Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Biol Cell ; 18(3): 768-80, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17182842

ABSTRACT

Hermansky-Pudlak syndrome (HPS) is a genetic disorder characterized by defects in the formation and function of lysosome-related organelles such as melanosomes. HPS in humans or mice is caused by mutations in any of 15 genes, five of which encode subunits of biogenesis of lysosome-related organelles complex (BLOC)-1, a protein complex with no known function. Here, we show that BLOC-1 functions in selective cargo exit from early endosomes toward melanosomes. BLOC-1-deficient melanocytes accumulate the melanosomal protein tyrosinase-related protein-1 (Tyrp1), but not other melanosomal proteins, in endosomal vacuoles and the cell surface due to failed biosynthetic transit from early endosomes to melanosomes and consequent increased endocytic flux. The defects are corrected by restoration of the missing BLOC-1 subunit. Melanocytes from HPS model mice lacking a different protein complex, BLOC-2, accumulate Tyrp1 in distinct downstream endosomal intermediates, suggesting that BLOC-1 and BLOC-2 act sequentially in the same pathway. By contrast, intracellular Tyrp1 is correctly targeted to melanosomes in melanocytes lacking another HPS-associated protein complex, adaptor protein (AP)-3. The results indicate that melanosome maturation requires at least two cargo transport pathways directly from early endosomes to melanosomes, one pathway mediated by AP-3 and one pathway mediated by BLOC-1 and BLOC-2, that are deficient in several forms of HPS.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , Vacuoles/metabolism , Adaptor Protein Complex 3/metabolism , Amino Acid Sequence , Animals , Endocytosis , Endosomes/ultrastructure , Humans , Lysosomes/ultrastructure , Melanins/biosynthesis , Melanosomes/ultrastructure , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred C57BL , Models, Biological , Molecular Sequence Data , Monophenol Monooxygenase/metabolism , Mutant Proteins/metabolism , Oxidoreductases/chemistry , Pigmentation/physiology , Protein Transport , Qa-SNARE Proteins/metabolism , Vacuoles/ultrastructure
2.
Mol Biol Cell ; 17(8): 3598-612, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16760433

ABSTRACT

Pmel17 is a pigment cell-specific integral membrane protein that participates in the formation of the intralumenal fibrils upon which melanins are deposited in melanosomes. The Pmel17 cytoplasmic domain is truncated by the mouse silver mutation, which is associated with coat hypopigmentation in certain strain backgrounds. Here, we show that the truncation interferes with at least two steps in Pmel17 intracellular transport, resulting in defects in melanosome biogenesis. Human Pmel17 engineered with the truncation found in the mouse silver mutant (hPmel17si) is inefficiently exported from the endoplasmic reticulum (ER). Localization and metabolic pulse-chase analyses with site-directed mutants and chimeric proteins show that this effect is due to the loss of a conserved C-terminal valine that serves as an ER exit signal. hPmel17si that exits the ER accumulates abnormally at the plasma membrane due to the loss of a di-leucine-based endocytic signal. The combined effects of reduced ER export and endocytosis significantly deplete Pmel17 within endocytic compartments and delay proteolytic maturation required for premelanosome-like fibrillogenesis. The ER export delay and cell surface retention are also observed for endogenous Pmel17si in melanocytes from silver mice, within which Pmel17 accumulation in premelanosomes is dramatically reduced. Mature melanosomes in these cells are larger, rounder, more highly pigmented, and less striated than in control melanocytes. These data reveal a dual sorting defect in a natural mutant of Pmel17 and support a requirement of endocytic trafficking in Pmel17 fibril formation.


Subject(s)
Endocytosis , Endoplasmic Reticulum/metabolism , Melanosomes/metabolism , Membrane Glycoproteins/metabolism , Mutation/genetics , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cells, Cultured , HeLa Cells , Humans , Melanosomes/ultrastructure , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Sorting Signals , Protein Transport , gp100 Melanoma Antigen
3.
Proc Natl Acad Sci U S A ; 102(31): 10964-9, 2005 Aug 02.
Article in English | MEDLINE | ID: mdl-16037214

ABSTRACT

In mammals, >100 genes regulate pigmentation by means of a wide variety of developmental, cellular, and enzymatic mechanisms. Nevertheless, genes that directly regulate pheomelanin production have not been described. Here, we demonstrate that the subtle gray (sut) mouse pigmentation mutant arose by means of a mutation in the Slc7a11 gene, encoding the plasma membrane cystine/glutamate exchanger xCT [Kanai, Y. & Endou, H. (2001) Curr. Drug Metab. 2, 339-354]. A resulting low rate of extracellular cystine transport into sut melanocytes reduces pheomelanin production. We show that Slc7a11 is a major genetic regulator of pheomelanin pigment in hair and melanocytes, with minimal or no effects on eumelanin. Furthermore, transport of cystine by xCT is critical for normal proliferation, glutathione production, and protection from oxidative stress in cultured cells. Thus, we have found that the Slc7a11 gene controls the production of pheomelanin pigment directly. Cells from sut mice provide a model for oxidative stress-related diseases and their therapies.


Subject(s)
Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Melanins/biosynthesis , Animals , Biological Transport, Active , Cell Proliferation , Cells, Cultured , Chromosome Mapping , Cystine/metabolism , Glutathione/metabolism , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/metabolism , Humans , Melanocytes/metabolism , Melanocytes/ultrastructure , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Mice, Transgenic , Oxidative Stress , Skin Pigmentation/genetics , Skin Pigmentation/physiology
4.
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
5.
Mamm Genome ; 15(10): 749-58, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15520878

ABSTRACT

The albino mouse was already known in ancient times and was apparently selectively bred in Egypt, China, and Japan. Thus, it is not surprising that the c or albino locus (now the Tyr locus) was among the first used to demonstrate Mendelian inheritance in mammals at the dawn of the past century. This locus is now known to encode tyrosinase, the rate-limiting enzyme in the production of melanin pigment, and the molecular basis of the albino ( Tyr(c)) mutation is known. Here we describe the congenic series of Tyr-locus alleles, from wild type to null ( albino). We compare eye and skin pigmentation phenotypes and the genetic lesions that cause each. We suggest that this panel of congenic mutants contains rich, untapped resources for the study of many questions of basic cell biological interest.


Subject(s)
Albinism/genetics , Alleles , Melanins/metabolism , Monophenol Monooxygenase/genetics , Mutation/genetics , Animals , Mice , Mice, Transgenic
7.
Am J Pathol ; 165(2): 491-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277223

ABSTRACT

The mi (microphthalmia) locus of mice encodes a transcription factor, MITF. B6-tg/tg mice that do not express any MITF have white coats and small eyes. Moreover, the number of mast cells decreased to one-third that of normal control (+/+) mice in the skin of B6-tg/tg mice. No mast cells were detectable in the stomach, mesentery, and peritoneal cavity of B6-tg/tg mice. Cultured mast cells derived from B6-tg/tg mice do not express a mast cell adhesion molecule, spermatogenic immunoglobulin superfamily (SgIGSF). To obtain in vivo evidence for the correlation of nonexpression of SgIGSF with decrease in mast cell number, we used another MITF mutant, B6-mi(vit)/mi(vit) mice that have a mild phenotype, ie, black coat with white patches and eyes of normal size. B6-mi(vit)/mi(vit) mice had a normal number of mast cells in the skin, stomach, and mesentery, but the number of peritoneal mast cells decreased to one-sixth that of +/+ mice. Cultured mast cells and peritoneal mast cells of B6-mi(vit)/mi(vit) mice showed a reduced but apparently detectable level of SgIGSF expression, demonstrating the parallelism between mast cell number and expression level of SgIGSF. The number of peritoneal mast cells appeared to be influenced by MITF through transcription of SgIGSF.


