RESUMO
The MC1R protein is a receptor found in melanocytes that plays a role in melanin synthesis. Mutations in this protein can impact hair color, skin tone, tanning ability, and increase the risk of skin cancer. The MC1R protein is activated by the alpha-melanocyte-stimulating hormone (α-MSH). Previous studies have shown that mutations affect the interaction between MC1R and α-MSH; however, the mechanism behind this process is poorly understood. Our study aims to shed light on this mechanism using molecular dynamics (MD) simulations to analyze the Asp84Glu and Asp294His variants. We simulated both the wild-type (WT) protein and the mutants with and without ligand. Our results reveal that mutations induce unique conformations during state transitions, hindering the switch between active and inactive states and decreasing cellular levels of cAMP. Interestingly, Asp294His showed increased ligand affinity but decreased protein activity, highlighting that tighter binding does not always lead to increased activation. Our study provides insights into the molecular mechanisms underlying the impact of MC1R mutations on protein activity.
Assuntos
AMP Cíclico , Mutação , Receptor Tipo 1 de Melanocortina , alfa-MSH , Humanos , alfa-MSH/química , alfa-MSH/metabolismo , alfa-MSH/genética , Sítios de Ligação , AMP Cíclico/metabolismo , AMP Cíclico/química , Ligantes , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 1 de Melanocortina/química , Receptor Tipo 1 de Melanocortina/metabolismoRESUMO
The prevalence of obesity in children and adolescents is increasing, and it is recognised as a complex disorder that often begins in early childhood and persists throughout life. Both polygenic and monogenic obesity are influenced by a combination of genetic predisposition and environmental factors. Rare genetic obesity forms are caused by specific pathogenic variants in single genes that have a significant impact on weight regulation, particularly genes involved in the leptin-melanocortin pathway. Genetic testing is recommended for patients who exhibit rapid weight gain in infancy and show additional clinical features suggestive of monogenic obesity as an early identification allows for appropriate treatment, preventing the development of obesity-related complications, avoiding the failure of traditional treatment approaches. In the past, the primary recommendations for managing obesity in children and teenagers have been focused on making multiple lifestyle changes that address diet, physical activity, and behaviour, with the goal of maintaining these changes long-term. However, achieving substantial and lasting weight loss and improvements in body mass index (BMI) through lifestyle interventions alone is rare. Recently the progress made in genetic analysis has paved the way for innovative pharmacological treatments for different forms of genetic obesity. By understanding the molecular pathways that contribute to the development of obesity, it is now feasible to identify specific patients who can benefit from targeted treatments based on their unique genetic mechanisms. Conclusion: However, additional preclinical research and studies in the paediatric population are required, both to develop more personalised prevention and therapeutic programs, particularly for the early implementation of innovative and beneficial management options, and to enable the translation of these novel therapy approaches into clinical practice. What is Known: ⢠The prevalence of obesity in the paediatric population is increasing, and it is considered as a multifaceted condition that often begins in early childhood and persists in the adult life. Particularly, rare genetic forms of obesity are influenced by a combination of genetic predisposition and environmental factors and are caused by specific pathogenic variants in single genes showing a remarkable impact on weight regulation, particularly genes involved in the leptin-melanocortin pathway. ⢠Patients who present with rapid weight gain in infancy and show additional clinical characteristics indicative of monogenic obesity should undergo genetic testing, which, by enabling a correct diagnosis, can prevent the development of obesity-related consequences through the identification for appropriate treatment. What is New: ⢠In recent years, advances made in genetic analysis has made it possible to develop innovative pharmacological treatments for various forms of genetic obesity. In fact, it is now achievable to identify specific patients who can benefit from targeted treatments based on their unique genetic mechanisms by understanding the molecular pathways involved in the development of obesity. ⢠As demonstrated over the last years, two drugs, setmelanotide and metreleptin, have been identified as potentially effective interventions in the treatment of certain rare forms of monogenic obesity caused by loss-of-function mutations in genes involved in the leptin-melanocortin pathway. Recent advancements have led to the development of novel treatments, including liraglutide, semaglutide and retatrutide, that have the potential to prevent the progression of metabolic abnormalities and improve the prognosis of individuals with these rare and severe forms of obesity. However, extensive preclinical research and, specifically, additional studies in the paediatric population are necessary to facilitate the translation of these innovative treatment techniques into clinical practice.
Assuntos
Obesidade Infantil , Criança , Adulto , Adolescente , Humanos , Pré-Escolar , Obesidade Infantil/tratamento farmacológico , Obesidade Infantil/genética , Leptina , Predisposição Genética para Doença , alfa-MSH/genética , Aumento de PesoRESUMO
Corneal transplantation is the most common form of tissue transplantation. The success of corneal transplantation mainly relies on the integrity of corneal endothelial cells (CEnCs), which maintain tissue transparency by pumping out excess water from the cornea. After transplantation, the rate of CEnC loss far exceeds that seen with normal aging, which can threaten sight. The underlying mechanisms are poorly understood. Alpha-melanocyte-stimulating hormone (α-MSH) is a neuropeptide that is constitutively found in the aqueous humor with both cytoprotective and immunomodulatory effects. The curent study found high expression of melanocortin 1 receptor (MC1R), the receptor for α-MSH, on CEnCs. The effect of α-MSH/MC1R signaling on endothelial function and allograft survival in vitro and in vivo was investigated using MC1R signaling-deficient mice (Mc1re/e mice with a nonfunctional MC1R). Herein, the results indicate that in addition to its well-known immunomodulatory effect, α-MSH has cytoprotective effects on CEnCs after corneal transplantation, and the loss of MC1R signaling significantly decreases long-term graft survival in vivo. In conclusion, α-MSH/MC1R signaling is critical for CEnC function and graft survival after corneal transplantation.
Assuntos
Córnea/imunologia , Transplante de Córnea , Células Endoteliais/imunologia , Sobrevivência de Enxerto/imunologia , Transdução de Sinais/imunologia , alfa-MSH/imunologia , Animais , Linhagem Celular Transformada , Córnea/patologia , Feminino , Sobrevivência de Enxerto/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 1 de Melanocortina/imunologia , Transdução de Sinais/genética , alfa-MSH/genéticaRESUMO
BACKGROUND: Hyperpigmentation, which causes excessive melanin synthesis and accumulation, is an important issue in the cosmetic industry. Since compounds developed against hyperpigmentation often come with side effects such as skin irritation and contact dermatitis, new studies focus on the use of natural agents that have no side effects. METHODS AND RESULTS: In this study, it was found that the effects of soybean cell culture extract (SCE) on alpha-melanocyte-stimulating hormone (α-MSH) induced melanogenesis in B16-F10 murine melanoma cells. The cells were incubated with SCE for 48 h after treatment with αMSH for 24 h to analysis the melanin content, cellular tyrosinase activity, and gene and protein expression. SCE at 1 mg/mL decreased melanin content and tyrosinase activity by 34% and 24%, respectively, compared to the α-MSH-treated group, which did not decrease cell viability. In addition, SCE (1 mg/mL) downregulated the expression of tyrosinase (TYR), tyrosinase-related protein (TRP)-1, tyrosinase-related protein (TRP)-2, and microphthalmia-associated transcription factor (MITF) genes 1.5-, 1.5-, 2-, and 2-fold, respectively. Furthermore, SCE inhibited the expression of TYR, TRP1, and TRP2 by decreasing the expression of MITF, as shown in a western blot assay. CONCLUSIONS: This study suggests that SCE reveals dose-dependent inhibition of melanin synthesis through the suppression of tyrosinase activity as well as molecular levels of TYR, TRP1, TRP2, and MITF in B16-F10 murine melanoma cells. Therefore, SCE has the potential to be an effective and natural skin-whitening agent for application in the cosmetic industry.
Assuntos
Hiperpigmentação , Melanoma Experimental , Animais , Técnicas de Cultura de Células , Extratos Celulares , Linhagem Celular Tumoral , Melaninas/metabolismo , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Monofenol Mono-Oxigenase , Extratos Vegetais/farmacologia , Glycine max/metabolismo , alfa-MSH/genética , alfa-MSH/metabolismo , alfa-MSH/farmacologiaRESUMO
As a key gene for balancing energy and regulating feeding behavior, MC4R is relevant to the growth of ruminants. In this presentation, a highly conserved c.612A>G site in the coding sequence (CDS) of MC4R has been selected during a selective sweep analysis of 35 Yiling goats and 20 other wild goats. This site mutation results in an amino acid change from Ile to Met. The genotyping analysis of the c.612A>G site revealed that the A allele was the dominant allele in the domestic goat populations, while the wild goat individuals only had the G allele. For a better understanding of the biological significance of this site, we examined the protein localization and signal detection to explain the function of the two MC4R receptors. The results showed that both the M204 and I204 receptors can normally localize on the membrane. When stimulating the M204 type without α-MSH, it was defective at the level of basal cAMP and decreased significantly against the I204 type. In contrast, the signaling capacity of the M204 receptor was also lower than that of I204 under the stimulation of α-MSH. In the ERK1/2 pathway, stimulating MC4R with NDP-α-MSH, both the M204 and I204 receptors had normal pERK1/2 levels. These results indicate that the p.I204M mutation may change the function by damaging the constitutive activity and signaling, and thus may regulate goats' appetite. This study has potential application for rearing domestic goats.
Assuntos
Receptor Tipo 4 de Melanocortina , alfa-MSH , Animais , Cabras/genética , Cabras/metabolismo , Mutação , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , Transdução de Sinais/fisiologia , alfa-MSH/química , alfa-MSH/genética , alfa-MSH/metabolismoRESUMO
The individuals carrying melanocortin-1 receptor (MC1R) variants, especially those associated with red hair color, fair skin, and poor tanning ability (RHC trait), are more prone to melanoma; however, the underlying mechanism is poorly defined. Here, we report that UVB exposure triggers phosphatase and tensin homolog (PTEN) interaction with wild-type (WT), but not RHC-associated MC1R variants, which protects PTEN from WWP2-mediated degradation, leading to AKT inactivation. Strikingly, the biological consequences of the failure of MC1R variants to suppress PI3K/AKT signaling are highly context dependent. In primary melanocytes, hyperactivation of PI3K/AKT signaling leads to premature senescence; in the presence of BRAF(V600E), MC1R deficiency-induced elevated PI3K/AKT signaling drives oncogenic transformation. These studies establish the MC1R-PTEN axis as a central regulator for melanocytes' response to UVB exposure and reveal the molecular basis underlying the association between MC1R variants and melanomagenesis.
Assuntos
Regulação da Expressão Gênica/efeitos da radiação , Melanócitos/metabolismo , Melanoma Experimental/patologia , PTEN Fosfo-Hidrolase/metabolismo , Receptor Tipo 1 de Melanocortina/metabolismo , Pigmentação da Pele/fisiologia , Raios Ultravioleta , Animais , Western Blotting , Células Cultivadas , Humanos , Técnicas Imunoenzimáticas , Melanócitos/efeitos da radiação , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Camundongos , Mutação/genética , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 1 de Melanocortina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Pigmentação da Pele/efeitos da radiação , alfa-MSH/genética , alfa-MSH/metabolismoRESUMO
The production of α-melanocyte-stimulating hormone (α-MSH), a peptide hormone composed of 13 amino acids, is attempted by recombinant expression using E. coli as the host. To achieve this aim, a synthetic gene containing eight tandem repeats of msh gene (8msh) was designed for ribosomal synthesis of 8 α-MSH. The merit of the strategy is to diminish the peptide toxicity against the host cell and to achieve a higher production yield. Pepsin cleavage sites are introduced between the peptides for enzymatic proteolysis to obtain the monomeric peptide of α-MSH. The constructed plasmid was transformed into different strains of E. coli hosts, and E. coli XL1-Blue with gene 8msh revealed the highest yield of 8 α-MSH. Although 8 α-MSH was fractionalized in the insoluble pellets after cell lysis, pepsin cleavage was able to produce soluble α-MSH peptide, as analyzed and confirmed by mass spectrometry and peptide activity assays. The production of α-MSH was quantified using HPLC with a yield of 42.9 mg/L of LB culture. This study demonstrates the feasibility of producing α-MSH using recombinant expression of tandem repeat gene. The production procedure involves minimal post-treatment and processing and can be scaled up for industrial application.
Assuntos
alfa-MSH/biossíntese , alfa-MSH/genética , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Linhagem Celular Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Genes Sintéticos , Melaninas/biossíntese , Melanoma Experimental , Camundongos , Pepsina A/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem , Sequências de Repetição em Tandem/genética , alfa-MSH/administração & dosagemRESUMO
The purpose of this study was to examine the melanoma targeting and imaging properties of 99mTc(CO)3-NOTA-GGNle-CycMSHhex {1,4,7-triazacyclononane-1,4,7-triyl-triacetic acid-GlyGlyNle-c[Asp-His-DPhe-Arg-Trp-Lys]-CONH2} and 99mTc(CO)3-NODAGA-GGNle-CycMSHhex {1,4,7-triazacyclononane,1-gluteric acid-4,7-acetic acid-GlyGlyNle-c[Asp-His-DPhe-Arg-Trp-Lys]-CONH2} on B16/F10 melanoma-bearing C57 mice to demonstrate the feasibility of NOTA/NODAGA as metal chelators for 99mTc(CO)3+ radiolabeling. NOTA/NODAGA-GGNle-CycMSHhex were synthesized using fluorenylmethoxycarbonyl (Fmoc) chemistry. The melanocortin-1 (MC1) receptor binding affinities of the peptides were determined on B16/F10 melanoma cells. The biodistribution of 99mTc(CO)3-NOTA-GGNle-CycMSHhex and 99mTc(CO)3-NODAGA-GGNle-CycMSHhex were determined on B16/F10 melanoma-bearing C57 mice at 2 h postinjection to select a lead peptide for further evaluation. The melanoma targeting and imaging properties of 99mTc(CO)3-NOTA-GGNle-CycMSHhex and 99mTc(CO)3-NODAGA-GGNle-CycMSHhex were determined on B16/F10 melanoma-bearing C57 mice. The IC50 values of NOTA/NODAGA-GGNle-CycMSHhex were 0.8 ± 0.1 and 0.9 ± 0.1 nM on B16/F10 cells. 99mTc(CO)3-NOTA-GGNle-CycMSHhex and 99mTc(CO)3-NODAGA-GGNle-CycMSHhex were readily prepared via the [99mTc(CO)3(OH2)3]+ intermediate and displayed MC1R-specific binding on B16/F10 cells. 99mTc(CO)3-NOTA-GGNle-CycMSHhex was further evaluated as a lead peptide because of its higher tumor uptake (19.76 ± 3.62% ID/g) and lower kidney uptake (1.59 ± 0.52% ID/g) at 2 h postinjection than 99mTc(CO)3-NODAGA-GGNle-CycMSHhex. The B16/F10 melanoma uptake of 99mTc(CO)3-NOTA-GGNle-CycMSHhex was 16.07 ± 4.47, 19.76 ± 3.62, 11.30 ± 2.81, and 3.16 ± 2.28% ID/g at 0.5, 2, 4, and 24 h postinjection, respectively. 99mTc(CO)3-NOTA-GGNle-CycMSHhex showed high tumor to normal organ uptake ratios after 2 h postinjection. The B16/F10 melanoma lesions were clearly visualized by SPECT/CT using 99mTc(CO)3-NOTA-GGNle-CycMSHhex as an imaging probe at 2 h postinjection. High tumor uptake, low kidney uptake, and fast urinary clearance of 99mTc(CO)3-NOTA-GGNle-CycMSHhex highlighted its potential for melanoma imaging and facilitated the evaluation of 188Re(CO)3-NOTA-GGNle-CycMSHhex for melanoma therapy.
Assuntos
Compostos Heterocíclicos com 1 Anel/química , Rim/metabolismo , Lactamas/química , Melanoma Experimental/metabolismo , Tecnécio/química , alfa-MSH/química , alfa-MSH/genética , Animais , Transporte Biológico/fisiologia , Linhagem Celular Tumoral , Quelantes/química , Quelantes/metabolismo , Ciclização/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Camundongos , Receptor Tipo 1 de Melanocortina/metabolismo , Distribuição Tecidual/fisiologia , alfa-MSH/metabolismoRESUMO
The purpose of this study was to understand the changes of metabolic pathway induced by alpha-melanocyte-stimulating hormone (α-MSH) in B16F10 melanoma cells in an untargeted metabolomics approach. Cells were treated with 100 nM of α-MSH and then incubated for 48 h. α-MSH increased tyrosinase activity and melanin content by 56.5 and 61.7%, respectively, compared to untreated cells after 48 h of cultivation. The clear separation between groups was observed in the principal component analysis score plot, indicating that the levels of metabolites of melanoma cells were altered by treatment with α-MSH. Metabolic pathways affected by α-MSH were involved in some amino acid metabolisms. The increased levels of fumaric acid, malic acid, oxaloacetic acid and citric acid related to the citric acid cycle pathway after α-MSH treatment suggested enhanced energy metabolism. Metabolic pathways altered by α-MSH treatment can provide useful information to develop new skin pigmentation inhibitors or anti-obesity drugs.
Assuntos
Melaninas/metabolismo , Melanoma Experimental/tratamento farmacológico , Metabolômica , alfa-MSH/genética , Animais , Linhagem Celular Tumoral , Humanos , Melaninas/genética , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Redes e Vias Metabólicas/genética , Camundongos , Pigmentação/genética , Transdução de Sinais/genética , alfa-MSH/metabolismo , alfa-MSH/farmacologiaRESUMO
The individual parts of Morus alba L. including root bark, branches, leaves, and fruits are used as a cosmetic ingredient in many Asian countries. This study identified several anti-melanogenesis constituents in a 70% ethanol extract of M. alba leaves. The ethyl acetate fraction of the initial ethanol extract decreased the activity of tyrosinase, a key enzyme in the synthetic pathway of melanin. Twelve compounds were isolated from this fraction and their structures were identified based on spectroscopic spectra. Then, the authors investigated the anti-melanogenesis effects of the isolated compounds in B16-F10 mouse melanoma cells. Compounds 3 and 8 significantly inhibited not only melanin production but also intracellular tyrosinase activity in alpha-melanocyte-stimulating-hormone (α-MSH)-induced B16-F10 cells in a dose-dependent manner. These same compounds also inhibited melanogenesis-related protein expression such as microphthalmia-associated transcription factor (MITF), tyrosinase, and tyrosinase-related protein-1 (TRP-1). Compound 3 modulated the cAMP-responsive element-binding protein (CREB) and p38 signaling pathways in α-MSH-activated B16-F10 melanoma cells, which resulted in the anti-melanogenesis effects. These results suggest that compound 3, isolated from M. alba leaves, could be used to inhibit melanin production via the regulation of melanogenesis-related protein expression.
Assuntos
Melaninas/biossíntese , Monofenol Mono-Oxigenase/antagonistas & inibidores , Morus/química , Extratos Vegetais/farmacocinética , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Melaninas/metabolismo , Melanoma Experimental/metabolismo , Camundongos , Monofenol Mono-Oxigenase/metabolismo , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Folhas de Planta/química , alfa-MSH/genéticaRESUMO
We previously found that ultraviolet B (UVB) could stimulate the paraventricular nucleus (PVN) with activation the systemic hypothalamic-pituitary- adrenal (HPA) axis. To investigate whether UVB can also stimulate other hypothalamic nuclei, we tested its effect on the proopiomelanocortin (POMC) related signalling system in the arcuate nucleus (ARC) of female C57BL/6 and FVB albino mice. The shaved back skin of the mice was irradiated with either 100 or 400 mJ/cm2 of UVB. After 1, 3, 6 and 12 h, blood and hypothalamus were collected and processed for gene and protein expression, and measurement of α-MSH and ß-endorphin (ß-END) levels. An in situ immunohistochemical examination was performed for melanocortin receptor 4 (MC4R) and POMC-derived α-MSH. The expression of Pomc and MC4R mRNAs was stimulated, whereas that of AgRP was inhibited after exposure to UVB. It was accompanied by an increased number of both α-MSH- and MC4R-immunoreactive neurons in the ARC, and by increased levels of α-MSH and ß-END (both found in the hypothalamus and plasma). This surprising discovery of UVB stimulating the POMC system in the ARC, accompanied by the increased plasma levels of α-MSH and ß-END, paves the way for exciting areas of research on the communication between the skin and the brain, as well as is suggesting a new role for UVB in regulation of body metabolism.
Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Pró-Opiomelanocortina/biossíntese , Pele/efeitos da radiação , Raios Ultravioleta , Proteína Relacionada com Agouti/biossíntese , Proteína Relacionada com Agouti/genética , Animais , Metabolismo Basal , Feminino , Regulação da Expressão Gênica/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Pró-Opiomelanocortina/genética , RNA Mensageiro/biossíntese , Receptor Tipo 4 de Melanocortina/biossíntese , Receptor Tipo 4 de Melanocortina/genética , Transdução de Sinais/fisiologia , Organismos Livres de Patógenos Específicos , alfa-MSH/biossíntese , alfa-MSH/sangue , alfa-MSH/genética , beta-Endorfina/biossíntese , beta-Endorfina/sangue , beta-Endorfina/genéticaRESUMO
Neuroinflammation constitutes a principal process involved in the progression of various central nervous system (CNS) disorders, including Parkinson's disease, Alzheimer's disease, ischemic stroke, and traumatic brain injury. The safety and efficacy of potential neuroprotective therapeutic agents is controversial and limited. Alpha-melanocyte-stimulating hormone (α-MSH) as a tridecapeptide derived from pro-opiomelanocortin displays potent anti-inflammatory and protective effects with a wide therapeutic window in brain damage. However, it is difficult to deliver effective concentrations of α-MSH into brain tissue via nondirect application. Besides, the half-life of the tridecapeptide is only a few minutes. In the present study, we generated a novel TAT-HSA-α-MSH by genetically fusing α-MSH with N-terminus 11-amino acid protein transduction domain of the human immunodeficiency virus Tat protein (TAT) and human serum albumin (HSA), which showed favorable pharmacokinetic properties and can effectively cross the blood brain barrier (BBB). The findings showed that TAT-HSA-α-MSH significantly inhibits NF-κB activation in human glioma cells A172 and tumor necrosis factor-α (TNF-α) production in experimental brain inflammation. These results indicate that TAT-HSA-α-MSH may be a potential therapeutic agent for treating neuroinflammation which plays a fundamental role in CNS disorders.
Assuntos
Encéfalo/metabolismo , Encefalite/tratamento farmacológico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , alfa-MSH/farmacologia , alfa-MSH/farmacocinética , Animais , Barreira Hematoencefálica , Encéfalo/efeitos dos fármacos , Encefalite/metabolismo , Regulação da Expressão Gênica , Meia-Vida , Humanos , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Albumina Sérica/química , Albumina Sérica/genética , Fator de Necrose Tumoral alfa/metabolismo , alfa-MSH/administração & dosagem , alfa-MSH/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genéticaRESUMO
Most of our understanding of G protein-coupled receptor (GPCR) activation has been focused on the direct interaction between diffusible ligands and their seven-transmembrane domains. However, a number of these receptors depend on their extracellular N-terminal domain for ligand recognition and activation. To dissect the molecular interactions underlying both modes of activation at a single receptor, we used the unique properties of the melanocortin-4 receptor (MC4R), a GPCR that shows constitutive activity maintained by its N-terminal domain and is physiologically activated by the peptide α-melanocyte stimulating hormone (αMSH). We find that activation by the N-terminal domain and αMSH relies on different key residues in the transmembrane region. We also demonstrate that agouti-related protein, a physiological antagonist of MC4R, acts as an inverse agonist by inhibiting N terminus-mediated activation, leading to the speculation that a number of constitutively active orphan GPCRs could have physiological inverse agonists as sole regulators.
Assuntos
Receptor Tipo 4 de Melanocortina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Membrana Celular , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Modelos Moleculares , Plasmídeos , Conformação Proteica , Estrutura Terciária de Proteína , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/genética , alfa-MSH/genética , alfa-MSH/metabolismoRESUMO
Mutations that perturb leptin-melanocortin signaling are known to cause hyperphagia and obesity, but energy expenditure has not been well studied outside rodents. We report on a common canine mutation in pro-opiomelanocortin (POMC), which prevents production of ß-melanocyte-stimulating hormone (ß-MSH) and ß-endorphin but not α-MSH; humans, similar to dogs, produce α-MSH and ß-MSH from the POMC propeptide, but rodents produce only α-MSH. We show that energy expenditure is markedly lower in affected dogs, which also have increased motivational salience in response to a food cue, indicating increased wanting or hunger. There was no difference in satiety at a modified ad libitum meal or in their hedonic response to food, nor disruption of adrenocorticotropic hormone (ACTH) or thyroid axes. In vitro, we show that ß-MSH signals comparably to α-MSH at melanocortin receptors. These data implicate ß-MSH and ß-endorphin as important in determining hunger and moderating energy expenditure and suggest that this role is independent of the presence of α-MSH.
Assuntos
beta-Endorfina , beta-MSH , Humanos , Cães , Animais , beta-Endorfina/genética , Metabolismo Basal , Pró-Opiomelanocortina/genética , Fome , alfa-MSH/genéticaRESUMO
The melanocortin 1 receptor (MC1R), a key regulator of melanogenesis, is known to control inflammation, acting in concert with the MC1R ligand α-melanocyte-stimulating hormone. Although cell migration is a key event in inflammation, few studies have addressed the function of MC1R in this context. Using highly motile melanoma cells, we found that the expression level of MC1R was associated with the extent of migration of mouse melanoma cells, suggesting that MC1R plays a functional role in controlling this migration. Overexpression of MC1R enhanced melanoma cell migration, whereas the opposite was true when MC1R levels were knocked down using small inhibitory RNAs. Interestingly, MC1R expression enhanced the synthesis of syndecan-2, a cell surface heparan sulfate proteoglycan known to be involved in melanoma cell migration. Knockdown of syndecan-2 expression decreased MC1R-mediated cell migration. Further, MC1R inhibited the activation of p38 MAPK, subsequently enhancing expression of sydnecan-2, in parallel with an increase in the extent of cell migration. Consistently, activation of p38 by H(2)O(2) inhibited syndecan-2 expression and cell migration, whereas inhibition of p38 activation enhanced syndecan-2 expression and cell migration. Finally, we found that α-melanocyte-stimulating hormone inhibited MC1R-mediated cell migration via activation of p38 and inhibition of syndecan-2 expression. Together, the data strongly suggest that MC1R regulates melanoma cell migration via inhibition of syndecan-2 expression.
Assuntos
Movimento Celular , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Receptor Tipo 1 de Melanocortina/metabolismo , Sindecana-2/biossíntese , Animais , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Técnicas de Silenciamento de Genes , Humanos , Peróxido de Hidrogênio/farmacologia , Melanoma/genética , Melanoma/patologia , Camundongos , Proteínas de Neoplasias/genética , Oxidantes/farmacologia , Receptor Tipo 1 de Melanocortina/genética , Sindecana-2/genética , alfa-MSH/genética , alfa-MSH/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismoRESUMO
Caffeic acid phenethyl ester (1), a natural compound found in various plants and propolis, is a well-known anti-inflammatory, immunomodulatory, and cytotoxic agent. The present study aimed to investigate the molecular events underlying the antimelanogenic activity of 1 in alpha-melanocyte stimulating hormone (α-MSH)-stimulated B16-F10 melanoma cells. In this investigation, 1 effectively reduced α-MSH-stimulated melanin synthesis by suppressing expression of melanogenic enzymes such as tyrosinase, tyrosinase-related protein-1 (TRP-1), and tyrosinase-related protein-2 (TRP-2), although this compound did not directly inhibit tyrosinase enzyme activity. On the other hand, the expression and nuclear translocation of microphthalmia-associated transcription factor (MITF) as a key transcription factor for tyrosinase expression regulating melanogenesis were not affected by treatment with 1. The upstream signaling pathways including cAMP response element-binding protein (CREB), glycogen synthase kinase-3ß (GSK-3ß), and Akt for activation and expression of MITF were also not influenced by 1. Interestingly, 1 inhibited transcriptional activity of a tyrosinase promoter by suppressing the interaction of MITF protein with an M-box containing a CATGTG motif on the tyrosinase promoter. Given the important role of MITF in melanogenesis, suppression of 1 on the function of MITF to transactivate tyrosinase promoter may present a novel therapeutic approach to treat hyperpigmentation disorders.
Assuntos
Ácidos Cafeicos/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fator de Transcrição Associado à Microftalmia/genética , Oxirredutases/metabolismo , Álcool Feniletílico/análogos & derivados , Animais , Western Blotting , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Interferon Tipo I , Oxirredutases Intramoleculares/metabolismo , Levodopa/metabolismo , Melaninas/biossíntese , Melaninas/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/metabolismo , Estrutura Molecular , Monofenol Mono-Oxigenase/metabolismo , Álcool Feniletílico/farmacologia , Proteínas da Gravidez , Ativação Transcricional , alfa-MSH/genética , alfa-MSH/metabolismoRESUMO
Natural human skin colour is determined both by environmental exposure to ultraviolet light and through inherited genetic variation in a very limited number of genes. Variation of a non-synonymous single-nucleotide polymorphism (nsSNP; rs1426654) in the gene (SLC24A5) encoding the NCKX5 protein is associated with differences in constitutive skin colour in South Asians. The nsSNP encodes the substitution of alanine for threonine at residue 111 (A111T) near a transmembrane region required for exchanger activity, a region which is highly conserved across different species and between NCKX family members. We have shown that NCKX5 is located at the trans-Golgi network of melanocytes and functions as a potassium-dependent sodium-calcium exchanger. When heterologously expressed, the 111T variant of NCKX5 shows significantly lower exchanger activity than the A111 variant. We have postulated that lower exchanger activity causes the reduced melanogenesis and lighter skin in Thr111-positive individuals. We used gene expression microarrays with qPCR replication and validation to assess the impact of siRNA-mediated knockdown of SLC24A5 on the transcriptome of cultured normal human melanocytes (NHM). Very few genes associated with melanogenesis were altered at the transcript level except for MC1R, suggesting that SLC24A5 interacts with at least one well-characterized melanogenic signalling pathway. More surprisingly, the expression of a number of cholesterol homeostatic genes was altered after SLC24A5 knockdown, and the total cholesterol content of NHM was increased. Cholesterol has previously been identified as a potential melanogenic regulator, and our data imply that NCKX5 exchanger function influences natural variation in skin pigmentation via a novel, unknown mechanism affecting cellular sterol levels.
Assuntos
Antiporters/metabolismo , Colesterol/metabolismo , Regulação da Expressão Gênica/fisiologia , Homeostase/fisiologia , Melanócitos/metabolismo , Receptor Tipo 1 de Melanocortina/biossíntese , Pigmentação da Pele/fisiologia , Pele/metabolismo , alfa-MSH/biossíntese , Substituição de Aminoácidos , Antiporters/genética , Colesterol/genética , Perfilação da Expressão Gênica , Humanos , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Estrutura Terciária de Proteína , Receptor Tipo 1 de Melanocortina/genética , alfa-MSH/genética , Rede trans-Golgi/genética , Rede trans-Golgi/metabolismoRESUMO
The ubiquitous overexpression of agouti-signaling protein (ASP), a paracrine-signaling molecule that regulates pigment-type switching in the hair follicle of the mouse, is responsible for the obesity and yellow pelage of the Yellow mouse (A(y)). Mahogany (Attractin, Atrn/mg) and mahoganoid (Mahogunin Ring Finger-1, Mgrn1/md) are mutations epistatic to A(y). These mutations have been described as suppressors of ASP action, blocking its antagonizing effects on the melanocortin 1 and 4 receptors (MC1R and MC4R) in the skin and the brain, respectively, via unknown mechanisms. Here, we describe the molecular bases for the md- and mg-dependent rescue of the A(y) phenotype at the MC4R. We show that overexpression of ASP inhibits the rise in cAMP levels in response to α-melanocyte-stimulating hormone, an MC4R agonist, by blocking ligand binding and by directing MC4R trafficking to the lysosome. Loss-of-function of either attractin or MGRN1 blocks ASP-dependent MC4R degradation and promotes increased trafficking of internalized MC4R to the cell surface, but it does not restore α-melanocyte-stimulating hormone-dependent cAMP signaling. We propose that MGRN1 and attractin are components of an evolutionarily conserved receptor trafficking pathway and that the md and mg mutations rescue the A(y) phenotypes by a primarily cAMP-independent mechanism promoting trafficking of MC4R and likely MC1R away from the lysosome toward the cell surface.
Assuntos
Endossomos/metabolismo , Proteínas de Membrana/metabolismo , Mutação , Obesidade/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteína Agouti Sinalizadora/genética , Proteína Agouti Sinalizadora/metabolismo , Animais , AMP Cíclico/genética , AMP Cíclico/metabolismo , Endossomos/genética , Expressão Gênica/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Obesidade/genética , Transporte Proteico/genética , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 1 de Melanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/genética , Transdução de Sinais/genética , Ubiquitina-Proteína Ligases/genética , alfa-MSH/genética , alfa-MSH/metabolismoAssuntos
Carboxipeptidases/química , Peptídeos e Proteínas de Sinalização Intercelular/química , Placenta/química , alfa-MSH/química , Sequência de Aminoácidos , Animais , Apelina , Carboxipeptidases/genética , Carboxipeptidases/isolamento & purificação , Carboxipeptidases/metabolismo , Colipases/química , Colipases/genética , Colipases/metabolismo , Metabolismo Energético/genética , Precursores Enzimáticos/química , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Feminino , Expressão Gênica , Grelina/química , Grelina/genética , Grelina/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Cinética , Placenta/enzimologia , Gravidez , Proteólise , Especificidade por Substrato , alfa-MSH/genética , alfa-MSH/metabolismoRESUMO
Alpha-melanocyte stimulating hormone (alpha-MSH) is a proopiomelanocortin derivative and a multi-function neuropeptide, well know for its pigment-inducing capacity, inhibitory action on proinflammatory cytokines and chemoattractant cytokines, and suppressive action on collagen synthesis. Human Tenon's capsule fibroblasts (HTF) are the main effector cells in the initiation and mediation of wound healing and fibrotic scar formation after trabeculectomy. In this study the effects of alpha-MSH on proliferation of HTF stimulated by transforming growth factor beta1 (TGF-beta1), have been investigated and discussed. Fibroblasts were cultured in Dulbecco's modified Eagle's medium (DMEM) in the control group, and in DMEM with TGF-beta1 at concentration of 10(-12) M in the TGF-beta1 group, and DMEM with 10(-12) M TGF-beta1 and alpha-MSH ranging from 0, 10(-8) to 10(-4) M in the TGF-beta1/alpha-MSH groups. Cell proliferation was assessed 48 h later by the CellTiter 96 Aqueous One Solution Cell Proliferation Assay. After administration of TGF-beta1 at a concentration of 10(-12) M, or TGF-beta1 at 10(-12) M plus alpha-MSH at 10(-6) M, the mRNA level of type I (alpha1) collagen, fibronectin, TNF-alpha, intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), MMP-1, MMP-2, TIMP-1, and TIMP-2 in HTF were analyzed using the real time reverse transcription polymerase chain reaction. Alpha-MSH demonstrated an inhibitory effect on the proliferation of HTF induced by TGF-beta1 in a dose-dependent manner, when the concentration was lower than 10(-5) M, and a suppressive effect on the mRNA expression of type I (alpha1) collagen, TNF-alpha, ICAM-1 and VCAM-1, which were up-regulated by TGF-beta1. Our results showed a reverse effect of alpha-MSH on the imbalance between MMPs and TIMPs compared with TGF-beta1. Based on all these results, we conclude that alpha-MSH could effectively suppress HTF proliferation and modulate correlative genes in collagen synthesis stimulated by TGF-beta1, which implies that alpha-MSH could be exploited in the treatment of conjunctival fibrotic scar disorder.