Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Aging (Albany NY) ; 13(8): 11889-11906, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33952720

RESUMEN

Sepiella maindroni ink, a flavoring and coloring agent in food, has attracted considerable attention due to its various pharmacological activities. Our previous study showed that the melanin of Sepiella maindroni ink (MSMI) can alleviate oxidative damage and delay aging in D-galactose(D-gal)-induced aging mice. This study aimed to reveal the possible mechanisms of the anti-aging effect of MSMI. In this article, a comprehensive analysis of gas chromatography-mass spectrometry (GC-MS)-based metabolomics and microarray-based transcriptomics revealed that 221 mRNAs were differentially expressed and 46 metabolites were significantly changed in the anti-aging progress of MSMI. Integrated analysis of transcript and metabolic profiles indicated that MSMI mainly altered carbohydrate metabolism, lipid metabolism, and insulin signaling pathway. MSMI achieved anti-aging effects not only by reducing oxidative damage and sorbitol toxicity but also by regulating lipid metabolism, improving insulin sensitivity, and reducing the formation of advanced glycation end products (AGEs). Moreover, our findings firstly demonstrated that MSMI could increase the expression of interferon-induced proteins and might be a potential antiviral compound.


Asunto(s)
Envejecimiento/efectos de los fármacos , Decapodiformes/química , Melaninas/administración & dosificación , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Metabolismo de los Hidratos de Carbono/genética , Galactosa/administración & dosificación , Galactosa/toxicidad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Productos Finales de Glicación Avanzada/metabolismo , Tinta , Insulina/metabolismo , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Masculino , Melaninas/aislamiento & purificación , Metabolómica , Ratones , Ratones Endogámicos ICR , Modelos Animales , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
2.
Sci Rep ; 10(1): 18025, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-33093538

RESUMEN

Sleep disturbances are commonly found in trauma-exposed populations. Additionally, trauma exposure results in fear-associated memory impairments. Given the interactions of sleep with learning and memory, we hypothesized that increasing sleep duration following trauma exposure would restore overall function and improve trauma-induced fear-associated memory dysfunction. Here, we utilized single prolonged stress, a validated rodent model of post-traumatic stress disorder, in combination with optogenetic activation of hypothalamic melanin-concentrating hormone containing cells to increase sleep duration. The goal of this work was to ascertain if post-trauma sleep increases are sufficient to improve fear-associated memory function. In our laboratory, optogenetic stimulation after trauma exposure was sufficient to increase REM sleep duration during both the Light and Dark Phase, whereas NREM sleep duration was only increased during the Dark Phase of the circadian day. Interestingly though, animals that received optogenetic stimulation showed significantly improved fear-associated memory processing compared to non-stimulated controls. These results suggest that sleep therapeutics immediately following trauma exposure may be beneficial and that post-trauma sleep needs to be further examined in the context of the development of post-traumatic stress disorder.


Asunto(s)
Miedo , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Trastornos de la Memoria/prevención & control , Optogenética , Hormonas Hipofisarias/administración & dosificación , Trastornos del Sueño-Vigilia/prevención & control , Sueño REM , Trastornos por Estrés Postraumático/complicaciones , Animales , Masculino , Consolidación de la Memoria , Trastornos de la Memoria/etiología , Ratas , Ratas Long-Evans , Trastornos del Sueño-Vigilia/etiología
3.
Cancer Immunol Immunother ; 69(12): 2501-2512, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32561966

RESUMEN

Peptide vaccines represent an attractive alternative to conventional anti-tumor therapies, but have not yet achieved significant clinical efficacy with commonly used formulations. Combination of short antigenic peptides, synthetic melanin and TLR9 agonist (Toll-like receptor 9, CpG-28) was reported as highly efficient to trigger strong CD8 + T-cell responses. We compared this vaccine approach to the standard adjuvant formulation that combines the incomplete Freund's adjuvant (IFA) and CpG-28, using either an ovalbumin epitope (pOVA30) or a spontaneously occurring tumor neoepitope (mAdpgk).Melanin-based vaccine induced significantly higher cytotoxic T lymphocytes (CTL) responses than IFA-based vaccine in both pOVA30- and mAdpgk-targeted vaccines. The anti-tumor efficacy of melanin-based vaccine was further assessed in mice, grafted either with E.G7-OVA cells (E.G7 cells transfected with ovalbumin) or with MC38 cells that spontaneously express the mAdpgk neoepitope. Melanin-based vaccine induced a major inhibition of E.G7-OVA tumor growth when compared to IFA-based vaccine (p < 0.001), but tumors eventually relapsed from day 24. In the MC38 tumor model, no significant inhibition of tumor growth was observed. In both cases, tumor escape appeared related to the loss of antigen presentation by tumor cells (loss of ovalbumin expression in E.G7-OVA model; poor presentation of mAdpgk in MC38 model), although the CTL responses displayed an effector memory phenotype, a high cytolytic potential and low programmed cell death-1 (PD1) expression.In conclusion, synthetic melanin can be efficiently used as an adjuvant to enhance T-cells response against subunit vaccine antigens and compared favorably to the classic combination of IFA and TLR9 agonist in mice.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cáncer/inmunología , Melaninas/inmunología , Neoplasias/terapia , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Femenino , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/inmunología , Humanos , Inmunogenicidad Vacunal , Lípidos/administración & dosificación , Lípidos/inmunología , Melaninas/administración & dosificación , Ratones , Neoplasias/inmunología , Neoplasias/patología , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/inmunología , Ovalbúmina/genética , Ovalbúmina/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología
4.
J Photochem Photobiol B ; 205: 111816, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32070822

