Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 125(Pt B): 111226, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37976597

RESUMO

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.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Animais , Camundongos , Caspase 3 , Caspase 9 , Inibidores de Poli(ADP-Ribose) Polimerases , Proteína X Associada a bcl-2 , Osteossarcoma/tratamento farmacológico , Encefalina Metionina/farmacologia , Encefalina Metionina/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico
2.
Int Immunopharmacol ; 124(Pt B): 110967, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37741126

RESUMO

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.


Assuntos
Neoplasias do Colo do Útero , Humanos , Feminino , Camundongos , Animais , Neoplasias do Colo do Útero/tratamento farmacológico , Encefalina Metionina/farmacologia , Transição Epitelial-Mesenquimal , Microambiente Tumoral , Receptor 1 Desencadeador da Citotoxicidade Natural , Linhagem Celular Tumoral , Movimento Celular
3.
Int Immunopharmacol ; 118: 110064, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36989897

RESUMO

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.


Assuntos
Neoplasias Pulmonares , Células Supressoras Mieloides , Animais , Camundongos , Transição Epitelial-Mesenquimal , Macrófagos Associados a Tumor/metabolismo , Encefalina Metionina/farmacologia , Encefalina Metionina/uso terapêutico , Encefalina Metionina/metabolismo , Microambiente Tumoral , Camundongos Nus , Linhagem Celular Tumoral , Movimento Celular
4.
Int Immunopharmacol ; 111: 109125, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35988519

RESUMO

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.


Assuntos
Neoplasias Colorretais , Encefalina Metionina , Animais , Antígeno B7-H1 , Neoplasias Colorretais/tratamento farmacológico , Encefalina Metionina/farmacologia , Encefalina Metionina/uso terapêutico , Fatores Imunológicos , Camundongos , Receptor de Morte Celular Programada 1 , Microambiente Tumoral
5.
Int Immunopharmacol ; 99: 107996, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34311187

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Macrófagos/efeitos dos fármacos , Células Supressoras Mieloides/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Encefalina Metionina/genética , Feminino , Humanos , Terapia de Imunossupressão , Janus Quinase 2/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Interferente Pequeno , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Int Immunopharmacol ; 99: 107999, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34315116

RESUMO

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.


Assuntos
Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Encefalina Metionina/genética , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno , Linfócitos T/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos
7.
Int Immunopharmacol ; 78: 106032, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31835089

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/farmacologia , Encefalina Metionina/farmacologia , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Adjuvantes Imunológicos/uso terapêutico , Animais , Encefalina Metionina/uso terapêutico , Humanos , Influenza Humana/imunologia , Influenza Humana/virologia , Interferon beta/metabolismo , Interleucina-6/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , Receptores Opioides mu/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia
8.
Int Immunopharmacol ; 68: 193-203, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30654309

RESUMO

The purpose of this study was to identify the modulatory effect of MENK on MDSCs and to investigate the relationship between this modulation and the expression of opioid receptors. Our results showed that MENK could inhibit the proliferation of MDSCs from both marine bone marrow and spleen. MENK also could promote the expression of opioid receptors MOR and DOR. MENK suppressed the PMN-MDSCs generated from splenocyte while up-regulated M-MDSCs. The stimulation of MENK increased the production of IL-4 secreted by MDSCs from slpenocytes. Our currently data indicated that MENK could suppress the accumulation of MDSCs in tumor-bearing mice via binding to and up-regulating expressions of subunits of opioid receptors. Therefore, it is concluded that MENK, through triggering opioid receptors could exert inhibiting modulation on MDSCs.


Assuntos
Encefalina Metionina/farmacologia , Células Supressoras Mieloides/efeitos dos fármacos , Receptores Opioides/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Feminino , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/metabolismo , Neoplasias/metabolismo , Baço/citologia
9.
Cancer Manag Res ; 10: 4773-4787, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30425572

RESUMO

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.

10.
Int Immunopharmacol ; 65: 312-322, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30343258

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Encefalina Metionina/uso terapêutico , Macrófagos/fisiologia , Neoplasias Gástricas/tratamento farmacológico , Animais , Apoptose , Diferenciação Celular , Citocinas/metabolismo , Feminino , Xenoenxertos , Humanos , Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Opioides/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Células Th2/imunologia , Evasão Tumoral
11.
Int Immunopharmacol ; 65: 76-83, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30290369

RESUMO

The goal of this work was to investigate how MENK could regulate the functions of CD8+T cells and to explore the relationship between this regulation and opioid receptor expression. Our results showed that the opioid receptors presented on the cell menbrane of CD8+T cells were MOR and DOR. MENK promoted the expression of opioid receptors as well as the elevation of the surface molecules such as CD28, PD-1, CTLA-4 and FasL and intracellular granzyme B. Selectively blocking the MOR by CTAP or DOR by NTI could result in inhibition of the corresponding CD8+T cells proliferation and the expressions of surface molecules. In addition, non-selectively blocking both MOR and DOR by NTX could further impair the functions and proliferation of CD8+T cells. Our currently data indicated that MENK could play a vital role in immune functions via precise regulation to subunits of opioid receptors.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Encefalina Metionina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Neurotransmissores/farmacologia , Receptores Opioides/metabolismo , Animais , Linfócitos T CD8-Positivos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Subunidades Proteicas , Receptores Opioides/genética
12.
Int Immunopharmacol ; 61: 178-184, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29885638

RESUMO

Naltrexone, a non-selective antagonist of opioid receptors, is mainly used as rehabilitation therapy for discharged opiate addicts to eliminate addiction in order to maintain a normal life and prevent or reduce relapse. In recent years, there have been some novel and significant findings on the off-label usage of naltrexone. Within a specific dosage window, LDN can act as an immunomodulator in multiple autoimmune diseases and malignant tumors as well as alleviate the symptoms of some mental disorders. The results of increasing studies indicate that LDN exerts its immunoregulatory activity by binding to opioid receptors in or on immune cells and tumor cells. These new discoveries indicate that LDN may become a promising immunomodulatory agent in the therapy for cancer and many immune-related diseases. In this article, we review the pharmacological functions and mechanisms of LDN as well as its clinical therapeutic potential as revealed by our team and other researchers.


Assuntos
Analgésicos Opioides/uso terapêutico , Doença de Crohn/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Autoimunidade/efeitos dos fármacos , Cálculos da Dosagem de Medicamento , Humanos , Uso Off-Label , Receptores Opioides/metabolismo , Centros de Tratamento de Abuso de Substâncias
13.
Int Immunopharmacol ; 55: 38-48, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29220721

RESUMO

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.


Assuntos
Antivirais/uso terapêutico , Encefalina Metionina/uso terapêutico , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/tratamento farmacológico , Pulmão/patologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Animais , Citocinas/metabolismo , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Receptores Opioides/metabolismo , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/metabolismo , Receptor 7 Toll-Like/metabolismo , Replicação Viral
14.
Oncol Rep ; 38(4): 2132-2140, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28849104

RESUMO

Melanoma is a common cutaneous malignancy, that is also found in specific mucosal sites, and is associated with a poor prognosis. The aim of the present study was to investigate the cytotoxicity of methionine enkephalin (MENK) for B16 melanoma cells in vivo and in vitro. The results of the present study allowed our conclusion that MENK regulates the proliferation of B16 cells, causing cell cycle arrest in the G0/G1 phase and a decrease in the percentage of cells in the S and G2/M phases. Reverse transcription-quantitative polymerase chain reaction demonstrated that MENK increased opioid receptor expression in the B16 cells. Furthermore, the tumor volume and weight in the MENK-treated group were lower than those in the control group (NS) and MENK and naltrexone (NTX)-treated groups. MENK exerted both significant antitumor activity on the growth of B16 cells and a longer survival time in mice. The mice treated with MENK exhibited an increased ratio of CD4+ to CD8+ T cells as tested by flow cytometry (FCM), resulting in a ratio of 2.03 in the control group, 3.69 in the MENK-treated group, and 2.65 in the MENK and NTX group. Furthermore, a significant increase in plasma levels of IL-2, IFN-γ and TNF-α was revealed as assessed by ELISA. In conclusion, the results of the present study indicate that MENK has a cytotoxic effect on B16 melanoma cells in vitro and in vivo, and suggest a potential mechanism for these bioactivities. Therefore, we posit that MENK should be investigated, not only as a primary therapy for melanoma, but also as an adjuvant therapy in combination with chemotherapies.


Assuntos
Encefalina Metionina/administração & dosagem , Interferon gama/genética , Interleucina-2/genética , Melanoma Experimental/tratamento farmacológico , Fator de Necrose Tumoral alfa/genética , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Naltrexona/administração & dosagem , Carga Tumoral/efeitos dos fármacos
15.
Int Immunopharmacol ; 44: 61-71, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28088065

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Células Dendríticas/efeitos dos fármacos , Encefalina Metionina/uso terapêutico , Imunoterapia/métodos , Neoplasias/terapia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/imunologia , Feminino , Humanos , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neoplasias/imunologia , Transdução de Sinais/efeitos dos fármacos
16.
Mol Med Rep ; 14(6): 5521-5527, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27878237

RESUMO

Melanoma is an aggressive cancer, the incidence of which is increasing worldwide. Limited therapies are currently available, particularly following metastasis. The aim of the present study was to investigate the inhibiting effect of methionine enkephalin (MENK) on human melanoma via opioid receptors. The results of the present study revealed that MENK markedly regulates the proliferation of A375 cells, causing cell cycle arrest in G0/G1 phase and a decrease in the percentage of cells in S and G2/M phases. Reverse transcription­quantitative polymerase chain reaction demonstrated that MENK treatment increased opioid receptor expression in A375 cells. Furthermore, the expression level of survivin, an inhibitory apoptotic protein, was 1.1% of the level in the control group in the MENK group following 48 h of treatment. In conclusion, the results of the present study revealed, to the best of our knowledge for the first time, that MENK may inhibit growth and induce apoptosis of A375 cells, and describes a potential mechanism underlying these effects. Therefore, MENK should be investigated as a primary therapy for human melanoma cancer and as an adjuvant to other chemotherapies. Further studies are required to develop an optimal strategy for the use of MENK for the treatment of human cancers.


Assuntos
Antineoplásicos/farmacologia , Encefalina Metionina/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Expressão Gênica , Humanos , Melanoma/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Opioides/genética , Receptores Opioides/metabolismo
17.
Int Immunopharmacol ; 39: 397-402, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27561742

RESUMO

Previously it was confirmed that naltrexone, a non-peptide δ-opioid receptor selective antagonist is mainly used for alcoholic dependence and opioid addiction treatment. However, there is increasing data on immune regulation of low dose naltrexone (LDN). The aim of this work was to explore the effect of LDN on the phenotype and function of macrophage. The changes of macrophage after treatment with LDN were examined using flow cytometry (FCM); FITC-dextran phagocytosis and enzyme-linked immunosorbent assay (ELISA). We have found that LDN enhances function of macrophage as confirmed by up-regulating MHC II molecule and CD64 on macrophage while down-regulating CD206 expression. Furthermore the productions of TNF-α, IL-6, IL-1ß, increased significantly. Macrophages in LDN treated group performed the enhanced phagocytosis. Therefore it is concluded that LDN could promote function of macrophage and this work has provided concrete data of impact on immune system by LDN. Especially the data would support interaction between CD4+T cell and macrophage in AIDS treatment with LDN in Africa (LDN has already been approved in Nigeria for the use in AIDS treatment).


Assuntos
Alcoolismo/tratamento farmacológico , Fatores Imunológicos/farmacologia , Macrófagos/efeitos dos fármacos , Naltrexona/farmacologia , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Analgésicos Opioides/efeitos adversos , Animais , Células Cultivadas , Humanos , Interleucina-12/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Receptores de IgG/metabolismo , Receptores Opioides delta/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
18.
Int Immunopharmacol ; 37: 59-64, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26927200

RESUMO

Methionine enkephalin (MENK), an endogenous neuropeptide has a crucial role in both neuroendocrine and immune systems. MENK is believed to have an immunoregulatory activity to have cancer biotherapy activity by binding to the opioid receptors on immune and cancer cells. Clinical trial studies in cancer patients have shown that MENK activates immune cells directly and by inhibiting regulatory T-cells (Tregs). MENK may also change the tumor microenvironment by binding to opioid receptor on or in cancer cells. All of these mechanisms of action have biologic significance and potential for use in cancer immunotherapy. Furthermore, they reveal a relationship between the endocrine and immune systems. Due to the apparent role of MENK in cancer therapy we reviewed herein, the research undertaken with MENK in recent years; which has advanced our understanding of the role MENK has in cancer progression and its relationship to immunity, supporting MENK as a new strategy for cancer immunotherapy.


Assuntos
Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Sistema Imunitário/metabolismo , Imunoterapia/métodos , Neoplasias/terapia , Sistemas Neurossecretores/metabolismo , Receptores Opioides/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Ensaios Clínicos como Assunto , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Humanos , Sistema Imunitário/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Neoplasias/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo
19.
Cancer Biol Ther ; 16(3): 450-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25701137

RESUMO

Methionine enkephalin (MENK), an endogenous neuropeptide, plays an crucial role in both neuroendocrine and immune systems. CD4+Foxp3+ regulatory T cells (Tregs) are identified as a major subpopulation of T lymphocytes in suppressing immune system to keep balanced immunity. The aim of this research work was to elucidate the mechanisms via which MENK interacts with Tregs in cancer situation. The influence of MENK on transforming growth factor-ß (TGF-ß) mediated conversion from naïve CD4+CD25- T cells to CD4+CD25+ Tregs was determined and the data from flow cytometry (FCM) analysis indicated that MENK effectively inhibited the expression of Foxp3 during the process of TGF-ßinduction. Furthermore, this inhibiting process was accompanied by diminishing phosphorylation and nuclear translocation of Smad2/3, confirmed by western blot (WB) analysis and immunofluorescence (IF) at molecular level. We established sarcoma mice model with S180 to investigate whether MENK could modulate Tregs in tumor circumstance. Our findings showed that MENK delayed the development of tumor in S180 tumor bearing mice and down-regulated level of Tregs. Together, these novel findings reached a conclusion that MENK could inhibit Tregs activity directly and retard tumor development through down-regulating Tregs in mice. This work advances the deepening understanding of the influence of MENK on Tregs in cancer situation, and relation of MENK with immune system, supporting the implication of MENK as a new strategy for cancer immunotherapy.


Assuntos
Encefalina Metionina/administração & dosagem , Fatores de Transcrição Forkhead/metabolismo , Imunoterapia , Sarcoma/tratamento farmacológico , Linfócitos T Reguladores/metabolismo , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Humanos , Camundongos , Sarcoma/metabolismo , Sarcoma/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
20.
Hum Vaccin Immunother ; 10(7): 1836-40, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25424790

RESUMO

MENK, a penta-peptide is considered as being involved in the regulatory feedback loop between the immune and neuroendocrine systems, with marked modulation of various functions of human immune cells. The aim of the present work was to investigate change of lymphocyte subpopulations in peripheral blood of 50 cancer patients before and after treatment with MENK. Peripheral blood mononuclear cells (PBMCs) of peripheral blood from 50 cancer patients were isolated by density gradient centrifugation using Ficoll-Paque solution and cultured with MENK. We measured proliferation of total nucleated cells, subpopulations of individual CD4+T cells, CD8+T cells, CD4+CD25+ regulatory T cells (Treg), natural killer cells (NK) before and after treatment with 10(-12)M MENK in cell culture by flow cytometry (FCM). Our results indicated that MENK showed a strong inhibiting effect on Treg cells while it stimulated marked proliferation of other lymphocyte subpopulations. All data obtained were of significance statistically. It was therefore concluded that MENK could work as a strong immune booster with great potential in restoring damaged human immune system and we could consider MENK as a drug to treat cancer patients, whose immune systems are damaged by chemotherapy or radiotherapy. Furthermore we could consider MENK as a chemotherapy additive, which would sustain immune system of cancer patients during the process of chemotherapy to get maximized efficacy with minimized side effect.


Assuntos
Encefalina Metionina/administração & dosagem , Fatores Imunológicos/administração & dosagem , Subpopulações de Linfócitos/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Sanguíneas , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Humanos , Linfócitos T Reguladores/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...