Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
BMC Cancer ; 16: 186, 2016 Mar 05.
Article in English | MEDLINE | ID: mdl-26944546

ABSTRACT

BACKGROUND: Intrinsic and acquired resistance to drug therapies remains a challenge for malignant melanoma patients. Intratumoral heterogeneities within the tumor microenvironment contribute additional complexity to the determinants of drug efficacy and acquired resistance. METHODS: We use 3D biomimetic platforms to understand dynamics in extracellular matrix (ECM) biogenesis following pharmaceutical intervention against mitogen-activated protein kinases (MAPK) signaling. We further determined temporal evolution of secreted ECM components by isogenic melanoma cell clones. RESULTS: We found that the cell clones differentially secrete and assemble a myriad of ECM molecules into dense fibrillar and globular networks. We show that cells can modulate their ECM biosynthesis in response to external insults. Fibronectin (FN) is one of the key architectural components, modulating the efficacy of a broad spectrum of drug therapies. Stable cell lines engineered to secrete minimal levels of FN showed a concomitant increase in secretion of Tenascin-C and became sensitive to BRAF(V600E) and ERK inhibition as clonally- derived 3D tumor aggregates. These cells failed to assemble exogenous FN despite maintaining the integrin machinery to facilitate cell- ECM cross-talk. We determined that only clones that increased FN production via p38 MAPK and ß1 integrin survived drug treatment. CONCLUSIONS: These data suggest that tumor cells engineer drug resistance by altering their ECM biosynthesis. Therefore, drug treatment may induce ECM biosynthesis, contributing to de novo resistance.


Subject(s)
Extracellular Matrix/metabolism , Melanoma/metabolism , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Cell Survival , Disease Models, Animal , Drug Resistance, Neoplasm , Extracellular Matrix Proteins/metabolism , Female , Fibronectins/metabolism , Heterografts , Humans , Melanoma/drug therapy , Melanoma/pathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplasm Metastasis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Tenascin/metabolism , Tumor Microenvironment
2.
Cancer Res ; 75(13): 2708-15, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25832654

ABSTRACT

The AMPK-related kinase NUAK2 has been implicated in melanoma growth and survival outcomes, but its therapeutic utility has yet to be confirmed. In this study, we show how its genetic amplification in PTEN-deficient melanomas may rationalize the use of CDK2 inhibitors as a therapeutic strategy. Analysis of array-CGH data revealed that PTEN deficiency is coupled tightly with genomic amplification encompassing the NUAK2 locus, a finding strengthened by immunohistochemical evidence that phospho-Akt overexpression was correlated with NUAK2 expression in clinical specimens of acral melanoma. Functional studies in melanoma cells showed that inactivation of the PI3K pathway upregulated p21 expression and reduced the number of cells in S phase. NUAK2 silencing and inactivation of the PI3K pathway efficiently controlled CDK2 expression, whereas CDK2 inactivation specifically abrogated the growth of NUAK2-amplified and PTEN-deficient melanoma cells. Immunohistochemical analyses confirmed an association of CDK2 expression with NUAK2 amplification and p-Akt expression in melanomas. Finally, pharmacologic inhibition of CDK2 was sufficient to suppress the growth of NUAK2-amplified and PTEN-deficient melanoma cells in vitro and in vivo. Overall, our results show how CDK2 blockade may offer a promising therapy for genetically defined melanomas, where NUAK2 is amplified and PTEN is deleted.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Melanoma/genetics , PTEN Phosphohydrolase/deficiency , Protein Serine-Threonine Kinases/genetics , Skin Neoplasms/genetics , Aged , Animals , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Gene Amplification , Humans , Male , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/pathology , Mice , Mice, Nude , Middle Aged , Molecular Targeted Therapy , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Roscovitine , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/enzymology , Skin Neoplasms/pathology
3.
Proc Natl Acad Sci U S A ; 108(16): 6597-602, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21460252

ABSTRACT

The identification of genes that participate in melanomagenesis should suggest strategies for developing therapeutic modalities. We used a public array comparative genomic hybridization (CGH) database and real-time quantitative PCR (qPCR) analyses to identify the AMP kinase (AMPK)-related kinase NUAK2 as a candidate gene for melanomagenesis, and we analyzed its functions in melanoma cells. Our analyses had identified a locus at 1q32 where genomic gain is strongly associated with tumor thickness, and we used real-time qPCR analyses and regression analyses to identify NUAK2 as a candidate gene at that locus. Associations of relapse-free survival and overall survival of 92 primary melanoma patients with NUAK2 expression measured using immunohistochemistry were investigated using Kaplan-Meier curves, log rank tests, and Cox regression models. Knockdown of NUAK2 induces senescence and reduces S-phase, decreases migration, and down-regulates expression of mammalian target of rapamycin (mTOR). In vivo analysis demonstrated that knockdown of NUAK2 suppresses melanoma tumor growth in mice. Survival analysis showed that the risk of relapse is greater in acral melanoma patients with high levels of NUAK2 expression than in acral melanoma patients with low levels of NUAK2 expression (hazard ratio = 3.88; 95% confidence interval = 1.44-10.50; P = 0.0075). These data demonstrate that NUAK2 expression is significantly associated with the oncogenic features of melanoma cells and with the survival of acral melanoma patients. NUAK2 may provide a drug target to suppress melanoma progression. This study further supports the importance of NUAK2 in cancer development and tumor progression, while AMPK has antioncogenic properties.


Subject(s)
Cell Movement , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Melanoma/enzymology , Melanoma/mortality , Neoplasm Proteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Animals , Cellular Senescence/genetics , Disease-Free Survival , Female , Gene Knockdown Techniques , Genetic Loci/genetics , Genome-Wide Association Study , Humans , Male , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Mice, Nude , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/genetics , S Phase/genetics , Survival Rate , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
4.
J Natl Cancer Inst ; 101(18): 1259-71, 2009 Sep 16.
Article in English | MEDLINE | ID: mdl-19704071

ABSTRACT

BACKGROUND: Malignant melanomas are intrinsically resistant to many conventional treatments, such as radiation and chemotherapy, for reasons that are poorly understood. Here we propose and test a model that explains drug resistance or sensitivity in terms of melanosome dynamics. METHODS: The growth and sensitivity to cisplatin of MNT-1 cells, which are melanotic and enriched with mature stage III and IV melanosomes, and SK-MEL-28 cells, which have only immature stage I and II melanosomes, were compared using clonogenic assays. Differences in pigmentation, melanosome stages, melanosome number, and cellular structures in different cell lines in response to various treatments were examined by electron microscopy. The relative numbers of melanosomes of different stages were compared after treatment with 1-phenyl-2-thiourea. The relationship between drug transporter function and endogenous melanogenic toxicity was assessed by treating cells with the cyclosporin analog PSC-833 and by assessing vacuole formation and cell growth inhibition. All statistical tests were two-sided. RESULTS: Endogenous melanogenic cytotoxicity, produced by damaged melanosomes, resulted in pronounced cell growth inhibition in MNT-1 cells compared with amelanotic SK-MEL-28 cells. The sensitivity to CDDP of MNT-1 cells was 3.8-fold higher than that of SK-MEL-28 cells (mean IC(50) for SK-MEL-28 and MNT-1 = 2.13 microM and 0.56 microM, respectively; difference = 1.57 microM, 95% confidence interval = 1.45 to 1.69; P = .0017). After treatment with 6.7 microM CDDP for 72 hours, the number of stage II-III melanosomes in surviving MNT-1 cells was 6.8-fold that of untreated cells. Modulation of MNT-1 cells to earlier-stage (II, II-III, III) melanosomes by treatment with the tyrosinase inhibitor 1-phenyl-2-thiourea dramatically increased CDDP resistance. Furthermore, PSC-833 principally suppressed MNT-1 melanotic cell growth via an elevation of autophagosome-like vacuolar structures, possibly by inhibiting melanosome membrane transporters. CONCLUSIONS: Melanosome dynamics (including their biogenesis, density, status, and structural integrity) regulate the drug resistance of melanoma cells. Manipulation of melanosome functions may be an effective way to enhance the therapeutic activity of anticancer drugs against melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm , Melanoma/drug therapy , Melanoma/pathology , Melanosomes/drug effects , Melanosomes/ultrastructure , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Animals , Cell Death/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Cyclosporins/pharmacology , Doxorubicin/pharmacology , Fluorescent Antibody Technique, Indirect , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Microscopy, Confocal , Microscopy, Electron , Verapamil/pharmacology , Vinblastine/pharmacology
5.
J Clin Invest ; 119(4): 954-63, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19273910

ABSTRACT

Treatments for primary and metastatic melanomas are rarely effective. Even therapeutics such as retinoic acid (RA) that are successfully used to treat several other forms of cancer are ineffective. Recent evidence indicates that the antiproliferative effects of RA are mediated by the transcription factor SOX9 in human cancer cell lines. As we have previously shown that SOX9 is expressed in normal melanocytes, here we investigated SOX9 expression and function in human melanomas. Although SOX9 was expressed in normal human skin, it was increasingly downregulated as melanocytes progressed to the premalignant and then the malignant and metastatic states. Overexpression of SOX9 in both human and mouse melanoma cell lines induced cell cycle arrest by increasing p21 transcription and restored sensitivity to RA by downregulating expression of PRAME, a melanoma antigen. Furthermore, SOX9 overexpression in melanoma cell lines inhibited tumorigenicity both in mice and in a human ex vivo model of melanoma. Treatment of melanoma cell lines with PGD2 increased SOX9 expression and restored sensitivity to RA. Thus, combined treatment with PGD2 and RA substantially decreased tumor growth in human ex vivo and mouse in vivo models of melanoma. The results of our experiments targeting SOX9 provide insight into the pathophysiology of melanoma. Further, the effects of SOX9 on melanoma cell proliferation and RA sensitivity suggest the encouraging possibility of a noncytotoxic approach to the treatment of melanoma.


Subject(s)
Melanoma/genetics , Melanoma/pathology , SOX9 Transcription Factor/genetics , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Humans , Melanoma/drug therapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Microphthalmia-Associated Transcription Factor/genetics , Nevus/genetics , Nevus/pathology , Prostaglandin D2/pharmacology , SOX9 Transcription Factor/physiology , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tretinoin/pharmacology , Up-Regulation/drug effects
7.
J Biol Chem ; 281(30): 21198-21208, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16682408

ABSTRACT

Over 125 pigmentation-related genes have been identified to date. Of those, PMEL17/GP100 has been widely studied as a melanoma-specific antigen as well as a protein required for the formation of fibrils in melanosomes. PMEL17 is synthesized, glycosylated, processed, and delivered to melanosomes, allowing them to mature from amorphous round vesicles to elongated fibrillar structures. In contrast to other melanosomal proteins such as TYR and TYRP1, the processing and sorting of PMEL17 is highly complex. Monoclonal antibody HMB45 is commonly used for melanoma detection, but has the added advantage that it specifically reacts with sialylated PMEL17 in the fibrillar matrix in melanosomes. In this study, we generated mutant forms of PMEL17 to clarify the subdomain of PMEL17 required for formation of the fibrillar matrix, a process critical to pigmentation. The internal proline/serine/threonine-rich repeat domain (called the RPT domain) of PMEL17 undergoes variable proteolytic cleavage. Deletion of the RPT domain abolished its recognition by HMB45 and its capacity to form fibrils. Truncation of the C-terminal domain did not significantly affect the processing or trafficking of PMEL17, but, in contrast, deletion of the N-terminal domain abrogated both. We conclude that the RPT domain is essential for its function in generating the fibrillar matrix of melanosomes and that the luminal domain is necessary for its correct processing and trafficking to those organelles.


Subject(s)
Melanosomes/metabolism , Membrane Glycoproteins/physiology , Amino Acid Sequence , Antigens, Neoplasm , Cell Line, Tumor , Gene Deletion , Golgi Apparatus/metabolism , HeLa Cells , Humans , Melanoma-Specific Antigens , Membrane Glycoproteins/chemistry , Models, Genetic , Molecular Sequence Data , Mutation , Neoplasm Proteins/chemistry , Protein Structure, Tertiary , Transfection , gp100 Melanoma Antigen
8.
J Cell Sci ; 119(Pt 6): 1080-91, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16492709

ABSTRACT

Adaptor proteins (AP) play important roles in the sorting of proteins from the trans-Golgi network, but how they function in the sorting of various melanosome-specific proteins such as Pmel17, an essential structural component of melanosomes, in melanocytes is unknown. We characterized the processing and trafficking of Pmel17 via adaptor protein complexes within melanocytic cells. Proteomics analysis detected Pmel17, AP1 and AP2, but not AP3 or AP4 in early melanosomes. Real-time PCR, immunolabeling and tissue in-situ hybridization confirmed the coexpression of AP1 isoforms mu1A and mu1B (expressed only in polarized cells) in melanocytes and keratinocytes, but expression of mu1B is missing in some melanoma cell lines. Transfection with AP1 isoforms (mu1A or mu1B) showed two distinct distribution patterns that involved Pmel17, and only mu1B was able to restore the sorting of Pmel17 to the plasma membrane in cells lacking mu1B expression. Finally, we established that expression of mu1B is regulated physiologically in melanocytes by UV radiation or DKK1. These results show that Pmel17 is sorted to melanosomes by various intracellular routes, directly or indirectly through the plasma membrane, and the presence of basolateral elements in melanocytes suggests their polarized nature.


Subject(s)
Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex 2/metabolism , Melanocytes/metabolism , Melanosomes/metabolism , Membrane Glycoproteins/metabolism , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 2/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Cell Polarity , Humans , Melanocytes/ultrastructure , Microscopy, Electron , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport , Skin/metabolism , gp100 Melanoma Antigen
9.
Biochem J ; 391(Pt 2): 249-59, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-15960609

ABSTRACT

Dopachrome tautomerase (Dct) is a type I membrane protein and an important regulatory enzyme that plays a pivotal role in the biosynthesis of melanin and in the rapid metabolism of its toxic intermediates. Dct-mutant melanocytes carrying the slaty or slaty light mutations were derived from the skin of newborn congenic C57BL/6J non-agouti black mice and were used to study the effect(s) of these mutations on the intracellular trafficking of Dct and on the pigmentation of the cells. Dct activity is 3-fold lower in slaty cells compared with non-agouti black melanocytes, whereas slaty light melanocytes have a surprisingly 28-fold lower Dct activity. Homology modelling of the active site of Dct suggests that the slaty mutation [R194Q (Arg194-->Gln)] is located in the active site and may alter the ability of the enzyme to transform the substrate. Transmembrane prediction methods indicate that the slaty light mutation [G486R (Gly486-->Arg)] may result in the sliding of the transmembrane domain towards the N-terminus, thus interfering with Dct function. Chemical analysis showed that both Dct mutations increase pheomelanin and reduce eumelanin produced by melanocytes in culture. Thus the enzymatic activity of Dct may play a role in determining whether the eumelanin or pheomelanin pathway is preferred for pigment biosynthesis.


Subject(s)
Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Melanins/biosynthesis , Melanocytes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cells, Cultured , Gene Expression Regulation, Enzymologic , Melanocytes/cytology , Mice , Molecular Sequence Data , Protein Conformation , Protein Transport , Sequence Homology, Amino Acid
10.
J Biol Chem ; 280(14): 14006-16, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15695812

ABSTRACT

More than 125 genes that regulate pigmentation have been identified to date. Of those, MART-1 has been widely studied as a melanoma-specific antigen and as a melanosome-specific marker. Whereas the functions of other melanosomal proteins, such as tyrosinase, tyrosinase-related protein-1, dopachrome tautomerase, and Pmel17, are known, the function of MART-1 in melanogenesis, is unclear. A role for MART-1 in pigmentation is expected because its expression pattern and subcellular distribution is quite similar to the other melanosomal proteins and usually correlates with melanin content. We investigated the function of MART-1 using a multidisciplinary approach, including the use of siRNA to inhibit MART-1 function and the use of transfection to re-express MART-1 in MART-1-negative cells. We show that MART-1 forms a complex with Pmel17 and affects its expression, stability, trafficking, and the processing which is required for melanosome structure and maturation. We conclude that MART-1 is indispensable for Pmel17 function and thus plays an important role in regulating mammalian pigmentation.


Subject(s)
Melanosomes/physiology , Neoplasm Proteins/metabolism , Proteins/metabolism , Animals , Antigens, Neoplasm , Cell Line , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Humans , MART-1 Antigen , Melanoma/metabolism , Melanosomes/ultrastructure , Membrane Glycoproteins , Neoplasm Proteins/genetics , Pigmentation/physiology , Protein Transport , Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Subcellular Fractions/metabolism , gp100 Melanoma Antigen
12.
J Biol Chem ; 279(9): 7971-81, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14634018

ABSTRACT

Pigmentation of the hair, skin, and eyes of mammals results from a number of melanocyte-specific proteins that are required for the biosynthesis of melanin. Those proteins comprise the structural and enzymatic components of melanosomes, the membrane-bound organelles in which melanin is synthesized and deposited. Tyrosinase (TYR) is absolutely required for melanogenesis, but other melanosomal proteins, such as TYRP1, DCT, and gp100, also play important roles in regulating mammalian pigmentation. However, pigmentation does not always correlate with the expression of TYR mRNA/protein, and thus its function is also regulated at the post-translational level. Thus, TYR does not necessarily exist in a catalytically active state, and its post-translational activation could be an important control point for regulating melanin synthesis. In this study, we used a multidisciplinary approach to examine the processing and sorting of TYR through the endoplasmic reticulum (ER), Golgi apparatus, coated vesicles, endosomes and early melanosomes because those organelles hold the key to understanding the trafficking of TYR to melanosomes and thus the regulation of melanogenesis. In pigmented cells, TYR is trafficked through those organelles rapidly, but in amelanotic cells, TYR is retained within the ER and is eventually degraded by proteasomes. We now show that TYR can be released from the ER in the presence of protonophore or proton pump inhibitors which increase the pH of intracellular organelles, after which TYR is transported correctly to the Golgi, and then to melanosomes via the endosomal sorting system. The expression of TYRP1, which facilitates TYR processing in the ER, is down-regulated in the amelanotic cells; this is analogous to a hypopigmentary disease known as oculocutaneous albinism type 3 and further impairs melanin production. The sum of these results shows that organellar pH, proteasome activity, and down-regulation of TYRP1 expression all contribute to the lack of pigmentation in TYR-positive amelanotic melanoma cells.


Subject(s)
Cysteine Endopeptidases/metabolism , Homeostasis , Melanoma, Amelanotic/enzymology , Monophenol Monooxygenase/metabolism , Multienzyme Complexes/metabolism , Oxidoreductases , Animals , Coated Vesicles/enzymology , Endoplasmic Reticulum/enzymology , Endosomes/enzymology , Enzyme Stability , Gene Expression Regulation, Enzymologic , Golgi Apparatus/enzymology , Humans , Hydrogen-Ion Concentration , Immunohistochemistry , Melanins/biosynthesis , Melanoma , Melanoma, Amelanotic/ultrastructure , Melanosomes/enzymology , Membrane Glycoproteins/genetics , Mice , Microscopy, Electron , Monophenol Monooxygenase/analysis , Monophenol Monooxygenase/genetics , Proteasome Endopeptidase Complex , RNA, Messenger/analysis , Tumor Cells, Cultured
13.
J Cell Sci ; 116(Pt 15): 3203-12, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12829739

ABSTRACT

Oculocutaneous albinism (OCA) type 4 is a newly identified human autosomal recessive hypopigmentary disorder that disrupts pigmentation in the skin, hair and eyes. Three other forms of OCA have been previously characterized, each resulting from the aberrant processing and/or sorting of tyrosinase, the enzyme critical to pigment production in mammals. The disruption of tyrosinase trafficking occurs at the level of the endoplasmic reticulum (ER) in OCA1 and OCA3, but at the post-Golgi level in OCA2. The gene responsible for OCA4 is the human homologue of the mouse underwhite (uw) gene, which encodes the membrane-associated transporter protein (MATP). To characterize OCA4, we investigated the processing and sorting of melanogenic proteins in primary melanocytes derived from uw/uw mice and from wild-type mice. OCA4 melanocytes were found to be constantly secreted into the medium dark vesicles that contain tyrosinase and two other melanogenic enzymes, Tyrp1 (tyrosinase-related protein 1) and Dct (DOPAchrome tautomerase); this secretory process is not seen in wild-type melanocytes. Although tyrosinase was synthesized at comparable rates in wild-type and in uw-mutant melanocytes, tyrosinase activity in uw-mutant melanocytes was only about 20% of that found in wild-type melanocytes, and was enriched only about threefold in melanosomes compared with the ninefold enrichment in wild-type melanocytes. OCA4 melanocytes showed a marked difference from wild-type melanocytes in that tyrosinase was abnormally secreted from the cells, a process similar to that seen in OCA2 melanocytes, which results from a mutation of the pink-eyed dilution (P) gene. The P protein and MATP have 12 transmembrane regions and are predicted to function as transporters. Ultrastructural analysis shows that the vesicles secreted from OCA4 melanocytes are mostly early stage melanosomes. Taken together, our results show that in OCA4 melanocytes, tyrosinase processing and intracellular trafficking to the melanosome is disrupted and the enzyme is abnormally secreted from the cells in immature melanosomes, which disrupts the normal maturation process of those organelles. This mechanism explains the hypopigmentary phenotype of these cells and provides new insights into the involvement of transporters in the normal physiology of melanocytes.


Subject(s)
Albinism, Oculocutaneous/physiopathology , Melanocytes/physiology , Monophenol Monooxygenase/physiology , Oxidoreductases , Albinism, Oculocutaneous/enzymology , Albinism, Oculocutaneous/genetics , Animals , Enzyme Activation/physiology , Intramolecular Oxidoreductases/physiology , Melanocytes/enzymology , Melanocytes/ultrastructure , Membrane Glycoproteins/physiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Protein Transport/physiology , Secretory Vesicles/physiology , Skin/physiopathology , Skin Pigmentation/physiology , Symporters
14.
Pigment Cell Res ; 16(3): 237-44, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12753396

ABSTRACT

Melanosomes provide an intriguing model for study at many levels. In part this is due to their unique structure and function, but also in part to their involvement in pigmentary diseases and as a model to study basic cellular mechanisms of organelle biogenesis. Recent studies have elucidated the full proteome of the melanosome and the metabolic and molecular lesions involved in a number of pigmentary diseases have been resolved. This paper summarizes recent advances in the field in these areas.


Subject(s)
Cell Differentiation , Melanosomes/metabolism , Melanosomes/physiology , Organelles/physiology , Animals , Blotting, Western , Humans , Melanins/chemistry , Melanocytes/cytology , Microscopy, Confocal , Models, Biological , Monophenol Monooxygenase/metabolism , Pigmentation , Proteome
15.
J Proteome Res ; 2(1): 69-79, 2003.
Article in English | MEDLINE | ID: mdl-12643545

ABSTRACT

Melanin is a heterogeneous biopolymer produced only by specific cells termed melanocytes, which synthesize and deposit the pigment in specialized membrane-bound organelles known as melanosomes. Although melanosomes have been suspected of being closely related to lysosomes and platelets, the total number of melanosomal proteins is still unknown. Thus far, six melanosome-specific proteins have been identified, and the challenge is to characterize the complete proteome of the melanosome to further understand its mechanism of biogenesis. In this report, we used mass spectrometry and subcellular fractionation to identify protein components of early melanosomes. Using this approach, we have identified all 6 of the known melanosome-specific proteins, 56 proteins that are shared with other organelles, and confirmed the presence of 6 novel melanosomal proteins using western blotting and by immunohistochemistry.


Subject(s)
Mass Spectrometry/methods , Melanosomes/metabolism , Melanosomes/physiology , Proteome , Blood Platelets/metabolism , Blotting, Western , Cell Line , Humans , Immunohistochemistry , Melanosomes/chemistry , Microscopy, Confocal , Microscopy, Electron , Microscopy, Fluorescence , Microscopy, Immunoelectron , Models, Biological , Subcellular Fractions/metabolism , Sucrose/pharmacology , Time Factors
16.
J Invest Dermatol ; 119(6): 1341-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12485437

ABSTRACT

In this study, we used melb-a melanoblasts as a model to study mechanisms involved in stimulating melanocyte function in vitiliginous skin following exposure to 8-methoxypsoralen (8MOP). Melanin content and tyrosinase activity increased 3- and 7-fold, respectively, in melanoblasts treated with 8MOP for 6 d compared with untreated controls. The intracellular signal pathways involved in 8MOP-induced effects on melanoblasts were investigated, particularly the roles of protein kinase A and protein kinase C. Forskolin, a protein kinase A activator, mimicked and enhanced the 8MOP stimulation of melanoblast pigmentation whereas a protein kinase C activator, 1-oleoyl-2-acetylglycerol, had no effect, indicating that the protein kinase A pathway is involved rather than the protein kinase C pathway. Those observations were confirmed using inhibitors of the protein kinase A or protein kinase C pathways. Western blot and semiquantitative reverse transcriptase polymerase chain reaction were performed to assess the protein and mRNA expression levels of microphthalmia-associated transcription factor and tyrosinase in melanoblasts treated with 8MOP for 3 h, 6 h, 1 d, 3 d, or 6 d. Incubation with 8MOP stimulated microphthalmia-associated transcription factor protein and mRNA levels within 3 h, but, in contrast, tyrosinase mRNA and protein levels did not increase following 8MOP treatment until 1 d after treatment. The proteasome inhibitor lactacystin blocked the proteasome-mediated proteolysis of tyrosinase, and its effect on proteasomal function was enhanced by 8MOP. Taken together, these results show that 8MOP functions by initially stimulating levels of microphthalmia-associated transcription factor expression via activation of the protein kinase A pathway, which thereby stimulates tyrosinase expression and function and eventually leads to dramatic increases in melanin production by melanoblasts.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cysteine Endopeptidases/metabolism , DNA-Binding Proteins/metabolism , Melanocytes/physiology , Methoxsalen/pharmacology , Multienzyme Complexes/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Gene Expression/physiology , Melanins/metabolism , Melanocytes/cytology , Mice , Mice, Inbred C57BL , Microphthalmia-Associated Transcription Factor , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Proteasome Endopeptidase Complex , RNA, Messenger/analysis , Skin Pigmentation/physiology , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
17.
Pigment Cell Res ; 15(6): 426-32, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12453184

ABSTRACT

It is known that the migration of melanocyte precursors (melanoblasts) from the outer root sheath of hair follicles into clinically depigmented epidermis is crucial to the repigmentation of vitiliginous skin treated with photochemotherapy (PUVA), but such migratory cells must penetrate extracellular matrix tissue barriers in vivo. To test the hypothesis that matrix metalloproteinases (MMPs) are required for this process, we determined whether cultured melb-a cells, an immortal line of melanoblasts isolated from neonatal mouse epidermis, express and secrete MMPs and whether a synthetic metalloproteinase inhibitor, GM6001 (Galardin), inhibits their migratory behavior in vitro. Reverse transcriptase-polymerase chain reaction and Western blotting were used to determine the patterns of MMP expression by melanoblasts at the mRNA and protein levels, respectively. The proteolytic activities of MMPs secreted into the culture medium were assessed by gelatin zymography. The capacity of melanoblasts to migrate on fibronectin, laminin or laminin-5 substrates was estimated using Transwell migration assays. The results show that MMP2, MMP9 and MT1-MMP transcripts are expressed by these melanoblasts, but only MMP2 is secreted and activated in the extracellular environment. Although the therapeutic efficacy of PUVA in stimulating repigmentation of vitiliginous skin might derive from direct effects of UVA and/or 8-methoxypsoralen (8MOP), recent studies have shown that keratinocyte-derived factors induced by ultraviolet radiation, especially alpha-melanocyte stimulating hormone (alpha MSH), play a major role in regulating melanocyte function. Therefore, we also examined whether 8MOP and/or alphaMSH are involved in the up-regulation of MMP2 expression in melanoblasts. Western blotting and zymographic analyses revealed that MMP2 synthesis and secretion were induced by 8MOP and/or by alpha MSH. This induction of MMP2 resulted in significant increases of migration by melanoblasts on laminin or on laminin-5 substrates, while concomitant treatment with GM6001 blocked that induced migration. Taken together, these results suggest the importance of MMP2 in melanoblast migration and in the response to PUVA therapy.


Subject(s)
Cell Movement/physiology , Matrix Metalloproteinase 2/metabolism , Melanocytes/cytology , Melanocytes/enzymology , PUVA Therapy , Vitiligo/drug therapy , Animals , Cell Adhesion Molecules/pharmacology , Cells, Cultured , Dipeptides/pharmacology , Enzyme Induction/drug effects , In Vitro Techniques , Laminin/pharmacology , Matrix Metalloproteinase 2/genetics , Methoxsalen/pharmacology , Mice , Mice, Inbred C57BL , Protease Inhibitors/pharmacology , Vitiligo/pathology , alpha-MSH/pharmacology , Kalinin
18.
Anal Biochem ; 305(2): 260-8, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12054455

ABSTRACT

Many melanocyte or skin equivalent models have been used to evaluate the potential efficacy of melanogenic compounds to regulate pigmentation, but there has been great variation in results, partially stemming from the use of different cell lines and diverse conditions for the melanogenic assays. In an earlier report, we optimized a microtiter format assay system to screen potential bioactive compounds using immortalized melan-a melanocytes. That assay system, termed the STOPR protocol, allowed effects on melanocyte proliferation and differentiation to be assessed in a highly sensitive, reproducible, and cost-effective manner. However, in the skin and hair, melanocytes interact with keratinocytes, fibroblasts, and other cell types, and testing of putative bioactive compounds on melanocytes alone in culture does not allow one to observe the interactions with those other cell types, such as would occur in vivo. Therefore, we developed a melanocyte-keratinocyte coculture protocol that allows testing of compounds for potential effects on pigmentation in a more physiologically relevant context. It is a sensitive, reproducible, and reliable model for testing melanogenic regulators, and we have standardized it with known melanogenic inhibitors (hydroquinone, arbutin, kojic acid, and niacinamide) and stimulators (alpha-melanocyte-stimulating hormone, 8-methoxypsoralen, and 3,4-dihydroxyphenylalanine). This coculture system allows for large-scale screening of candidate compounds in conjunction with the STOPR protocol and provides a more physiologically relevant system to study melanocyte-keratinocyte interactions and to elucidate the regulatory mechanisms of melanogenic compounds.


Subject(s)
Keratinocytes/metabolism , Melanocytes/metabolism , Models, Biological , Pigmentation/physiology , Animals , Arbutin/pharmacology , Coculture Techniques/methods , Dihydroxyphenylalanine/metabolism , Hydroquinones/pharmacology , Keratinocytes/drug effects , Melanocyte-Stimulating Hormones/metabolism , Melanocytes/drug effects , Mice , Monophenol Monooxygenase/antagonists & inhibitors , Niacinamide/pharmacology , Pyrones/pharmacology
19.
Pigment Cell Res ; 15(3): 217-24, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12028586

ABSTRACT

Oculocutaneous albinism (OCA) is caused by reduced or deficient melanin pigmentation in the skin, hair, and eyes. OCA has different phenotypes resulting from mutations in distinct pigmentation genes involved in melanogenesis. OCA type 2 (OCA2), the most common form of OCA, is an autosomal recessive disorder caused by mutations in the P gene, the function(s) of which is controversial. In order to elucidate the mechanism(s) involved in OCA2, our group used several antibodies specific for various melanosomal proteins (tyrosinase, Tyrp1, Dct, Pmel17 and HMB45), including a specific set of polyclonal antibodies against the p protein. We used confocal immunohistochemistry to compare the processing and distribution of those melanosomal proteins in wild type (melan-a) and in p mutant (melan-p1) melanocytes. Our results indicate that the melanin content of melan-p1 melanocytes was less than 50% that of wild type melan-a melanocytes. In contrast, the tyrosinase activities were similar in extracts of wild type and p mutant melanocytes. Confocal microscopy studies and pulse-chase analyses showed altered processing and sorting of tyrosinase, which is released from melan-p1 cells to the medium. Processing and sorting of Tyrp1 was also altered to some extent. However, Dct and Pmel17 expression and subcellular localization were similar in melan-a and in melan-p1 melanocytes. In melan-a cells, the p protein showed mainly a perinuclear pattern with some staining in the cytoplasm where some co-localization with HMB45 antibody was observed. These findings suggest that the p protein plays a major role in modulating the intracellular transport of tyrosinase and a minor role for Tyrp1, but is not critically involved in the transport of Dct and Pmel17. This study provides a basis to understand the relationship of the p protein with tyrosinase function and melanin synthesis, and also provides a rational approach to unveil the consequences of P gene mutations in the pathogenesis of OCA2.


Subject(s)
Albinism, Oculocutaneous/etiology , Albinism, Oculocutaneous/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Iron-Binding Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Oxidoreductases , Albinism, Oculocutaneous/genetics , Animals , Cation Transport Proteins/metabolism , Cytoplasm/metabolism , Humans , Immunohistochemistry , Melanocytes/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Precipitin Tests , Protein Transport , Proteins/metabolism , Time Factors , gp100 Melanoma Antigen
SELECTION OF CITATIONS
SEARCH DETAIL
...