Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 380
Filtrar
1.
Adv Neurobiol ; 35: 45-85, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38874718

RESUMO

It has become apparent that endogenous opioids act not only as neurotransmitters and neuromodulators, but have multiple functions in the body. Activation of the opioid system by opiate drugs is associated with a risk of cancer development through direct stimulation of tumor cell proliferation and through immunosuppression. In contrast, the endogenous peptide opioid [Met5]-enkephalin, now commonly referred to as Opioid Growth Factor (OGF), negatively regulates cell proliferation in a wide number of cells during development, homeostasis, and neoplasia. This action is mediated through the opioid growth factor receptor, originally designated the zeta (ζ) opioid receptor. Further, contrary to the traditional notion of opiates as immunosuppressive, endogenous OGF has been shown to possess a number of positive immunomodulatory properties and may provide a beneficial effect in cancer by augmenting the activity of cells involved in both innate and acquired immunity. Taken together, the evidence supports consideration of opioid peptides such as OGF as new strategies for cancer therapy.


Assuntos
Neoplasias , Receptores Opioides , Animais , Humanos , Proliferação de Células/efeitos dos fármacos , Encefalina Metionina/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Peptídeos Opioides/metabolismo , Receptores Opioides/metabolismo
2.
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
3.
Int J Mol Sci ; 23(19)2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36232883

RESUMO

Pain is a worldwide public health problem and its treatment is still a challenge since clinically available drugs do not completely reverse chronic painful states or induce undesirable effects. Crotalphine is a 14 amino acids synthetic peptide that induces a potent and long-lasting analgesic effect on acute and chronic pain models, peripherally mediated by the endogenous release of dynorphin A and the desensitization of the transient receptor potential ankyrin 1 (TRPA1) receptor. However, the effects of crotalphine on the central nervous system (CNS) and the signaling pathway have not been investigated. Thus, the central effect of crotalphine was evaluated on the partial sciatic nerve ligation (PSNL)-induced chronic neuropathic pain model. Crotalphine (100 µg/kg, p.o.)-induced analgesia on the 14th day after surgery lasting up to 24 h after administration. This effect was prevented by intrathecal administration of CB1 (AM251) or CB2 (AM630) cannabinoid receptor antagonists. Besides that, crotalphine-induced analgesia was reversed by CTOP, nor-BNI, and naltrindole, antagonists of mu, kappa, and delta-opioid receptors, respectively, and also by the specific antibodies for ß-endorphin, dynorphin-A, and met-enkephalin. Likewise, the analgesic effect of crotalphine was blocked by the intrathecal administration of minocycline, an inhibitor of microglial activation and proliferation. Additionally, crotalphine decreased the PSNL-induced IL-6 release in the spinal cord. Importantly, in vitro, crotalphine inhibited LPS-induced CD86 expression and upregulated CD206 expression in BV-2 cells, demonstrating a polarization of microglial cells towards the M2 phenotype. These results demonstrated that crotalphine, besides activating opioid and cannabinoid analgesic systems, impairs central neuroinflammation, confirming the neuromodulatory mechanism involved in the crotalphine analgesic effect.


Assuntos
Analgesia , Canabinoides , Neuralgia , Aminoácidos/metabolismo , Analgésicos/metabolismo , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Analgésicos Opioides/metabolismo , Anquirinas/metabolismo , Antagonistas de Receptores de Canabinoides/uso terapêutico , Canabinoides/uso terapêutico , Dinorfinas/metabolismo , Encefalina Metionina/metabolismo , Humanos , Interleucina-6/metabolismo , Lipopolissacarídeos/metabolismo , Microglia/metabolismo , Minociclina/uso terapêutico , Neuralgia/metabolismo , Peptídeos , Fenótipo , Receptores Opioides/metabolismo , Medula Espinal , beta-Endorfina/metabolismo
4.
Int Immunopharmacol ; 110: 108933, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35738090

RESUMO

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.


Assuntos
Carcinoma , Células Supressoras Mieloides , Neoplasias do Colo do Útero , Animais , Apoptose , Linhagem Celular Tumoral , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Encefalina Metionina/uso terapêutico , Feminino , Humanos , Fatores Imunológicos/metabolismo , Camundongos , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/metabolismo
5.
Int J Biol Sci ; 18(7): 2882-2897, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35541895

RESUMO

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.


Assuntos
Cardiotoxicidade , Doxorrubicina , Encefalina Metionina , Animais , Apoptose/efeitos dos fármacos , Cardiotoxicidade/etiologia , Cardiotoxicidade/metabolismo , Cardiotoxicidade/patologia , Proliferação de Células , Doxorrubicina/toxicidade , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Humanos , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Proteínas de Ligação a RNA/genética
6.
Int Immunopharmacol ; 99: 107713, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34426103

RESUMO

Methionine enkephalin (MENK) has an important role in both neuroendocrine and immune systems. MENK was known as an opioid growth factor (OGF) for its growth regulatory characteristics. OGF interacts with the OGF receptor (OGFr) to inhibit DNA synthesis by upregulating p16 and/or p21, which delays the cell cycle transition from G0/G1 to S phase, and inhibits cell proliferation. In addition, OGF combines with OGFr in immune cells to exert its immunomodulatory activity and regulate immune function. OGF has been studied as an immunomodulator in a variety of autoimmune diseases, including multiple sclerosis, inflammatory bowel disease, diabetes and viral infections, and has been proven to relieve symptoms of certain diseases in animal and in vitro experiments. Also, OGF and OGFr have various anti-tumor molecular mechanisms. OGF can be used as the primary therapy alone or combined with other drugs to treat tumors. This article summarizes the research progress of OGF in immune-related diseases and cancer diseases.


Assuntos
Encefalina Metionina/metabolismo , Doenças do Sistema Imunitário/metabolismo , Neoplasias/metabolismo , Animais , Humanos , Doenças do Sistema Imunitário/tratamento farmacológico , Antagonistas de Entorpecentes/uso terapêutico , Neoplasias/tratamento farmacológico
7.
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
8.
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
9.
Int Immunopharmacol ; 98: 107837, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34116288

RESUMO

The aim of this study was to investigate how methionine enkephalin (MENK) regulates the biological behavior of lung cancer cells and to further explore its anti-lung cancer mechanisms in vitro and in vivo. The results showed that MENK enhanced the expression of opioid receptor (OGFr) and induced apoptosis of lung cancer cells by activating the Bcl-1/Bax/caspase-3 signaling pathway in vitro and in vivo. However, the regulatory effects of MENK disappeared after blockade of the OGFr. This confirmed that a prerequisite for the anti-tumor action of MENK is binding to OGFr. Additionally, we observed that MENK treatment enhanced the immunogenicity of lung cancer by enhancing the exposure of calreticulin and high mobility group box 1, and increasing the expression of NKG2D ligands. Further studies showed that MENK treatment increased the expression of natural killer (NK) cell-related cytokines such as granzyme B and interferon-γ and NK cell activation. Thus, we concluded that MENK might inhibit the proliferation of lung cancer cells by activating the Bcl-2/Bax/caspase-3 signaling pathway and enhancing immunogenicity and NK cell-driven tumor immunity.


Assuntos
Antineoplásicos/metabolismo , Caspase 3/metabolismo , Encefalina Metionina/metabolismo , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Células A549 , Animais , Humanos , Imunidade Celular , Neoplasias Pulmonares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Transdução de Sinais
10.
Mol Med Rep ; 22(5): 4351-4359, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33000241

RESUMO

Endometriosis (EMS) is a common disease in women aged 25­45 years, and pain is the main clinical symptom. The primary clinical treatment is surgical excision and drug therapy targeting the ectopic lesions, but these have not been very effective. Botulinum neurotoxin serotype A (BTX­A) has been reported to be useful in the treatment of pain in a variety of diseases. Based on this, the aim of the present study was to explore the therapeutic effect and mechanism of BTX­A on EMS. A model of nerve injury induced by oxygen glucose deprivation (OGD) was constructed in PC12 cells and EMS mice. Model cells and mice were treated with different concentrations of BTX­A to observe the changes in pain behavior, to detect cell viability and the secretion of norepinephrine (NE) and methionine enkephalin (M­EK) in cells and the spinal cord, and to evaluate the expression of apoptosis­related molecules in spinal cord nerves. The results revealed that BTX­A significantly reduced the amount of writhing in model mice, enhanced the activity of PC12 OGD cells, increased the secretion of NE and M­EK in model cells and the spinal cord of mice, and decreased the apoptosis of neural cells in the spinal cord of the model mice. Therefore, it was hypothesized that BTX­A may alleviate the pain induced by EMS by increasing the secretion of analgesic substances and promoting the repair of nerve injury. The present study provided a theoretical basis for the treatment of pain induced by EMS.


Assuntos
Toxinas Botulínicas Tipo A/administração & dosagem , Endometriose/tratamento farmacológico , Encefalina Metionina/metabolismo , Norepinefrina/metabolismo , Dor/tratamento farmacológico , Adulto , Animais , Toxinas Botulínicas Tipo A/farmacologia , Sobrevivência Celular , Modelos Animais de Doenças , Endometriose/complicações , Endometriose/etiologia , Endometriose/metabolismo , Encefalina Metionina/líquido cefalorraquidiano , Feminino , Glucose/efeitos adversos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Norepinefrina/líquido cefalorraquidiano , Células PC12 , Dor/etiologia , Dor/metabolismo , Ratos
11.
Proc Natl Acad Sci U S A ; 117(34): 20696-20705, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32769209

RESUMO

Regulatory T (Treg) cells, expressing CD25 (interleukin-2 receptor α chain) and Foxp3 transcription factor, maintain immunological self-tolerance and suppress various immune responses. Here we report a feature of skin Treg cells expanded by ultraviolet B (UVB) exposure. We found that skin Treg cells possessing a healing function are expanded by UVB exposure with the expression of an endogenous opioid precursor, proenkephalin (PENK). Upon UVB exposure, skin Treg cells were expanded with a unique TCR repertoire. Also, they highly expressed a distinctive set of genes enriched in "wound healing involved in inflammatory responses" and the "neuropeptide signaling pathway," as indicated by the high expression of Penk. We found that not only was PENK expression at the protein level detected in the UVB-expanded skin Treg (UVB-skin Treg) cells, but that a PENK-derived neuropeptide, methionine enkephalin (Met-ENK), from Treg cells promoted the outgrowth of epidermal keratinocytes in an ex vivo skin explant assay. Notably, UVB-skin Treg cells also promoted wound healing in an in vivo wound closure assay. In addition, UVB-skin Treg cells produced amphiregulin (AREG), which plays a key role in Treg-mediated tissue repair. Identification of a unique function of PENK+ UVB-skin Treg cells provides a mechanism for maintaining skin homeostasis.


Assuntos
Encefalinas/metabolismo , Precursores de Proteínas/metabolismo , Linfócitos T Reguladores/metabolismo , Cicatrização/fisiologia , Anfirregulina/metabolismo , Animais , Células Cultivadas , Encefalina Metionina/metabolismo , Encefalinas/efeitos da radiação , Feminino , Homeostase/fisiologia , Humanos , Tolerância Imunológica/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Precursores de Proteínas/efeitos da radiação , Tolerância a Antígenos Próprios/imunologia , Pele/metabolismo , Raios Ultravioleta , Cicatrização/imunologia
12.
Int Immunopharmacol ; 75: 105723, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31408839

RESUMO

The standard treatments for neoplasia include surgery, chemotherapy, hormone antagonists and radiotherapy, which can prolong survival, but rarely cure the tumors of gynecological cancer patients. OGF - OGFr expression, in various gynecologic cells and tissues, is an intersection point between cell development, neuroendocrine function and immune modulation. It has been identified that OGF and OGFr expression differs between gynecological tumor and normal cells. Further, exogenous or endogenous OGF and OGFr antagonists have been known to have a role in regulating cell viability and apoptosis. Moreover, the expression of proteins in the OGF - OGFr axis modulate differentiation and membrane expression of immune cells, which can enhance the immune response. In vivo and in vitro assays have shown that OGF and OGFr antagonists inhibit mitosis as well as induce apoptosis in gynecologic cancer cells. Although immune augmentation combination therapies can intensify cytotoxic activity, OGF or OGFr antagonists do not increase toxicities associated with dual-immune regulation. In conclusion, the OGF - OGFr axis provides significant strategies for antitumor efficiency in gynecological cancer.


Assuntos
Encefalina Metionina/metabolismo , Neoplasias dos Genitais Femininos/metabolismo , Receptores Opioides/metabolismo , Animais , Feminino , Humanos
13.
Amino Acids ; 51(8): 1201-1207, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31302778

RESUMO

Rat sialorphin (Gln-His-Asn-Pro-Arg) is a natural blocker of neprilysin (NEP) that belongs to the family of endogenous opioid peptide-degrading enzymes. Studies have confirmed the efficiency of sialorphin in blocking the activity of NEP, both in vitro and in vivo. It has been demonstrated that this inhibitor has a strong analgesic, anti-inflammatory, immunological and metabolic effect either directly or indirectly by affecting the level of Met/Leu-enkephalins. In this work, sialorphin and their 12 analogues were synthesised using the solid-phase method. The effect of the peptides on the degradation of Met-enkephalin by NEP and metabolic degradation in human plasma was investigated in vitro. We show that the change in the N-terminal amino acid configuration from L to D in almost all peptides, except D-Arg-His-Asn-Pro-Arg (peptide XI), led to the abolition of their inhibitory activity. With molecular modelling technique we explained the structural properties of the L and D-arginine located on the N-terminal part of the peptide. The detailed analysis of the protein binding pocket allowed us to explain why D-arginine is so unique among all D residues. Peptide XI showed the highest stability among the tested peptides in human plasma. For instance sialorphin after a 2-hour incubation in human plasma was almost completely decomposed, while the level of peptide XI dropped to 45% after 48 h under these conditions.


Assuntos
Encefalina Metionina/metabolismo , Modelos Moleculares , Neprilisina/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Humanos , Técnicas In Vitro
14.
PLoS One ; 13(10): e0204830, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30286124

RESUMO

The objective of this study was to evaluate the effect of low-dose naltrexone (LDN) as a carboplatin chemotherapy-associated drug in female dogs with mammary carcinoma in benign mixed tumors (MC-BMT) after mastectomy and to assess its association with quality of life and survival rates. Sixty female dogs were included in this study, all of which had histopathological diagnosis of MC-BMT and were divided into three groups: G1 (control), consisting of animals submitted only to mastectomy with or without regional metastasis; G2, composed of treated animals that did not present with metastasis; and G3, treated dogs that presented with metastasis. G2 and G3 were also subdivided according to the treatment administered: chemotherapy alone (MC-BMT(-) C/MC-BMT(+) C) or LDN and chemotherapy (MC-BMT(-) C+LDN/MC-BMT(+) C+LDN). All animals were subjected to clinical evaluation, mastectomy, peripheral blood lymphocyte immunophenotyping, beta-endorphin and met-enkephalin quantification, and evaluation of survival rates and quality of life scores. The results showed higher serum concentrations of beta-endorphin and met-enkephalin, fewer chemotherapy-related side effects, and better quality of life and survival rates in the LDN-treated groups than in LDN-untreated groups (P < 0.05). Evaluation of clinical and pathological parameters indicated a significant association between the use of LDN and both prolonged survival and enhanced quality of life. These results indicate that LDN is a viable chemotherapy-associated treatment in female dogs with MC-BMT, maintaining their quality of life and prolonging survival rates.


Assuntos
Carboplatina/administração & dosagem , Carboplatina/farmacologia , Linfócitos/metabolismo , Neoplasias Mamárias Animais/tratamento farmacológico , Naltrexona/administração & dosagem , Animais , Cães , Sinergismo Farmacológico , Encefalina Metionina/metabolismo , Feminino , Imunofenotipagem , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/cirurgia , Mastectomia/veterinária , Naltrexona/farmacologia , Metástase Neoplásica , Qualidade de Vida , Análise de Sobrevida , Resultado do Tratamento , beta-Endorfina/metabolismo
15.
Amino Acids ; 50(8): 1083-1088, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29752565

RESUMO

Enkephalins are involved in a number of physiological processes. However, these peptides are quickly degraded by peptidases, e.g. the neutral endopeptidase (NEP). Inhibition of the enzymatic degradation of enkephalins is one of the possible approaches to prolong their activity. Selective inhibitor of NEP, sialorphin, is the attractive lead compound for enkephalins degradation studies. In this work, an alanine scan of sialorphin and a series of its hybrids with opiorphin, synthesised by the solid phase method, were performed. The effect of the peptides on degradation of Met-enkephalin by NEP in vitro was investigated. Molecular modelling technique was used to identify residues responsible for protein-ligand interactions. We showed that substitution of amino acids Gln1, Pro4 and Arg5 of sialorphin for Ala significantly reduced the half-life of Met-enkephalin in the presence of NEP. [Ala3]sialorphin displayed a higher inhibitory potency against NEP than sialorphin. Substitution of His2 for Ala led to a compound which was as active as lead compound. Sialorphin has a structure which hardly tolerates substitution in its sequence at positions 1, 4 and 5. The conversion of His2 for alanine in sialorphin is tolerated very well. The higher inhibitory potency of [Ala3]sialorphin than sialorphin against NEP is caused by removal of the hydrophilic residue (Asn) and a better fit of the peptide to the enzyme-binding pocket. The role of side chains of sialorphin in degradation of enkephalin by NEP has been explored. This study also provides an important SAR information essential for further drug design.


Assuntos
Alanina/química , Encefalina Metionina/metabolismo , Encefalinas/metabolismo , Neprilisina/antagonistas & inibidores , Oligopeptídeos/farmacologia , Peptídeos/farmacologia , Proteínas e Peptídeos Salivares/farmacologia , Acetilação , Sequência de Aminoácidos , Encefalinas/química , Modelos Moleculares , Oligopeptídeos/química , Peptídeos/química , Ligação Proteica , Conformação Proteica , Proteólise/efeitos dos fármacos , Proteínas e Peptídeos Salivares/química , Técnicas de Síntese em Fase Sólida
16.
Int Immunopharmacol ; 56: 1-8, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29324390

RESUMO

The aim of the work was to investigate the effect and possible mechanism of MENK on the growth of rat C6 glioma in vivo or in vitro. Our findings showed that MENK could inhibit the growth of rat C6 glioma, prolong median survival times in tumor-bearing rats, and induce glioma cell apoptosis. Moreover, MENK could increase the activities of caspase-3, caspase-8 and caspase-9. It also increased the expression of Fas, FasL, Bax, while decreased the expression of Bcl-2. We further confirmed that MENK could increase opioid receptors MOR and DOR expressions, Ca2+ influx into the cytoplasm, and a substantial increase of NFAT1accumulation in the nuclei in C6 glioma cell. When we specifically knocked down NFAT1, there was no effect of MENK on the cell viability and FasL up-regulation in NFAT1 knocked-down cell. These results demonstrate that MENK could bind to opioid receptors MOR and DOR on C6 glioma cells and trigger a Ca2+ influx into the cytoplasm, resulting in translocation of NFAT1 into the nucleus. The hyper-activation of NFAT1 may regulate transcription of downstream gene, such as FasL, and induce apoptosis of rat C6 glioma cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Encefalina Metionina/metabolismo , Glioma/metabolismo , Fatores de Transcrição NFATC/metabolismo , Animais , Apoptose , Neoplasias Encefálicas/genética , Sinalização do Cálcio , Caspases/metabolismo , Linhagem Celular Tumoral , Proteína Ligante Fas/metabolismo , Glioma/genética , Fatores de Transcrição NFATC/genética , Transplante de Neoplasias , RNA Interferente Pequeno/genética , Ratos , Ratos Wistar , Ativação Transcricional , Regulação para Cima , Proteína X Associada a bcl-2/metabolismo
17.
Photodermatol Photoimmunol Photomed ; 33(2): 84-91, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28039905

RESUMO

BACKGROUND: We previously reported that ultraviolet (UV) A eye irradiation reduces the ulcerative colitis induced by dextran sodium sulfate (DSS). This study examined the effects of UVA on colon carcinoma induced by azoxymethane (AOM) and DSS. METHODS: We irradiated the eyes of ICR mice with UVA at a dose of 110 kJ/m2 using an FL20SBLB-A lamp for the experimental period. RESULTS: In mice treated with these drugs, the symptom of colon carcinoma was reduced by UVA eye irradiation. The levels of interleukin (IL)-6 and tumor necrosis factor (TNF)-α in the blood were increased in AOM + DSS-treated mice; however, those levels were reduced by UVA eye irradiation. The expression of ß-endorphin, methionine-enkephalin (OGF), µ-opioid receptor, and opioid growth factor receptor (OGFR) of the colon was increased in the AOM + DSS-treated mice, and these levels were increased further following UVA eye irradiation. When ß-endorphin inhibitor was administered, the ameliorative effect of UVA eye irradiation was reduced, and the effect of eye irradiation disappeared entirely following the administration of naltrexone (inhibitor of both opioid receptor and OGFR). CONCLUSIONS: These results suggested that UVA eye irradiation exerts major effects on AOM + DSS-induced colon carcinoma.


Assuntos
Colo/metabolismo , Neoplasias do Colo/metabolismo , Encefalina Metionina/metabolismo , Olho/efeitos da radiação , Raios Ultravioleta , beta-Endorfina/metabolismo , Aminoquinolinas/farmacologia , Animais , Azoximetano , Benzamidas/farmacologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia , Sulfato de Dextrana , Feminino , Interleucina-6/sangue , Antígeno Ki-67/metabolismo , Camundongos Endogâmicos ICR , Naltrexona/farmacologia , Receptores Opioides/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo , Fator de Necrose Tumoral alfa/sangue , beta-Endorfina/antagonistas & inibidores
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.
J Orthop Res ; 34(7): 1195-205, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26687326

RESUMO

Chronic opioid therapy is associated with bone loss. This led us to hypothesize that the opioid antagonists, that include naloxone, would stimulate bone formation by regulating MSC differentiation. The opioid growth factor receptor (OGFR) is a non-canonical opioid receptor that binds naloxone with high affinity whereas the native opioid growth factor, met5-enkephalin (met5), binds both the OGFR and the canonical delta opioid receptor (OPRD). Naloxone and an shRNA OGFR lentivirus were employed to disrupt the OGFR-signaling axis in cultured MSC. In parallel, naloxone was administered to bone marrow using a mouse unicortical defect model. OPRD, OGFR, and the met5-ligand were highly expressed in MSC and osteoblasts. A pulse-dose of naloxone increased mineral formation in MSC cultures in contrast to MSC treated with continuous naloxone or OGFR deficient MSC. Importantly, SMAD1 and SMAD8/9 expression increased after a pulse dose of naloxone whereas SMAD1, SMAD7, and ID1 were increased in the OGFR deficient MSC. Inhibited OGFR signaling decreased proliferation and increased p21 expression. The addition of naloxone to the unicortical defect resulted in increased bone formation within the defect. Our data suggest that novel mechanism through which signaling through the OGFR regulates osteogenesis via negative regulation of SMAD1 and p21. © 2015 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1195-1205, 2016.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Osteoblastos , Animais , Células Cultivadas , Encefalina Metionina/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Receptores Opioides/metabolismo , Proteínas Smad Reguladas por Receptor/metabolismo , Quinases Ativadas por p21/metabolismo
20.
J Pharmacol Sci ; 125(2): 117-24, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24882143

RESUMO

Nicotine (NIC) is an exogenous ligand of the nicotinic acetylcholine receptor (nAChR), and it influences various functions in the central nervous system. Systemic administration of NIC elicits the release of endogenous opioids (endorphins, enkephalins, and dynorphins) in the supraspinal cord. Additionally, systemic NIC administration induces the release of methionine-enkephalin in the spinal dorsal horn. NIC has acute neurophysiological actions, including antinociceptive effects, and the ability to activate the hypothalamic-pituitary-adrenal (HPA) axis. The endogenous opioid system participates in NIC-induced antinociception, but not HPA axis activation. Moreover, NIC-induced antinociception is mediated by α4ß2 and α7 nAChRs, while NIC-induced HPA axis activation is mediated by α4ß2, not α7, suggesting that the effects of NIC on the endogenous opioid system are mediated by α7, not α4ß2. NIC has substantial physical dependence liability. The opioid-receptor antagonist naloxone (NLX) elicits NIC withdrawal after repeated NIC administration, and NLX-induced NIC withdrawal is inhibited by concomitant administration of an opioid-receptor antagonist. NLX-induced NIC withdrawal is also inhibited by concomitant administration of an α7 antagonist, but not an α4ß2 antagonist. Taken together, these findings suggest that NIC-induced antinociception and the development of physical dependence are mediated by the endogenous opioid system, via the α7 nAChR.


Assuntos
Analgésicos , Nicotina/farmacologia , Peptídeos Opioides/fisiologia , Tabagismo/etiologia , Receptor Nicotínico de Acetilcolina alfa7/fisiologia , Animais , Encefalina Metionina/metabolismo , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Naloxona/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Nicotina/efeitos adversos , Peptídeos Opioides/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Medula Espinal/metabolismo , Síndrome de Abstinência a Substâncias , Tabagismo/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA