Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int J Mol Sci ; 24(6)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36982309

ABSTRACT

N-propionyl-4-S-cysteaminylphenol (N-Pr-4-S-CAP) is a substrate for tyrosinase, which is a melanin biosynthesis enzyme and has been shown to be selectively incorporated into melanoma cells. It was found to cause selective cytotoxicity against melanocytes and melanoma cells after selective incorporation, resulting in the induction of anti-melanoma immunity. However, the underlying mechanisms for the induction of anti-melanoma immunity remain unclear. This study aimed to elucidate the cellular mechanism for the induction of anti-melanoma immunity and clarify whether N-Pr-4-S-CAP administration could be a new immunotherapeutic approach against melanoma, including local recurrence and distant metastasis. A T cell depletion assay was used for the identification of the effector cells responsible for N-Pr-4-S-CAP-mediated anti-melanoma immunity. A cross-presentation assay was carried out by using N-Pr-4-S-CAP-treated B16-OVA melanoma-loaded bone marrow-derived dendritic cells (BMDCs) and OVA-specific T cells. Administration of N-Pr-4-S-CAP induced CD8+ T cell-dependent anti-melanoma immunity and inhibited the growth of challenged B16F1 melanoma cells, indicating that the administration of N-Pr-4-S-CAP can be a prophylactic therapy against recurrence and metastasis of melanoma. Moreover, intratumoral injection of N-Pr-4-S-CAP in combination with BMDCs augmented the tumor growth inhibition when compared with administration of N-Pr-4-S-CAP alone. BMDCs cross-presented a melanoma-specific antigen to CD8+ T cells through N-Pr-4-S-CAP-mediated melanoma cell death. Combination therapy using N-Pr-4-S-CAP and BMDCs elicited a superior anti-melanoma effect. These results suggest that the administration of N-Pr-4-S-CAP could be a new strategy for the prevention of local recurrence and distant metastasis of melanoma.


Subject(s)
CD8-Positive T-Lymphocytes , Melanoma, Experimental , Animals , Mice , Phenols/pharmacology , Cysteamine/pharmacology , Melanoma, Experimental/drug therapy , Mice, Inbred C57BL , Melanoma, Cutaneous Malignant
2.
Cancers (Basel) ; 14(22)2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36428680

ABSTRACT

Malignant melanoma is one of the most malignant of all cancers. Melanoma occurs at the epidermo-dermal interface of the skin and mucosa, where small vessels and lymphatics are abundant. Consequently, from the onset of the disease, melanoma easily metastasizes to other organs throughout the body via lymphatic and blood circulation. At present, the most effective treatment method is surgical resection, and other attempted methods, such as chemotherapy, radiotherapy, immunotherapy, targeted therapy, and gene therapy, have not yet produced sufficient results. Since melanogenesis is a unique biochemical pathway that functions only in melanocytes and their neoplastic counterparts, melanoma cells, the development of drugs that target melanogenesis is a promising area of research. Melanin consists of small-molecule derivatives that are always synthesized by melanoma cells. Amelanosis reflects the macroscopic visibility of color changes (hypomelanosis). Under microscopy, melanin pigments and their precursors are present in amelanotic melanoma cells. Tumors can be easily targeted by small molecules that chemically mimic melanogenic substrates. In addition, small-molecule melanin metabolites are toxic to melanocytes and melanoma cells and can kill them. This review describes our development of chemo-thermo-immunotherapy based on the synthesis of melanogenesis-based small-molecule derivatives and conjugation to magnetite nanoparticles. We also introduce the other melanogenesis-related chemotherapy and thermal medicine approaches and discuss currently introduced targeted therapies with immune checkpoint inhibitors for unresectable/metastatic melanoma.

3.
Int J Mol Sci ; 23(12)2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35742905

ABSTRACT

A major advance in drug discovery and targeted therapy directed at cancer cells may be achieved by the exploitation and immunomodulation of their unique biological properties. This review summarizes our efforts to develop novel chemo-thermo-immunotherapy (CTI therapy) by conjugating a melanogenesis substrate, N-propionyl cysteaminylphenol (NPrCAP: amine analog of tyrosine), with magnetite nanoparticles (MNP). In our approach, NPrCAP provides a unique drug delivery system (DDS) because of its selective incorporation into melanoma cells. It also functions as a melanoma-targeted therapeutic drug because of its production of highly reactive free radicals (melanoma-targeted chemotherapy). Moreover, the utilization of MNP is a platform to develop thermo-immunotherapy because of heat shock protein (HSP) expression upon heat generation in MNP by exposure to an alternating magnetic field (AMF). This comprehensive review covers experimental in vivo and in vitro mouse melanoma models and preliminary clinical trials with a limited number of advanced melanoma patients. We also discuss the future directions of CTI therapy.


Subject(s)
Magnetite Nanoparticles , Melanoma , Animals , Drug Delivery Systems , Humans , Immunotherapy , Magnetic Fields , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Melanoma/metabolism , Mice
4.
Int J Mol Sci ; 21(17)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854423

ABSTRACT

Melanogenesis is the biological and biochemical process of melanin and melanosome biosynthesis. Melanin is formed by enzymic reactions of tyrosinase family proteins that convert tyrosine to form brown-black eumelanin and yellow-red pheomelanin within melanosomal compartments in melanocytes, following the cascades of events interacting with a series of autocrine and paracrine signals. Fully melanized melanosomes are delivered to keratinocytes of the skin and hair. The symbiotic relation of a melanocyte and an associated pool of keratinocytes is called epidermal melanin unit (EMU). Microphthalmia-associated transcription factor (MITF) plays a vital role in melanocyte development and differentiation. MITF regulates expression of numerous pigmentation genes for promoting melanocyte differentiation, as well as fundamental genes for maintaining cell homeostasis. Diseases involving alterations of EMU show various forms of pigmentation phenotypes. This review introduces four major topics of melanogenesis cascade that include (1) melanocyte development and differentiation, (2) melanogenesis and intracellular trafficking for melanosome biosynthesis, (3) melanin pigmentation and pigment-type switching, and (4) development of a novel therapeutic approach for malignant melanoma by elucidation of melanogenesis cascade.


Subject(s)
Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Pigmentation Disorders/metabolism , Cell Differentiation , Humans , Melanins/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Melanoma/drug therapy , Pigmentation Disorders/drug therapy
5.
J Invest Dermatol ; 138(1): 150-158, 2018 01.
Article in English | MEDLINE | ID: mdl-28887108

ABSTRACT

Induced pluripotent stem (iPS) cells have the ability to differentiate into multiple cell types in the body and have an unlimited growth potential. However, iPS cell-derived melanocytes produced by existing protocols have significant limitations in developing novel strategies for regenerative medicine and cell therapies of pigmentation disorders in humans because they involve culture in media containing fetal bovine serum and nonphysiological agents. In this study, we established an in vitro approach to generate iPS cell-derived human melanocytes that have higher proliferation rates and increased melanin production compared with melanocytes prepared by previously reported approaches. Importantly, our iPS cell-derived human melanocytes are prepared in fetal bovine serum-free culture conditions that do not contain any nonphysiological agents. We designed two original methods, transferring black colonies by pipette and recovering black cell pellets from centrifuged medium, and numerous human iPS cell-derived melanocytes proliferated in gelatinous dishes coated with Matrigel after 12 days. We also succeeded in inducing melanin pigmentation in the nude mouse skin in vivo using those human iPS cell-derived melanocytes. We propose that this method using iPS cells established from T cells in the blood of normal human volunteers could be applied clinically to develop regenerative medicine and cell therapies for various forms of human pigmentation disorders.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Melanocytes/physiology , Pigmentation Disorders/therapy , Adult , Animals , Cell Proliferation , Cell Transplantation/methods , Cells, Cultured , Culture Media, Serum-Free/chemistry , Healthy Volunteers , Humans , Male , Melanins/metabolism , Melanocytes/transplantation , Mice , Mice, Nude , Models, Animal , Regenerative Medicine/methods , Skin/cytology , Skin/metabolism , T-Lymphocytes/physiology
6.
Nanomedicine (Lond) ; 9(11): 1715-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25321171

ABSTRACT

Magnetic nanoparticle-mediated hyperthermia (MNHT) generates heat to a local tumor tissue of above 43°C without damaging surrounding normal tissues. By applying MNHT, a significant amount of heat-shock proteins is expressed within and around the tumor tissues, inducing tumor-specific immune responses. In vivo experiments have indicated that MNHT can induce the regression of not only a local tumor tissue exposed to heat, but also distant metastatic tumors unexposed to heat. In this article, we introduce recent progress in the application of MNHT for antitumor treatments and summarize the mechanisms and processes of its biological effects during antitumor induction by MNHT. Several clinical trials have been conducted indicating that the MNHT system may add a promising and novel approach to antitumor therapy.


Subject(s)
Antineoplastic Agents/chemistry , Hyperthermia, Induced/methods , Magnetite Nanoparticles/chemistry , Nanomedicine/methods , Neoplasms/therapy , Animals , Antigen-Presenting Cells , Clinical Trials as Topic , HSP70 Heat-Shock Proteins/metabolism , Humans , Immune System , Magnetics , Neoplasm Metastasis , Neoplasms/immunology , Temperature
7.
J Invest Dermatol ; 134(6): 1693-1700, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24480879

ABSTRACT

Melanoma is highly metastatic, but the mechanism of melanoma cell migration is still unclear. We found that melanoma cells expressed the nicotinamide adenine dinucleotide-dependent protein deacetylase SIRT1 in the cytoplasm. Cell membrane extension and migration of melanoma cells were inhibited by SIRT1 inhibitors or SIRT1 knockdown, whereas SIRT1 activators enhanced elongation of protrusion and cellular motility. In B16F1 cells, growth factor stimulation induced lamellipodium extension, a characteristic feature at the leading edge of migrating cells, and SIRT1 was found in the lamellipodium. SIRT1 inhibitor nicotinamide (NAM) or SIRT1 small interfering RNAs suppressed the lamellipodium extension by serum or platelet-derived growth factor (PDGF). The lamellipodium formation by dominant-active Rac1 was also inhibited by NAM, a SIRT1 inhibitor. NAM inhibited the accumulation of phosphorylated Akt at the submembrane by serum or PDGF. Using fluorescence resonance energy transfer, we found that NAM impaired PDGF-dependent increase in the phosphatidylinositol-3,4,5-trisphosphate level at the leading edge. NAM inhibited the abdominal metastasis of transplanted B16F1 melanoma cells in C57BL6/J mice and improved survival. Finally, SIRT1-knockdown B16F1 cells showed significantly reduced metastasis in transplanted mice compared with that in control B16F1 cells. These results indicate that SIRT1 inhibition is a strategy to suppress metastasis of melanoma cells.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Pseudopodia/metabolism , Sirtuin 1/metabolism , Skin Neoplasms/metabolism , Animals , Female , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Niacinamide/chemistry , Platelet-Derived Growth Factor/metabolism , RNA, Small Interfering/metabolism
8.
J Dermatol Sci ; 73(3): 209-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24315204

ABSTRACT

BACKGROUND: N-propionyl cysteaminylphenol-maleimide-dextran (NPCMD) is a toxic tyrosinase substrate developed to treat melanoma. OBJECTIVE: We investigated the effect of NPCMD on innate immune responses in monocytes. METHODS: CD14⁺ monocytes and a monocytic cell line, THP-1, were stimulated with NPCMD in vitro. Cytokines in the culture supernatants were determined by ELISA and flow cytometry. RESULTS: NPCMD stimulated CD14⁺ monocytes and THP-1 cells to secrete TNFα, IL-6 and IL-8, but not IL-10 or IL-12. TNFα secretion from THP-1 cells stimulated with NPCMD was inhibited by addition of an anti-TLR4 mAb in culture. Moreover, NPCMD stimulated production of pro-IL-1ß in CD14⁺ monocytes and monocytic cell line THP-1 cells and activated the NLRP3-inflammasome, resulting in production of mature IL-1ß. Use of ASC and NLRP3-deficient THP-1 cell lines established involvement of the NLRP3 inflammasome in an IL-1ß secretion in treatment with NPCMD. Inhibition of IL-1ß secretion by an endocytosis inhibitor, cytochalasin B, and a lysosomal enzyme cathepsin B inhibitor, CA-074 Me, suggested the involvement of lysosomal rupture and leakage of cathepsin B into the cytosol in NLRP3 activation by NPCMD. CONCLUSION: The immunopotentiating effect of NPCMD mediated by TLR4 and NLRP3 inflammasome activation could be useful for eliciting effective adaptive immune responses against melanoma and other tumors.


Subject(s)
Carrier Proteins/physiology , Cystamine/analogs & derivatives , Dextrans/pharmacology , Inflammasomes/physiology , Maleimides/pharmacology , Monocytes/physiology , Phenols/pharmacology , Toll-Like Receptor 4/physiology , Cell Line, Tumor , Cystamine/pharmacology , Humans , Interleukin-1beta/metabolism , Monocytes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein
9.
J Skin Cancer ; 2013: 742925, 2013.
Article in English | MEDLINE | ID: mdl-23533767

ABSTRACT

Exploitation of biological properties unique to cancer cells may provide a novel approach to overcome difficult challenges to the treatment of advanced melanoma. In order to develop melanoma-targeted chemothermoimmunotherapy, a melanogenesis substrate, N-propionyl-4-S-cysteaminylphenol (NPrCAP), sulfur-amine analogue of tyrosine, was conjugated with magnetite nanoparticles. NPrCAP was exploited from melanogenesis substrates, which are expected to be selectively incorporated into melanoma cells and produce highly reactive free radicals through reacting with tyrosinase, resulting in chemotherapeutic and immunotherapeutic effects by oxidative stress and apoptotic cell death. Magnetite nanoparticles were conjugated with NPrCAP to introduce thermotherapeutic and immunotherapeutic effects through nonapoptotic cell death and generation of heat shock protein (HSP) upon exposure to alternating magnetic field (AMF). During these therapeutic processes, NPrCAP was also expected to provide melanoma-targeted drug delivery system.

10.
Nanomedicine (Lond) ; 8(6): 891-902, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23066648

ABSTRACT

AIM: Accumulating evidence has indicated that hyperthermia using magnetite nanoparticles induces antitumor immunity. This study investigated the diversity of T-cell receptors (TCRs) in tumor-infiltrating lymphocytes after hyperthermia using magnetite nanoparticles. MATERIALS & METHODS: Functionalized magnetite nanoparticles, N-propionyl-4-S-cysteaminylphenol (NPrCAP)/magnetite, were synthesized by conjugating the melanogenesis substrate NPrCAP with magnetite nanoparticles. NPrCAP/magnetite nanoparticles were injected into B16 melanomas in C57BL/6 mice, which were subjected to an alternating magnetic field for hyperthermia treatment. RESULTS: Enlargement of the tumor-draining lymph nodes was observed after hyperthermia. The TCR repertoire was restricted in tumor-infiltrating lymphocytes, and expansion of Vß11(+) T cells was preferentially found. DNA sequences of the third complementaritydetermining regions revealed the presence of clonally expanded T cells. CONCLUSION: These results indicate that the T-cell response in B16 melanomas after hyperthermia is dominated by T cells directed toward a limited number of epitopes and that epitope-specific T cells frequently use a restricted TCR repertoire.


Subject(s)
Hyperthermia, Induced/methods , Lymphocytes, Tumor-Infiltrating/immunology , Magnetite Nanoparticles/therapeutic use , Melanoma, Experimental/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Female , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Magnetic Fields , Magnetite Nanoparticles/chemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/pathology
11.
Biotechnol Prog ; 29(1): 135-42, 2013.
Article in English | MEDLINE | ID: mdl-23073999

ABSTRACT

A three-dimensional (3D) multicellular tumor spheroid culture array has been fabricated using a magnetic force-based cell patterning method, analyzing the effect of stromal fibroblast on the invasive capacity of melanoma. Formation of spheroids was observed when array-like multicellular patterns of melanoma were developed using a pin-holder device made of magnetic soft iron and an external magnet, which enables the assembly of the magnetically labeled cells on the collagen gel-coated surface as array-like cell patterns. The interaction of fibroblast on the invasion of melanoma was investigated using three types of cell interaction models: (i) fibroblasts were magnetically labeled and patterned together in array with melanoma spheroids (direct-interaction model), (ii) fibroblasts coexisting in the upper collagen gel (indirect-interaction model) of melanoma spheroids, and (iii) fibroblast-sheets coexisting under melanoma spheroids (fibroblast-sheet model). The fibroblast-sheet model has largely increased the invasive capacity of melanoma, and the promotion of adhesion, migration, and invasion were also observed. In the fibroblast-sheet model, the expression of IL-8 and MMP-2 increased by 24-fold and 2-fold, respectively, in real time RT-PCR compared to the absence of fibroblasts. The results presented in this study demonstrate the importance of fibroblast interaction to invasive capacity of melanoma in the 3D in vitro bioengineered tumor microenvironment.


Subject(s)
Cell Culture Techniques , Fibroblasts/metabolism , Liposomes/chemistry , Magnetite Nanoparticles/chemistry , Melanoma/genetics , Stromal Cells/metabolism , Animals , Cells, Cultured , Fibroblasts/cytology , Gene Expression Profiling , Humans , Melanoma/metabolism , Melanoma/pathology , Mice , NIH 3T3 Cells , Real-Time Polymerase Chain Reaction , Stromal Cells/cytology
12.
Biochem Pharmacol ; 84(5): 646-53, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22728921

ABSTRACT

Metastatic melanoma is resistant to conventional therapies. N-propionyl-4-S-cysteaminylphenol (NPrCAP), an N-protected sulfur-amine analog of tyrosine, is a good substrate for tyrosinase and is selectively incorporated into melanoma cells, causing cytotoxicity in vitro and in vivo. We have recently shown that intratumoral injections of NPrCAP suppress not only the growth of primary B16F1 melanoma tumors but also of secondary, re-challenged tumors. The participation of CD8(+) T cells has been suggested for the NPrCAP-mediated anti-B16 melanoma immunity. In this study, the molecular mechanism of the NPrCAP cytotoxicity and immunogenicity was examined. The phenol NPrCAP was shown to be activated by mushroom tyrosinase to the ortho-quinone N-propionyl-4-S-cysteaminyl-1,2-benzoquinone (NPrCAQ), and the structure was confirmed by reducing it to the corresponding catechol. NPrCAQ reacted rapidly with biologically relevant sulfhydryl compounds such as cysteine, glutathione and bovine serum albumin. The NPrCAQ-thiol adduct formation was proven with a model thiol N-acetylcysteine by spectroscopic methods. The production and release of NPrCAQ-protein adducts was verified in B16F1 melanoma cells in vitro and in B16F1 melanoma-bearing mice in vivo through the detection of 5-S-cysteaminyl-3-S-cysteinylcatechol after acid hydrolysis of the protein fraction. These results suggest that the phenol NPrCAP, acting as a prohapten, can be activated in melanoma cells by tyrosinase to the quinone-hapten NPrCAQ, which binds to melanosomal proteins through their cysteine residues to form possible neo-antigens, thus triggering the immunological response. NPrCAP thus represents a potential new approach to immunotherapy against metastatic melanoma.


Subject(s)
Antigens, Neoplasm/biosynthesis , Cystamine/analogs & derivatives , Melanoma, Experimental/immunology , Monophenol Monooxygenase/metabolism , Phenols/metabolism , Animals , Chromatography, High Pressure Liquid , Cystamine/metabolism , Magnetic Resonance Spectroscopy , Melanoma, Experimental/enzymology , Mice , Oxidation-Reduction , Spectrophotometry, Ultraviolet , Substrate Specificity
13.
J Dermatol Sci ; 67(1): 51-60, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22622238

ABSTRACT

BACKGROUND: N-propionyl-4-S-cysteaminylphenol (NPr-4-S-CAP) is selectively incorporated into melanoma cells and degrades them. However, it remains unclear whether NPr-4-S-CAP can induce cell death associated with the induction of host immune responses and tumor suppression in vivo. OBJECTIVE: To examine the molecular mechanism of NPr-4-S-CAP-mediated cytotoxicity toward melanoma cells and to test whether NPr-4-S-CAP can suppress transplanted primary and secondary B16F1 melanomas. METHODS: Cytotoxicity and apoptosis of melanoma cells were assessed by cell counting, flow cytometry, and detection of reactive oxygen species (ROS) and apoptotic molecules. NPr-4-S-CAP-associated host immunity was studied using a B16F1 mouse melanoma model through the application of CD4- and CD8-specific antibodies and tetramer assay. RESULTS: NPr-4-S-CAP suppressed growth of pigmented melanoma cells associated with an increase of intracellular ROS, activation of caspase 3 and DNA fragmentation, suggesting that NPr-4-S-CAP mediated ROS production, eliciting apoptosis of melanoma cells. Growth of transplanted B16F1 melanomas was inhibited after the consecutive intratumoral injections of NPr-4-S-CAP, and the tumor growth after rechallenge of B16F1 was significantly suppressed in the treated mice. This suppression occurred when the treated mice were given the anti-CD4 antibody, but not the anti-CD8 antibody. Tetramer assay demonstrated increased TYRP-2-specific CD8(+) T cells in the lymph node and spleen cells prepared from NPr-4-S-CAP-treated B16F1-bearing mice. CONCLUSIONS: These suggest that NPr-4-S-CAP induces apoptosis in melanoma cells through ROS production and generates CD8(+) cell immunity resulting in the suppression of rechallenged B16F1 melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cystamine/analogs & derivatives , Immunity, Cellular/drug effects , Melanoma, Experimental/drug therapy , Phenols/pharmacology , Animals , Antibodies/administration & dosage , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cystamine/pharmacology , DNA Fragmentation , Dose-Response Relationship, Drug , Enzyme Activation , Female , Flow Cytometry , Humans , Intramolecular Oxidoreductases/metabolism , Melanins/biosynthesis , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Reactive Oxygen Species/metabolism , Time Factors , Tumor Burden/drug effects
14.
J Dermatol ; 38(5): 432-41, 2011 May.
Article in English | MEDLINE | ID: mdl-21352276

ABSTRACT

How melanosomal proteins such as enzymic proteins (tyrosinase and tyrosinase-related proteins, Tyrps) and structural protein (gp100) are transported from Golgi to melanosomal compartments is not yet fully understood. A number of small GTPases have been found to be associated with melanosomes and we have identified one of them, Rab7, a regulator of vesicular transport, organelle motility, phospholipid signaling and cytosolic degradative machinery, as being involved in the transport of Tyrp1 from Golgi to stage I melanosomes. This study further characterizes the role of Rab7 as a regulator of differential sorting of melanosomal proteins in this process. Murine melanocytes were transiently transfected with a plasmid encoding either wild-type (Rab7WT), constitutively active (Rab7Q67L) or dominant-negative (Rab7N125I and Rab7T22N) Rab7. Through immunocytostaining and confocal laser scanning microscopy, we quantitatively compared the bio-distribution of melanosomal proteins between Rab7WT-expressing cells and mutant Rab7-expressing cells. We also characterized their differential elimination from melanosomal compartments by Rab7 by utilizing a proteasome inhibitor, MG132. Our findings indicate that Rab7 plays an important role in differential sorting of tyrosinase, Tyrp1 and gp100 in early melanogenesis cascade, and that it is more specifically involved with Tyrp1 than tyrosinase and gp100 in the trafficking from Golgi to melanosomes and the specific exit from the degradative process.


Subject(s)
Melanocytes/metabolism , Oxidoreductases/metabolism , rab GTP-Binding Proteins/physiology , Animals , Cells, Cultured , Golgi Apparatus/metabolism , Melanins/biosynthesis , Melanosomes/metabolism , Mice , Monophenol Monooxygenase/metabolism , Protein Transport , Transfection , gp100 Melanoma Antigen/metabolism , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
15.
Cancer Sci ; 101(9): 1939-46, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20594194

ABSTRACT

Melanogenesis substrate, N-propionyl-4-S-cysteaminylphenol (NPrCAP) is specifically taken up by melanoma cells and inhibits their growth by producing cytotxic free radicals. By taking advantage of this unique chemical agent, we have established melanoma-targeting intracellular hyperthermia by conjugating NPrCAP with magnetite nanoparticles (NPrCAP/M) upon exposure to an alternating magnetic field (AMF). This treatment causes cytotoxic reaction as well as heat shock responses, leading to elicitation of antitumor immune response, which was proved by tumor rechallenge test and CTL induction. We found the level of heat shock protein 72 (Hsp72) to be increased in the cell lysate and culture supernatant after intracellular hyperthermia. Melanoma-specific CD8(+) T-cell response to dendritic cells loaded with hyperthermia-treated tumor lysate was enhanced when compared with non-treated tumor lysate. When heat shock protein, particularly Hsp72, was immuno-depleted from hyperthermia-treated tumor cell lysate, specific CD8(+) T-cell response was abolished. Thus, it is suggested that antitumor immune response induced by hyperthermia using NPrCAP/M is derived from the release of HSP-peptide complex from degraded tumor cells. Therefore, this chemo-thermo-immuno (CTI)-therapy might be effective not only for primary melanoma but also for distant metastasis because of induction of systemic antimelanoma immune responses.


Subject(s)
Cystamine/analogs & derivatives , Heat-Shock Proteins/immunology , Magnetite Nanoparticles/therapeutic use , Melanoma, Experimental/therapy , Phenols/chemistry , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Cross-Priming/immunology , Cystamine/chemistry , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Electromagnetic Fields , Female , HSP72 Heat-Shock Proteins/immunology , HSP72 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Hot Temperature , Hyperthermia, Induced , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/chemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Peptides/immunology , Peptides/metabolism , Protein Binding , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
16.
J Interferon Cytokine Res ; 30(5): 349-57, 2010 May.
Article in English | MEDLINE | ID: mdl-20187765

ABSTRACT

Interferon (IFN) is believed to be one of the most effective anti-melanoma agents. Specifically, IFN-beta has the ability to induce apoptosis of melanoma cells. Induction of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has also been suggested to have a critical role in IFN-beta-induced apoptosis. To characterize the signaling pathway involved in IFN-beta-induced apoptosis, we analyzed the biological effects of IFN-beta on the cell death and caspase activation of melanoma cells. IFN-sensitive cell lines, MM418, SK-mel-23, and SK-mel-118, showed increased apoptotic populations correlated with the activation of caspase-2 and caspase-3 by IFN-beta. IFN-beta-induced apoptosis was significantly suppressed by inhibitors for caspase-2 or caspase-3, but not by inhibitors for caspase-8 or caspase-9 in these cell lines. TRAIL expression was observed in IFN-beta-treated cells of SK-mel-23 and SK-mel-118, but not in those cells of MM418, which showed massive IFN-beta-induced apoptosis and resistance to exogenous TRAIL-mediated apoptosis. G361 was resistant to IFN-beta-induced apoptosis but sensitive to exogenous TRAIL-mediated apoptosis. Furthermore, IFN-beta pretreatment significantly increased the sensitivity against exogenous TRAIL-mediated apoptosis and activation of caspase-2 in G361. These results suggested that caspase-2 activation is commonly associated with induction of IFN-beta-induced apoptosis in IFN-beta-sensitive melanoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Caspase 2/metabolism , Interferon-beta/pharmacology , Melanoma/enzymology , Melanoma/immunology , Apoptosis/drug effects , Caspase 2/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Humans , Melanoma/drug therapy , Melanoma/pathology , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism
17.
J Biomed Biotechnol ; 2009: 457936, 2009.
Article in English | MEDLINE | ID: mdl-19830247

ABSTRACT

Melanogenesis substrate, N-propionyl-cysteaminylphenol (NPrCAP), is selectively incorporated into melanoma cells and inhibits their growth by producing cytotoxic free radicals. Magnetite nanoparticles also disintegrate cancer cells and generate heat shock protein (HSP) upon exposure to an alternating magnetic field (AMF). This study tested if a chemo-thermo-immunotherapy (CTI therapy) strategy can be developed for better management of melanoma by conjugating NPrCAP on the surface of magnetite nanoparticles (NPrCAP/M). We examined the feasibility of this approach in B16 mouse melanoma and evaluated the impact of exposure temperature, frequency, and interval on the inhibition of re-challenged melanoma growth. The therapeutic protocol against the primary transplanted tumor with or without AMF exposure once a day every other day for a total of three treatments not only inhibited the growth of the primary transplant but also prevented the growth of the secondary, re-challenge transplant. The heat-generated therapeutic effect was more significant at a temperature of 43 degrees C than either 41 degrees C or 46 degrees C. NPrCAP/M with AMF exposure, instead of control magnetite alone or without AMF exposure, resulted in the most significant growth inhibition of the re-challenge tumor and increased the life span of the mice. HSP70 production was greatest at 43 degrees C compared to that with 41 degrees C or 46 degrees C. CD8(+)T cells were infiltrated at the site of the re-challenge melanoma transplant.


Subject(s)
Cystamine/analogs & derivatives , Ferrosoferric Oxide/pharmacology , Melanoma/therapy , Neoplasms, Experimental/therapy , Phenols/pharmacology , Animals , Cystamine/pharmacology , Female , Free Radicals/metabolism , Hot Temperature , Immunotherapy , Melanoma/metabolism , Mice , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Time Factors
18.
Pigment Cell Melanoma Res ; 22(5): 623-34, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19493315

ABSTRACT

Melanocortin-1 receptor (MC1R) and its ligands, alpha-melanocyte stimulating hormone (alphaMSH) and agouti signaling protein (ASIP), regulate switching between eumelanin and pheomelanin synthesis in melanocytes. Here we investigated biological effects and signaling pathways of ASIP. Melan-a non agouti (a/a) mouse melanocytes produce mainly eumelanin, but ASIP combined with phenylthiourea and extra cysteine could induce over 200-fold increases in the pheomelanin to eumelanin ratio, and a tan-yellow color in pelletted cells. Moreover, ASIP-treated cells showed reduced proliferation and a melanoblast-like appearance, seen also in melanocyte lines from yellow (A(y)/a and Mc1r(e)/ Mc1r(e)) mice. However ASIP-YY, a C-terminal fragment of ASIP, induced neither biological nor pigmentary changes. As, like ASIP, ASIP-YY inhibited the cAMP rise induced by alphaMSH analog NDP-MSH, and reduced cAMP level without added MSH, the morphological changes and depigmentation seemed independent of cAMP signaling. Melanocytes genetically null for ASIP mediators attractin or mahogunin (Atrn(mg-3J/mg-3J) or Mgrn1(md-nc/md-nc)) also responded to both ASIP and ASIP-YY in cAMP level, while only ASIP altered their proliferation and (in part) shape. Thus, ASIP-MC1R signaling includes a cAMP-independent pathway through attractin and mahogunin, while the known cAMP-dependent component requires neither attractin nor mahogunin.


Subject(s)
Agouti Signaling Protein/metabolism , Cyclic AMP/metabolism , Melanins/biosynthesis , Melanocytes/physiology , Membrane Proteins/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Agouti Signaling Protein/genetics , Animals , Cell Proliferation , Cell Shape , Cells, Cultured , Hair Color/physiology , Melanins/metabolism , Melanocytes/cytology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Pigmentation/physiology , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Ubiquitin-Protein Ligases/genetics
19.
J Invest Dermatol ; 129(9): 2233-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19295615

ABSTRACT

A magnetite nanoparticle, NPrCAP/M, was produced for intracellular hyperthermia treatment of melanoma by conjugating N-propionyl-cysteaminylphenol (NPrCAP) with magnetite and used for the study of selective targeting and degradation of melanoma cells. NPrCAP/M, like NPrCAP, was integrated as a substrate in the oxidative reaction by mushroom tyrosinase. Melanoma, but not non-melanoma, cells incorporated larger amounts of iron than magnetite from NPrCAP/M. When mice bearing a B16F1 melanoma and a lymphoma on opposite flanks were given NPrCAP/M, iron was observed only in B16F1 melanoma cells and iron particles (NPrCAP/M) were identified within late-stage melanosomes by electron microscopy. When cells were treated with NPrCAP/M or magnetite and heated to 43 degrees C by an external alternating magnetic field (AMF), melanoma cells were degraded 1.7- to 5.4-fold more significantly by NPrCAP/M than by magnetite. Growth of transplanted B16 melanoma was suppressed effectively by NPrCAP/M-mediated hyperthermia, suggesting a clinical application of NPrCAP/M to lesional therapy for melanoma. Finally, melanoma cells treated with NPrCAP/M plus AMF showed little sub-G1 fraction and no caspase 3 activation, suggesting that the NPrCAP/M-mediated hyperthermia induced non-apoptotic cell death. These results suggest that NPrCAP/M may be useful in targeted therapy for melanoma by inducing non-apoptotic cell death after appropriate heating by the AMF.


Subject(s)
Cystamine/analogs & derivatives , Ferrosoferric Oxide/pharmacology , Melanoma, Experimental/drug therapy , Nanoparticles , Phenols/pharmacology , Animals , Cell Line, Tumor , Cell Survival , Cystamine/metabolism , Cystamine/pharmacology , Female , Ferrosoferric Oxide/metabolism , Humans , Hyperthermia, Induced , Magnetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Monophenol Monooxygenase/metabolism , Phenols/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...