RESUMEN

A melanin producer bacteria Halomonas venusta was isolated from a marine sponge Callyspongia sp. and optimized for melanin production. The optimized fermented media supplemented with 1% tyrosine yielded 4.92 mg/ml of melanin. The melanin incorporated cream was formulated and fortified with concentrates of seaweed Gelidium spinosum. Melanin and seaweed concentrate were found to be rich in antioxidant activity and were effectively inhibited the growth of S. aureus (MTCC 96) and S. pyogenes (MTCC 442). Various combinations of the cream were optimized and selected a formula containing 0.25% of melanin and 0.75% of seaweed concentrate which showed improved texture quality of cream. The formulated cream showed a pH of 5.52, spreadability 23 mm, and smooth and homogeneous texture. On application over skin provides a cooling effect and immediate disappearance without formation of white or oily film. Texture analysis of newly formulated cream showed similar results with that of control cream in terms of firmness, cohesiveness, index of viscosity and consistency. The formulated cream showed significant reduction of reactive oxygen species generated on exposure to direct sunlight. The cream showed protective effect on photohemolysis thus protecting the skin from lysis of red blood cells. The sun protection factor of the formulated cream F3 was found to be 18.373 ± 1.45. The combined antimicrobial and antioxidant effect of melanin and seaweed concentrate increased the shelf life of cream over the control. This study was the first report on photoprotective cream formulation using melanin and seaweed concentrate, which improved antioxidant and wound healing properties. The antimicrobial effect of the formulated natural cream could reduce the emergence of drug resistant bacteria and side effects of synthetic creams.


Asunto(s)
Antibacterianos/administración & dosificación , Antioxidantes/administración & dosificación , Melaninas/administración & dosificación , Nanopartículas/administración & dosificación , Protectores contra Radiación/administración & dosificación , Rhodophyta , Algas Marinas , Crema para la Piel/administración & dosificación , Animales , Línea Celular , Halomonas/metabolismo , Melaninas/metabolismo , Ratones , Staphylococcus/efectos de los fármacos , Staphylococcus/crecimiento & desarrollo , Luz Solar , Cicatrización de Heridas/efectos de los fármacos
5.
J Psychopharmacol ; 34(4): 478-489, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31909693

RESUMEN

BACKGROUND: Identifying neural substrates that are differentially affected by drugs of abuse and natural rewards is key to finding a target for an efficacious treatment for substance abuse. Melanin-concentrating hormone is a polypeptide with an inhibitory effect on the mesolimbic dopamine system. Here we test the hypothesis that melanin-concentrating hormone in the lateral hypothalamus and nucleus accumbens shell is differentially involved in the regulation of morphine and food-rewarded behaviors. METHODS: Male Sprague-Dawley rats were trained with morphine (5.0 mg/kg, subcutaneously) or food pellets (standard chow, 10-14 g) to induce a conditioned place preference, immediately followed by extinction training. Melanin-concentrating hormone (1.0 µg/side) or saline was infused into the nucleus accumbens shell or lateral hypothalamus before the reinstatement primed by morphine or food, and locomotor activity was simultaneously monitored. As the comparison, melanin-concentrating hormone was also microinjected into the nucleus accumbens shell or lateral hypothalamus before the expression of food or morphine-induced conditioned place preference. RESULTS: Microinfusion of melanin-concentrating hormone into the nucleus accumbens shell (but not into the lateral hypothalamus) prevented the reinstatement of morphine conditioned place preference but had no effect on the reinstatement of food conditioned place preference. In contrast, microinfusion of melanin-concentrating hormone into the lateral hypothalamus (but not in the nucleus accumbens shell) inhibited the reinstatement of food conditioned place preference but had no effect on the reinstatement of morphine conditioned place preference. CONCLUSIONS: These results suggest a clear double dissociation of melanin-concentrating hormone in morphine/food rewarding behaviors and melanin-concentrating hormone in the nucleus accumbens shell. Melanin-concentrating hormone could be a potential target for therapeutic intervention for morphine abuse without affecting natural rewards.


Asunto(s)
Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Morfina/farmacología , Núcleo Accumbens/metabolismo , Hormonas Hipofisarias/farmacología , Animales , Condicionamiento Operante/efectos de los fármacos , Hormonas Hipotalámicas/administración & dosificación , Masculino , Melaninas/administración & dosificación , Microinyecciones , Hormonas Hipofisarias/administración & dosificación , Ratas , Ratas Sprague-Dawley , Recompensa
6.
ACS Appl Mater Interfaces ; 11(45): 42671-42679, 2019 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-31663328

RESUMEN

Melanin and its synthetic analogs (i.e., polydopamine nanomaterials) are able to transform a near-infrared (NIR) light energy source to heat for the selective killing of cancer cells. Although many of the effects on these nontoxic photothermal agents have been well documented, a concern has arisen that the extended usage of these natural and synthetic melanins might be hindered by their limited photothermal effects under low-density light irradiation. To address this issue, herein, we propose a rational and green fabrication strategy toward a new class of synthetic melanin nanoparticles (SMNPs) with superior photothermal effects via the one-pot copolymerization of two kinds of naturally occurring monomers (arginine and dopamine). The total photothermal efficiencies of these arginine-doped SMNPs could be significantly improved (i.e., ∼60% increase) by enhancing 808 nm NIR light absorption via the construction of donor-acceptor microstructures within SMNPs and decreasing nonthermal radiative transition processes via the increase of free radical concentrations within SMNPs. The resulting SMNPs demonstrated higher photothermal therapy efficiencies in both killing 4T1 cancer cells in vitro and suppressing tumor growth and recurrence compared with conventional agents. This work offers new opportunities in the structural and functional tailoring of melanin-inspired nanomaterials for cancer treatment via green fabrication strategies.


Asunto(s)
Hipertermia Inducida , Melaninas/administración & dosificación , Neoplasias/terapia , Fototerapia , Animales , Arginina/química , Femenino , Humanos , Melaninas/química , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Nanopartículas/química , Neoplasias/diagnóstico por imagen
7.
Biomater Sci ; 7(10): 4060-4074, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31475710

RESUMEN

Combined photothermal-chemotherapy guided by multimodal imaging is a promising strategy for cancer diagnosis and treatment. Multifunctional nanoparticles, such as those comprising organic and inorganic compounds, have been extensively investigated for combined photothermal-chemotherapy; however, their application is still limited by their potential long-term toxicity and lack of contrast properties. To solve these problems, in this study, a new type of multifunctional nanoparticle for combined photothermal-chemotherapy guided by dual-modality imaging was prepared with endogenous melanin by multistep emulsification to enhance tumor ablation. The nanoparticles were coated with poly(lactide-co-glycolic acid) (PLGA) and loaded with paclitaxel (PTX), encapsulated melanin and perfluoropentane (PFP). The materials in the nanoparticles were endogenous, ensuring high stability, biocompatibility, and biosafety. Nanoparticles irradiated with a laser, which induced their phase transformation into microbubbles, exhibited high photothermal conversion efficiency, thereby achieving photoacoustic (PA)/ultrasound (US) dual-modality imaging to determine tumor location, boundary, and size and to monitor drug distribution. Furthermore, optical droplet vaporization (ODV) of the nanoparticles could trigger the release of PTX; thus, these nanoparticles are a useful drug carrier. In vivo and in vitro experiments revealed that a strong synergistic antitumor effect was achieved by combining the photothermal properties of the nanoparticles with a chemotherapy drug. Importantly, the cavitation, thermoelastic expansion, and sonoporation caused by the phase transformation of the nanoparticles could directly damage the tumors. These processes also promoted the release, penetration and absorption of the drug, further enhancing the effect of combined photothermal-chemotherapy on tumor suppression. Therefore, the multifunctional nanoparticles prepared in this study provide a new strategy of using endogenous materials for controlled near-infrared (NIR)-responsive drug release and combined photothermal-chemotherapy guided by multimodal imaging.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Fluorocarburos/administración & dosificación , Melaninas/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias/terapia , Paclitaxel/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Animales , Antineoplásicos Fitogénicos/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Preparaciones de Acción Retardada/administración & dosificación , Femenino , Fluorocarburos/farmacocinética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Melaninas/farmacocinética , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Paclitaxel/farmacocinética , Técnicas Fotoacústicas , Fototerapia , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacocinética , Distribución Tisular , Ultrasonografía
8.
Mol Neurobiol ; 56(12): 8076-8086, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31183806

RESUMEN

Melanin-concentrating hormone (MCH) is a highly conserved neuropeptide known to exhibit important functions in the brain. Some studies have reported that MCH improves memory by promoting memory retention. However, the precise molecular mechanisms by which MCH enhances memory impairment have yet to be fully elucidated. In this study, MCH was administered to the scopolamine-induced memory-impaired mice via the nasal cavity to examine the acute effects of MCH and Alzheimer's disease (AD) mouse models to evaluate the chronic effects of MCH. MCH improved memory impairment in both models and reduced soluble amyloid beta in the cerebral cortex of APP/PS1 transgenic mice. In vitro assays also showed that MCH inhibits amyloid beta-induced cytotoxicity. Furthermore, MCH increased long-term potentiation (LTP) in the hippocampus of wild-type and 5XFAD AD mouse model. To further elucidate the mechanisms of the chronic effect of MCH, the levels of phosphorylated CREB and GSK3ß, and the expression of BDNF, TrkB and PSD95 were examined in the cerebral cortex and hippocampus. Our findings indicate that MCH might have neuroprotective effects via downstream pathways associated with the enhancement of neuronal synapses and LTP. This suggests a therapeutic potential of MCH for the treatment of neurodegenerative diseases such as AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Modelos Animales de Enfermedad , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Hormonas Hipofisarias/administración & dosificación , Administración Intranasal , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cavidad Nasal/efectos de los fármacos , Cavidad Nasal/metabolismo , Embarazo
9.
Forensic Sci Med Pathol ; 15(4): 555-563, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31250256

RESUMEN

The purpose of this study was to evaluate the ability of alternate light source illumination to enhance bruises in pigmented skin. Previous work was limited to simulating bruises in non-pigmented (Caucasoid type) skin by injecting blood into pigskin. In this study, it was investigated if adding a layer of melanin to the surface of the skin would simulate pigmented skin. The study included evaluating the use of a filter that transmitted infrared light (wavelength greater than 720 nm) in place of the recommended visible light filters for the alternate light sources. The results obtained using pigskin with a layer of melanin were almost the same as results using the naturally pigmented goat ear. This indicated adding a layer of melanin could be used as a model for pigmented skin in this simulation of fresh bruising. Comparing the pigskin without melanin with pigskin with melanin revealed that the optimal light source to enhance the appearance of bruising, simulated by injection of blood, changed from violet to blue-green. Using the infrared transmitting filter resulted in greater enhancement than using the alternate light sources with their recommended visible light filter. The advantage of using the infrared transmitting filter was greater with the pigskin coated with melanin and the naturally pigmented goat ears than in the non-pigmented pigskin, however, the results remain to be validated using real bruises in naturally pigmented human skin.


Asunto(s)
Contusiones/patología , Rayos Infrarrojos , Luz , Fotograbar , Pigmentación de la Piel , Piel/patología , Animales , Patologia Forense , Cabras , Melaninas/administración & dosificación , Modelos Animales , Piel/lesiones , Porcinos
10.
Neuropeptides ; 74: 70-81, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30642579

RESUMEN

Melanin-concentrating hormone (MCH) is a neuropeptide present in neurons located in the hypothalamus that densely innervate serotonergic cells in the dorsal raphe nucleus (DRN). MCH administration into the DRN induces a depressive-like effect through a serotonergic mechanism. To further understand the interaction between MCH and serotonin, we used primary cultured serotonergic neurons to evaluate the effect of MCH on serotonergic release and metabolism by HPLC-ED measurement of serotonin (5-HT) and 5-hydroxyindolacetic acid (5-HIAA) levels. We confirmed the presence of serotonergic neurons in the E14 rat rhombencephalon by immunohistochemistry and showed for the first time evidence of MCHergic fibers reaching the area. Cultures obtained from rhombencephalic tissue presented 2.2 ±â€¯0.7% of serotonergic and 48.9 ±â€¯5.4% of GABAergic neurons. Despite the low concentration of serotonergic neurons, we were able to measure basal cellular and extracellular levels of 5-HT and 5-HIAA without the addition of any serotonergic-enhancer drug. As expected, 5-HT release was calcium-dependent and induced by depolarization. 5-HT extracellular levels were significantly increased by incubation with serotonin reuptake inhibitors (citalopram and nortriptyline) and a monoamine-oxidase inhibitor (clorgyline), and were not significantly modified by a 5-HT1A autoreceptor agonist (8-OHDPAT). Even though serotonergic cells responded as expected to these pharmacological treatments, MCH did not induce significant modifications of 5-HT and 5-HIAA extracellular levels in the cultures. Despite this unexpected result, we consider that assessment of 5-HT and 5-HIAA levels in primary serotonergic cultures may be an adequate approach to study the effect of other drugs and modulators on serotonin release, uptake and turnover.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Núcleos del Rafe/metabolismo , Serotonina/metabolismo , Animales , Neuronas GABAérgicas/citología , Hormonas Hipotalámicas/administración & dosificación , Hipotálamo/citología , Melaninas/administración & dosificación , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Hormonas Hipofisarias/administración & dosificación , Cultivo Primario de Células , Núcleos del Rafe/citología , Núcleos del Rafe/efectos de los fármacos , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/metabolismo
11.
Sci Rep ; 8(1): 707, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29335480

RESUMEN

The melanin-concentrating hormone (MCH) is a peptidergic neuromodulator synthesized by neurons in the lateral hypothalamus and zona incerta. MCHergic neurons project throughout the central nervous system, indicating the involvements of many physiological functions, but the role in pain has yet to be determined. In this study, we found that pMCH-/- mice showed lower baseline pain thresholds to mechanical and thermal stimuli than did pMCH+/+ mice, and the time to reach the maximum hyperalgesic response was also significantly earlier in both inflammatory and neuropathic pain. To examine its pharmacological properties, MCH was administered intranasally into mice, and results indicated that MCH treatment significantly increased mechanical and thermal pain thresholds in both pain models. Antagonist challenges with naltrexone (opioid receptor antagonist) and AM251 (cannabinoid 1 receptor antagonist) reversed the analgesic effects of MCH in both pain models, suggesting the involvement of opioid and cannabinoid systems. MCH treatment also increased the expression and activation of CB1R in the medial prefrontal cortex and dorsolateral- and ventrolateral periaqueductal grey. The MCH1R antagonist abolished the effects induced by MCH. This is the first study to suggest novel analgesic actions of MCH, which holds great promise for the application of MCH in the therapy of pain-related diseases.


Asunto(s)
Analgésicos/farmacología , Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Percepción del Dolor/efectos de los fármacos , Hormonas Hipofisarias/farmacología , Administración Intranasal , Analgésicos/administración & dosificación , Animales , Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/deficiencia , Melaninas/administración & dosificación , Melaninas/deficiencia , Ratones , Ratones Noqueados , Hormonas Hipofisarias/administración & dosificación , Hormonas Hipofisarias/deficiencia
12.
Food Funct ; 9(2): 880-889, 2018 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-29299589

RESUMEN

In the present study, we investigated the hypolipidemic properties of melanin from Lachnum YM226 (LM) in high-fat diet induced hyperlipidemic mice. After the hyperlipidemic model was established, mice were randomly divided into six groups, as follows: normal control group (NC), hyperlipidemic control group (HC), positive control group (7 mg kg-1 d-1 simvastatin) (PC) and LM groups (50, 100 and 200 mg kg-1 d-1 denoted as LM-50, LM-100 and LM-200, respectively). Subsequently, the body weight, organ indices, lipid metabolism, antioxidant properties and liver-kidney functions of the mice were examined. Moreover, the activities of lipoprotein metabolism enzymes in serum and liver tissue were examined to study the feasible mechanism. The results imply that LM could effectively reduce body weight, total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C) and atherogenic index (AI), and increase high density lipoprotein cholesterol (HDL-C). Moreover, treatment with LM also increased the antioxidant enzymes activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) and reduced malondialdehyde (MDA) content relative to the HC group. In addition, the liver and kidney damage indices such as alanine aminotransferase (ALT), aspartate aminotransaminase (AST), alkaline phosphatase (ALP), creatinine (CRE), blood urea nitrogen (BUN) and uric acid were lowered. LM administration also significantly corrected disturbances of liver-kidney functions with no fatty deposits in the liver, resulting in a protective effect against renal histological alteration. The hypolipidemic effect occurred partly due to the regulation of hepatic lipase (HL) and lipoprotein lipase (LPL) in serum and liver to markedly decrease TG. This confirms the important role of LM in the prevention of hyperlipidemia.


Asunto(s)
Ascomicetos/química , Hiperlipidemias/tratamiento farmacológico , Hipolipemiantes/administración & dosificación , Riñón/fisiopatología , Hígado/fisiopatología , Melaninas/administración & dosificación , Extractos Vegetales/administración & dosificación , Alanina Transaminasa/metabolismo , Animales , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Dieta Alta en Grasa/efectos adversos , Glutatión Peroxidasa/metabolismo , Humanos , Hiperlipidemias/metabolismo , Hiperlipidemias/fisiopatología , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Superóxido Dismutasa/metabolismo , Triglicéridos/metabolismo , Verduras/química
13.
Neuropharmacology ; 130: 62-70, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29191753

RESUMEN

Melanin-Concentrating Hormone (MCH) is one of the most relevant orexigenic factors specifically located in the lateral hypothalamic area (LHA), with its physiological relevance demonstrated in studies using several genetically manipulated mice models. However, the central mechanisms controlling MCH-induced hyperphagia remain largely uncharacterized. Here, we show that central injection of MCH in mice deficient for kappa opoid receptor (k-OR) failed to stimulate feeding. To determine the hypothalamic area responsible for this MCH/k-OR interaction, we performed virogenetic studies and found that downregulation of k-OR by adeno-associated viruses (shOprk1-AAV) in LHA, but not in other hypothalamic nuclei, was sufficient to block MCH-induced food intake. Next, we sought to investigate the molecular signaling pathway within the LHA that mediates acute central MCH stimulation of food intake. We found that MCH activates k-OR and that increased levels of phosphorylated extracellular signal regulated kinase (ERK) are associated with downregulation of phospho-S6 Ribosomal Protein. This effect was prevented when a pharmacological inhibitor of k-OR was co-administered with MCH. Finally, the specific activation of the direct upstream regulator of S6 (p70S6K) in the LHA attenuated MCH-stimulated food consumption. Our results reveal that lateral hypothalamic k-OR system modulates the orexigenic action of MCH via the p70S6K/S6 pathway.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Hormonas Hipofisarias/administración & dosificación , Receptores Opioides kappa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Animales , Depresores del Apetito/administración & dosificación , Depresores del Apetito/metabolismo , Dependovirus , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Hormonas Hipotalámicas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Hormonas Hipofisarias/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides kappa/metabolismo , Proteínas Quinasas S6 Ribosómicas/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas/metabolismo
14.
BMC Microbiol ; 17(1): 147, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28673241

RESUMEN

BACKGROUND: By the search for new natural compounds with beneficial health effects, cephalopod ink has been considered as an attempt to develop new drugs and functional foods, which is an especially active field in Asia, where cephalopods are a major fishery catch, for which ink sacs are a bi-product and where homeopathic medicine has deep roots. There is a demand to evaluate the safety and influence to the organism. The specific composition and relative abundance of the gut microbiota, which is potentially a major modulator of host metabolism, drives the interaction between functional foods and host health. We explore the effects of melanin from Sepiella Maindroni, most common cuttlefish in China, on the intestinal microbiome of mice. RESULTS: ICR mice were randomly divided four groups, which were normal group (S), low melanin dose group (D; 120 mg/kg), medium melanin dose group (Z; 240 mg/kg), and high melanin dose group (G; 480 mg/kg). Melanin was delivered for 28 consecutive days. Fecal samples were used to generate 7715 operational taxonomic units (OTUs) via high-throughput sequencing. There were significant shifts in relative abundance of the dominant taxa at the phylum, class, order, family, and genus levels following melanin treatment. CONCLUSIONS: MSMI had no significant effect on the structure of intestinal flora in mice. The main effect was in the proportion of dominant bacterial communities. The effect positively correlated with the dose. From a health point of view, the use of melanin does not cause intestinal flora disorder. Our results may have important implications for MSMI as functional food component and potential therapeutic for manipulating gut microbiota.


Asunto(s)
Bacterias/clasificación , Decapodiformes/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Melaninas/administración & dosificación , Animales , Bacterias/genética , Relación Dosis-Respuesta a Droga , Heces/microbiología , Secuenciación de Nucleótidos de Alto Rendimiento , Melaninas/farmacología , Ratones , Ratones Endogámicos ICR , Filogenia , ARN Bacteriano/genética , ARN Ribosómico 16S/genética , Distribución Aleatoria , Análisis de Secuencia de ARN
15.
Biomed Mater ; 12(2): 025010, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28181477

RESUMEN

Melanin function in the skin has been associated with pigmentation but other properties such as electrical conductance, photoprotection, and antioxidant and antimicrobial activity have also been recognized. Nonetheless, the use of melanin in a skin wound healing context has never been considered. In this sense, eumelanin particles with a typical round and nano-sized morphology and electrical conductivity of 2.09 × 10-8 S cm-1 were extracted from the ink of Sepia officinalis. The ability of primary human keratinocytes (hKCs) to phagocyte eumelanin, which was then accumulated in cytosolic vesicles and nuclei surroundings, was demonstrated. Keratinocyte viability and maturation was not affected by eumelanin contact, but at eumelanin amounts higher than 0.1 mg l-1 cell morphology was altered and cell proliferation was inhibited. A time and eumelanin amount-dependent reduction of reactive oxygen species (ROS) released by eumelanin-containing ultraviolet (UV)-irradiated keratinocytes was observed. Eumelanin-containing gellan gum (GG) spongy-like hydrogels allowed a sustained release of eumelanin in the range of 0.1 to 5 mg l-1, which was shown in vitro to not be harmful to hKCs, and the absence of a strong host reaction after subcutaneous implantation in mice. Herein, we propose spongy-like hydrogels as sustained release matrices of S. officinalis eumelanin for predicting a beneficial role in skin wound healing through a direct effect over keratinocytes.


Asunto(s)
Queratinocitos/efectos de los fármacos , Melaninas/administración & dosificación , Repitelización/efectos de los fármacos , Animales , Materiales Biocompatibles , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Sistemas de Liberación de Medicamentos , Humanos , Hidrogeles , Queratinocitos/citología , Queratinocitos/fisiología , Ensayo de Materiales , Melaninas/farmacocinética , Melaninas/fisiología , Ratones , Fagocitosis , Polisacáridos Bacterianos , Repitelización/fisiología , Piel/efectos de los fármacos , Piel/lesiones , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
16.
J Virol ; 91(6)2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28077650

RESUMEN

Prion diseases are progressive fatal neurodegenerative illnesses caused by the accumulation of transmissible abnormal prion protein (PrP). To find treatments for prion diseases, we searched for substances from natural resources that inhibit abnormal PrP formation in prion-infected cells. We found that high-molecular-weight components from insect cuticle extracts reduced abnormal PrP levels. The chemical nature of these components was consistent with that of melanin. In fact, synthetic melanin produced from tyrosine or 3-hydroxy-l-tyrosine inhibited abnormal PrP formation. Melanin did not modify cellular or cell surface PrP levels, nor did it modify lipid raft or cellular cholesterol levels. Neither did it enhance autophagy or lysosomal function. Melanin was capable of interacting with PrP at two N-terminal domains. Specifically, it strongly interacted with the PrP region of amino acids 23 to 50 including a positively charged amino acid cluster and weakly interacted with the PrP octarepeat peptide region of residues 51 to 90. However, the in vitro and in vivo data were inconsistent with those of prion-infected cells. Abnormal PrP formation in protein misfolding cyclic amplification was not inhibited by melanin. Survival after prion infection was not significantly altered in albino mice or exogenously melanin-injected mice compared with that of control mice. These data suggest that melanin, a main determinant of skin color, is not likely to modify prion disease pathogenesis, even though racial differences in the incidence of human prion diseases have been reported. Thus, the findings identify an interaction between melanin and the N terminus of PrP, but the pathophysiological roles of the PrP-melanin interaction remain unclear.IMPORTANCE The N-terminal region of PrP is reportedly important for neuroprotection, neurotoxicity, and abnormal PrP formation, as this region is bound by many factors, such as metal ions, lipids, nucleic acids, antiprion compounds, and several proteins, including abnormal PrP in prion disease and the Aß oligomer in Alzheimer's disease. In the present study, melanin, a main determinant of skin color, was newly found to interact with this N-terminal region and inhibits abnormal PrP formation in prion-infected cells. However, the data for prion infection in mice lacking melanin production suggest that melanin is not associated with the prion disease mechanism, although the incidence of prion disease is reportedly much higher in white people than in black people. Thus, the roles of the PrP-melanin interaction remain to be further elucidated, but melanin might be a useful competitive tool for evaluating the functions of other ligands at the N-terminal region.


Asunto(s)
Melaninas/metabolismo , Enfermedades por Prión/prevención & control , Priones/metabolismo , Animales , Línea Celular , Melaninas/administración & dosificación , Ratones , Neuronas/metabolismo , Enfermedades por Prión/tratamiento farmacológico , Unión Proteica , Mapeo de Interacción de Proteínas , Análisis de Supervivencia
17.
Macromol Biosci ; 17(5)2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27906510

RESUMEN

Melanin is an effective absorber of light and can extend to near infrared (NIR) regions. In this study, a natural melanin is presented as a photothermal therapeutic agent (PTA) because it provides a good photothermal conversion efficiency, shows biodegradability, and does not induce long-term toxicity during retention in vivo. Poloxamer solution containing melanin (Pol-Mel) does not show any precipitation and shows sol-gel transition at body temperature. After irradiation from 808 nm NIR laser at 1.5 W cm-2 for 3 min, the photothermal conversion efficiency of Pol-Mel is enough to kill cancer cells in vitro and in vivo. The tumor growth of mice bearing CT26 tumors treated with Pol-Mel injection and laser irradiation is suppressed completely without recurrence postirradiation. All these results indicate that Pol-Mel can become an attractive PTA for photothermal cancer therapy.


Asunto(s)
Hidrogeles/química , Hipertermia Inducida/métodos , Melaninas/uso terapéutico , Neoplasias/terapia , Fototerapia/métodos , Animales , Línea Celular Tumoral , Humanos , Rayos Infrarrojos , Masculino , Melaninas/administración & dosificación , Melaninas/química , Ratones , Ratones Endogámicos BALB C , Poloxámero , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Mol Neurobiol ; 54(10): 7706-7721, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27844281

RESUMEN

Acupuncture has shown the therapeutic effect on various neurodegenerative disorders including Parkinson's disease (PD). While investigating the neuroprotective mechanism of acupuncture, we firstly found the novel function of melanin-concentrating hormone (MCH) as a potent neuroprotective candidate. Here, we explored whether hypothalamic MCH mediates the neuroprotective action of acupuncture. In addition, we aimed at evaluating the neuroprotective effects of MCH and elucidating underlying mechanism in vitro and in vivo PD models. First, we tested whether hypothalamic MCH mediates the neuroprotective effects of acupuncture by challenging MCH-R1 antagonist (i.p.) in mice PD model. We also investigated whether MCH has a beneficial role in dopaminergic neuronal protection in vitro primary midbrain and human neuronal cultures and in vivo MPTP-induced, Pitx3-/-, and A53T mutant mice PD models. Transcriptomics followed by quantitative PCR and western blot analyses were performed to reveal the neuroprotective mechanism of MCH. We first found that hypothalamic MCH biosynthesis was directly activated by acupuncture treatment and that administration of an MCH-R1 antagonist reverses the neuroprotective effects of acupuncture. A novel finding is that MCH showed a beneficial role in dopaminergic neuron protection via downstream pathways related to neuronal survival. This is the first study to suggest the novel neuroprotective action of MCH as well as the involvement of hypothalamic MCH in the acupuncture effects in PD, which holds great promise for the application of MCH in the therapy of neurodegenerative diseases.


Asunto(s)
Terapia por Acupuntura/métodos , Hormonas Hipotalámicas/biosíntesis , Melaninas/biosíntesis , Fármacos Neuroprotectores/metabolismo , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/prevención & control , Hormonas Hipofisarias/biosíntesis , Animales , Células Cultivadas , Humanos , Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/antagonistas & inhibidores , Hipotálamo/metabolismo , Masculino , Melaninas/administración & dosificación , Melaninas/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Hormonas Hipofisarias/administración & dosificación , Hormonas Hipofisarias/antagonistas & inhibidores , Resultado del Tratamiento
19.
Peptides ; 87: 28-33, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27845162

RESUMEN

Neuropeptideglutamic acid-isoleucine (NEI) as well as melanin concentrating hormone (MCH) is cleaved from the 165 amino acid protein, prepro-melanin concentrating hormone (prepro-MCH). Among many physiological roles of MCH, we demonstrated that intracerebroventricular (icv) injection of MCH induced increases in REM sleep episodes as well as in non REM sleep episodes. However, there are no studies on the effect of NEI on the sleep-wake cycle. As for the sites of action of MCH for induction of REM sleep, the ventrolateral periaqueductal gray (vlPAG) has been reported to be one of its site of action. Although MCH neurons contain NEI, GABA, MCH, and other neuropeptides, we do not know which transmitter(s) might induce REM sleep by acting on the vlPAG. Thus, we first examined the effect of icv injection of NEI on the sleep-wake cycle, and investigated how microinjection of either NEI, MCH, or GABA into the vlPAG affected REM sleep in rats. Icv injection of NEI (0.61µg/5µl: n=7) significantly increased the time spent in REM episodes compared to control (saline: 5µl; n=6). Microinjection of either NEI (61ng/0.2µl: n=7), MCH (100ng/0.2µl: n=6) or GABA (250mM/0.2µl: n=7) into the vlPAG significantly increased the time spent in REM episodes and the AUC. Precise hourly analysis of REM sleep also revealed that after those microinjections, NEI and MCH increased REM episodes at the latter phase, compared to GABA which increased REM episodes at the earlier phase. This result suggests that NEI and MCH may induce sustained REM sleep, while GABA may initiate REM sleep. In conclusion, our findings demonstrate that NEI, a cleaved peptide from the same precursor, prepro-MCH, as MCH, induce REM sleep at least in part through acting on the vlPAG.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Neuropéptidos/administración & dosificación , Hormonas Hipofisarias/metabolismo , Sueño REM/efectos de los fármacos , Animales , Ácido Glutámico/administración & dosificación , Ácido Glutámico/metabolismo , Hormonas Hipotalámicas/administración & dosificación , Hormonas Hipotalámicas/química , Isoleucina/administración & dosificación , Isoleucina/metabolismo , Melaninas/administración & dosificación , Melaninas/química , Microinyecciones , Neuronas/efectos de los fármacos , Neuropéptidos/metabolismo , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Sustancia Gris Periacueductal/fisiología , Hormonas Hipofisarias/administración & dosificación , Hormonas Hipofisarias/química , Ratas , Sueño REM/fisiología , Ácido gamma-Aminobutírico/administración & dosificación
20.
Curr Eye Res ; 42(5): 748-753, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27854122

RESUMEN

PURPOSE: Several studies showed that repeated topical administration of brimonidine tartrate ophthalmic solution reached the human vitreous concentration above 2 nM, which is the concentration necessary to activate the α2-adrenergic receptor. The purpose of this study was to elucidate the relationship of the brimonidine concentration in the vitreous body to the free concentration in the retina/choroid which is the target site of brimonidine on neuroprotective effect after topical administration. MATERIALS AND METHODS: Brimonidine concentrations in the eye tissues of pigmented rabbits were determined following single ocular administration of 0.1% brimonidine tartrate ophthalmic solution at pH 7.3. Binding affinity of brimonidine to melanin and melanin content in the retina/choroid of pigmented rabbits was also examined. The concentration of free brimonidine which did not bind to melanin in the retina/choroid was calculated using the binding parameters to melanin. RESULTS: Topically applied brimonidine rapidly distributed to intraocular tissues. The elimination rate from melanin-containing tissues such as the iris/ciliary body and retina/choroid was slower than the aqueous humor and vitreous body in pigmented rabbits. In both the anterior and posterior retina/choroid, the free brimonidine concentrations were over 100-fold lower than the total concentrations. The concentrations in the vitreous body closely matched to the free concentrations in the posterior retina/choroid. Simulated free concentrations in the posterior retina/choroid were gradually increased when 0.1% solution was instilled twice daily. CONCLUSION: The present data indicated that the brimonidine concentration in the vitreous body was comparable to the free concentration in the posterior retina/choroid. This suggests that the vitreous concentration can be a surrogate indicator of the free brimonidine concentration in the posterior retina/choroid. From the present findings, it is expected that multiple instillation of brimonidine tartrate ophthalmic solution may produce the sufficient free concentration for activation of the α2-adrenergic receptor in the retina/choroid in human.


Asunto(s)
Tartrato de Brimonidina/farmacocinética , Glaucoma/tratamiento farmacológico , Retina/metabolismo , Cuerpo Vítreo/metabolismo , Administración Tópica , Animales , Antihipertensivos/administración & dosificación , Antihipertensivos/farmacocinética , Tartrato de Brimonidina/administración & dosificación , Cromatografía Liquida , Modelos Animales de Enfermedad , Glaucoma/metabolismo , Glaucoma/patología , Masculino , Melaninas/administración & dosificación , Melaninas/farmacocinética , Soluciones Oftálmicas , Conejos , Retina/patología , Espectrometría de Masas en Tándem , Cuerpo Vítreo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...