Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Biother Radiopharm ; 35(6): 459-473, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32013538

RESUMEN

An α particle-emitting nanodrug that is a potent and specific antitumor agent and also prompts significant remodeling of local immunity in the tumor microenvironment (TME) has been developed and may impact the treatment of melanoma. Biocompatible ultrasmall fluorescent core-shell silica nanoparticles (C' dots, diameter ∼6.0 nm) have been engineered to target the melanocortin-1 receptor expressed on melanoma through α melanocyte-stimulating hormone peptides attached to the C' dot surface. Actinium-225 is also bound to the nanoparticle to deliver a densely ionizing dose of high-energy α particles to cancer. Nanodrug pharmacokinetic properties are optimal for targeted radionuclide therapy as they exhibit rapid blood clearance, tumor-specific accumulation, minimal off-target localization, and renal elimination. Potent and specific tumor control, arising from the α particles, was observed in a syngeneic animal model of melanoma. Surprisingly, the C' dot component of this drug initiates a favorable pseudopathogenic response in the TME generating distinct changes in the fractions of naive and activated CD8 T cells, Th1 and regulatory T cells, immature dendritic cells, monocytes, MΦ and M1 macrophages, and activated natural killer cells. Concomitant upregulation of the inflammatory cytokine genome and adaptive immune pathways each describes a macrophage-initiated pseudoresponse to a viral-shaped pathogen. This study suggests that therapeutic α-particle irradiation of melanoma using ultrasmall functionalized core-shell silica nanoparticles potently kills tumor cells, and at the same time initiates a distinct immune response in the TME.


Asunto(s)
Partículas alfa/uso terapéutico , Portadores de Fármacos/química , Melanoma Experimental/radioterapia , Radiofármacos/administración & dosificación , Neoplasias Cutáneas/radioterapia , Microambiente Tumoral/efectos de la radiación , Actinio/administración & dosificación , Actinio/farmacocinética , Animales , Línea Celular Tumoral/trasplante , Biología Computacional , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Inmunidad Celular/genética , Inmunidad Celular/efectos de la radiación , Masculino , Dosis Máxima Tolerada , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Terapia Molecular Dirigida/métodos , Nanopartículas/química , RNA-Seq , Radiofármacos/farmacocinética , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/metabolismo , Dióxido de Silicio/química , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Distribución Tisular , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
2.
ACS Chem Neurosci ; 9(12): 3015-3023, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29924583

RESUMEN

The melanocortin-3 and melanocortin-4 receptors (MC3R and MC4R), endogenous agonists derived from the proopiomelanocortin gene transcript, and naturally occurring antagonists agouti and agouti-related protein (AGRP) have been linked to biological pathways associated with energy homeostasis. The active tripeptide sequence of AGRP, Arg111-Phe112-Phe113, is located on a hypothesized ß-hairpin loop. Herein, stereochemical modifications of the Arg-Phe-Phe sequence were examined in the octapeptide AGRP-derived macrocyclic scaffold c[Pro-Arg-Phe-Phe-Xxx-Ala-Phe-DPro], where Xxx was Asn or diaminopropionic acid (Dap). Macrocyclic peptides were synthesized with one, two, or three residues of the Arg-Phe-Phe sequence substituted with the corresponding d-isomer(s), generating a 14 compound library. While l-to-d inversions of the Arg-Phe-Phe sequence in a 20-residue AGRP-derived ligand previously resulted in agonist activity at the MC1R, MC3R, MC4R, and MC5R, only the MC1R was consistently stimulated by the macrocyclic ligands in the present study, with varying ligand potencies and efficacies observed at the MC1R. A general trend of increased MC4R antagonist potency was observed for Dap-containing compounds, while MC5R inverse agonist activity was observed for select ligands. It was observed that stereochemical modification of the Arg-Phe-Phe active tripeptide sequence was insufficient to convert melanocortin antagonist into agonists. Overall, these observations are important in the design of melanocortin ligands possessing potent and selective agonist and antagonist activities.


Asunto(s)
Proteína Relacionada con Agouti/química , Aminoácidos/química , Péptidos/química , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Agonismo Inverso de Drogas , Humanos , Ligandos , Compuestos Macrocíclicos , Receptor de Melanocortina Tipo 1/efectos de los fármacos , Receptor de Melanocortina Tipo 3/efectos de los fármacos , Receptor de Melanocortina Tipo 4/efectos de los fármacos , Receptores de Melanocortina/efectos de los fármacos , Estereoisomerismo
3.
Arch Pharm Res ; 41(3): 324-332, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29397551

RESUMEN

Finasteride is a well-known 5α-reductase inhibitor used for treatment of alopecia and prostate cancer. But the effect of finasteride in regulating melanogenesis is still unclear. In the present study the role of finasteride on melanogenesis was investigated. Finasteride decrease melanin level in melanocyte melan-a cells and B16F10 melanoma cells without inducing cytotoxicity. MC1R (melanocortin 1 receptor) protein expression was also inhibited by finasteride thereby decreasing the expression of adenylate cyclase, MITF (Melanogenesis associated transcription factor), tyrosinases, TRP (tyrosinase-related protein) -1 and -2. Thus our study suggest that finasteride inhibits melanogenesis in melanocyte and melanoma cells by inhibiting MC1R.


