Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.508
Filter
1.
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
2.
BMC Immunol ; 24(1): 38, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828468

ABSTRACT

Novel prophylactic drugs and vaccination strategies for protection against influenza virus should induce specific effector T-cell immune responses in pulmonary airways and peripheral lymphoid organs. Designing approaches that promote T-cell-mediated responses and memory T-cell differentiation would strengthen host resistance to respiratory infectious diseases. The results of this study showed that pulmonary delivery of MENK via intranasal administration reduced viral titres, upregulated opioid receptor MOR and DOR, increased the proportions of T-cell subsets including CD8+ T cells, CD8+ TEM cells, NP/PA-effector CD8+ TEM cells in bronchoalveolar lavage fluid and lungs, and CD4+/CD8+ TCM cells in lymph nodes to protect mice against influenza viral challenge. Furthermore, we demonstrated that, on the 10th day of infection, the proportions of CD4+ TM and CD8+ TM cells were significantly increased, which meant that a stable TCM and TEM lineage was established in the early stage of influenza infection. Collectively, our data suggested that MENK administered intranasally, similar to the route of natural infection by influenza A virus, could exert antiviral activity through upregulating T-cell-mediated adaptive immune responses against influenza virus.


Subject(s)
Influenza A virus , Influenza, Human , Mice , Animals , Humans , CD8-Positive T-Lymphocytes , Enkephalin, Methionine/pharmacology , Memory T Cells , Immunologic Memory , Mice, Inbred C57BL
3.
Int Immunopharmacol ; 124(Pt B): 110967, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37741126

ABSTRACT

This study was to study the role of methionine enkephalin (menk) in cell invasion and migration as well as NK cells activation of tumor microenvironment in cervical cancer. The results showed that menk inhibited cervical cancer migration and invasion. In addition, we found menk affected epithelial to mesenchymal transition (EMT) related indicators, with increasing E-cadherin level, decreasing N-cadherin and vimentin level. Through in vivo mouse model, we found that menk IFNγ and NKP46 expression was upregulated in tumor tissues by menk compared with controls, while LAG3 expression was inhibited by menk, besides, there was an upregulation of CD11b+ NCR1+ NKs of tumor microenvironment in cervical cancer. Therefore, we concluded that menk inhibited cancer migration and invasion via affecting EMT related indicators and activated CD11b+ NCR1+ NKs of tumor microenvironment in cervical cancer, laying a theoretical foundation for the further clinical treatment of menk.


Subject(s)
Uterine Cervical Neoplasms , Humans , Female , Mice , Animals , Uterine Cervical Neoplasms/drug therapy , Enkephalin, Methionine/pharmacology , Epithelial-Mesenchymal Transition , Tumor Microenvironment , Natural Cytotoxicity Triggering Receptor 1 , Cell Line, Tumor , Cell Movement
4.
Gen Comp Endocrinol ; 342: 114353, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37536461

ABSTRACT

Although methionine-enkephalin (M-ENK) is implicated in the regulation of reproductive functions in vertebrates, its function in reptiles is little understood. This study aims to elucidate the role of M-ENK on seasonal and follicle stimulating hormone (FSH)-induced ovarian recrudescence in the gecko Hemidactylus frenatus. In the first experiment, administration of 5 µg M-ENK did not affect germinal bed activity or follicular developmental stages I, II, and III (previtellogenic) and IV (vitellogenic), but there were no stage V (vitellogenic) follicles in the ovary. However, there was a significant decrease in the mean numbers of oogonia and primary oocytes in the germinal bed associated with the complete absence of stage IV and V follicles in 25 µg M-ENK-treated lizards in contrast to experimental controls. Furthermore, there was a significant decrease in gonadotropin-releasing hormone - immunoreactive (GnRH-ir) content in the median eminence (ME) and pars distalis (PD) of the pituitary gland and sparse labelling of hypothalamic GnRH-ir neurons in 25 µg M-ENK-treated lizards. In the second experiment, treatment with FSH during the regression phase of the ovarian cycle resulted in the appearance of stage IV and V follicles, in contrast to their absence in the initial controls and treatment controls. However, treatment with 25 µg M-ENK + FSH did not result in the appearance of these follicles, indicating the inhibitory effect of M-ENK on FSH-induced ovarian recrudescence. These findings suggest that M-ENK inhibits the germinal bed and vitellogenic follicular growth in a dose-dependent manner, possibly mediated through the suppression of GnRH release in the ME and PD. In addition, M-ENK may also act at the level of the ovary in the gecko.


