Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Toxicol Appl Pharmacol ; 359: 12-23, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30222981

ABSTRACT

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.


Subject(s)
Adipose Tissue, Brown/drug effects , Diet, High-Fat/adverse effects , Enkephalin, Methionine/therapeutic use , Metabolic Syndrome/drug therapy , Adipocytes, Brown/drug effects , Adipose Tissue, Brown/growth & development , Adipose Tissue, Brown/pathology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/metabolism , Glucose Intolerance/drug therapy , Insulin Resistance , Male , Metabolic Syndrome/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Opioid, delta/drug effects
2.
J Neurosci ; 32(32): 10797-808, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22875915

ABSTRACT

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.


Subject(s)
Hyperalgesia/drug therapy , Medulla Oblongata/metabolism , Opioid Peptides/metabolism , Pain/drug therapy , Peroxynitrous Acid/administration & dosage , Signal Transduction/drug effects , Analysis of Variance , Animals , CD11b Antigen/metabolism , Carrageenan/adverse effects , Cell Line, Transformed , Chromatography, Liquid , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Electrochemical Techniques , Enkephalin, Methionine/therapeutic use , Glial Fibrillary Acidic Protein/metabolism , Humans , Hyperalgesia/chemically induced , Hyperalgesia/pathology , Injections, Spinal , Male , Medulla Oblongata/drug effects , Metalloporphyrins/therapeutic use , Microinjections , Neuroglia/metabolism , Neurons/metabolism , Pain Measurement , Phosphopyruvate Hydratase/metabolism , Protein Binding/drug effects , Radioligand Assay , Rats , Rats, Sprague-Dawley , Tyrosine/analogs & derivatives , Tyrosine/metabolism
3.
Invest New Drugs ; 31(4): 1066-70, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23275062

ABSTRACT

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.


Subject(s)
Enkephalin, Methionine/therapeutic use , Hepatoblastoma/drug therapy , Opioid Peptides/therapeutic use , Chemotherapy, Adjuvant , Child , Child, Preschool , Female , Hepatoblastoma/diagnostic imaging , Humans , Infant , Infant, Newborn , Pregnancy , Ultrasonography
4.
Int Immunopharmacol ; 125(Pt B): 111226, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37976597

ABSTRACT

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.


Subject(s)
Bone Neoplasms , Osteosarcoma , Animals , Mice , Caspase 3 , Caspase 9 , Poly(ADP-ribose) Polymerase Inhibitors , bcl-2-Associated X Protein , Osteosarcoma/drug therapy , Enkephalin, Methionine/pharmacology , Enkephalin, Methionine/therapeutic use , Bone Neoplasms/drug therapy
5.
Int Immunopharmacol ; 118: 110064, 2023 May.
Article in English | MEDLINE | ID: mdl-36989897

ABSTRACT

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.


Subject(s)
Lung Neoplasms , Myeloid-Derived Suppressor Cells , Animals , Mice , Epithelial-Mesenchymal Transition , Tumor-Associated Macrophages/metabolism , Enkephalin, Methionine/pharmacology , Enkephalin, Methionine/therapeutic use , Enkephalin, Methionine/metabolism , Tumor Microenvironment , Mice, Nude , Cell Line, Tumor , Cell Movement
6.
J Pain ; 24(5): 840-859, 2023 05.
Article in English | MEDLINE | ID: mdl-36586660

ABSTRACT

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.


Subject(s)
Hyperalgesia , Pain , Rats , Mice , Animals , Pain/drug therapy , Hyperalgesia/drug therapy , Analgesics/pharmacology , Analgesics/therapeutic use , Enkephalins/metabolism , Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Enkephalin, Methionine/therapeutic use , Ganglia, Spinal/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism
7.
Cancer Immunol Immunother ; 61(10): 1755-68, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22419372

ABSTRACT

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.


Subject(s)
Cell Polarity/drug effects , Enkephalin, Methionine/therapeutic use , Macrophages/immunology , Neoplasms/drug therapy , Animals , Arginase/biosynthesis , Cell Line, Tumor , Cytokines/biosynthesis , Cytokines/metabolism , Down-Regulation/drug effects , Histocompatibility Antigens Class II/biosynthesis , Lectins, C-Type/biosynthesis , Macrophages/drug effects , Male , Mannose Receptor , Mannose-Binding Lectins/biosynthesis , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Nitric Oxide Synthase Type II/biosynthesis , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, IgG/biosynthesis , Up-Regulation
8.
Int Immunopharmacol ; 111: 109125, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35988519

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms , Enkephalin, Methionine , Animals , B7-H1 Antigen , Colorectal Neoplasms/drug therapy , Enkephalin, Methionine/pharmacology , Enkephalin, Methionine/therapeutic use , Immunologic Factors , Mice , Programmed Cell Death 1 Receptor , Tumor Microenvironment
9.
Int Immunopharmacol ; 110: 108933, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35738090

ABSTRACT

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.


Subject(s)
Carcinoma , Myeloid-Derived Suppressor Cells , Uterine Cervical Neoplasms , Animals , Apoptosis , Cell Line, Tumor , Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Enkephalin, Methionine/therapeutic use , Female , Humans , Immunologic Factors/metabolism , Mice , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism
10.
Gynecol Oncol ; 122(2): 382-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21531450

ABSTRACT

OBJECTIVE: The opioid growth factor (OGF) and its receptor, OGFr, serve as a tonically active inhibitory axis regulating cell proliferation in normal cells and a variety of cancers, including human ovarian cancer. Blockade of OGF and OGFr with the nonselective opioid receptor antagonist naltrexone (NTX) upregulates expression of OGF and OGFr. Administration of a low dosage of NTX (LDN) blocks endogenous opioids from opioid receptors for a short period of time (4-6 h) each day, providing a window of 18-20 h for the upregulated opioids and receptors to interact. The present study investigated the repercussions of upregulating the OGF-OGFr axis by treatment with OGF or LDN on human ovarian tumorigenesis in vivo. METHODS: Female nude mice were transplanted intraperitoneally with SKOV-3 human ovarian cancer cells and treated on a daily basis with OGF (10 mg/kg), LDN (0.1 mg/kg), or an equivalent volume of vehicle (saline). Tumor burden, as well as DNA synthesis, apoptosis, and angiogenesis was assessed in tumor tissue following 40 days of treatment. RESULTS: OGF and LDN markedly reduced ovarian tumor burden (tumor nodule number and weight). The mechanism of action was targeted to an inhibition of tumor cell proliferation and angiogenesis; no changes in cell survival were noted. CONCLUSIONS: This study shows that a native opioid pathway can suppress human ovarian cancer in a xenograft model, and provides novel non-toxic therapies for the treatment of this lethal neoplasia.


Subject(s)
Enkephalin, Methionine/therapeutic use , Naltrexone/therapeutic use , Narcotic Antagonists/therapeutic use , Ovarian Neoplasms/pathology , Receptors, Opioid/physiology , Animals , Apoptosis/drug effects , Cell Line, Tumor , DNA/biosynthesis , Disease Progression , Enkephalin, Methionine/analysis , Female , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , Ovarian Neoplasms/blood supply , Receptors, Opioid/analysis , Xenograft Model Antitumor Assays
11.
Gut ; 59(3): 348-56, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19828468

ABSTRACT

OBJECTIVE: To examine the role of CD10, a characteristic marker of liver metastasis of colorectal cancers (CRCs). DESIGN: The effect of CD10 and Met-enkephalin (MENK) in CD10-positive and -negative human CRC cells was investigated under in vitro and in vivo conditions. Human CRC samples were examined. MAIN OUTCOME MEASURE: CD10-positive and CD10-knockdown HT29 cells and CD10-negative and CD10-transfected Colo320 cells in nude mice were treated with MENK and/or the CD10 inhibitor (thiorphan). Intracellular signalling of MENK and delta-opioid receptor (DOR) was examined by immunoblotting. RESULTS: MENK inhibited the growth, invasion and survival of CRC cells following thiorphan-induced CD10 inactivation. Thiorphan suppressed liver metastasis of CD10-positive CRC cells. Inoculation of mice with CRC cells induced MENK expression in the liver. Inhibition of hepatic MENK expression by cholesterol-conjugated antisense S-oligodeoxynucleotide increased liver metastasis of CRC cells even when the cells did not express CD10. DOR activation by MENK decreased the phosphorylation of epidermal growth factor receptor and extracellular signal-regulated kinase and increased p38-dependent apoptosis. Nitric oxide was found to induce DOR expression in CRC cells. Co-treatment with thiorphan and a nitric oxide donor had a marked anti-tumour effect on liver metastasis of HT29 cells. Of 68 CRC patients, 19 (28%) showed CD10 expression, which was dependent on the extent of liver metastasis. MENK concentration in metastasis-positive human liver was higher than that in the normal liver. CONCLUSION: CD10 expression in CRC cells abrogates the anti-tumour effect of hepatic MENK by degrading it, which enhances liver metastasis of CD10-positive CRC cells.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/pathology , Enkephalin, Methionine/pharmacology , Liver Neoplasms/secondary , Neprilysin/physiology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Enkephalin, Methionine/therapeutic use , ErbB Receptors/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Lymphatic Metastasis , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Neprilysin/metabolism , Nitric Oxide/physiology , Receptors, Opioid, delta/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Eur J Pharmacol ; 882: 173253, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32535097

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Enkephalin, Methionine/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Enkephalin, Methionine/pharmacology , Humans , Neoplasms/metabolism
13.
J Leukoc Biol ; 108(1): 215-228, 2020 07.
Article in English | MEDLINE | ID: mdl-31994797

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/metabolism , Enkephalin, Methionine/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Cyclophosphamide/pharmacology , Cytokines/metabolism , Enkephalin, Methionine/therapeutic use , Glycolipids/metabolism , Glycolysis/drug effects , Immunosuppression Therapy , Inflammation/pathology , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
14.
Int Immunopharmacol ; 78: 106032, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31835089

ABSTRACT

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.


Subject(s)
Adjuvants, Immunologic/pharmacology , Enkephalin, Methionine/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/drug therapy , Macrophages/drug effects , Adjuvants, Immunologic/therapeutic use , Animals , Enkephalin, Methionine/therapeutic use , Humans , Influenza, Human/immunology , Influenza, Human/virology , Interferon-beta/metabolism , Interleukin-6/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , RAW 264.7 Cells , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects , Virus Replication/drug effects , Virus Replication/immunology
15.
Int Immunopharmacol ; 75: 105785, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31404891

ABSTRACT

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.


Subject(s)
Enkephalin, Methionine/therapeutic use , Naltrexone/therapeutic use , Narcotic Antagonists/therapeutic use , Neoplasms/drug therapy , Animals , Enkephalin, Methionine/pharmacology , Humans , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neoplasms/immunology , Neoplasms/metabolism
16.
Int Immunopharmacol ; 55: 38-48, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29220721

ABSTRACT

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.


Subject(s)
Antiviral Agents/therapeutic use , Enkephalin, Methionine/therapeutic use , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/drug therapy , Lung/pathology , Orthomyxoviridae Infections/drug therapy , Animals , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Lung/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Receptors, Opioid/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 7/metabolism , Virus Replication
17.
Int Immunopharmacol ; 65: 312-322, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30343258

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Enkephalin, Methionine/therapeutic use , Macrophages/physiology , Stomach Neoplasms/drug therapy , Animals , Apoptosis , Cell Differentiation , Cytokines/metabolism , Female , Heterografts , Humans , Macrophages/drug effects , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Opioid/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Th2 Cells/immunology , Tumor Escape
18.
Int Immunopharmacol ; 44: 61-71, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28088065

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Dendritic Cells/drug effects , Enkephalin, Methionine/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/immunology , Female , Humans , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasms/immunology , Signal Transduction/drug effects
19.
J Clin Oncol ; 6(4): 663-8, 1988 Apr.
Article in English | MEDLINE | ID: mdl-3282034

ABSTRACT

Diarrhea commonly occurs following the administration of cisplatin. BW942C, a pentapeptide, is a synthetic enkephalin shown to control castor oil-induced and traveler's diarrhea. To assess the safety and efficacy of BW942C in controlling diarrhea caused by cisplatin, 30 adults with lung cancer who had already experienced diarrhea (three or more loose bowel movements) during the 24-hour period following a prior cisplatin administration were randomized to receive either BW942C or placebo during the next cisplatin course. All patients received a concomitant antiemetic regimen including metoclopramide, dexamethasone, and lorazepam during all courses. Patients administered BW942C experienced less diarrhea (27% v 67%, P = .02). Twenty-seven percent of patients given the pentapeptide had loose bowel movements as opposed to 93% who received placebo (P = .0002). There were no significant differences in the incidence and degree of vomiting and other treatment-related side effects observed between the placebo and treatment groups. We conclude that oral BW942C is more effective than placebo in controlling diarrhea following cisplatin chemotherapy.


Subject(s)
Antidiarrheals/therapeutic use , Cisplatin/adverse effects , Diarrhea/prevention & control , Enkephalin, Methionine/analogs & derivatives , Enkephalins , Neoplasms/drug therapy , Adult , Cisplatin/therapeutic use , Clinical Trials as Topic , Diarrhea/chemically induced , Double-Blind Method , Enkephalin, Methionine/therapeutic use , Female , Humans , Male , Middle Aged , Placebos , Random Allocation
20.
Cancer Chemother Pharmacol ; 56(5): 510-20, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15947928

ABSTRACT

Gemcitabine is the standard of care for advanced pancreatic neoplasia, and exerts its effect through inhibition of DNA synthesis. However, gemcitabine has limited survival benefits. Opioid growth factor (OGF) is an autocrine-produced peptide that interacts with the nuclear receptor, OGFr, to inhibit cell proliferation but is not cytotoxic or apoptotic. The present study was designed to examine whether a combination of chemotherapy with gemcitabine and biotherapy with OGF is more effective than either agent alone in inhibiting pancreatic cancer growth in vitro and in vivo. The combination of OGF (10(-6) M) and gemcitabine (10(-8) M) reduced MIA PaCa-2 cell number from control levels by 46% within 48 h, and resulted in a growth inhibition greater than that of the individual compounds. OGF in combination with 5-fluorouracil also depressed cell growth more than either agent alone. The action of OGF, but not gemcitabine, was mediated by a naloxone-sensitive receptor, and was completely reversible. OGF, but no other endogenous or exogenous opioids, altered pancreatic cancer growth in tissue culture. The combination of OGF and gemcitabine also repressed the growth of another pancreatic cancer cell line, PANC-1. MIA PaCa-2 cells transplanted into athymic mice received 10 mg/kg OGF daily, 120 mg/kg gemcitabine every 3 days; 10 mg/kg OGF daily and 120 mg/kg gemcitabine every 3rd day, or 0.1 ml of sterile saline daily. Tumor incidence, and latency times to tumor appearance, of mice receiving combined therapy with OGF and gemcitabine, were significantly decreased from those of the control, OGF, and gemcitabine groups. Tumor volumes in the OGF, gemcitabine, and OGF/gemcitabine groups were markedly decreased from controls beginning on days 14, 12, and 8, respectively, after tumor cell inoculation. Tumor weight and tumor volume were reduced from control levels by 36-85% in the OGF and/or gemcitabine groups on day 45 (date of termination), and the group of mice exposed to a combination of OGF and gemcitabine had decreases in tumor size of 70% and 63% from the OGF or the gemcitabine alone groups, respectively. This preclinical evidence shows that combined chemotherapy (e.g. gemcitabine) and biotherapy (OGF) provides an enhanced therapeutic benefit for pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Enkephalin, Methionine/therapeutic use , Growth Substances/therapeutic use , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/pathology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/therapeutic use , Drug Therapy, Combination , Enkephalin, Methionine/administration & dosage , Fluorouracil/pharmacology , Growth Substances/administration & dosage , Growth Substances/blood , Humans , Male , Mice , Mice, Nude , Naloxone/pharmacology , Narcotics/pharmacology , Pancreatic Neoplasms/pathology , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL