Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Curr Oncol Rep ; 24(10): 1337-1349, 2022 10.
Article in English | MEDLINE | ID: mdl-35648340

ABSTRACT

PURPOSE OF REVIEW: Antagonists of mu-opioid receptor role in cancer progression remains to be elucidated. The objective of this review was to summarize the available evidence on antagonists of mu-opioid receptor effect on tumor progression and prognosis in different types of cancers and an evaluation of the available findings on their mechanism of action. RECENT FINDINGS: We have found studies related to methylnaltrexone (MNTX) and naltrexone (NTX) usage in cancer outcomes-related setting. We found consistent preclinical evidence of a potential action of MNTX and NTX on cancer growth and spread mediated mainly by effect on the opioid growth factor receptor (OGFr) axis, which results in depressed cell replication. However, clinical results are scarce and limited to poor-quality evidence. Further high-quality studies are warranted to study antagonists of mu-opioid receptor role as a therapeutic option in different types of cancer, especially in patients where the classical treatment causes unacceptable side effects.


Subject(s)
Narcotic Antagonists , Neoplasms , Cell Proliferation , Humans , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Quaternary Ammonium Compounds , Receptors, Opioid/metabolism
2.
Connect Tissue Res ; 60(6): 521-529, 2019 11.
Article in English | MEDLINE | ID: mdl-30931654

ABSTRACT

Purpose: This research investigated the presence and integrity of the opioid growth factor (OGF)-opioid growth factor receptor (OGFr) regulatory pathway in type 1 diabetic (T1D) rats, and investigated whether modulation of this axis by naltrexone (NTX) altered the composition of normal bone or fractured femurs. Materials and Methods: Diabetes was induced by streptozotocin; controls rats received buffer. Hyperglycemic animals were subjected to femur osteotomy, with randomized cohorts receiving either topical NTX or sterile saline in calcium carbonate. In experiment 2, hyperglycemic rats were injected daily for 3 weeks with either 30 mg/kg NTX or sterile saline. Expression levels of OGF and OGFr were measured by immunohistochemistry, bone composition was assessed by histomorphometry, and bone integrity was evaluated by µCT and 3-point bending. Results: Relative to normoglycemic bones, OGF and OGFr expression levels were increased 95% and 84%, respectively, in T1D bone; serum levels of OGF in T1D rats were elevated 23%. Hyperglycemia decreased the strength (26%), osteocalcin expression (17%), and number of proliferative (Ki67+) cells (32%) in intact femur. Topical NTX treatment of fractured femurs reduced the percentage of granulation tissue and increased cartilage. Systemic NTX treatment of diabetic rats increased strength by 21% and energy absorbed by105% in bone relative to measurements in saline-treated diabetic rats. Conclusions: The OGF-OGFr pathway appears to be dysregulated in the bone of T1D rats. Topical NTX treatment of T1D fractured bone accelerated some aspects of delayed diabetic fracture repair, and systemic NTX protected against some elements of compromised bone composition.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Femur/metabolism , Naltrexone/pharmacology , Receptors, Opioid/metabolism , Signal Transduction/drug effects , Animals , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Femur/pathology , Male , Rats , Rats, Sprague-Dawley
3.
Free Radic Biol Med ; 223: 398-412, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39122201

ABSTRACT

Enkephalins are reportedly correlated with heart function. However, their regulation in the heart remains unexplored. This study revealed a substantial increase in circulating levels of opioid growth factor (OGF) (also known as methionine enkephalin) and myocardial expression levels of both OGF and its receptor (OGFR) in subjects treated with doxorubicin (Dox). Silencing OGFR through gene knockout or using adeno-associated virus serotype 9 carrying small hairpin RNA effectively alleviated Dox-induced cardiotoxicity (DIC) in mice. Conversely, OGF supplementation exacerbated DIC manifestations, which could be abolished by administration of the OGFR antagonist naltrexone (NTX). Mechanistically, the previously characterized OGF/OGFR/P21 axis was identified to facilitate DIC-related cardiomyocyte apoptosis. Additionally, OGFR was observed to dissociate STAT1 from the promoters of ferritin genes (FTH and FTL), thereby repressing their transcription and exacerbating DIC-related cardiomyocyte ferroptosis. To circumvent the compromised therapeutic effects of Dox on tumors owing to OGFR blockade, SiO2-based modifiable lipid nanoparticles were developed for heart-targeted delivery of NTX. The pretreatment of tumor-bearing mice with the assembled NTX nanodrug successfully provided cardioprotection against Dox toxicity without affecting Dox therapy in tumors. Taken together, this study provides a novel understanding of Dox cardiotoxicity and sheds light on the development of cardioprotectants for patients with tumors receiving Dox treatment.

4.
Int J Gen Med ; 16: 5527-5548, 2023.
Article in English | MEDLINE | ID: mdl-38034898

ABSTRACT

Introduction: The relationship between the expression of opioid-associated receptors and cancer outcomes is complex and varies among studies. Methods: This study focused on six opioid-related receptors (OPRM1, OPRD1, OPRK1, OPRL1, OGFR, and TLR4) and their impact on cancer patient survival. Bioinformatics analysis was conducted on 33 cancer types from The Cancer Genome Atlas database to examine their expression, clinical correlations, mechanisms in the tumor microenvironment, and potential for immunotherapy. Due to significantly lower expression of OPRM1, OPRD1, and OPRK1 compared to OGFR and TLR4, the analysis concentrated on the latter two genes. Results: OGFR was highly expressed in 16 tumor types, while TLR4 showed low expression in 13. Validation from external samples, the Gene Expression Omnibus, and the Human Protein Atlas supported these findings. The diagnostic value of these two genes was demonstrated using the Genotype-Tissue Expression database. Univariate Cox regression models and Kaplan-Meier curves confirmed OGFR's impact on prognosis in a cancer type-specific manner, while high TLR4 expression was associated with a favorable prognosis. Analysis of the tumor microenvironment using a deconvolution algorithm linked OGFR to CD8+ T cells and TLR4 to macrophages. Single-cell datasets further validated this correlation. In 25 immune checkpoint blockade treatment cohorts, TLR4 expression showed promise as an immunotherapy efficacy predictor in non-small cell lung cancer, urothelial carcinoma, and melanoma. Conclusion: In a pan-cancer analysis of 33 tissues, OGFR was consistently highly expressed, while TLR4 had low expression. Both genes have diagnostic and prognostic significance and are linked to immune cells in the tumor microenvironment. TLR4 has potential as an immunotherapeutic response marker.

5.
Int J Biol Sci ; 18(7): 2882-2897, 2022.
Article in English | MEDLINE | ID: mdl-35541895

ABSTRACT

Cold-inducible RNA-binding protein (CIRBP) is documented to be required for maintaining cardiac function, however, its role in chemotherapy-induced cardiotoxicity remains obscured. Herein, we report that CIRBP decreases cardiomyocyte apoptosis and attenuates cardiotoxicity through disrupting OGF-OGFR signal. CIRBP deficiency is involved in diverse chemotherapeutic agents induced cardiomyocyte apoptosis. Delivery of exogenous CIRBP to the mouse myocardium significantly mitigated doxorubicin-induced cardiac apoptosis and dysfunction. Specifically, OGFR was identified as a downstream core effector responsible for chemotherapy-induced cardiomyocyte apoptosis. CIRBP was shown to interact with OGFR mRNA and to repress OGFR expression by reducing mRNA stability. CIRBP-mediated cytoprotection against doxorubicin-induced cardiac apoptosis was demonstrated to largely involve OGFR repression by CIRBP. NTX as a potent antagonist of OGFR successfully rescued CIRBP ablation-rendered susceptibility to cardiac dyshomeostasis upon exposure to doxorubicin, whereas another antagonist ALV acting only on opioid receptors did not. Taken together, our results demonstrate that CIRBP confers myocardium resistance to chemotherapy-induced cardiac apoptosis and dysfunction by dampening OGF/OGFR axis, shedding new light on the mechanisms of chemo-induced cardiotoxicity and providing insights into the development of an efficacious cardioprotective strategy for cancer patients.


Subject(s)
Cardiotoxicity , Doxorubicin , Enkephalin, Methionine , Animals , Apoptosis/drug effects , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Cell Proliferation , Doxorubicin/toxicity , Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Humans , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , RNA-Binding Proteins/genetics
6.
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
7.
Int Immunopharmacol ; 96: 107733, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33965882

ABSTRACT

Cutaneous squamous cell carcinoma (CSCC) is a common skin tumour. Due to weak immunogenicity, recurrence is frequent after treatment. In this study, we explored the effects and mechanisms of methionine enkephalin (MENK), an endogenous opioid peptide and negative growth regulator, in CSCC. MENK inhibited A431 cell proliferation and promoted apoptosis through the opioid growth factor receptor (OGFr). Importantly, MENK also induced autophagy in CSCC and stimulated the emission of DAMPs in A431 cells, which resulted in enhanced activation of dendritic cells (DC).In conclusion, MENK provides an effective method with therapeutic potential to modulate the CSCC microenvironment by utilizing autophagy in the cancer cells.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Dendritic Cells/immunology , Enkephalin, Methionine/pharmacology , Histocompatibility Antigens Class II/immunology , Skin Neoplasms/drug therapy , Animals , Autophagy/drug effects , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Female , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Mice, Inbred BALB C , Neurotransmitter Agents/pharmacology , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Int Immunopharmacol ; 99: 107996, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34311187

ABSTRACT

The antitumor effects of methionine enkephalin (MENK), also known as opioid growth factor (OGF), including its inhibitory effects on cutaneous squamous cell carcinoma (CSCC), have been established. In this study, we determined the precise mechanism by which MENK suppresses CSCC cell growth. In particular, MENK induced G0/G1 cell cycle arrest and promoted apoptosis in CSCC cells via the Bcl-2/Bax/Caspase-3 signaling pathway. Moreover, MENK reduced immunosuppression by downregulating the number of myeloid-derived suppressor cells (MDSCs) and regulating the polarization of tumor-associated macrophages from M2 to M1 in vivo. Furthermore, JAK2/STAT3, an important tumor-promotion and immunosuppression signaling pathway that is involved in MDSC expansion in tumors and macrophage polarization, was inhibited. These findings highlight the potential of the JAK2/STAT3 signaling pathway as a therapeutic target and suggest the clinical application of MENK for CSCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Macrophages/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Polarity/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Enkephalin, Methionine/genetics , Female , Humans , Immunosuppression Therapy , Janus Kinase 2/metabolism , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
9.
Front Oncol ; 11: 792290, 2021.
Article in English | MEDLINE | ID: mdl-35004315

ABSTRACT

Opioids are administered to cancer patients in the period surrounding tumour excision, and in the management of cancer-associated pain. The effects of opioids on tumour growth and metastasis, and their consequences on disease outcome, continue to be the object of polarised, discrepant literature. It is becoming clear that opioids contribute a range of direct and indirect effects to the biology of solid tumours, to the anticancer immune response, inflammation, angiogenesis and importantly, to the tumour-promoting effects of pain. A common misconception in the literature is that the effect of opioid agonists equates the effect of the mu-opioid receptor, the major target of the analgesic effect of this class of drugs. We review the evidence on opioid receptor expression in cancer, opioid receptor polymorphisms and cancer outcome, the effect of opioid antagonists, especially the peripheral antagonist methylnaltrexone, and lastly, the evidence available of a role for opioids through non-opioid receptor mediated actions.

10.
Exp Biol Med (Maywood) ; 246(5): 629-636, 2021 03.
Article in English | MEDLINE | ID: mdl-33203224

ABSTRACT

The opioid growth factor (OGF)-OGF receptor (OGFr) pathway is present in the ocular surface and functions to maintain homeostasis of the epithelium. The OGF-OGFr pathway has been reported to be dysregulated in diabetic individuals and animal models, and is reflected in elevations of the inhibitory growth factor, OGF, chemically termed [Met5]-enkephalin. Recently, our laboratory reported elevated levels of OGF and OGFr in the serum and corneal epithelium of type 1 diabetic rats, suggesting that dysregulation of the OGF-OGFr axis may lead to dry eye, abnormal corneal surface sensitivity, and delayed re-epithelialization. Blockade of OGF-OGFr pathway using naltrexone, a potent opioid receptor antagonist, reverses dry eye symptoms and restores corneal surface sensitivity in diabetic rats when used as a therapy. Based on the evidence that both OGF and OGFr are elevated in type 1 diabetic rats, this study examined whether systemic or topical naltrexone treatment initiated at the time of induction of hyperglycemia could protect against the development of diabetic ocular surface complications. Diabetic male Sprague-Dawley rats treated systemically or topically with naltrexone had a delayed onset of dry eye and altered corneal surface sensitivity, and an improved healing rate for corneal wounds, that were comparable to non-diabetic rats. Serum levels of OGF were normal for rats receiving systemic naltrexone, and OGF tissue levels were normal for type 1 diabetic rats receiving twice daily naltrexone drops. OGFr levels remained elevated. These data support the role of the OGF-OGFr axis in regulation of ocular surface complications, and suggest that naltrexone therapy may be beneficial for pre-diabetic and early diabetic individuals.


Subject(s)
Diabetes Complications/pathology , Eye/pathology , Receptors, Opioid/metabolism , Severity of Illness Index , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cornea/drug effects , Cornea/pathology , Diabetes Complications/blood , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/pathology , Enkephalin, Methionine/blood , Enkephalin, Methionine/metabolism , Eye/drug effects , Male , Naltrexone/pharmacology , Rats, Sprague-Dawley , Receptors, Opioid/blood , Time Factors
11.
Exp Biol Med (Maywood) ; 245(15): 1414-1421, 2020 09.
Article in English | MEDLINE | ID: mdl-32640891

ABSTRACT

IMPACT STATEMENT: This research extends our knowledge about the presence and role of the OGF-OGFr regulatory axis in type 1 diabetes (T1D) and demonstrates specific targets within the pathway that are dysregulated. Serum levels of OGF, an inhibitory growth factor, are significantly elevated in male T1D rats, and OGFr serum values are increased in T1D. The onset of elevated OGF corresponds to the onset of ocular surface complications including dry eye, delayed corneal epithelial repair, and abnormal corneal surface sensitivity in T1D. Systemic insulin does not protect against elevated OGF levels or the onset of dry eye and sensitivity. These data are the first to associate some ocular surface defects in T1D with alterations in the OGF-OGFr pathway.


Subject(s)
Analgesics, Opioid/blood , Analgesics, Opioid/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Eye/pathology , Receptors, Opioid/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Epithelium, Corneal/pathology , Male , Neprilysin/blood , Rats, Sprague-Dawley , Re-Epithelialization , Receptors, Opioid/blood , Time Factors
12.
Int Immunopharmacol ; 78: 106068, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31835085

ABSTRACT

AIMS: Cancer is a major worldwide health problem. Cancer cells express opioid growth factor (OGF) which controls their growth. Naltrexone in low dose (LDN) blocks opioid receptors intermittently and controls the replication of cancer cells. The aim of this study was to investigate the effect of LDN and its chemotherapeutic additive effect on the growth of solid Ehrlich carcinoma in mice with focus on the OGFr and immune responses. MAIN METHODS: Sixty female Swiss albino mice were assigned into 5 groups (n: 12 mice each): (i): normal control, (ii): Solid Ehrlich carcinoma (SEC), (iii): SEC treated with LDN, (iv): SEC treated with 5-fluorouracil (5-FU), (v): SEC treated with LDN + 5-FU. All drugs were started when the tumor became palpable on 9th day. At the end of the study animals were sacrificed, blood and tissue samples were collected. Tumor weight and volume were measured. Splenocytes and myeloid derived suppressor cells (MDSC) were counted. Tumor expression of opioid growth factor receptors (OGFr), serum level of IFN-γ, tumor histopathology (H&E) and immunohistochemistry staining of p21, p53, Bcl2 were assessed. KEY FINDINGS: All drug-treated groups showed reduction in tumor weight and volume, significant increase of splenocyte with tendency to reduce MDSC cell counts. LDN led to significant increase in OGFr both in solo and in combination with 5FU. Serum IFN-γ is significantly increased by LDN but decreased by 5-FU. Also, LDN and 5FU increased immunehistochemical staining of p21 while decreased immunostaining of Bcl2. In animals treated with a combination of LDN and 5FU a maximal downregulation of the antiapoptotic mediator BCL2 was observed. SIGNIFICANCE: The current study suggested that LDN may play a role in inhibiting cancer cell growth and highlights the possibility of promising combination with cancer chemotherapeutics, which guarantee further clinical studies for approval.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Ehrlich Tumor/drug therapy , Naltrexone/administration & dosage , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Opioid/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Ehrlich Tumor/immunology , Carcinoma, Ehrlich Tumor/pathology , Cell Line, Tumor/transplantation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Female , Fluorouracil/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunologic Factors/administration & dosage , Mice , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Off-Label Use
13.
Int Immunopharmacol ; 86: 106718, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32585612

ABSTRACT

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


Subject(s)
Antineoplastic Agents/pharmacology , Naltrexone/pharmacology , Uterine Cervical Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytokines/blood , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , Humans , Macrophage Activation/drug effects , Mice, Inbred BALB C , Mice, Nude , Naltrexone/therapeutic use , Receptors, Opioid/biosynthesis , Receptors, Opioid/drug effects , Receptors, Opioid/genetics , Snail Family Transcription Factors/metabolism , Tumor-Associated Macrophages/drug effects , Uterine Cervical Neoplasms/pathology , Vimentin/metabolism , Xenograft Model Antitumor Assays
14.
Int Immunopharmacol ; 83: 106388, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32171145

ABSTRACT

The incidence of colorectal cancer (CRC) is increasing annually worldwide. However, traditional chemotherapy has obvious side effects. Low-dose naltrexone (LDN) has been reported to delay tumor progression, but the mechanism remains unclear. Therefore, the aim of this study was to explore the mechanisms underlying the inhibitory effect of LDN on CRC progression in vivo and in vitro. We found that expression of macrophage markers (F4/80, CD68) was increased in nude mice treated with LDN compared with the control group (p < 0.05). Additionally, levels of M1 macrophage phenotypic markers (CD80) and cytokines (tumor necrosis factor-α, TNF-α) were higher than in the control group (p < 0.05). LDN was able to upregulate expression of the opioid growth factor receptor (OGFr) and apoptosis-related factors Bax, caspase-9, caspase-3, and PARP and downregulate expression of Bcl-2, Survivin, and Ki67 to promote tumor cell apoptosis. Therefore, we speculate that LDN reduces tumor size by increasing levels of M1-like macrophages and activating the Bax/Bcl-2/caspase-3/PARP signaling pathway to induce apoptosis. We suggest that LDN has potential for the treatment of CRC.


Subject(s)
Antineoplastic Agents/therapeutic use , Caspase 3/metabolism , Colorectal Neoplasms/drug therapy , Macrophages/immunology , Naltrexone/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis , Carcinogenesis , Cell Differentiation , Cytokines/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mice , Mice, Nude , Receptors, Opioid/metabolism , Signal Transduction , Th1 Cells/immunology
15.
Onco Targets Ther ; 12: 6145-6156, 2019.
Article in English | MEDLINE | ID: mdl-31496722

ABSTRACT

PURPOSE: The long noncoding RNA LINC00673 has emerged as an important regulator of cancer development and progression. However, the clinical significance and biological roles of LINC00673 in epithelial ovarian cancer (EOC) remain unclear. In this study, we aimed to explore the oncogenic roles and underlying molecular mechanisms of LINC00673 in EOC. PATIENTS AND METHODS: The expression levels of LINC00673 in EOC tissues and cell lines were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Real-time cellular analysis (RTCA), flow cytometry, and transwell assays were conducted to investigate cell proliferation, apoptosis, migration and invasion in vitro. Subcutaneous transplanted tumors were established to explore the oncogenic role of LINC00673 in vivo. Differentially expressed genes were analyzed using transcriptome sequencing. Protein levels were determined by Western blot assays. RESULTS: LINC00673 was upregulated in EOC tissues and cell lines compared to their corresponding normal controls. High expression of LINC00673 was associated with advanced International Federation of Gynecology and Obstetrics (FIGO) stage, serous histological subtype, lymph node metastasis and poor prognosis in patients with EOC. LINC00673 was also identified as an independent prognostic factor for EOC. In addition, LINC00673 promoted cell migration, invasion and proliferation and inhibited cell apoptosis in vitro and induced tumor growth in vivo. Mechanistically, opioid growth factor receptor (OGFR) was found to be a potential downstream target gene that mediated the oncogenic effect of LINC00673 in EOC. CONCLUSION: LINC00673 contributes to EOC proliferation and metastasis and may be a promising prognostic biomarker for EOC patients.

16.
Cancer Manag Res ; 10: 4773-4787, 2018.
Article in English | MEDLINE | ID: mdl-30425572

ABSTRACT

BACKGROUND: Gastric cancer (GC) is the second cause of cancer-related deaths. Methionine enkephalin (MENK), an endogenous opioid peptide, has immunological and antitumor activity. PURPOSE: The aim of this work was to investigate whether MENK could exhibit activity against human GC in vitro and in vivo. MATERIALS AND METHODS: Human GC cells were treated with MENK. Cell viability, colony formation, cell morphology, cell cycle, and apoptosis were assessed. The effects of MENK on gene expression of OGFr, Bax, BCL-2, caspase-3, PARP, Ki67, cyclin D1, c-myc, survivin were quantifed by qRT-PCR. Western blot was used to analyze the effects of MENK on protein expression of OGFr, Bax, BCL-2, caspase-3, PARP. The anti-tumor activity of MENK in gastic carcinoma was also investigated with animal experiments. RESULTS: The results indicate that MENK could significantly inhibit the growth of human GC cells SGC7901 and HGC27 in a concentration- and time-dependent manner, decrease the number of cell colonies, and arrest cell cycle in the G0/G1 phase by causing a decrease in Ki67, cyclin D1, and c-myc mRNA. Furthermore, MENK could induce tumor cell apoptosis associated with the upregulation of Bax, a corresponding downregulation of BCL-2 and survivin, and activation of caspase-3 and PARP. Moreover, MENK upregulated the expression of opioid receptors (OGFr) in SGC7901 and HGC27 cells. The interaction between MENK and OGFr in SGC7901 and HGC27 cells appears to be essential for the antitumor activity of MENK. CONCLUSION: We conclude that MENK may be a potential drug for the treatment of GC.

17.
Am J Clin Exp Urol ; 6(4): 164-171, 2018.
Article in English | MEDLINE | ID: mdl-30246052

ABSTRACT

Opioid receptors are G protein-coupled receptors that bind opioid ligands including endorphins and enkephalins. The existence of a number of opioid receptors, including the mu-opioid receptor (OPRM1), delta-opioid receptor (OPRD1), kappa-opioid receptor (OPRK1) and zeta-opioid receptor (OGFR) have been reported. However, the potential expression and role of these receptors on human prostate carcinogenesis is unknown. In the present study, we examined opioid receptor expression in human prostate cancer cell lines and in prostate cancer tissue. We observed using quantitative real-time PCR analysis that OGFR and OGFRL1 mRNA is expressed in all examined prostate cancer cell lines as well as in an immortalized, non-tumorigenic prostate epithelial cell line (RWPE-1). Conversely, OPRK1 mRNA expression was detected in a more limited number of cell lines (LNCaP and VCaP), while OPRD1 and OPRM1 mRNA expression was undetectable in all examined prostate cell lines. Interestingly, androgen sensitive LNCaP cells expressed high amounts of OPRK1, OGFR and OGFRL1 compared to other cell lines. Therefore, we investigated the effect of androgen on the mRNA expression of OPRK1, OGFR, OGFRL1 in the LNCaP cell line. Our results demonstrated that the synthetic androgen (R1881) represses mRNA of OPRK1, OGFR and OGFRL1 in a time-dependent manner. Furthermore, immunohistochemistry demonstrated OGFR is expressed at high levels in prostate cancer tissue compared to benign tissue, and that OGFR expression is high in undifferentiated and aggressive prostate cancer tissue. This is the first study showing OGFR and OGFRL1 are androgen repressed genes, and these results suggest a role for the opioid signaling axis in prostate cancer.

18.
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
19.
Exp Biol Med (Maywood) ; 241(3): 273-81, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26429201

ABSTRACT

Opioid growth factor receptor (OGFr) facilitates growth inhibition in the presence of its specific ligand opioid growth factor (OGF), chemically termed [Met(5)]-enkephalin. The function of the OGF-OGFr axis requires the receptor to translocate to the nucleus. However, the mechanism of nuclear export of OGFr is unknown. In this study, endogenous OGFr, as well as exogenously expressed OGFr-EGFP, demonstrated significant nuclear accumulation in response to leptomycin B (LMB), an inhibitor of CRM1-dependent nuclear export, suggesting that OGFr is exported in a CRM1-dependent manner. One consensus sequence for a nuclear export signal (NES) was identified. Mutation of the associated leucines, L217 L220 L223 and L225, to alanine resulted in decreased nuclear accumulation. NES-EGFP responded to LMB, indicating that this sequence is capable of functioning as an export signal in isolation. To determine why the sequence functions differently in isolation than as a full length protein, the localization of subNES was evaluated in the presence and absence of MG132, a potent inhibitor of proteosomal degradation. MG132 had no effect of subNES localization. The role of tandem repeats located at the C-terminus of OGFr was examined for their role in nuclear trafficking. Six of seven tandem repeats were removed to form deltaTR. DeltaTR localized exclusively to the nucleus indicating that the tandem repeats may contribute to the localization of the receptor. Similar to the loss of cellular proliferation activity (i.e. inhibition) recorded with subNES, deltaTR also demonstrated a significant loss of inhibitory activity indicating that the repeats may be integral to receptor function. These experiments reveal that OGFr contains one functional NES, L217 L220 L223 and L225 and can be exported from the nucleus in a CRM1-dependent manner.


Subject(s)
Active Transport, Cell Nucleus , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Opioid/metabolism , Animals , COS Cells , Chlorocebus aethiops , Nuclear Export Signals , Receptors, Opioid/genetics , Exportin 1 Protein
20.
Exp Biol Med (Maywood) ; 238(5): 579-87, 2013 May.
Article in English | MEDLINE | ID: mdl-23856908

ABSTRACT

The opioid growth factor (OGF) - opioid growth factor receptor (OGFr) axis is a biological pathway that is present in human ovarian cancer cells and tissues. OGF, chemically termed [Met(5)]-enkephalin, is an endogenous opioid peptide that interfaces with OGFr to delay cells moving through the cell cycle by upregulation of cyclin-dependent inhibitory kinase pathways. OGF inhibitory activity is dose dependent, receptor mediated, reversible, protein and RNA dependent, but not related to apoptosis or necrosis. The OGF-OGFr axis can be targeted for treatment of human ovarian cancer by (i) administration of exogenous OGF, (ii) genetic manipulation to over-express OGFr and (iii) use of low dosages of naltrexone, an opioid antagonist, which stimulates production of OGF and OGFr for subsequent interaction following blockade of the receptor. The OGF-OGFr axis may be a feasible target for treatment of cancer of the ovary (i) in a prophylactic fashion, (ii) following cytoreduction or (iii) in conjunction with standard chemotherapy for additive effectiveness. In summary, preclinical data support the transition of these novel therapies for treatment of human ovarian cancer from the bench to bedside to provide additional targets for treatment of this devastating disease.


Subject(s)
Naltrexone/therapeutic use , Narcotic Antagonists/therapeutic use , Neoplasm Proteins , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Receptors, Opioid , Apoptosis/drug effects , Cell Cycle/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Necrosis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Opioid Peptides/metabolism , Ovarian Neoplasms/pathology , Receptors, Opioid/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL