefore, it is concluded that MENK might skew macrophage toward M2 phenotype from M1 phenotype within tumor and induce cells apoptosis though blocking OGFr/PI3K/AKT/mTOR signaling pathway.


Asunto(s)
Antineoplásicos/uso terapéutico , Encefalina Metionina/uso terapéutico , Macrófagos/fisiología , Neoplasias Gástricas/tratamiento farmacológico , Animales , Apoptosis , Diferenciación Celular , Citocinas/metabolismo , Femenino , Xenoinjertos , Humanos , Macrófagos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Opioides/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Células Th2/inmunología , Escape del Tumor
11.
Toxicol Appl Pharmacol ; 359: 12-23, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30222981

RESUMEN

Obesity and its related metabolic disorders including insulin resistance and fatty liver become major public health concerns in both developed and developing countries. Brown adipose tissue (BAT), a critical organ of energy expenditure due to thermogenesis, has been considered as an attractive target for prevention or treatment of obesity and obesity related diseases. Previous studies indicate Met-enkephalin (MetEnk) has the potential on adipocyte browning, however, whether MetEnk displays the impact on adipocyte browning in vivo to improve obesity associated morbidities is still unclear. In the present study, we showed that MetEnk effectively prevented high fat diet (HFD) induced C57BL/6J mice weight gain, clearly enhanced glucose tolerance and insulin sensitivity, and dramatically reduced hepatic steatosis in HFD fed mice. Mechanically, MetEnk restored protein kinase A (PKA) signaling pathway in HFD challenged mice and promoted subcutaneous white adipose tissue (WAT) browning. Our study suggests that MetEnk can be considered as a potential therapeutic peptide for diet-induced obesity and metabolic disorders.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Encefalina Metionina/uso terapéutico , Síndrome Metabólico/tratamiento farmacológico , Adipocitos Marrones/efectos de los fármacos , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Pardo/patología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocinas/metabolismo , Intolerancia a la Glucosa/tratamiento farmacológico , Resistencia a la Insulina , Masculino , Síndrome Metabólico/patología , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Receptores Opioides delta/efectos de los fármacos
12.
Int Immunopharmacol ; 55: 38-48, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29220721

RESUMEN

The morbidity and mortality associated with influenza A virus infections, have stimulated the search for novel prophylactic and therapeutic drugs. The purpose of this study was to investigate the prophylactic and therapeutic effect of synthetic methionine enkephalin (MENK) on mice infected by A/PR/8/34 influenza virus (H1N1) in vivo. The results showed that MENK could exert both prophylactic and therapeutic influences on infected mice, significantly improve the survival rate, relieve acute lung injury and decrease cytokine (IFN-α, IFN-ß, TNF-α, IL-6, and IL-1ß) levels. MENK also inhibited virus replication on day 4 post infection (p.i.) through upregulating opioid receptors (MOR, DOR) and suppressing TLR7-MyD88-TRAF6-NF-κB p65 signaling pathways. These results suggest that MENK, given via intranasal administration, could provide a novel drug with a new mode of action as a nonspecific anti-influenza agent or vaccine adjuvant.


Asunto(s)
Antivirales/uso terapéutico , Encefalina Metionina/uso terapéutico , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/tratamiento farmacológico , Pulmón/patología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Citocinas/metabolismo , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Receptores Opioides/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 7/metabolismo , Replicación Viral
13.
Int Immunopharmacol ; 44: 61-71, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28088065

RESUMEN

MENK, an endogenous opioid peptide has been reported to have many immunological and antitumor activities. So far the detailed mechanisms of antitumor through regulating DCs by MENK have not been elucidated yet. The aim of this work was to investigate the antitumor mechanisms of MENK via regulating DC. The monitoring methods, such as ELISA, MTS assay, CFSE, Real-time PCR and Western blot were included in our research. We found bone marrow derived dendritic cells (BMDCs) in 36 female C57BL/6 mice treated with MENK enhanced expression of key surface molecules, increased production of critical cytokines reduced endocytosis of FITC-dextran, upregulated TLR4 through MyD88/NF-κB signaling pathway and mounted higher antitumor activity. These observations were further supported by an enhancement of nuclear translocation of the p65NF-κB subunit involved in this process. Surprisingly, mu-opioid receptors were the main participants of this kind of activation, not delta-opioid receptors nor kappa-opioid receptors, and these interactions could be partly blocked by Naltrexone (a kind of opioid antagonist). In vivo study the activated CD4+, CD8+T cells and decreased ability to induce differentiation of Foxp3+ regulatory T cells were detected post treatment of MENK. Thus, it is concluded that MENK could exert antitumor effect through precisely regulating opioid receptor mediated functions of DCs. In addition, MENK treated DCs may serve as a new immunotherapy approach against tumor.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Dendríticas/efectos de los fármacos , Encefalina Metionina/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/inmunología , Femenino , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Neoplasias/inmunología , Transducción de Señal/efectos de los fármacos
14.
World J Gastroenterol ; 20(9): 2218-23, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24605021

RESUMEN

Opioid growth factor (OGF), chemically termed [Met5]-enkephalin, and its receptor, OGF receptor (OGFr), form a biological axis that tonically regulates cell proliferation by delaying the G1/S interface of the cell cycle under homeostatic conditions or in neoplasia. Modulation of the OGF-OGFr pathway mediates the course of pancreatic cancer, with exogenous OGF or upregulation of OGFr repressing growth of human pancreatic cancer cells in culture and in nude mice. OGF therapy alone or in combination with standard chemotherapies such as gemcitabine and 5-fluorouracil results in enhanced inhibition of DNA synthesis and tumor growth. Molecular manipulation of OGFr confirms that the receptor is specific for OGF's inhibitory action. Preclinical studies have warranted Phase I and Phase II clinical trials using OGF infusions as a treatment for patients with advanced, unresectable pancreatic cancers. OGF, an endogenous neuropeptide, is a safe, non-toxic, and effective biotherapy that utilizes the OGF-OGFr axis to mediate pancreatic tumor progression.


Asunto(s)
Antineoplásicos/uso terapéutico , Encefalina Metionina/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalina Metionina/metabolismo , Humanos , Ratones , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Receptores Opioides/agonistas , Receptores Opioides/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
15.
Invest New Drugs ; 31(4): 1066-70, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23275062

RESUMEN

Hepatoblastoma is the most common liver malignancy in children, typically diagnosed before age 2. The survival rate for hepatoblastoma has increased dramatically in the last 30 years, but the typical chemotherapeutic agents used for treatment are associated with significant toxicity. In this report, the authors present two cases of hepatoblastoma treated with surgical resection and a novel biotherapeutic regimen that included opioid growth factor (OGF). Case #1 is an infant diagnosed with a large mass on prenatal ultrasound. After subsequent diagnosis of hepatoblastoma, she was treated with one course of neoadjuvant chemotherapy at approximately 1 week of age. Following significant complications from the chemotherapy (neutropenic fever, pneumonia and sepsis), the patient's parents declined further chemotherapy, and the infant was treated with surgical resection and opioid growth factor (OGF)/low dose naltrexone (LDN). She is currently at close to 10 years disease-free survival. Case #2 is a child diagnosed with a liver mass on ultrasound at 20 months of age, later biopsy-proven to represent hepatoblastoma. Due to existing co-morbidities including autosomal recessive polycystic kidney disease and hypertension, and indications from the biopsy that the tumor might be insensitive to chemotherapy, the parents elected not to proceed with neoadjuvant chemotherapy. The patient was treated with surgical resection and OGF/LDN, and is currently at more than 5 years disease-free survival. This case series highlights the need for less toxic treatment options than conventional chemotherapy. Modulation of the OGF-OGF receptor axis represents a promising safe and therapeutic avenue for effective treatment of hepatoblastoma.


Asunto(s)
Encefalina Metionina/uso terapéutico , Hepatoblastoma/tratamiento farmacológico , Péptidos Opioides/uso terapéutico , Quimioterapia Adyuvante , Niño , Preescolar , Femenino , Hepatoblastoma/diagnóstico por imagen , Humanos , Lactante , Recién Nacido , Embarazo , Ultrasonografía
16.
J Neurosci ; 32(32): 10797-808, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22875915

RESUMEN

Peroxynitrite (PN, ONOO(-)) is a potent oxidant and nitrating agent that contributes to pain through peripheral and spinal mechanisms, but its supraspinal role is unknown. We present evidence here that PN in the rostral ventromedial medulla (RVM) is essential for descending nociceptive modulation in rats during inflammatory and neuropathic pain through PN-mediated suppression of opioid signaling. Carrageenan-induced thermal hyperalgesia was associated with increased 3-nitrotyrosine (NT), a PN biomarker, in the RVM. Furthermore, intra-RVM microinjections of the PN decomposition catalyst Fe(III)-5,10,15,20-tetrakis(N-methyl-pyridinium-4-yl)porphyrin (FeTMPyP(5+)) dose-dependently reversed this thermal hyperalgesia. These effects of FeTMPyP(5+) were abrogated by intra-RVM naloxone, implicating potential interplay between PN and opioids. In support, we identified NT colocalization with the endogenous opioid enkephalin (ENK) in the RVM during thermal hyperalgesia, suggesting potential in situ interactions. To address the functional significance of such interactions, we exposed methionine-enkephalin (MENK) to PN and identified the major metabolite, 3-nitrotyrosine-methionine-sulfoxide (NSO)-MENK, using liquid chromatography-mass spectrometry. Next, we isolated, purified, and tested NSO-MENK for opioid receptor binding affinity and analgesic effects. Compared to MENK, this NSO-MENK metabolite lacked appreciable binding affinity for δ, µ, and κ opioid receptors. Intrathecal injection of NSO-MENK in rats did not evoke antinociception, suggesting that PN-mediated chemical modifications of ENK suppress opioid signaling. When extended to chronic pain, intra-RVM FeTMPyP(5+) produced naloxone-sensitive reversal of mechanical allodynia in rats following chronic constriction injury of the sciatic nerve. Collectively, our data reveal the central role of PN in RVM descending facilitation during inflammatory and neuropathic pain potentially through anti-opioid activity.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Bulbo Raquídeo/metabolismo , Péptidos Opioides/metabolismo , Dolor/tratamiento farmacológico , Ácido Peroxinitroso/administración & dosificación , Transducción de Señal/efectos de los fármacos , Análisis de Varianza , Animales , Antígeno CD11b/metabolismo , Carragenina/efectos adversos , Línea Celular Transformada , Cromatografía Liquida , Enfermedad Crónica , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Técnicas Electroquímicas , Encefalina Metionina/uso terapéutico , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/patología , Inyecciones Espinales , Masculino , Bulbo Raquídeo/efectos de los fármacos , Metaloporfirinas/uso terapéutico , Microinyecciones , Neuroglía/metabolismo , Neuronas/metabolismo , Dimensión del Dolor , Fosfopiruvato Hidratasa/metabolismo , Unión Proteica/efectos de los fármacos , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Tirosina/análogos & derivados , Tirosina/metabolismo
17.
Hum Vaccin Immunother ; 8(9): 1236-42, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22906944

RESUMEN

The aim of this investigation is to look at whether MENK could improve antitumor effect of CD8+T cell elicited by BMDCs. We investigated the effects of MENK on the differentiation, maturation, and functions of murine BMDC loaded with Rac-1 antigens (RG) and CTL of tumor specific immune response elicited by the BMDC in vitro and in vivo. The production of cytokine IL-12 and TNF-α secreted by BMDCs in the presence of MENK was assayed with ELISA and key surface markers of CD40, CD86, CD83 and MHC-II on the BMDCs were analyzed with use of flow cytometry (FCM). In addition, the activities to induce CD8+ T cell proliferation, along with displayed cytotoxicity of the CD8+T cells(CTL) by the BMDCs after treatment with MENK were determined with use of FCM as well as MTS. Our results indicated that MENK induced phenotypic and functional maturation of BMDC loaded with RG antigen, as evidenced by higher level of expression of key surface markers and more production of cytokines. Subsequently, the BMDC activated by MENK intensified immune responses mounted by CTL, resulting in stronger antitumor activity. Our results suggest that MENK could be working as an effective immune adjuvant in vaccine preparation for cancer fight and other immune related diseases. We concluded that MENK could be a positive immune modulator in the improved functions of BMDCs loaded with antigen as well as in CD8+T cell mediated anti-tumor responses.


Asunto(s)
Antígenos/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Encefalina Metionina/uso terapéutico , Animales , Células de la Médula Ósea/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Ratones
18.
Biochem Pharmacol ; 84(6): 746-55, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22687282

RESUMEN

The opioid growth factor (OGF), chemically termed [Met(5)]-enkephalin, is an endogenous opioid peptide that interacts with the OGF receptor (OGFr) to delay the G(1)/S interface of the cell cycle by modulating cyclin-dependent inhibitory kinase (CKI) pathways. The OGF-OGFr axis is a tonically active, inhibitory pathway that is an important regulator during homeostasis and re-epithelialization, and plays a role in the onset and progression of autoimmune diseases and cancer. Modulation of the OGF-OGFr axis can be accomplished by a variety of pharmacological and molecular approaches including use of intermittent or continuous exposure to the opioid antagonist naltrexone, genetic manipulation of OGFr expression, and antibody neutralization of OGF. Clinically, OGF is a biological therapy that has potential application for treatment of cancer. Currently, naltrexone at low dosages is being evaluated for treatment of autoimmune diseases such as Crohn's and multiple sclerosis. High dosages of naltrexone are effective in reversing dry eye and accelerating the repair of corneal abrasions in normal and diabetic rats; these studies are under investigation in the clinical setting. Naltrexone also enhances full-thickness wound closure in animal models of Type 1 or Type 2 diabetes, and translation of this knowledge to the clinic is planned. In summary, understanding the OGF-OGFr axis as a homeostatic regulator of proliferation has substantial implications for maintaining human health and treatment of disease.


Asunto(s)
Encefalina Metionina/fisiología , Receptores Opioides/fisiología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Ciclo Celular/fisiología , Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Complicaciones de la Diabetes/tratamiento farmacológico , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Homeostasis , Humanos , Linfocitos/citología , Linfocitos/fisiología , Naltrexona/farmacología , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Cicatrización de Heridas/efectos de los fármacos
19.
Cancer Immunol Immunother ; 61(10): 1755-68, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22419372

RESUMEN

The aim of this study is to investigate macrophages polarization induced by methionine enkephalin (MENK) that promotes tumoricidal responses in vivo and in vitro. Both phenotypic and functional activities of macrophages were assessed by the quantitative analysis of key surface molecules on macrophages with flow cytometry, immunofluorescent staining, and the production of cytokines with enzyme-linked immunosorbent assay and reverse transcriptase-polymerase chain reaction. Our results showed that MENK could down-regulate the expression of CD206 and the production of arginase-1 (the markers of alternatively activated (M2) macrophage) in tumor-associated macrophages in vivo, meanwhile it could significantly up-regulate the expression of CD64, MHC-II, and the production of induced nitric oxide synthase (the markers of classically activated (M1) macrophages). Furthermore, the studies on bone marrow-derived macrophages treated with MENK (10(-12) M) in vitro had demonstrated that MENK could markedly increase tumoricidal activity. MENK could also enhance the release of reactive oxidant species and the production of interleukin-12p40, tumor necrosis factor-α, while decrease the production of interleukin-10. In conclusion, MENK could effectively induce M2 macrophages polarizing to M1 macrophages, sequentially to modulate the Th1 responses of the host immune system. Our results suggest that MENK might have great potential as a new therapeutic agent for cancer.


Asunto(s)
Polaridad Celular/efectos de los fármacos , Encefalina Metionina/uso terapéutico , Macrófagos/inmunología , Neoplasias/tratamiento farmacológico , Animales , Arginasa/biosíntesis , Línea Celular Tumoral , Citocinas/biosíntesis , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Antígenos de Histocompatibilidad Clase II/biosíntesis , Lectinas Tipo C/biosíntesis , Macrófagos/efectos de los fármacos , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/biosíntesis , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Receptores de Superficie Celular/biosíntesis , Receptores de IgG/biosíntesis , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA