Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Mol Imaging ; 16: 1536012117737919, 2017.
Article in English | MEDLINE | ID: mdl-29182034

ABSTRACT

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.


Subject(s)
Melanoma/diagnostic imaging , Melanoma/drug therapy , Radiopharmaceuticals/therapeutic use , Receptor, Melanocortin, Type 1/antagonists & inhibitors , alpha-MSH/analogs & derivatives , Animals , Early Detection of Cancer , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/metabolism , Molecular Imaging/methods , Radiopharmaceuticals/pharmacology , Tomography, Emission-Computed, Single-Photon , Up-Regulation/drug effects , alpha-MSH/chemistry
2.
Arterioscler Thromb Vasc Biol ; 35(9): 1936-44, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26112010

ABSTRACT

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.


Subject(s)
Brain Ischemia/prevention & control , Gene Expression Regulation , Neutrophil Infiltration/genetics , RNA, Messenger/genetics , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 3/genetics , Reperfusion Injury/complications , Animals , Brain Ischemia/etiology , Brain Ischemia/metabolism , Disease Models, Animal , Humans , Male , Melanocyte-Stimulating Hormones/pharmacology , Mice , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/biosynthesis , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/biosynthesis , Reperfusion Injury/metabolism
3.
Amino Acids ; 47(4): 813-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25557051

ABSTRACT

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.


Subject(s)
Kidney/metabolism , Melanoma/drug therapy , Oligopeptides/chemistry , Radiopharmaceuticals/pharmacokinetics , Technetium/chemistry , alpha-MSH/chemical synthesis , alpha-MSH/pharmacokinetics , Animals , Cell Line, Tumor , Female , Humans , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Oligopeptides/metabolism , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Technetium/metabolism , Tissue Distribution , alpha-MSH/administration & dosage , alpha-MSH/chemistry
4.
Biochemistry ; 52(4): 752-64, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23276279

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Peptides, Cyclic/pharmacology , Receptor, Melanocortin, Type 1/antagonists & inhibitors , gamma-MSH/pharmacology , Amino Acid Sequence , Cell Line, Tumor/drug effects , Humans , Hydrogen Bonding , Inhibitory Concentration 50 , Melanocyte-Stimulating Hormones/chemistry , Melanocyte-Stimulating Hormones/pharmacology , Molecular Dynamics Simulation , Molecular Targeted Therapy , Protein Binding , Protein Structure, Secondary , Receptor, Melanocortin, Type 1/metabolism , Structure-Activity Relationship
5.
Biochem Biophys Res Commun ; 438(2): 375-81, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23892040

ABSTRACT

[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.


Subject(s)
Catechols/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Fatty Alcohols/pharmacology , MAP Kinase Signaling System , Melanins/biosynthesis , Melanoma/metabolism , Monophenol Monooxygenase/metabolism , Animals , Cell Line, Tumor , Cell Survival , Down-Regulation , Gene Expression Regulation, Enzymologic , Intramolecular Oxidoreductases/antagonists & inhibitors , Melanoma, Experimental , Mice , Microphthalmia-Associated Transcription Factor/antagonists & inhibitors , Monophenol Monooxygenase/antagonists & inhibitors , Reactive Oxygen Species , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Signal Transduction
6.
Mol Biol Rep ; 40(2): 1987-96, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23090482

ABSTRACT

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.


Subject(s)
Dermatitis, Atopic/metabolism , Receptor, Melanocortin, Type 1/metabolism , Animals , Cell Line , Cell Survival/drug effects , Cyclic AMP/metabolism , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/immunology , Disease Models, Animal , Ficusin/pharmacology , Gene Expression , Gene Expression Regulation/drug effects , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Picryl Chloride , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/genetics , Signal Transduction , Skin/drug effects , Skin/pathology , Tumor Necrosis Factor-alpha/metabolism
7.
Biochemistry (Mosc) ; 78(11): 1228-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24460937

ABSTRACT

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.


Subject(s)
Melanoma/metabolism , Receptor, Melanocortin, Type 1/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Endocytosis , Humans , Melanocyte-Stimulating Hormones/chemistry , Melanocyte-Stimulating Hormones/genetics , Melanocyte-Stimulating Hormones/metabolism , Melanocytes/metabolism , Melanoma/diagnosis , Melanoma/therapy , Radiopharmaceuticals/therapeutic use , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 107(47): 20164-71, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-20980662

ABSTRACT

Background adaptation is used by teleosts as one of a variety of camouflage mechanisms for avoidance of predation. Background adaptation is known to involve light sensing by the retina and subsequent regulation of melanophore dispersion or contraction in melanocytes, mediated by α-melanocyte-stimulating hormone and melanin-concentrating hormone, respectively. Here, we demonstrate that an agouti gene unique to teleosts, agrp2, is specifically expressed in the pineal and is required for up-regulation of hypothalamic pmch and pmchl mRNA and melanosome contraction in dermal melanocytes in response to a white background. floating head, a mutant with defective pineal development, exhibits defective up-regulation of mch mRNAs by white background, whereas nrc, a blind mutant, exhibits a normal response. These studies identify a role for the pineal in background adaptation in teleosts, a unique physiological function for the agouti family of proteins, and define a neuroendocrine axis by which environmental background regulates pigmentation.


Subject(s)
Adaptation, Physiological/genetics , Agouti-Related Protein/metabolism , Pigmentation/genetics , Pineal Gland/metabolism , Zebrafish/metabolism , Animals , Gene Expression Regulation/physiology , Melanosomes/metabolism , Pigmentation/physiology , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Zebrafish/genetics
9.
J Biol Inorg Chem ; 17(4): 491-505, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22286955

ABSTRACT

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.


Subject(s)
Melanoma, Experimental/diagnosis , Organotechnetium Compounds , Peptides , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/metabolism , alpha-MSH/analogs & derivatives , Animals , Female , Humans , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Molecular Imaging , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/pharmacokinetics , Peptides/chemistry , Peptides/pharmacokinetics , Receptor, Melanocortin, Type 1/chemistry , Tissue Distribution , Tumor Cells, Cultured , alpha-MSH/pharmacokinetics
10.
J Biol Chem ; 284(46): 31714-25, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19737927

ABSTRACT

Mahogunin ring finger-1 (MGRN1) is a RING domain-containing ubiquitin ligase mutated in mahoganoid, a mouse mutation causing coat color darkening, congenital heart defects, high embryonic lethality, and spongiform neurodegeneration. The melanocortin hormones regulate pigmentation, cortisol production, food intake, and body weight by signaling through five G protein-coupled receptors positively coupled to the cAMP pathway (MC1R-MC5R). Genetic analysis has shown that mouse Mgrn1 is an accessory protein for melanocortin signaling that may inhibit MC1R and MC4R by unknown mechanisms. These melanocortin receptors (MCRs) regulate pigmentation and body weight, respectively. We show that human melanoma cells express 4 MGRN1 isoforms differing in the C-terminal exon 17 and in usage of exon 12. This exon contains nuclear localization signals. MGRN1 isoforms decreased MC1R and MC4R signaling to cAMP, without effect on beta(2)-adrenergic receptor. Inhibition was independent on receptor plasma membrane expression, ubiquitylation, internalization, or stability and occurred upstream of Galpha(s) binding to/activation of adenylyl cyclase. MGRN1 co-immunoprecipitated with MCRs, suggesting a physical interaction of the proteins. Significantly, overexpression of Galpha(s) abolished the inhibitory effect of MGRN1 and decreased co-immunoprecipitation with MCRs, suggesting competition between MGRN1 and Galpha(s) for binding to MCRs. Although all MGRN1s were located in the cytosol in the absence of MCRs, exon 12-containing isoforms accumulated in the nuclei upon co-expression with the receptors. Therefore, MGRN1 inhibits MCR signaling by a new mechanism involving displacement of Galpha(s), thus accounting for key features of the mahoganoid phenotype. Moreover, MGRN1 might provide a novel pathway for melanocortin signaling from the cell surface to the nucleus.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Melanoma/metabolism , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 4/genetics , Ubiquitin-Protein Ligases/metabolism , Binding, Competitive , Blotting, Western , Cells, Cultured , Cyclic AMP/pharmacology , GTP-Binding Protein alpha Subunits/genetics , Humans , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Melanoma/genetics , Melanoma/pathology , Molecular Sequence Data , Protein Isoforms , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/metabolism , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/metabolism , Subcellular Fractions , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
11.
Anesthesiology ; 112(1): 181-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19996949

ABSTRACT

BACKGROUND: N-Methyl-D-aspartate receptor antagonists reverse hyperalgesia during morphine infusion in male mice only. Because the melanocortin-1 receptor can act as a female-specific counterpart to N-methyl-D-aspartate receptors in kappa-opioid analgesic mechanisms, the authors assessed the contribution of melanocortin-1 receptors to the sex-specific mechanisms underlying morphine hyperalgesia. METHODS: The tail-withdrawal test was used to compare the nociceptive responses of male and female C57BL/6J (B6) mice with those of C57BL/6J-Mc(1r(e/e)) mice, spontaneous mutants of the B6 background lacking functional melanocortin-1 receptors, during continuous morphine infusion (1.6 and 40.0 mgkg(-1) . 24 h(-1)). Separate groups of hyperalgesic B6 and outbred CD-1 mice were injected with MK-801 or MSG606, selective N-methyl-D-aspartate and melanocortin-1 receptor antagonists, respectively. RESULTS: Morphine infusion (40.0 mg . kg(-1) . 24 h(-1)) reduced baseline withdrawal latencies by 45-55% in B6 mice of both sexes, indicating hyperalgesia; this increased nociception was manifest in male e/e mice only. Although MK-801 reversed hyperalgesia in male mice only, increasing latencies by 72%, MSG606 increased latencies by approximately 60% exclusively in females. A lower morphine infusion dose (1.6 mg . kg(-1) . 24 h(-1)) reduced baseline withdrawal latencies by 45-52% in B6 and e/e mice of both sexes, which was reversed by MK-801, but not MSG606, in both male and female B6 mice. CONCLUSIONS: The data indicate the sex-specific mediation of high-dose morphine-induced hyperalgesia by N-methyl-d-aspartate and melanocortin-1 receptors in male and female mice, respectively, suggesting a broader relevance of this known sexual dimorphism. The data further indicate that the neural substrates contributing to hyperalgesia are morphine dose-dependent.


Subject(s)
Analgesics, Opioid/toxicity , Hyperalgesia/chemically induced , Receptor, Melanocortin, Type 1/drug effects , Animals , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Female , Hyperalgesia/psychology , Infusions, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/toxicity , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/genetics , Receptors, N-Methyl-D-Aspartate/drug effects , Sex Characteristics
12.
Cancer Biother Radiopharm ; 35(6): 459-473, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32013538

ABSTRACT

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.


Subject(s)
Alpha Particles/therapeutic use , Drug Carriers/chemistry , Melanoma, Experimental/radiotherapy , Radiopharmaceuticals/administration & dosage , Skin Neoplasms/radiotherapy , Tumor Microenvironment/radiation effects , Actinium/administration & dosage , Actinium/pharmacokinetics , Animals , Cell Line, Tumor/transplantation , Computational Biology , Disease Models, Animal , Dose-Response Relationship, Radiation , Female , Gene Expression Regulation, Neoplastic/immunology , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Immunity, Cellular/genetics , Immunity, Cellular/radiation effects , Male , Maximum Tolerated Dose , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Molecular Targeted Therapy/methods , Nanoparticles/chemistry , RNA-Seq , Radiopharmaceuticals/pharmacokinetics , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/metabolism , Silicon Dioxide/chemistry , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tissue Distribution , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
13.
Arch Pharm Res ; 41(3): 324-332, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29397551

ABSTRACT

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.


Subject(s)
Adenylyl Cyclases/physiology , Finasteride/pharmacology , Melanocytes/drug effects , Melanocytes/enzymology , Melanoma, Experimental/enzymology , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Melanoma, Experimental/drug therapy , Mice , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/metabolism
14.
ACS Chem Neurosci ; 9(12): 3015-3023, 2018 12 19.
Article in English | MEDLINE | ID: mdl-29924583

ABSTRACT

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.


Subject(s)
Agouti-Related Protein/chemistry , Amino Acids/chemistry , Peptides/chemistry , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Drug Inverse Agonism , Humans , Ligands , Macrocyclic Compounds , Receptor, Melanocortin, Type 1/drug effects , Receptor, Melanocortin, Type 3/drug effects , Receptor, Melanocortin, Type 4/drug effects , Receptors, Melanocortin/drug effects , Stereoisomerism
15.
Photochem Photobiol ; 93(1): 245-258, 2017 01.
Article in English | MEDLINE | ID: mdl-27645605

ABSTRACT

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.


Subject(s)
DNA Repair , Melanocortins/metabolism , Melanocytes/metabolism , Pyrimidine Dimers/metabolism , Signal Transduction , Ultraviolet Rays/adverse effects , Cyclic AMP/metabolism , Humans , Melanoma/epidemiology , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/metabolism , United States/epidemiology , Xeroderma Pigmentosum/etiology
16.
Oncotarget ; 7(18): 26331-45, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27028866

ABSTRACT

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.


Subject(s)
Agouti Signaling Protein/metabolism , Melanoma, Experimental/pathology , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Agouti Signaling Protein/genetics , Animals , Cell Line, Tumor , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL
17.
Peptides ; 26(11): 2302-13, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15893405

ABSTRACT

We identified a large number of peptide mimotopes of the adrenocorticotropic hormone (ACTH) and the alpha-melanocyte stimulating hormone (alpha-MSH) to analyze better the structure-function relationships of these hormones with the human MC1 receptor (hMC1R). We have investigated the use of phage-display technology to isolate specific peptides of this receptor by using three monoclonal anti-ACTH antibodies (mAbs). A library of 10(8) phage-peptides displaying randomized decapeptides was constructed and used to select phage-peptides that bind to mAbs. Forty-five phage-peptides have been isolated and from their amino acid sequences, we have identified two consensus sequences, EXFRWGKPA and WGXPVGKP, corresponding to the regions 5-13 and 9-16 of ACTH, respectively. A biological assay on cells expressing the hMC1-R was developed to determine the capacity of phage-peptides to stimulate the receptor. Only two phage-peptides showed detectable activity. Thirty-one peptides were synthesized to analyze their biological effect. We identified two weak agonists, EC50=16 and 11 microM, two strong agonists, EC50=25 and 14 nM and a partial antagonist, IC50=36 microM. This work confirmed the modulator agonist role of the regions 11-12 of alpha-MSH and ACTH, and the importance of the methionine residue at position 4 for the stimulation of the hMC1-R. We also identified analogues of the regions 8-17 of ACTH that exhibited a weak activator effect, and of one analogue of the N-terminal regions 1-9 of ACTH and alpha-MSH having a partial antagonist effect. These results may be useful in the development of potential agonists or antagonists of the hMC1R.


Subject(s)
Oligopeptides/pharmacology , Peptide Library , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/antagonists & inhibitors , alpha-MSH/pharmacology , Cell Line , Dose-Response Relationship, Drug , Humans , Oligopeptides/genetics , alpha-MSH/genetics
18.
Mutat Res ; 571(1-2): 133-52, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15748644

ABSTRACT

The constitutive color of our skin plays a dramatic role in our photoprotection from solar ultraviolet radiation (UVR) that reaches the Earth and in minimizing DNA damage that gives rise to skin cancer. More than 120 genes have been identified and shown to regulate pigmentation, one of the key genes being melanocortin 1 receptor (MC1R) that encodes the melanocortin 1 receptor (MC1R), a seven-transmembrane G protein-coupled receptor expressed on the surface of melanocytes. Modulation of MC1R function regulates melanin synthesis by melanocytes qualitatively and quantitatively. The MC1R is regulated by the physiological agonists alpha-melanocyte-stimulating hormone (alphaMSH) and adrenocorticotropic hormone (ACTH), and antagonist agouti signaling protein (ASP). Activation of the MC1R by binding of an agonist stimulates the synthesis of eumelanin primarily via activation of adenylate cyclase. The significance of cutaneous pigmentation lies in the photoprotective effect of melanin, particularly eumelanin, against sun-induced carcinogenesis. Epidermal melanocytes and keratinocytes respond to UVR by increasing their expression of alphaMSH and ACTH, which up-regulate the expression of MC1R, and consequently enhance the response of melanocytes to melanocortins. Constitutive skin pigmentation dramatically affects the incidence of skin cancer. The pigmentary phenotype characterized by red hair, fair complexion, inability to tan and tendency to freckle is an independent risk factor for all skin cancers, including melanoma. The MC1R gene is highly polymorphic in human populations, and allelic variation at this locus accounts, to a large extent, for the variation in pigmentary phenotypes and skin phototypes (SPT) in humans. Several allelic variants of the MC1R gene are associated with the red hair and fair skin (RHC) phenotype, and carrying one of these variants is thought to diminish the ability of the epidermis to respond to DNA damage elicited by UVR. The MC1R gene is considered a melanoma susceptibility gene, and its significance in determining the risk for skin cancer is of tremendous interest.


Subject(s)
Melanocytes/radiation effects , Receptor, Melanocortin, Type 1/physiology , Ultraviolet Rays , Alleles , Humans , Melanins/physiology , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/genetics
19.
PLoS One ; 10(4): e0123232, 2015.
Article in English | MEDLINE | ID: mdl-25860801

ABSTRACT

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.


Subject(s)
Erythroblasts/cytology , Erythroblasts/metabolism , Melanocortins/metabolism , Receptor, Melanocortin, Type 1/metabolism , Receptor, Melanocortin, Type 2/metabolism , Receptors, Melanocortin/metabolism , Adrenocorticotropic Hormone/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Antibodies, Neutralizing , Cell Differentiation/physiology , Cells, Cultured , Erythropoiesis/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 2/antagonists & inhibitors , Receptor, Melanocortin, Type 2/genetics , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction
20.
Physiol Behav ; 147: 364-72, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25982086

ABSTRACT

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.


Subject(s)
Brain Stem/metabolism , Hyperalgesia/pathology , Receptor, Melanocortin, Type 1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Sex Characteristics , Spinal Cord/metabolism , Analgesics, Opioid/toxicity , Animals , Brain Stem/drug effects , Dose-Response Relationship, Drug , Drug Administration Routes , Female , Hyperalgesia/chemically induced , Male , Mice , Morphine/toxicity , Naltrexone/administration & dosage , Ovariectomy , Pain Measurement , Progesterone/pharmacology , Progestins/pharmacology , Reaction Time/drug effects , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Spinal Cord/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL