Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
Proteomics ; 18(7): e1700408, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29406625

RESUMO

Adverse life experiences increase the lifetime risk to several stress-related psychopathologies, such as anxiety or depressive-like symptoms following stress in adulthood. However, the neurochemical modulations triggered by stress have not been fully characterized. Neuropeptides play an important role as signaling molecules that contribute to physiological regulation and have been linked to neurological and psychiatric diseases. However, little is known about the influence of stress on neuropeptide regulation in the brain. Here, we have performed an exploratory study of how neuropeptide expression at adulthood is modulated by experiencing a period of multiple stressful experiences. We have targeted hippocampus and prefrontal cortex (PFC) brain areas, which have previously been shown to be modulated by stressors, employing a targeted liquid chromatography-mass spectrometry (LC-MS) based approach that permits broad peptide coverage with high sensitivity. We found that in the hippocampus, Met-enkephalin, Met-enkephalin-Arg-Phe, and Met-enkephalin-Arg-Gly-Leu were upregulated, while Leu-enkephalin and Little SAAS were downregulated after stress. In the PFC area, Met-enkephalin-Arg-Phe, Met-enkephalin-Arg-Gly-Leu, peptide PHI-27, somatostatin-28 (AA1-12), and Little SAAS were all downregulated. This systematic evaluation of neuropeptide alterations in the hippocampus and PFC suggests that stressors impact neuropeptides and that neuropeptide regulation is brain-area specific. These findings suggest several potential peptide candidates, which warrant further investigations in terms of correlation with depression-associated behaviors.


Assuntos
Regulação da Expressão Gênica , Hipocampo/metabolismo , Neuropeptídeos/genética , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/metabolismo , Animais , Cromatografia Líquida , Encefalina Metionina/genética , Hipocampo/fisiologia , Masculino , Espectrometria de Massas , Córtex Pré-Frontal/fisiologia , Proteômica , Ratos , Somatostatina-28/genética , Estresse Psicológico/genética
4.
Sci Rep ; 6: 32799, 2016 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-27605249

RESUMO

Neuropathic pain often results from peripheral nerve damage, which can involve immune response. Local leukocyte-derived opioid peptides or exogenous opioid agonists inhibit neuropathy-induced mechanical hypersensitivity in animal models. Since neuropathic pain can also be augmented by heat, in this study we investigated the role of opioids in the modulation of neuropathy-evoked heat hypersensitivity. We used a chronic constriction injury of the sciatic nerve in wild-type and opioid peptide-knockout mice, and tested opioid effects in heat and mechanical hypersensitivity using Hargreaves and von Frey tests, respectively. We found that although perineural exogenous opioid agonists, including peptidergic ligands, were effective, the endogenous opioid peptides ß-endorphin, Met-enkephalin and dynorphin A did not alleviate heat hypersensitivity. Specifically, corticotropin-releasing factor, an agent triggering opioid peptide secretion from leukocytes, applied perineurally did not attenuate heat hypersensitivity in wild-type mice. Exogenous opioids, also shown to release opioid peptides via activation of leukocyte opioid receptors, were equally analgesic in wild-type and opioid peptide-knockout mice, indicating that endogenous opioids do not contribute to exogenous opioid analgesia in heat hypersensitivity. Furthermore, exogenously applied opioid peptides were ineffective as well. Conversely, opioid peptides relieved mechanical hypersensitivity. Thus, both opioid type and sensory modality may determine the outcome of neuropathic pain treatment.


Assuntos
Analgésicos Opioides/farmacologia , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Peptídeos Opioides/metabolismo , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Hormônio Liberador da Corticotropina/farmacologia , Dinorfinas/genética , Dinorfinas/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Encefalina Metionina/genética , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Temperatura Alta/efeitos adversos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuralgia/genética , Peptídeos Opioides/genética , Sistema Nervoso Periférico/metabolismo , Receptores Opioides/agonistas , beta-Endorfina/genética , beta-Endorfina/metabolismo , beta-Endorfina/farmacologia
5.
Zhongguo Zhen Jiu ; 35(9): 917-21, 2015 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-26721148

RESUMO

OBJECTIVE: To observe the analgesic effect of electroacupuncture (EA) on collagen-induced arthritis (CIA) rats and its regulating effect on inflammation reaction and the endogenous opioid system of synovial tissues. Methods A total of 30 healthy male Wistar rats were randomly divided into a control group, a model group and an EA group, 10 rats in each one. The chronic pain model of CIA rats was made by cattle type-II collagen in the model group and EA group. Rats in the EA group were treated with EA at "Zusanli" (ST 36) and "Kunlun" (BL 60) for 30 min from 16th day after model establishment, once a day for consecutive 10 days. Rats in the control group did not receive any treatment. Rats in the model group were treated with fixation as the EA group. Threshold of pain, arthritis index, paw swelling were measured before model establishment and 16 d, 20 d, 23 d and 25 d after model establishment. The levels of beta-endorphin (ß-END), met-enkephalin (met-ENK), dynorphin A (Dyn A) were measured by radioimmunoassay; the mRNA expressions of mu opioid receptor (MOR), kappa opioid receptor (KOR) and delta opioid receptor (DOR) in synovial tissues of CIA rats were detected by I quantitative polymerase chain reaction (qPCR). RESULTS: Compared with the control group, threshold of pain was reduced (all P<0. 01), arthritis index was increased (all P<0. 01) and paw swelling was increased (all P<0. 01) in the model group on the 16th day, 20th day, 23rd day, 25th day after model establishment. Compared with the model group, the threshold of pain was increased in the EA group (all P<0. 01), arthritis index and paw swelling were reduced (all P<0. 01) on the 23rd day and 25th day after model establishment. Compared with the control group, the level of Dyn A in synovial tissues of CIA rats was increased in the model group (P<0. 01); the mRNA expressions of MOR, KOR and DOR were down-regulated lower than 0. 5 fold of normal level. Compared with the model group, the level of ß-END in synovial tissues of the knee joint was increased in the EA group (P<0. 05), and the mRNA expressions of MOR, KOR and DOR in synovial tissues of CIA rats were up-regulated more than 2 folds of normal level. CONCLUSION: The intervention of EA on chronic pain of CIA rats is superior, which is likely to be related with effects of EA on anti-inflammation and up-regulation of synovial tissue ß-END and MOR, KOR, DOR.


Assuntos
Analgésicos Opioides/imunologia , Artrite Reumatoide/terapia , Dor Crônica/terapia , Eletroacupuntura , Líquido Sinovial/imunologia , Analgesia por Acupuntura , Pontos de Acupuntura , Animais , Artrite Reumatoide/imunologia , Bovinos , Dor Crônica/imunologia , Dinorfinas/genética , Dinorfinas/imunologia , Encefalina Metionina/genética , Encefalina Metionina/imunologia , Humanos , Masculino , Ratos , Ratos Wistar , Receptores Opioides mu/genética , Receptores Opioides mu/imunologia , beta-Endorfina/genética , beta-Endorfina/imunologia
6.
Dev Neurosci ; 35(4): 326-37, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23838073

RESUMO

Approximately 500,000 infants are born prematurely each year in the United States. These infants typically require an extensive stay in the neonatal intensive care unit (NICU), where they experience on average 14 painful and invasive procedures each day. These procedures, including repeated heel lance, insertion of intravenous lines, and respiratory and gastric suctioning, typically result in an inflammatory response, inducing pain and stress in the newborn. Remarkably, the majority of these procedures are performed in the complete absence of pre- or post-emptive analgesics. Recent clinical studies report that former NICU patients have increased thresholds for pain and stress later in life as compared with term-born infants. However, to date, the mechanisms whereby early-life inflammation alters later-life response to stress and pain are not known. The present studies were conducted to determine if neonatal injury impairs adult responses to anxiety- and stress-provoking stimuli. As we have previously reported that early-life pain results in a significant increase in opioid peptide expression within the midbrain periaqueductal gray, the role of endogenous opioids in our behavioral studies was also examined. Male and female rats received an intraplantar injection of the inflammatory agent carrageenan (1%) on the day of birth. In adulthood, animals were assessed for changes in response to anxiety- and stress-provoking stimuli using the open field and forced swim tests, respectively. Injury-induced changes in sucrose preference and stress-induced analgesia were also assessed. As adults, neonatally injured animals displayed a blunted response to both anxiety- and stress-provoking stimuli, as indicated by significantly more time spent in the inner area of the open field and a 2-fold increase in latency to immobility in the forced swim test as compared to controls. No change in sucrose preference was observed. Using in situ hybridization and immunohistochemistry, we observed a 2-fold increase in enkephalin mRNA and protein expression, respectively, in stress-related brain regions including the central amygdala and lateral septum. Administration of the opioid receptor antagonist naloxone reversed the attenuated responses to forced swim stress and stress-induced analgesia, suggesting the changes in stress-related behavior were opioid-dependent. Together, these data contribute to mounting evidence that neonatal injury in the absence of analgesics has adverse effects that are both long-term and polysystemic.


Assuntos
Animais Recém-Nascidos/fisiologia , Estresse Psicológico/psicologia , Tonsila do Cerebelo/fisiologia , Analgesia , Anedonia , Animais , Ansiedade/psicologia , Comportamento Animal/efeitos dos fármacos , Densitometria , Encefalina Metionina/biossíntese , Encefalina Metionina/genética , Encefalinas/biossíntese , Encefalinas/genética , Feminino , Preferências Alimentares/efeitos dos fármacos , Imuno-Histoquímica , Hibridização In Situ , Peptídeos Opioides/fisiologia , Substância Cinzenta Periaquedutal/fisiologia , Gravidez , Precursores de Proteínas/biossíntese , Precursores de Proteínas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Septo do Cérebro/fisiologia , Estresse Psicológico/fisiopatologia , Sacarose/farmacologia , Natação/psicologia
7.
Brain Res Bull ; 90: 43-51, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22985690

RESUMO

Astrocytes become activated and proliferate in response to autoimmune diseases involving the CNS. In many cases, elevated numbers of astrocytes may compound inflammatory responses and exacerbate neuronal degeneration. The regulation of astrocytes in disease states has been shown as an important corollary to disease progression and recovery. This study explores the role of the opioid growth factor (OGF)-OGF receptor (OGFr) axis in the proliferation of primary cultures of mouse cerebral astrocytes, as well as in spinal cord astrocytes of mice with experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis. OGF was found to inhibit purified cultures of cortical astrocytes in a dose-related, receptor-mediated, and reversible manner. Examination of a number of endogenous opioid peptides supported that OGF was the most effective inhibitor of astrocyte proliferation and that endogenous production of this peptide was neutralized by OGF antibodies. The specificity and requirement for OGFr to be present and functional was indicated by studies using siRNA technology to knockdown the classical µ, δ, and κ opioid receptors, as well as OGFr. Only knockdown of OGFr resulted in changes in astrocyte cell number with OGF rendered ineffective without the presence of OGFr. Astrocyte migration as studied by a scratch model, as well as apoptosis and necrosis pathways, were not altered by OGF treatment. However, BrdU incorporation during DNA synthesis phases of the cell cycle was significantly repressed in activated cultured astrocytes by OGF exposure. Overall activation of the astrocytes measured by nitric oxide levels was reduced, but proportional to the reduction in cell number. Examination of spinal cord tissues from mice with EAE revealed fewer astrocytes within 10 days of OGF treatment, with the mechanism of cell number reduction being repression of DNA synthesis. In conclusion, these studies delineate a novel role for OGF in the proliferation of mouse cortical astrocytes in vitro and spinal cord astrocytes in vivo, and support the use of OGF as a therapy to inhibit astrogliosis within a broad spectrum of autoimmune diseases.


Assuntos
Astrócitos/fisiologia , Proliferação de Células , Encefalomielite Autoimune Experimental/patologia , Encefalina Metionina/metabolismo , Receptores Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Astrócitos/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/patologia , Modelos Animais de Doenças , Interações Medicamentosas , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/complicações , D-Penicilina (2,5)-Encefalina/farmacologia , Encefalina Metionina/genética , Encefalina Metionina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito/química , Antagonistas de Entorpecentes/farmacologia , Necrose/etiologia , Necrose/metabolismo , Necrose/patologia , Óxido Nítrico/metabolismo , Peptídeos/toxicidade , RNA Interferente Pequeno/farmacologia , Receptores Opioides/genética , Fatores de Tempo
8.
Neuroscience ; 178: 56-67, 2011 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21241776

RESUMO

The heptapeptide Met-enkephalin-Arg6-Phe7 (MERF) with the sequence of YGGFMRF is a potent endogenous opioid located at the C-terminus of proenkephalin-A (PENK), the common polypeptide precursor of Met- and Leu-enkephalin. Our systematic bioinformatic survey revealed considerable sequence polymorphism at the heptapeptide region of different PENK prepropeptides among 56 vertebrate animals. Four orthologous heptapeptides with single or double amino acid replacements were identified among 15 animals, such as YGGFMGY (zebrafish), YGGFMRY (newt), YGGFMKF (hedgehog tenrek) and YGGFMRI (mudpuppy). Each novel heptapeptide, together with the mammalian consensus MERF and Met-enkephalin, were chemically synthesized and subjected to functionality studies, using radioligand binding competition and G-protein activation assays in rat brain membranes. Equilibrium binding affinities changed from good to modest as measured by receptor type selective [3H]opioid radioligands. The relative affinities of the heptapeptides reveal slight mu-receptor (MOP) preference over the delta-receptors (DOP). [35S]GTPγS assay, which measures the agonist-mediated G-protein activation, has demonstrated that all the novel heptapeptides were also potent in stimulating the regulatory G-proteins. All peptides were effective in promoting the agonist induced internalization of the green fluorescence protein-tagged human mu-opioid receptor (hMOP-EGFP) stably expressed in HEK293 cells. Thus, the C-terminally processed PENK heptapeptide orthologs exhibited satisfactory bioactivities, moreover they represent further members of the so-called "natural combinatorial neuropeptide library" emerged by evolution.


Assuntos
Encefalina Metionina/análogos & derivados , Encefalinas/genética , Oligopeptídeos/genética , Fragmentos de Peptídeos/genética , Filogenia , Receptores Opioides/agonistas , Animais , Encefalina Metionina/genética , Encefalina Metionina/metabolismo , Cobaias , Células HEK293 , Humanos , Oligopeptídeos/farmacologia , Polimorfismo Genético , Ensaio Radioligante/métodos , Ratos , Ratos Wistar , Receptores Opioides/metabolismo , Análise de Sequência/métodos , Radioisótopos de Enxofre/metabolismo , Vertebrados/genética , Vertebrados/metabolismo
9.
Neuropeptides ; 43(5): 355-62, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19716174

RESUMO

Endogenous peptide, Met-enkephalin-Arg-Phe (Tyr-Gly-Gly-Phe-Met-Arg-Phe; MERF) induces effects like antinociception, inhibit contraction of guinea pig ileum, mouse vas deferens and anti-tussive action. However, results regarding its functional efficiency and selectivity are controversial. Therefore, present study was undertaken to investigate whether MERF on systemic (intra-peritoneal, i.p.) route of administration induce any antinociception or not; to scrutinize the effect of 6 days chronic i.p. treatment of MERF on expression of mu (MOR1), delta (DOR1) and kappa (KOR1) opioid receptors; and finally, the antinociceptive effect of two synthetic peptides, MERFamide and (D-Ala(2))-MERFamide was compared with MERF on intracerebroventricular administration in order to understand the role of FMRF moiety in analgesic effect of MERF. Pharmacological results revealed that only 68.4 and 91.2 micromol/kg dose induce significant antinociception among various doses. Further, on 6 days chronic treatment, MERF induced significant antinociception in comparison to saline. Differential expression of MOR1 and KOR1 showed continuous up-regulation throughout the treatment whereas DOR1 showed down-regulation in initial 3 days followed by subsequently up-regulation during the latter observable period. Moreover, variation in opioid receptors expression had not affected the MERF antinociception. In conclusion, present study discursively demonstrates that MERF during chronic treatment interacts with all three opioid receptors (mu, delta and kappa) in rats and differently regulates their expression. Further, the interaction was such that the induction was mainly observed at molecular/expression level and not at pharmacological level to affect antinociception.


Assuntos
Analgésicos Opioides/metabolismo , Encefalina Metionina/metabolismo , Receptores Opioides/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Encefalina Metionina/administração & dosagem , Encefalina Metionina/genética , Humanos , Masculino , Camundongos , Medição da Dor , Ratos , Ratos Wistar , Receptores Opioides/genética
11.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 40(6): 1053-5, 2009 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-20067118

RESUMO

OBJECTIVE: To investigate the Methionine Enkephalin (M-Enk) expression in lamina II of cat spinal cord and the atypical complex terminal (ACT) after complete dorsal rhizotomy. METHODS: 10 male adult cats were divided into 2 groups after carrying out the complete dorsal rhizotomy: acute group, which survived 2 weeks; chronic group, which survived 2 months. At maturity, all cats were perfused after anaesthesia, the L6 segments of the spinal cord were removed and prepared for light and electron microscopy of M-Enk immunohistochemistry and for observing of ACT. RESULTS: M-Enk immuno-reactivity can be seen in laminal II lateral sides both in operation and control sides in acute group. They were both decreased in chronic group on the photos by naked eyes, and little intensive in operation side on the photos. The immuno-positive ACTs have been founded in the Lamina II of chronic operation side beside some M-Enk immunopositive simple terminals, they are round or ellipse in shape and usually form flat or convex two synapse with two post-compounds. ACTs contained both clear vesicles and dense core vesicles. CONCLUSION: M-Enk express in operation side after complete dorsal rhizotomy is mainly on the lateral side in lamina II. The M-Enk immuno-positive ACT may imply the mechanism of spinal cord plasticity.


Assuntos
Encefalina Metionina/metabolismo , Gânglios Espinais/metabolismo , Plasticidade Neuronal/fisiologia , Medula Espinal/fisiologia , Animais , Gatos , Encefalina Metionina/genética , Gânglios Espinais/cirurgia , Masculino , Rizotomia , Medula Espinal/metabolismo
12.
Mol Biol Cell ; 20(1): 319-27, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18923142

RESUMO

Opioid growth factor (OGF) is an endogenous opioid peptide ([Met(5)]enkephalin) that interacts with the OGF receptor (OGFr) and serves as a tonically active negative growth factor in cell proliferation of normal cells. To clarify the mechanism by which OGF inhibits cell replication in normal cells, we investigated the effect of the OGF-OGFr axis on cell cycle activity in human umbilical vein endothelial cells (HUVECs) and human epidermal keratinocytes (NHEKs). OGF markedly depressed cell proliferation of both cell lines by up to 40% of sterile water controls. Peptide treatment induced cyclin-dependent kinase inhibitor (CKI) p16(INK4a) protein expression and p21(WAF1/CIP1) protein expression in HUVECs and NHEKs, but had no effect on p15, p18, p19, or p27 protein expression in either cell type. Inhibition of either p16(INK4a) or p21(WAF1/CIP1) activation by specific siRNAs blocked OGF inhibitory action. Human dermal fibroblasts and mesenchymal stem cells also showed a similar dependence of OGF action on p16(INK4a) and p21(WAF1/CIP1). Collectively, these results indicate that both p16(INK4a) and p21(WAF1/CIP1) are required for the OGF-OGFr axis to inhibit cell proliferation in normal cells.


Assuntos
Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Encefalina Metionina/metabolismo , Neurotransmissores/metabolismo , Receptores Opioides/metabolismo , Transdução de Sinais/fisiologia , Animais , Ciclo Celular/fisiologia , Linhagem Celular , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Encefalina Metionina/genética , Humanos , Queratinócitos/citologia , Queratinócitos/fisiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Opioides/genética
13.
Peptides ; 29(12): 2266-75, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18930087

RESUMO

Chimeric peptide of Met-enkephalin and FMRFa (YGGFMKKKFMRFa-YFa), a kappa-opioid receptor specific peptide, did not induce tolerance and cross-tolerance effects to its analgesic action on day 5 after pretreatment with either YFa or morphine for 4 days. However, pretreatment with YFa for 4 days led to the development of cross-tolerance to the analgesic effects of morphine and also 4 days of pretreatment of morphine resulted in the expression of tolerance to its own analgesic effects. Similar expression of tolerance and cross-tolerance were also observed when YFa was compared with the kappa receptor agonist peptide dynorphin A(1-13) [DynA(1-13)]. Cross-tolerance effects between YFa and DynA(1-13) analgesia were also not observed on day 5. Interestingly, when YFa and DynA(1-13) were tested for their analgesic effects for 5 days, reduction in analgesia on day 3 was observed in case of DynA(1-13) whereas YFa maintained its analgesia for 5 days. Thus, chimeric peptide YFa may serve as a useful probe to understand pain modulation and expression of tolerance and cross-tolerance behavior with other opioids.


Assuntos
Analgésicos/farmacologia , Tolerância a Medicamentos , Encefalina Metionina/genética , Morfina/farmacologia , Oligopeptídeos/genética , Proteínas Recombinantes de Fusão/farmacologia , Analgesia/métodos , Analgésicos/administração & dosagem , Animais , Dinorfinas/administração & dosagem , Dinorfinas/farmacologia , Masculino , Camundongos , Morfina/administração & dosagem , Medição da Dor/métodos , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética
14.
J Comput Chem ; 27(11): 1177-95, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16752367

RESUMO

True ab initio prediction of protein 3D structure requires only the protein primary structure, a physicochemical free energy model, and a search method for identifying the free energy global minimum. Various characteristics of evolutionary algorithms (EAs) mean they are in principle well suited to the latter. Studies to date have been less than encouraging, however. This is because of the limited consideration given to EA design and control parameter issues. A comprehensive study of these issues was, therefore, undertaken for ab initio protein fold prediction using a full atomistic protein model. The performance and optimal control parameter settings of twelve EA designs where first established using a 15-residue polyalanine molecule-design aspects varied include the encoding alphabet, crossover operator, and replacement strategy. It can be concluded that real encoding and multipoint crossover are superior, while both generational and steady-state replacement strategies have merits. The scaling between the optimal control parameter settings and polyalanine size was also identified for both generational and steady-state designs based on real encoding and multipoint crossover. Application of the steady-state design to met-enkephalin indicated that these scalings are potentially transferable to real proteins. Comparison of the performance of the steady state design for met-enkephalin with other ab initio methods indicates that EAs can be competitive provided the correct design and control parameter values are used.


Assuntos
Algoritmos , Simulação por Computador , Encefalina Metionina/metabolismo , Evolução Molecular , Peptídeos/metabolismo , Dobramento de Proteína , Cromossomos/genética , Encefalina Metionina/química , Encefalina Metionina/genética , Modelos Moleculares , Mutação/genética , Peptídeos/química , Peptídeos/genética , Probabilidade , Estrutura Terciária de Proteína
15.
Cell Transplant ; 15(3): 225-30, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16719057

RESUMO

We have reported that transplantation of adrenal medullary chromaffin cells that release endogenous opioid peptides into pain modulatory regions in the CNS produce significant antinociceptive effects in patients with terminal cancer pain. However, the usefulness of this procedure is minimal because the availability of human adrenal tissue is very limited. Alternative xenogeneic materials, such as porcine and bovine adrenal chromaffin cells present problems of immune rejection and possible pathogenic contamination. In an attempt to develop opioid peptide-producing cells of autologous origin, we have transfected human mesenchymal stem cells (hMeSCs) with a mammalian expression vector containing a fusion gene of green fluorescent protein (GFP) and human preproenkephalin (hPPE), a precursor protein for enkephalin opioid peptides. Enkephalins are major neurotransmitters that play an important role in analgesia by activating peripheral opioid receptors. Following the establishment of stable transfection of hMeSCs, the expressions of hPPE and GFP were confirmed and the production of methionine enkephalin (Met-enkephalin) was significantly increased compared to control naive hMeSCs (p < 0.05). Our in vitro data demonstrated that genetically engineered hMeSCs with transfected hPPE gene can constitutively produce opioid peptide Met-enkephalin at an augmented high level. hMeSCs are relatively easy to isolate from a patient's bone marrow aspirates and expand in culture by repeated passages. Autologous hMeSCs would not require immunosuppression when transplanted back into the same patient. Through targeted gene manipulation such as hPPE gene transfection, this may offer a virtually unlimited safe cell supply for the treatment of opioid-sensitive pain in humans.


Assuntos
Analgésicos/metabolismo , Encefalina Metionina/genética , Encefalina Metionina/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Analgésicos/uso terapêutico , Analgésicos Opioides/uso terapêutico , Proliferação de Células , Células Cultivadas , DNA/genética , Encefalinas/genética , Regulação da Expressão Gênica/genética , Fusão Gênica , Engenharia Genética , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Humanos , Células-Tronco Mesenquimais/citologia , Dor/tratamento farmacológico , Precursores de Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
16.
J Hepatol ; 44(6): 1141-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16466825

RESUMO

BACKGROUND/AIMS: Cholestasis is associated with naloxone reversible antinociception and opiate receptor antagonists are used clinically to treat pruritus. Pain and pruritus are closely interrelated and opioids modulate both sensations. Therefore, we undertook a series of experiments to characterize opioid-mediated antinociception in cholestasis and determine if it occurs inside or outside the CNS. METHODS: Antinociception scores to both thermal and mechanical stimuli were determined in mice with cholestasis due to bile duct resection vs sham controls. RESULTS: Cholestatic mice demonstrated significant antinociception to both stimuli compared to controls, which was reversible by the opiate receptor antagonist naloxone. The experiments were repeated with a naloxone derivative, which does not cross the blood-brain-barrier (i.e. naloxone methiodide) with similar results, indicating an opioid antinociceptive effect mediated outside of the CNS. Experiments with intraplantar injections of low dose naloxone methiodide confirmed that cholestasis-associated antinociception occurs at the level of cutaneous nerve endings. These findings were supported by findings of increased dermal met-enkephalin expression in cholestatic mice. CONCLUSIONS: Cholestasis in mice is associated with antinociception due to local effects of endogenous opioids (i.e. met-enkephalin) at the level of sensory nerve endings. These findings may have direct implications in the management of cholestasis associated pruritus.


Assuntos
Colestase/fisiopatologia , Peptídeos Opioides/metabolismo , Limiar da Dor , Prurido/fisiopatologia , Receptores Opioides/metabolismo , Animais , Colestase/complicações , Encefalina Metionina/análise , Encefalina Metionina/genética , Encefalina Metionina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos Opioides/análise , Peptídeos Opioides/genética , Prurido/etiologia , RNA Mensageiro/metabolismo , Receptores Opioides/agonistas , Células Receptoras Sensoriais/química , Células Receptoras Sensoriais/metabolismo , Pele/química
17.
Neurosci Lett ; 370(1): 1-6, 2004 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-15489007

RESUMO

Transplantation of adrenal medullary tissue for terminal cancer pain has been tested clinically, but this approach is not practical for routine use because of the shortage of organ donors and lack of tissue homogeneity. As a first alternative step, we have generated immortalized chromaffin cells over-expressing opioid peptides, namely met-enkephalin. Rat chromaffin cells have been genetically modified with vectors containing expression cassettes with either synthetic met-enkephalin or pro-enkephalin gene coding regions, fused with the nerve growth factor signal peptide for secretion. After stable transfection and differentiation in vitro, met-enkephalin and pro-enkephalin cells had higher met-enkephalin immunoreactivity and secreted met-enkephalin levels, compared to control cells containing the expression vector only. In the formalin hindpaw-injection model, 15 days after subarachnoid transplant of cells, grafts of met-enkephalin and pro-enkephalin cells significantly reduced the number of formalin-evoked c-fos immunoreactive spinal neurons in the spinal cord, compared to grafts of vector-alone chromaffin cells. The use of such expandable cell lines, for chronic spinal delivery of opiates, could offer an attractive and safe alternative strategy based on ex vivo gene therapy for the control of opioid-sensitive chronic pain.


Assuntos
Células Cromafins/transplante , Encefalina Metionina/metabolismo , Formaldeído/farmacologia , Dor/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Medula Espinal/metabolismo , Análise de Variância , Animais , Contagem de Células/métodos , Linhagem Celular Transformada , Células Cromafins/metabolismo , Células Cromafins/fisiologia , Encefalina Metionina/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Engenharia Genética , Sobrevivência de Enxerto/fisiologia , Humanos , Imuno-Histoquímica/métodos , Masculino , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , Dor/induzido quimicamente , Feniletanolamina N-Metiltransferase/metabolismo , RNA Mensageiro/biossíntese , Radioimunoensaio/métodos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Medula Espinal/efeitos dos fármacos , Transfecção/métodos
18.
Neuroendocrinology ; 79(4): 185-96, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15153752

RESUMO

In mammals the opioids Met-enkephalin and Leu-enkephalin are derived from a common precursor, proenkephalin, and as a result these neuropeptides are co-localized in enkephalinergic neurons. The mammalian scheme for enkephalinergic networks is not universal for all classes of sarcopterygian vertebrates. In an earlier study, distinct Met- and Leu-enkephalin-positive neurons were detected in the central nervous system (CNS) of the African lungfish, Protopterus annectens. More recently, characterization of proenkephalin cDNAs separately cloned from the CNS of P. annectens and the Australian lungfish, Neoceratodus forsteri, revealed that the proenkephalin gene in these species encodes only Met-enkephalin-related opioids. In the current study a full-length prodynorphin cDNA (accession No. AY 445637) was cloned and sequenced from the CNS of N. forsteri. In addition to encoding alpha-neoendorphin, dynorphin A and dynorphin B sequences unique to the lungfish, two Leu-enkephalin sequences, flanked by paired basic amino acid proteolytic cleavage sites, were detected in this precursor. The partial sequence of a P. annectens prodynorphin cDNA (accession No. AY445638) also encoded a Leu-enkephalin sequence and a novel YGGFF sequence. The presence of the Leu-enkephalin sequence in the lungfish prodynorphin precursors would explain the origin of the distinct Leu-enkephalin-positive neurons found in the African lungfish CNS. The realization that Met-enkephalin and Leu-enkephalin can be derived from distinct opioid-coding precursor genes calls into question the interpretation of comparative immunohistochemical studies that have mapped 'enkephalinergic' networks in non-mammalian vertebrates.


Assuntos
Evolução Biológica , Encéfalo/fisiologia , DNA Complementar , Encefalinas/genética , Peixes/genética , Precursores de Proteínas/genética , África , Sequência de Aminoácidos , Animais , Austrália , Sequência de Bases , Clonagem Molecular , Encefalina Leucina/genética , Encefalina Metionina/genética , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Mod Pathol ; 16(11): 1086-94, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14614047

RESUMO

Mucinous cystic neoplasms and serous microcystic adenomas account for the majority of cystic tumors of pancreas. Mucinous cystic neoplasms and serous microcystic adenomas have different frequencies of progression to malignancy. The genetic and epigenetic alterations of these tumors have not been studied in detail. In this study, we compared methylation status of p16, p14, VHL, and ppENK genes by methylation-specific PCR (MSP), and genetic alterations including K-ras and beta-catenin gene mutations, chromosome 3p loss, and microsatellite instability in 15 mucinous cystic neoplasms (10 benign and 5 borderline) and 16 serous microcystic adenomas. There were no significant differences between mucinous cystic neoplasms and serous microcystic adenomas in methylation of p16 (14%, 2/14 and 12%, 2/16), p14 (15%, 2/13 and 37%, 6/16), VHL (0/14 and 7%, 1/14), and ppENK (0/14 and 0/13), respectively. K-ras mutation was present only in mucinous cystic neoplasms but not in serous microcystic adenomas (33%, 5/15 versus 0/16; P =.004). In addition, LOH at 3p25, the chromosomal location of VHL gene, was present in 57% (8/14) of serous microcystic adenomas compared with in 17% (2/12) of mucinous cystic neoplasms (P =.03). No beta-catenin mutation, microsatellite instability, or mutation of transforming growth factor beta type II receptor was present in either type of tumors. In conclusion, K-ras mutations and allelic loss of VHL locus at 3p25, but not methylation, distinguished mucinous cystic neoplasms and serous microcystic adenomas. The differences in genetic alterations but not epigenetic alterations may explain the pathogenesis and progression to malignancy of these cystic tumors of pancreas.


Assuntos
Adenoma/genética , Adenoma/patologia , Cistadenoma Mucinoso/genética , Cistadenoma Mucinoso/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Cromossomos Humanos Par 3/genética , Metilação de DNA , Encefalina Metionina/genética , Genes p16 , Genes ras , Proteínas de Helminto/genética , Humanos , Perda de Heterozigosidade , Proteínas Musculares/genética , Mutação , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/genética , Proteína Supressora de Tumor Von Hippel-Lindau
20.
J Biomol Struct Dyn ; 21(1): 65-87, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12854960

RESUMO

We have developed an iterative hybrid algorithm (HA) to predict the 3D structure of peptides starting from their amino acid sequence. The HA is made of a modified genetic algorithm (GA) coupled to a local optimizer. Each HA iteration is carried out in two phases. In the first phase several GA runs are performed upon the entire peptide conformational space. In the second phase we used the manifestation of what we have called conformational memories, that arises at the end of the first phase, as a way of reducing the peptide conformational space in subsequent HA iterations. Use of conformational memories speeds up and refines the localization of the structure at the putative Global Energy Minimum (GEM) since conformational barriers are avoided. The algorithm has been used to predict successfully the putative GEM for Met- and Leu-enkephalin, and to obtain useful information regarding the 3D structure for the 8mer of polyglycine and the 16 residue (AAQAA)(3)Y peptide. The number of fitness function evaluations needed to locate the putative GEMs are fewer than those reported for other heuristic methods. This study opens the possibility of using Genetic Algorithms in high level predictions of secondary structure of polypeptides.


Assuntos
Algoritmos , Genética , Peptídeos/química , Peptídeos/genética , Conformação Proteica , Sequência de Aminoácidos , Biologia Computacional , Encefalina Leucina/química , Encefalina Leucina/genética , Encefalina Metionina/química , Encefalina Metionina/genética , Modelos Moleculares , Estrutura Molecular , Valor Preditivo dos Testes , Dobramento de Proteína , Análise Espectral Raman , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...