Subject(s)
Lizards , Ovary , Female , Animals , Follicle Stimulating Hormone/pharmacology , Analgesics, Opioid/pharmacology , Ovarian Follicle , Enkephalin, Methionine/pharmacology , Seasons , Gonadotropin-Releasing Hormone/pharmacology , Lizards/physiology , Methionine/pharmacology
5.
Acta Neurobiol Exp (Wars) ; 83(1): 84-96, 2023.
Article in English | MEDLINE | ID: mdl-37078817

ABSTRACT

In the central nervous system, long­term effects of a vagotomy include disturbance of monoaminergic activity of the limbic system. Since low vagal activity is observed in major depression and autism spectrum disorder, the study aimed to determine whether animals fully recovered after subdiaphragmatic vagotomy demonstrates neurochemical indicators of altered well­being and social component of sickness behavior. Bilateral vagotomy or sham surgery was performed in adult rats. After one month of recovery, rats were challenged with lipopolysaccharide or vehicle to determine the role of central signaling upon sickness. Striatal monoamines and met­enkephalin concentrations were evaluated using HPLC and RIA methods. We also defined a concentration of immune­derived plasma met­enkephalin to establish a long­term effect of vagotomy on peripheral analgesic mechanisms. The data indicate that 30 days after vagotomy procedure, striatal dopaminergic, serotoninergic, and enkephalinergic neurochemistry was altered, both under physiological and inflammatory conditions. Vagotomy prevented inflammation­induced increases of plasma met­enkephalin - an opioid analgesic. Our data suggest that in a long perspective, vagotomized rats may be more sensitive to pain and social stimuli during peripheral inflammation.


Subject(s)
Autism Spectrum Disorder , Enkephalin, Methionine , Rats , Animals , Enkephalin, Methionine/pharmacology , Vagotomy , Vagus Nerve/physiology , Inflammation , Amines
6.
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
7.
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
8.
Life Sci ; 312: 121189, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36396109

ABSTRACT

AIMS: Obesity and its related metabolic disorders, including insulin resistance and fatty liver, have become a serious global public health problem. Previous studies have shown Methionine Enkephalin (MetEnk) has the potential on adipocyte browning, however, its effects on the potential mechanisms of its regulation in browning as well as its improvement in energy metabolic homeostasis remain to be deciphered. MAIN METHODS: C57BL/6J male mice were fed with high-fat diet (HFD) to induce obesity model, and MetEnk was injected subcutaneously to detect changes in the metabolic status of mice, adipocytes and HepG2 cells were also treated with MetEnk, and transcriptomic, metabolomic were used to detect the changes of lipid metabolism, mitochondrial function, inflammation and other related factors. KEY FINDINGS: We found that MetEnk effectively protected against obesity weight gain in HFD-induced C57BL/6J mice, significantly improved glucose tolerance and insulin sensitivity, reduced the expression levels of interleukin 6 (IL-6), promoted white fat browning, moreover, using a combination of transcriptomic, metabolomic and inhibitors, it was found that MetEnk improved mitochondrial function, promoted thermogenesis and lipolysis by activating cAMP/PKA pathway in adipocytes, further analysis found that MetEnk also promoted lipolysis and alleviated inflammation through AMP-activated protein kinase (AMPK) pathway in mice liver and HepG2 cells. SIGNIFICANCE: Our study provides profound evidence for the role of MetEnk in improving lipid metabolism disorders. This study provides a mechanical foundation for investigating the potential of MetEnk to improve obesity and its associated metabolic disorders.


Subject(s)
Enkephalin, Methionine , Insulin Resistance , Male , Mice , Animals , Enkephalin, Methionine/pharmacology , Mice, Inbred C57BL , Adipose Tissue, White/metabolism , Thermogenesis , Diet, High-Fat/adverse effects , Obesity/metabolism , Inflammation/metabolism , Adipose Tissue, Brown/metabolism
9.
Cell Rep ; 40(13): 111440, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36170833

ABSTRACT

Low dopamine D2 receptor (D2R) availability in the striatum can predispose for cocaine abuse; though how low striatal D2Rs facilitate cocaine reward is unclear. Overexpression of D2Rs in striatal neurons or activation of D2Rs by acute cocaine suppresses striatal Penk mRNA. Conversely, low D2Rs in D2-striatal neurons increases striatal Penk mRNA and enkephalin peptide tone, an endogenous mu-opioid agonist. In brain slices, met-enkephalin and inhibition of enkephalin catabolism suppresses intra-striatal GABA transmission. Pairing cocaine with intra-accumbens met-enkephalin during place conditioning facilitates acquisition of preference, while mu-opioid receptor antagonist blocks preference in wild-type mice. We propose that heightened striatal enkephalin potentiates cocaine reward by suppressing intra-striatal GABA to enhance striatal output. Surprisingly, a mu-opioid receptor antagonist does not block cocaine preference in mice with low striatal D2Rs, implicating other opioid receptors. The bidirectional regulation of enkephalin by D2R activity and cocaine offers insights into mechanisms underlying the vulnerability for cocaine abuse.


Subject(s)
Cocaine-Related Disorders , Cocaine , Analgesics, Opioid/pharmacology , Animals , Cocaine/pharmacology , Cocaine-Related Disorders/metabolism , Corpus Striatum/metabolism , Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Enkephalins/metabolism , Enkephalins/pharmacology , Mice , Narcotic Antagonists/metabolism , Narcotic Antagonists/pharmacology , RNA, Messenger/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Reward , gamma-Aminobutyric Acid/metabolism
10.
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
11.
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
12.
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
13.
J Neurosci Res ; 100(2): 551-563, 2022 02.
Article in English | MEDLINE | ID: mdl-34821408

ABSTRACT

Multiple sclerosis (MS) is a progressive disease of the central nervous system (CNS) that primarily affects women during the second or third decade of life. The mechanism is hypothesized to involve unregulated peripheral inflammation resulting in blood-brain barrier damage, and eventual axonal damage and demyelination. Based on this understanding, the animal model of MS, experimental autoimmune encephalomyelitis (EAE), often is utilized to study lymphocyte activation. Therapeutic paradigms of exogenous opioid growth factor (OGF) or low-dose naltrexone (LDN) treatment can modulate EAE, but little is reported regarding OGF or LDN effects on peripheral inflammation, microglia activation, and/or macrophage proliferation. Moreover, little is known about differential responses to LDN or OGF relative to the duration and timing of treatment. Utilizing a female mouse model of EAE, two treatment regimens were established to investigate differences between prophylactic treatment and traditional therapy initiated at the time of disease presentation. Prophylactic OGF or LDN treatment delayed the onset of behavior, suppressed neutrophil replication, and curtailed lymphocyte proliferation which ultimately improved behavioral outcome. Traditional therapy with OGF or LDN reversed behavioral deficits, restored OGF and IL-17 serum levels, and inhibited microglial activation within 8 days. Reduced serum OGF levels in untreated EAE mice correlated with increased microglia activation within lumbar spinal cords. Both treatment regimens of OGF or LDN reduced activated microglia, whereas only prophylactic treatment prevented CNS macrophage aggregation. These data demonstrate that the timing of LDN or OGF treatment initiation alters outcomes and can prevent or reverse behavioral deficits, cytokine activation, and spinal cord pathology.


Subject(s)
Analgesics, Opioid , Encephalomyelitis, Autoimmune, Experimental , Analgesics, Opioid/pharmacology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Enkephalin, Methionine/pharmacology , Female , Humans , Immunity , Mice , Mice, Inbred C57BL , Microglia/pathology , Spinal Cord/pathology
14.
Int Immunopharmacol ; 99: 107999, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34315116

ABSTRACT

This study examined the antitumor effect of methionine enkephalin (MENK) against lung cancer in vivo and in vitro and explored the underlying mechanisms. Changes in the immune status of the tumor microenvironment (TME) in response to MENK administration were examined in mice. MENK significantly inhibited the proliferation of lung cancer cells in vivo and in vitro by regulating the Wnt/ß-catenin pathway and causing cell cycle arrest at the G0/G1 phase. Knockdown of opioid growth factor receptor abolished the effect of MENK on lung cancer cells. The immune status of the TME of mice differed between the MENK and control groups. MENK increased the infiltration of M1-type macrophages, natural killer cells, CD8+ T cells, CD4+ T cells, and dendritic cells into the TME, and decreased the proportion of myeloid inhibitory cells and M2-type macrophages. Immunohistochemical analysis of the expression of cytokines in the TME showed that MENK upregulated IL-15, IL-21, IFN-γ, and granzyme B and downregulated IL-10 and TGF-ß1 in mice. Taken together, these finding indicate that MENK may be a potential agent for lung cancer treatment in the future, especially for overcoming immune escape and immune resistance.


Subject(s)
Enkephalin, Methionine/metabolism , Enkephalin, Methionine/pharmacology , Lung Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/drug effects , Dendritic Cells/drug effects , Enkephalin, Methionine/genetics , Female , Humans , Lung Neoplasms/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , RNA, Small Interfering , T-Lymphocytes/drug effects , Wnt Signaling Pathway/drug effects
15.
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
16.
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
17.
Food Funct ; 11(11): 10231-10241, 2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33169751

ABSTRACT

The emergence of the plasmid-mediated colistin resistance mechanism (mcr-1) makes bacterial resistance to colistin increasingly serious. This mcr-1 mediated bacterial resistance to colicin is conferred primarily through modification of lipid A in lipopolysaccharides (LPS). In our previous research, antimicrobial peptide F1 was derived from Tibetan kefir and has been shown to effectively inhibit the growth of Gram-negative bacteria (E. coli), Gram-positive bacteria (Staphylococcus aureus), and other pathogenic bacteria. Based on this characteristic of antibacterial peptide F1, we speculated that it could inhibit the growth of the colicin-resistant E. coli SHP45 (mcr-1) and not easily produce drug resistance. Studies have shown that antimicrobial peptide F1 can destroy the liposome structure of the phospholipid bilayer by destroying the inner and outer membranes of bacteria, thereby significantly inhibiting the growth of E. coli SHP45 (mcr-1), but without depending on LPS. The results of this study confirmed our hypothesis, and we anticipate that antimicrobial peptide F1 will become a safe antibacterial agent that can assist in solving the problem of drug resistance caused by colistin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Enkephalin, Methionine/analogs & derivatives , Escherichia coli/drug effects , Protein Precursors/pharmacology , Colistin/pharmacology , Enkephalin, Methionine/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Microbial Sensitivity Tests
18.
Brain Res Bull ; 165: 246-252, 2020 12.
Article in English | MEDLINE | ID: mdl-33141073

ABSTRACT

Multiple sclerosis is a chronic progressive neurological disorder that has few distinctive biomarkers associated with disease progression or response to therapy. This research investigated whether non-invasive imaging correlated with animal behavior and morphological indicators of disease in response to serum levels of [Met5]-enkephalin. Using the experimental autoimmune encephalomyelitis (EAE) model, adult female C57BL/6 J mice were randomized to receive daily injections of 0.1 mg/kg naltrexone (NTX) (= low dose naltrexone, LDN), 10 mg/kg Opioid Growth Factor (OGF) (chemically termed [Met5]-enkephalin) or saline beginning at the time of disease induction. Daily composite behavior scores were recorded over a 30-day period based on tail tone, gait, righting reflex, and limb strength. Prior to disease onset (day 7), and at peak disease (day 18), mice were imaged and tissues (blood and spinal cord) collected at day 30 for serum analyses of OGF and morphology. Serum OGF levels of EAE mice treated with saline were significantly reduced from baseline and from normal mice. Longitudinal cohort data demonstrated an increase in fractional anisotropy in all cohorts by day 18. There was a significant decrease in radial diffusivity in the saline group seen at day 18 whereas the axial diffusivity was not altered amongst treatment groups. Treatment with OGF or LDN resulted in mean diffusivity rates that were comparable to baseline (normal) levels at days 7 and 18. Luxol fast blue staining of the lumbar spinal cords demonstrated a 16 % reduction in myelin staining in saline treated EAE animals when compared to OGF and LDN treated EAE mice. Immunohistochemistry with Olig2 (pan-oligodendrocyte marker) and myelin basic protein (MBP) revealed that OGF and LDN treatment restored the area (%) of MBP and number of oligodendrocytes to that of normal spinal cord (∼75 %). Saline treated EAE mice had more demyelination and fewer oligodendrocytes than normal mice. Collectively, these data suggest that a panel of biomarkers including imaging, serum biomarker levels, and behavior correlate with progression of disease, and may begin to validate use of specific non-invasive markers for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Enkephalin, Methionine/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Spinal Cord/drug effects , Animals , Behavior, Animal/drug effects , Diffusion Tensor Imaging , Encephalomyelitis, Autoimmune, Experimental/blood , Enkephalin, Methionine/blood , Female , Gait/drug effects , Magnetic Resonance Imaging , Mice , Oligodendroglia/drug effects , Spinal Cord/diagnostic imaging
19.
Int J Mol Sci ; 21(17)2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32872204

ABSTRACT

Opioids and their antagonists alter vitamin C metabolism. Morphine binds to glutathione (l-γ-glutamyl-l-cysteinyl-glycine), an intracellular ascorbic acid recycling molecule with a wide range of additional activities. The morphine metabolite morphinone reacts with glutathione to form a covalent adduct that is then excreted in urine. Morphine also binds to adrenergic and histaminergic receptors in their extracellular loop regions, enhancing aminergic agonist activity. The first and second extracellular loops of adrenergic and histaminergic receptors are, like glutathione, characterized by the presence of cysteines and/or methionines, and recycle ascorbic acid with similar efficiency. Conversely, adrenergic drugs bind to extracellular loops of opioid receptors, enhancing their activity. These observations suggest functional interactions among opioids and amines, their receptors, and glutathione. We therefore explored the relative binding affinities of ascorbic acid, dehydroascorbic acid, opioid and adrenergic compounds, as well as various control compounds, to glutathione and glutathione-like peptides derived from the extracellular loop regions of the human beta 2-adrenergic, dopamine D1, histamine H1, and mu opioid receptors, as well as controls. Some cysteine-containing peptides derived from these receptors do bind ascorbic acid and/or dehydroascorbic acid and the same peptides generally bind opioid compounds. Glutathione binds not only morphine but also naloxone, methadone, and methionine enkephalin. Some adrenergic drugs also bind to glutathione and glutathione-like receptor regions. These sets of interactions provide a novel basis for understanding some ways that adrenergic, opioid and antioxidant systems interact during anesthesia and drug abuse and may have utility for understanding drug interactions.


Subject(s)
Analgesics, Opioid/pharmacology , Ascorbic Acid/pharmacology , Glutathione/metabolism , Peptides/pharmacology , Dehydroascorbic Acid/pharmacology , Enkephalin, Methionine/pharmacology , Humans , Hydromorphone/analogs & derivatives , Hydromorphone/urine , Methadone/pharmacology , Morphine/pharmacology , Naloxone/pharmacology , Receptors, Adrenergic, beta-2/chemistry , Receptors, Dopamine D1/chemistry , Receptors, Histamine H1/chemistry , Receptors, Opioid/chemistry , Receptors, Opioid, mu/chemistry
20.
J Pharmacol Exp Ther ; 375(1): 104-114, 2020 10.
Article in English | MEDLINE | ID: mdl-32759368

ABSTRACT

This dose-response study investigated the effects of sialorphin on [Met5]enkephalin (ME)-induced inhibition of contractions in mouse vas deferens and antinociception in male rats. Differences were compared among combinations of three chemical peptidase inhibitors: amastatin, captopril, and phosphoramidon. The ratio of potencies of ME in mouse vas deferens pretreated with both sialorphin (100 µM) and a mixture of the three peptidase inhibitors (1 µM each) was higher than that with the mixture of peptidase inhibitors alone at any dose. Intrathecal administration of sialorphin (100-400 nmol) significantly and dose dependently increased ME (3 nmol)-induced antinociception with the mixture of three peptidase inhibitors (10 nmol each). The degree of antinociception with a combination of any two of the peptidase inhibitors (10 nmol each) in the absence of sialorphin was less than that in the presence of sialorphin (200 nmol). Pretreatment with both sialorphin (200 nmol) and the mixture of three peptidase inhibitors (10 nmol each) produced an approximately 100-fold augmentation in ME (10 nmol)-induced antinociception, but without signs of toxicity such as motor dysfunction in rats. Radioligand receptor binding assay revealed that sialorphin did not affect either binding affinity or maximal binding capacity of [d-Ala2,N-MePhe4,Gly-ol5]enkephalin. These results indicate that sialorphin potentiates the effects of ME without toxicity by a mechanism other than peptidase inhibition and with no effect on its affinity to µ-opioid receptors. SIGNIFICANCE STATEMENT: Sialorphin is regarded as an endogenous peptidase inhibitor that interacts with enkephalin-degrading enzymes. The results of these in vitro and in vivo studies confirm that sialorphin potentiates the effects of [Met5]enkephalin without toxicity by an action other than peptidase inhibition. This suggests that sialorphin offers the advantage of reducing or negating the side effects of opioid drugs and endogenous opioid peptides.


Subject(s)
Analgesics/pharmacology , Enkephalin, Methionine/pharmacology , Peptides/pharmacology , Protease Inhibitors/pharmacology , Vas Deferens/drug effects , Analgesics/administration & dosage , Animals , Dose-Response Relationship, Drug , Drug Synergism , Enkephalin, Methionine/administration & dosage , In Vitro Techniques , Injections, Spinal , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Nociceptive Pain/drug therapy , Nociceptive Pain/metabolism , Pain Measurement , Peptides/administration & dosage , Protease Inhibitors/administration & dosage , Protein Binding , Radioligand Assay , Rats, Wistar , Receptors, Opioid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...