Asunto(s)
Adenilil Ciclasas/fisiología , Finasterida/farmacología , Melanocitos/efectos de los fármacos , Melanocitos/enzimología , Melanoma Experimental/enzimología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Melanoma Experimental/tratamiento farmacológico , Ratones , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/metabolismo
4.
Mol Imaging ; 16: 1536012117737919, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29182034

RESUMEN

Melanoma is a deadly disease at late metastatic stage, and early diagnosis and accurate staging remain the key aspects for managing melanoma. The melanocortin 1 receptor (MC1 R) is overexpressed in primary and metastatic melanomas, and its endogenous ligand, the α-melanocyte-stimulating hormone (αMSH), has been extensively studied for the development of MC1 R-targeted molecular imaging and therapy of melanoma. Natural αMSH is not well suited for this purpose due to low stability in vivo. Unnatural amino acid substitutions substantially stabilized the peptide, while cyclization via lactam bridge and metal coordination further improved binding affinity and stability. In this study, we summarized the development and the in vitro and in vivo characteristics of the radiolabeled αMSH analogues, including 99mTc-, 111In-, 67 Ga-, or 125I-labeled αMSH analogues for imaging with single-photon emission computed tomography; 68Ga-, 64Cu-, or 18F-labeled αMSH analogues for imaging with positron emission tomography; and 188Re-, 177Lu-, 90Y-, or 212Pb-labeled αMSH analogues for radionuclide therapy. These radiolabeled αMSH analogues showed promising results with high tumor uptake and rapid normal tissue activity clearance in the preclinical model of B16F1 and B16F10 mouse melanomas. These results highlight the potential of using radiolabeled αMSH analogues in clinical applications for molecular imaging and radionuclide therapy of melanoma.


Asunto(s)
Melanoma/diagnóstico por imagen , Melanoma/tratamiento farmacológico , Radiofármacos/uso terapéutico , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , alfa-MSH/análogos & derivados , Animales , Detección Precoz del Cáncer , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melanoma/metabolismo , Imagen Molecular/métodos , Radiofármacos/farmacología , Tomografía Computarizada de Emisión de Fotón Único , Regulación hacia Arriba/efectos de los fármacos , alfa-MSH/química
5.
Photochem Photobiol ; 93(1): 245-258, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27645605

RESUMEN

Melanoma is the deadliest form of skin cancer because of its propensity to spread beyond the primary site of disease and because it resists many forms of treatment. Incidence of melanoma has been increasing for decades. Although ultraviolet radiation (UV) has been identified as the most important environmental causative factor for melanoma development, UV-protective strategies have had limited efficacy in melanoma prevention. UV mutational burden correlates with melanoma development and tumor progression, underscoring the importance of UV in melanomagenesis. However, besides amount of UV exposure, melanocyte UV mutational load is influenced by the robustness of nucleotide excision repair, the genome maintenance pathway charged with removing UV photoproducts before they cause permanent mutations in the genome. In this review, we highlight the importance of the melanocortin hormonal signaling axis on regulating efficiency of nucleotide excision repair in melanocytes. By understanding the molecular mechanisms by which nucleotide excision repair can be increased, it may be possible to prevent many cases of melanoma by reducing UV mutational burden over time.


Asunto(s)
Reparación del ADN , Melanocortinas/metabolismo , Melanocitos/metabolismo , Dímeros de Pirimidina/metabolismo , Transducción de Señal , Rayos Ultravioleta/efectos adversos , AMP Cíclico/metabolismo , Humanos , Melanoma/epidemiología , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/metabolismo , Estados Unidos/epidemiología , Xerodermia Pigmentosa/etiología
6.
Oncotarget ; 7(18): 26331-45, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27028866

RESUMEN

Melanoma risk is increased in patients with mutations of melanocortin 1 receptor (MC1R) yet the basis for the increased risk remains unknown. Here we report in vivo evidence supporting a critical role for MC1R in regulating melanoma tumor growth and determining overall survival time. Inhibition of MC1R by its physiologically relevant competitive inhibitor, agouti signaling protein (ASIP), reduced melanin synthesis and morphological heterogeneity in murine B16-F10 melanoma cells. In the lungs of syngeneic C57BL/6 mice, mCherry-marked, ASIP-secreting lung tumors inhibited MC1R on neighboring tumors lacking ASIP in a dose dependent manner as evidenced by a proportional loss of pigment in tumors from mice injected with 1:1, 3:1 and 4:1 mixtures of parental B16-F10 to ASIP-expressing tumor cells. ASIP-expressing B16-F10 cells formed poorly pigmented tumors in vivo that correlated with a 20% longer median survival than those bearing parental B16-F10 tumors (p=0.0005). Mice injected with 1:1 mixtures also showed survival benefit (p=0.0054), whereas injection of a 4:1 mixture showed no significant difference in survival. The longer survival time of mice bearing ASIP-expressing tumors correlated with a significantly slower growth rate than parental B16-F10 tumors as judged by quantification of numbers of tumors and total tumor load (p=0.0325), as well as a more homogeneous size and morphology of ASIP-expressing lung tumors. We conclude that MC1R plays an important role in regulating melanoma growth and morphology. Persistent inhibition of MC1R provided a significant survival advantage resulting in part from slower tumor growth, establishing MC1R as a compelling new molecular target for metastatic melanoma.


Asunto(s)
Proteína de Señalización Agouti/metabolismo , Melanoma Experimental/patología , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Proteína de Señalización Agouti/genética , Animales , Línea Celular Tumoral , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL
7.
Arterioscler Thromb Vasc Biol ; 35(9): 1936-44, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26112010

RESUMEN

OBJECTIVE: Neutrophil recruitment is a key process in the pathogenesis of stroke, and may provide a valuable therapeutic target. Targeting the melanocortin (MC) receptors has previously shown to inhibit leukocyte recruitment in peripheral inflammation, however, it is not known whether treatments are effective in the unique cerebral microvascular environment. Here, we provide novel research highlighting the effects of the MC peptides on cerebral neutrophil recruitment, demonstrating important yet discrete roles for both MC1 and MC3. APPROACH AND RESULTS: Using intravital microscopy, in 2 distinct murine models of cerebral ischemia-reperfusion (I/R) injury, we have investigated MC control for neutrophil recruitment. After global I/R, pharmacological treatments suppressed pathological neutrophil recruitment. MC1 selective treatment rapidly inhibited neutrophil recruitment while a nonselective MC agonist provided protection even when coadministered with an MC3/4 antagonist, suggesting the importance of early MC1 signaling. However, by 2-hour reperfusion, MC1-mediated effects were reduced, and MC3 anti-inflammatory circuits predominated. Mice bearing a nonfunctional MC1 displayed a transient exacerbation of neutrophil recruitment after global I/R, which diminished by 2 hours. However importantly, enhanced inflammatory responses in both MC1 mutant and MC3 (-/-) mice resulted in increased infarct size and poor functional outcome after focal I/R. Furthermore, we used an in vitro model of leukocyte recruitment to demonstrate these anti-inflammatory actions are also effective in human cells. CONCLUSIONS: These studies reveal for the first time MC control for neutrophil recruitment in the unique pathophysiological context of cerebral I/R, while also demonstrating the potential therapeutic value of targeting multiple MCs in developing effective therapeutics.


Asunto(s)
Isquemia Encefálica/prevención & control , Regulación de la Expresión Génica , Infiltración Neutrófila/genética , ARN Mensajero/genética , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 3/genética , Daño por Reperfusión/complicaciones , Animales , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/biosíntesis , Receptor de Melanocortina Tipo 3/antagonistas & inhibidores , Receptor de Melanocortina Tipo 3/biosíntesis , Daño por Reperfusión/metabolismo
8.
Physiol Behav ; 147: 364-72, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25982086

RESUMEN

Morphine elicits a paradoxical state of increased pain sensitivity, known as morphine-induced hyperalgesia (MIH), which complicates its clinical efficacy. We have previously shown that systemic injections of N-methyl-d-aspartate receptor (NMDAR) and melanocortin-1 receptor (MC1R) antagonists sex-dependently reverse MIH during morphine infusion (40mg/kg/24h) in male and female mice, respectively. This qualitative sex difference is ovarian hormone dependent, as NMDAR antagonists reverse MIH in ovariectomized females but are rendered ineffective following progesterone injection in OVX mice. Here, we utilized intrathecal and intracerebroventricular injection paradigms to assess the contribution of spinal and supraspinal receptors to this sex difference in male and female CD-1 mice. Specifically, we injected NMDAR and MC1R selective antagonists, MK-801 and MSG606 respectively, during morphine infusion. Results illustrated that both spinal and supraspinal MK-801 and MSG606 selectively reversed MIH in males and females, respectively, during morphine infusion. Furthermore, while MK-801 reversed MIH in ovariectomized (OVX) females, MSG606 was most effective in doing so in this same group following an acute subcutaneous progesterone injection. The present studies thus indicate that both spinal and supraspinal NMDARs and MC1Rs underlie the qualitative sex difference observed during morphine infusion in mice, and that the receptors in these loci are also sensitive to sex steroidal modulation.


Asunto(s)
Tronco Encefálico/metabolismo , Hiperalgesia/patología , Receptor de Melanocortina Tipo 1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Caracteres Sexuales , Médula Espinal/metabolismo , Analgésicos Opioides/toxicidad , Animales , Tronco Encefálico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Femenino , Hiperalgesia/inducido químicamente , Masculino , Ratones , Morfina/toxicidad , Naltrexona/administración & dosificación , Ovariectomía , Dimensión del Dolor , Progesterona/farmacología , Progestinas/farmacología , Tiempo de Reacción/efectos de los fármacos , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Médula Espinal/efectos de los fármacos
9.
PLoS One ; 10(4): e0123232, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25860801

RESUMEN

In this study, we showed that adrenocorticotropic hormone (ACTH) promoted erythroblast differentiation and increased the enucleation ratio of erythroblasts. Because ACTH was contained in hematopoietic medium as contamination, the ratio decreased by the addition of anti-ACTH antibody (Ab). Addition of neutralizing Abs (nAbs) for melanocortin receptors (MCRs) caused erythroblast accumulation at specific stages, i.e., the addition of anti-MC2R nAb led to erythroblast accumulation at the basophilic stage (baso-E), the addition of anti-MC1R nAb caused accumulation at the polychromatic stage (poly-E), and the addition of anti-MC5R nAb caused accumulation at the orthochromatic stage (ortho-E). During erythroblast differentiation, ERK, STAT5, and AKT were consecutively phosphorylated by erythropoietin (EPO). ERK, STAT5, and AKT phosphorylation was inhibited by blocking MC2R, MC1R, and MC5R, respectively. Finally, the phosphorylation of myosin light chain 2, which is essential for the formation of contractile actomyosin rings, was inhibited by anti-MC5R nAb. Taken together, our study suggests that MC2R and MC1R signals are consecutively required for the regulation of EPO signal transduction in erythroblast differentiation, and that MC5R signal transduction is required to induce enucleation. Thus, melanocortin induces proliferation and differentiation at baso-E, and polarization and formation of an actomyosin contractile ring at ortho-E are required for enucleation.


Asunto(s)
Eritroblastos/citología , Eritroblastos/metabolismo , Melanocortinas/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Receptor de Melanocortina Tipo 2/metabolismo , Receptores de Melanocortina/metabolismo , Hormona Adrenocorticotrópica/antagonistas & inhibidores , Hormona Adrenocorticotrópica/metabolismo , Anticuerpos Neutralizantes , Diferenciación Celular/fisiología , Células Cultivadas , Eritropoyesis/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Modelos Biológicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 2/antagonistas & inhibidores , Receptor de Melanocortina Tipo 2/genética , Receptores de Melanocortina/antagonistas & inhibidores , Receptores de Melanocortina/genética , Factor de Transcripción STAT5/metabolismo , Transducción de Señal
10.
Amino Acids ; 47(4): 813-23, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25557051

RESUMEN

The purpose of this study was to examine whether the replacement of the positively-charged Lys or Arg linker with a neutral linker could reduce the renal uptake of Arg-Gly-Asp (RGD)-conjugated alpha-melanocyte stimulating hormone (α-MSH) hybrid peptide. The RGD motif {cyclic(Arg-Gly-Asp-DTyr-Asp)} was coupled to [Cys(3,4,10), D-Phe(7), Arg(11)]α-MSH3-13 {(Arg(11))CCMSH} through the neutral ßAla or Ahx {aminohexanoic acid} linker (replacing the Lys or Arg linker) to generate novel RGD-ßAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH hybrid peptides. The receptor-binding affinity and cytotoxicity of RGD-ßAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH were determined in B16/F1 melanoma cells. The melanoma targeting and imaging properties of (99m)Tc-RGD-ßAla-(Arg(11))CCMSH and (99m)Tc-RGD-Ahx-(Arg(11))CCMSH were determined in B16/F1 melanoma-bearing C57 mice. The replacement of the Lys or Arg linker with the ßAla or Ahx linker retained nanomolar receptor-binding affinities and remarkable cytotoxicity of RGD-ßAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH. The receptor-binding affinities of RGD-ßAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH were 0.8 ± 0.05 and 1.3 ± 0.1 nM. Three-hour incubation with 0.1 µM of RGD-ßAla-(Arg(11))CCMSH and RGD-Ahx-(Arg(11))CCMSH decreased the survival percentages of B16/F1 cells by 71 and 67 % as compared to the untreated control cells 5 days post the treatment. The replacement of the Arg linker with the ßAla or Ahx linker reduced the non-specific renal uptake of (99m)Tc-RGD-ßAla-(Arg(11))CCMSH and (99m)Tc-RGD-Ahx-(Arg(11))CCMSH by 62 and 61 % at 2 h post-injection. (99m)Tc-RGD-ßAla-(Arg(11))CCMSH displayed higher melanoma uptake than (99m)Tc-RGD-Ahx-(Arg(11))CCMSH at 0.5, 2, 4, and 24 h post-injection. Enhanced tumor to kidney uptake ratio of (99m)Tc-RGD-ßAla-(Arg(11))CCMSH warranted the further evaluation of (188)Re-labeled RGD-ßAla-(Arg(11))CCMSH as a novel MC1 receptor-targeting therapeutic peptide for melanoma treatment in the future.


Asunto(s)
Riñón/metabolismo , Melanoma/tratamiento farmacológico , Oligopéptidos/química , Radiofármacos/farmacocinética , Tecnecio/química , alfa-MSH/síntesis química , alfa-MSH/farmacocinética , Animales , Línea Celular Tumoral , Femenino , Humanos , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/metabolismo , Radiofármacos/administración & dosificación , Radiofármacos/síntesis química , Radiofármacos/química , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 1/metabolismo , Tecnecio/metabolismo , Distribución Tisular , alfa-MSH/administración & dosificación , alfa-MSH/química
11.
Biochem Biophys Res Commun ; 438(2): 375-81, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23892040

RESUMEN

[8]-Gingerol is an active component of Zinger and shows several pharmacological activities, such as antipyretic and anti-inflammation characteristics. To identify a potential skin-whitening agent, the inhibitory effects of [8]-gingerol on melanogenesis and its mechanism of action were investigated. In the present study, the effects of [8]-gingerol on mushroom tyrosinase, tyrosinase activity and melanin content were determined spectrophotometrically; the expression of melanogenesis-related proteins in B16F10 and B16F1 melanoma cells were determined by Western blotting. Furthermore, the possible signaling pathways involved in [8]-gingerol-mediated depigmentation were also investigated using specific inhibitors. The results revealed that [8]-gingerol (5-100µM) effectively suppressed intracellular tyrosinase activity and decreased the amount of melanin in B16F10 and B16F1 cells. In addition, [8]-gingerol also effectively decreased intracellular reactive species (RS) and reactive oxygen species (ROS) levels at the same dose range. Our results indicated that [8]-gingerol inhibited melanogenesis in B16F10 and B16F1 cells by down-regulation of both mitogen-activated protein kinases (MAPK) and protein kinase A (PKA) signaling pathways or through its antioxidant properties. Hence, [8]-gingerol could be used as an effective skin-whitening agent.


Asunto(s)
Catecoles/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Alcoholes Grasos/farmacología , Sistema de Señalización de MAP Quinasas , Melaninas/biosíntesis , Melanoma/metabolismo , Monofenol Monooxigenasa/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Melanoma Experimental , Ratones , Factor de Transcripción Asociado a Microftalmía/antagonistas & inhibidores , Monofenol Monooxigenasa/antagonistas & inhibidores , Especies Reactivas de Oxígeno , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Transducción de Señal
12.
Chem Biol ; 20(6): 784-95, 2013 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-23790489

RESUMEN

The ß-defensins are a class of small, cationic proteins first recognized as antimicrobial components of the innate and adaptive immune system. More recently, one of the major ß-defensins produced in skin, ß-defensin 3, has been discovered to function as a melanocortin receptor ligand in vivo and in vitro, but its biophysical and pharmacological basis of action has been enigmatic. Here, we report functional and biochemical studies focused on human ß-defensin 3 (HBD3) and melanocortin receptors 1 and 4. Genetic and pharmacologic studies indicate that HBD3 acts as a neutral melanocortin receptor antagonist capable of blocking the action of either stimulatory agonists such as α-melanocyte stimulating hormone or inhibitory inverse agonists such as Agouti signaling protein (ASIP) and Agouti-related protein (AGRP). A comprehensive structure-function analysis demonstrates that two patches of positively charged residues, located on opposite poles of HBD3 and spatially organized by the compact ß-defensin fold, are primarily responsible for high-affinity binding to melanocortin receptors. These findings identify a distinct mode of melanocortin receptor-ligand interactions based primarily on electrostatic complementarity, with implications for designing ligands that target melanocortin and potentially other seven transmembrane receptors.


Asunto(s)
Receptor de Melanocortina Tipo 1/metabolismo , beta-Defensinas/metabolismo , Proteína de Señalización Agouti/agonistas , Proteína de Señalización Agouti/genética , Proteína de Señalización Agouti/metabolismo , Proteína Relacionada con Agouti/agonistas , Proteína Relacionada con Agouti/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 4/química , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Electricidad Estática , beta-Defensinas/genética
13.
Biochemistry ; 52(4): 752-64, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23276279

RESUMEN

γ-MSH (γ-melanocyte-stimulating hormone, H-Tyr-Val-Met-Gly-His-Phe-Arg-Trp-Asp-Arg-Phe-Gly-OH), with its exquisite specificity and potency, has recently created much excitement as a drug lead. However, this peptide is like most peptides susceptible to proteolysis in vivo, which potentially decreases its beneficial activities. In our continued effort to design a proteolytically stable ligand with specific receptor binding, we have engineered peptides by cyclizing γ-MSH using a thioether bridge. A number of novel cyclic truncated γ-MSH analogues were designed and synthesized, in which a thioether bridge was incorporated between a cysteine side chain and an N-terminal bromoacyl group. One of these peptides, cyclo-[(CH(2))(3)CO-Gly(1)-His(2)-D-Phe(3)-Arg(4)-D-Trp(5)-Cys(S-)(6)]-Asp(7)-Arg(8)-Phe(9)-Gly(10)-NH(2), demonstrated potent antagonist activity and receptor selectivity for the human melanocortin 1 receptor (hMC1R) (IC(50) = 17 nM). This novel peptide is the most selective antagonist for the hMC1R to date. Further pharmacological studies have shown that this peptide can specifically target melanoma cells. The nuclear magnetic resonance analysis of this peptide in a membrane-like environment revealed a new turn structure, specific to the hMC1R antagonist, at the C-terminus, where the side chain and backbone conformation of D-Trp(5) and Phe(9) of the peptide contribute to hMC1R selectivity. Cyclization strategies represent an approach for stabilizing bioactive peptides while keeping their full potencies and should boost applications of peptide-based drugs in human medicine.


Asunto(s)
Antineoplásicos/farmacología , Melanoma/tratamiento farmacológico , Péptidos Cíclicos/farmacología , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , gamma-MSH/farmacología , Secuencia de Aminoácidos , Línea Celular Tumoral/efectos de los fármacos , Humanos , Enlace de Hidrógeno , Concentración 50 Inhibidora , Hormonas Estimuladoras de los Melanocitos/química , Hormonas Estimuladoras de los Melanocitos/farmacología , Simulación de Dinámica Molecular , Terapia Molecular Dirigida , Unión Proteica , Estructura Secundaria de Proteína , Receptor de Melanocortina Tipo 1/metabolismo , Relación Estructura-Actividad
14.
Mol Biol Rep ; 40(2): 1987-96, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23090482

RESUMEN

The alpha melanocyte stimulating hormone receptor (MC1R) is one of five G-protein coupled receptors belonging to the melanocortin subfamily, MC1R gene has been known to play a major role in regulating of fur color in mammals, and α-MSH and ACTH are endogenous nonselective agonists for MC1R. However, we found that MC1R was highly expressed in Raw 264.7 cells which were important inflammatory cells involved in the initiation of inflammatory responses. In addition, Cyclic AMP is not only a key molecule in the MC1R signal transduction pathway, but dampen innate immune-mediated responses. These intriguing biological results triggered the further conformation studies; it suggested that MC1R was very likely to be an important role in immunoregulation. In this study, we were to investigate the immunosuppressive effects of MC1R on inflammation in lipopolysaccharide (LPS) stimulated Raw 264.7 cells and LPS induced vivo 2-chloro-1,3,5-trinitrobenzene (TNCB)-induced atopic dermatitis (AD) model. The effects of the MC1R antagonist psoralen on pro-inflammatory cytokines and signaling pathways were analyzed by enzyme-linked immunosorbent assay, western blot, real-time fluorescence quantitative PCR and Histological analysis. Our results show a consistent and marked effect of high concentrations of MC1R antagonist psoralen increased the level of MC1R mRNA in Raw 264.7 cells by cumulative feedback regulation through preferential binding of MC1R. Moreover, as evidenced by inhibiting the LPS-induced TNF-α, IL-6 and enhancing the expression level of cyclic AMP protein in vitro. In vivo study it was also observed that psoralen promoted on histopathologic changes in the skin tissue of TNCB-induced AD mice. Taken together, our results suggest that MC1R decrease the inflammation in vitro and vivo, and might be a therapeutic signaling pathway to against inflammatory diseases.


Asunto(s)
Dermatitis Atópica/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/inmunología , Modelos Animales de Enfermedad , Ficusina/farmacología , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos BALB C , Cloruro de Picrilo , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/genética , Transducción de Señal , Piel/efectos de los fármacos , Piel/patología , Factor de Necrosis Tumoral alfa/metabolismo
15.
Biochemistry (Mosc) ; 78(11): 1228-37, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24460937

RESUMEN

The conventional chemotherapeutic treatment of malignant melanoma still remains poorly efficient in most cases. Thus the use of specific features of these tumors for development of new therapeutic modalities is highly needed. Melanocortin 1 receptor (MC1R) overexpression on the cell surface of the vast majority of human melanomas, making MC1R a valuable marker of these tumors, is one of these features. Naturally, MC1R plays a key role in skin protection against damaging ultraviolet radiation by regulating eumelanin production. MC1R activation is involved in regulation of melanocyte cell division. This article reviews the peculiarities of regulation and expression of MC1R, melanocytes, and melanoma cells, along with the possible connection of MC1R with signaling pathways regulating proliferation of tumor cells. MC1R is a cell surface endocytic receptor, thus considered perspective for diagnostics and targeted drug delivery. A number of new therapeutic approaches that utilize MC1R, including endoradiotherapy with Auger electron and α- and ß-particle emitters, photodynamic therapy, and gene therapy are now being developed.


Asunto(s)
Melanoma/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Endocitosis , Humanos , Hormonas Estimuladoras de los Melanocitos/química , Hormonas Estimuladoras de los Melanocitos/genética , Hormonas Estimuladoras de los Melanocitos/metabolismo , Melanocitos/metabolismo , Melanoma/diagnóstico , Melanoma/terapia , Radiofármacos/uso terapéutico , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapéutico , Transducción de Señal
16.
J Biomol Screen ; 17(8): 1096-101, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22674933

RESUMEN

Cyclic adenosine monophosphate (cAMP) is a second messenger of many G-protein-coupled receptors (GPCRs) and a useful readout molecule to estimate the biological activity of various GPCR-specific agents. Here we report the development and use of a Förster resonance energy transfer (FRET) biosensor for cAMP (Epac2-camps) combined with a baculovirus-based BacMam transduction system. The constructed BacMam-Epac2-camps viral transduction system is a simple and robust tool for ligand screening at the second-messenger level in a variety of mammalian cell lines. The level of biosensor protein expression can easily be adjusted in a dose-dependent manner depending on the multiplicity of viral infection. For setting up the assay, we used a B16F10 murine melanoma cell line with endogenous expression of melanocortin-1 receptor (MC(1)R). The receptor activation was characterized by a set of MC(1)R full and partial agonists. Bivalent ions Ca(2+) as well as Mg(2+) modulated ligand potencies, whereas the effect was ligand and ion specific. Results obtained for MC(1)R indicate that the BacMam-Epac2-camps system may also be applicable for studying the activation of other GPCRs and may be implemented in routine analysis as well as in high-throughput screening.


Asunto(s)
Técnicas Biosensibles , AMP Cíclico/análisis , Transferencia Resonante de Energía de Fluorescencia/métodos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Receptor de Melanocortina Tipo 1/agonistas , Animales , Baculoviridae , Línea Celular Tumoral , AMP Cíclico/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Proteínas Fluorescentes Verdes , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores
17.
J Invest Dermatol ; 132(9): 2255-62, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22572817

RESUMEN

The melanocortin 1 receptor (MC1R), a G(s) protein-coupled receptor, has an important role in human pigmentation. We investigated the regulation of expression and activity of the MC1R in primary human melanocyte cultures. Human ß-defensin 3 (HBD3) acted as an antagonist for MC1R, inhibiting the α-melanocortin (α-melanocyte-stimulating hormone (α-MSH))-induced increase in the activities of adenylate cyclase and tyrosinase, the rate-limiting enzyme for melanogenesis. α-Melanocortin and forskolin, which activate adenylate cyclase, and 12-O-tetradecanoylphorbol-13-acetate, which activates protein kinase C, increased, whereas exposure to UV radiation reduced, MC1R gene and membrane protein expression. Brief treatment with α-MSH resulted in MC1R desensitization, whereas continuous treatment up to 3 hours caused a steady rise in cAMP, suggesting receptor recycling. Pretreatment with agouti signaling protein or HBD3 prohibited responsiveness to α-MSH, but not forskolin, suggesting receptor desensitization by these antagonists. Melanocytes from different donors expressed different levels of the G protein-coupled receptor kinases (GRKs) 2, 3, 5, and 6, as well as ß-arrestin 1. Therefore, in addition to the MC1R genotype, regulation of MC1R expression and activity is expected to affect human pigmentation and the responses to UV.


Asunto(s)
Proteína de Señalización Agouti/farmacología , Melanocortinas/farmacología , Melanocitos/efectos de los fármacos , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , alfa-MSH/farmacología , beta-Defensinas/farmacología , Adenilil Ciclasas/metabolismo , Arrestinas/biosíntesis , Células Cultivadas , Colforsina/farmacología , Quinasas de Receptores Acoplados a Proteína-G/biosíntesis , Humanos , Melanocitos/metabolismo , Melanocitos/efectos de la radiación , Monofenol Monooxigenasa/metabolismo , Proteína Quinasa C/metabolismo , Receptor de Melanocortina Tipo 1/biosíntesis , Pigmentación de la Piel/efectos de los fármacos , Pigmentación de la Piel/fisiología , Pigmentación de la Piel/efectos de la radiación , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Rayos Ultravioleta , beta-Arrestina 1 , beta-Arrestinas
18.
J Control Release ; 161(3): 959-66, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22617522

RESUMEN

Targeted nanoparticle-based delivery systems have been used extensively to develop effective cancer theranostics. However, how targeting ligands affect extravascular transport of nanoparticles in solid tumors remains unclear. Here, we show, using B16/F10 melanoma cells expressing melanocortin type-1 receptor (MC1R), that the nature of targeting ligands, i.e., whether they are agonists or antagonists, directs tumor uptake and intratumoral distribution after extravasation of nanoparticles from tumor vessels into the extravascular fluid space. Pegylated hollow gold nanospheres (HAuNS, diameter=40 nm) coated with MC1R agonist are internalized upon ligand-receptor binding, whereas MC1R antagonist-conjugated HAuNS remain attached on the cell surface. Transcellular transport of agonist-conjugated HAuNS was confirmed by a multilayer tumor cell model and by transmission electron microscopy. MC1R agonist- but not MC1R antagonist-conjugated nanoparticles exhibit significantly higher tumor uptake than nontargeted HAuNS and are quickly dispersed from tumor vessels via receptor-mediated endocytosis and subsequent transcytosis. These results confirm an active transport mechanism that can be used to overcome one of the major biological barriers for efficient nanoparticle delivery to solid tumors.


Asunto(s)
Oro/administración & dosificación , Melanoma Experimental/metabolismo , Nanosferas/administración & dosificación , Receptor de Melanocortina Tipo 1/metabolismo , Transcitosis/fisiología , Animales , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/metabolismo , Femenino , Oro/química , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Nanosferas/química , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores
19.
J Biol Inorg Chem ; 17(4): 491-505, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22286955

RESUMEN

Aiming to apply the multivalency concept to melanoma imaging, we have assessed the in vivo melanocortin type 1 receptor (MC1R)-targeting properties of (99m)Tc(I)-labeled homobivalent peptide conjugates which contain copies of the α-melanocyte-stimulating hormone (α-MSH) analog [Ac-Nle(4), Asp(5), D-Phe(7), Lys(11)]α-MSH4-11 separated by linkers of different length (L(2) nine atoms and L(3) 14 atoms). The MC1R-binding affinity of L(2) and L(3) is significantly higher than that of the monovalent conjugate L(1). Metallation of these conjugates yielded the complexes fac-[M(CO)(3)(k(3)-L)](+) (M is (99m)Tc/Re; 1/1a, L is L(1); 2/2a, L is L(2); 3/3a, L is L(3)), with IC(50) values in the subnanomolar and nanomolar range. The MC1R-mediated internalization of 2 and 3 is higher than that of 1 in B16F1 melanoma cells. Biodistribution studies in melanoma-bearing mice have shown low nonspecific accumulation with a tumor uptake that correlates with IC(50) values. However, no correlation between tumor uptake and valency was found. Nevertheless, 2 displayed the highest tumor retention, and the best tumor to nontarget organ ratios.


Asunto(s)
Melanoma Experimental/diagnóstico , Compuestos de Organotecnecio , Péptidos , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/metabolismo , alfa-MSH/análogos & derivados , Animales , Femenino , Humanos , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Imagen Molecular , Compuestos de Organotecnecio/química , Compuestos de Organotecnecio/farmacocinética , Péptidos/química , Péptidos/farmacocinética , Receptor de Melanocortina Tipo 1/química , Distribución Tisular , Células Tumorales Cultivadas , alfa-MSH/farmacocinética
20.
J Dermatol Sci ; 63(1): 23-32, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21602033

RESUMEN

BACKGROUND: The melanocortin receptor-5 (MC5R) is present in human sebaceous glands, where it is expressed in differentiated sebocytes only. The targeted disruption of MC5R in mice resulted in reduced sebaceous lipid production and a severe defect in water repulsion. OBJECTIVE: To investigate the physiological function of MC5R in human sebaceous glands. METHODS: A novel MC1R and MC5R antagonist (JNJ-10229570) was used to treat primary human sebaceous cells or human skins grafted onto severe combined immunodeficient (SCID) mice. Transcription profiling, lipid analyses, and histological and immunohistochemical staining were used to analyze the effect of MC5R inhibition on sebaceous gland differentiation and sebum production. RESULTS: JNJ-10229570 dose dependently inhibited the production of sebaceous lipids in cultured primary human sebocytes. Topical treatment with JNJ-10229570 of human skins transplanted onto SCID mice resulted in a marked decrease in sebum-specific lipid production, sebaceous gland's size and the expression of the sebaceous differentiation marker epithelial-membrane antigen (EMA). Treatment with flutamide, a known inhibitor of sebum production, gave similar results, validating the human skin/SCID mouse experimental system for sebaceous secretion studies. CONCLUSION: Our data suggest that antagonists of MC1R and MC5R could be effective sebum suppressive agents and might have a potential for the treatment of acne and other sebaceous gland pathologies.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptores de Melanocortina/antagonistas & inhibidores , Glándulas Sebáceas/efectos de los fármacos , Sebo/metabolismo , Administración Cutánea , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Flutamida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Antagonistas de Hormonas/administración & dosificación , Humanos , Masculino , Ratones , Ratones SCID , Mucina-1/metabolismo , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 1/metabolismo , Receptores de Melanocortina/genética , Receptores de Melanocortina/metabolismo , Glándulas Sebáceas/metabolismo , Trasplante de Piel , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...