Subject(s)
Cell Adhesion , DNA-Binding Proteins/physiology , Immunoglobulins/metabolism , Mast Cells/cytology , Membrane Proteins/metabolism , Peritoneal Cavity/cytology , Transcription Factors/physiology , Transcription, Genetic , Animals , Cell Adhesion Molecule-1 , Cell Adhesion Molecules , Cell Count , Cells, Cultured , DNA-Binding Proteins/genetics , Female , Fibroblasts/metabolism , Gene Expression Regulation , Immunoglobulins/genetics , Male , Mast Cells/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microphthalmia-Associated Transcription Factor , Mutation , NIH 3T3 Cells , Phenotype , Transcription Factors/genetics
8.
Blood ; 104(10): 3181-9, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15265785

ABSTRACT

Hermansky-Pudlak syndrome (HPS), a disorder of organelle biogenesis, affects lysosomes, melanosomes, and platelet dense bodies. Seven genes cause HPS in humans (HPS1-HPS7) and at least 15 nonallelic mutations cause HPS in mice. Where their function is known, the HPS proteins participate in protein trafficking and vesicle docking/fusion events during organelle biogenesis. HPS-associated genes participate in at least 4 distinct protein complexes: the adaptor complex AP-3; biogenesis of lysosome-related organelles complex 1 (BLOC-1), consisting of 4 HPS proteins (pallidin, muted, cappuccino, HPS7/sandy); BLOC-2, consisting of HPS6/ruby-eye, HPS5/ruby-eye-2, and HPS3/cocoa; and BLOC-3, consisting of HPS1/pale ear and HPS4/light ear. Here, we report the cloning of the mouse HPS mutation reduced pigmentation (rp). We show that the wild-type rp gene encodes a novel, widely expressed 195-amino acid protein that shares 87% amino acid identity with its human orthologue and localizes to punctate cytoplasmic structures. Further, we show that phosphorylated RP is part of the BLOC-1 complex. In mutant rp/rp mice, a premature stop codon truncates the protein after 79 amino acids. Defects in all the 5 known components of BLOC-1, including RP, cause severe HPS in mice, suggesting that the subunits are nonredundant and that BLOC-1 plays a key role in organelle biogenesis.


Subject(s)
Carrier Proteins/genetics , Hermanski-Pudlak Syndrome/genetics , Hermanski-Pudlak Syndrome/physiopathology , Pigmentation/genetics , Adaptor Protein Complex 3 , Adaptor Protein Complex beta Subunits , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Chromosome Mapping , Cloning, Molecular , Disease Models, Animal , Female , Fibroblasts/cytology , Humans , Lysosomes/physiology , Male , Melanocytes/cytology , Melanocytes/physiology , Melanoma , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Nerve Tissue Proteins , Phenotype , Transcription Factors/metabolism
9.
Pigment Cell Res ; 16(4): 333-44, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12859616

ABSTRACT

Color loci in mammals are those genetic loci in which mutations can affect pigmentation of the hair, skin, and/or eyes. In the mouse, over 800 phenotypic alleles are now known, at 127 identified color loci. As the number of color loci passed 100 only recently, we celebrate this 'century' with an overview of these loci, especially the 59 that have been cloned and sequenced. These fall into a number of functional groups representing melanocyte development and differentiation, melanosomal components, organelle biogenesis, organelle transport, control of pigment-type switching, and some systemic effects. A human ortholog has been identified in all cases, and the majority of these human genes are found to be loci for human disorders, often affecting other body systems as well as pigmentation. We expect that a significant number of color loci remain to be identified. Nonetheless, the large number known already provide a treasury of resources for reconstruction of the mechanisms, at the subcellular, cellular and tissue levels, that produce a functional pigmentary system and contribute to the normal development and functioning of many other organ systems. The mutant mice also provide valuable models for the study of human disease.


Subject(s)
Melanosomes/genetics , Mice/genetics , Pigmentation/genetics , Animals , Cell Differentiation , Genotype , Humans , Melanins/genetics , Melanocytes/cytology , Mice/anatomy & histology , Mice, Mutant Strains , Pigments, Biological/genetics
10.
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
11.
Genetics ; 163(1): 267-76, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12586714

ABSTRACT

Mutations at the mouse microphthalmia locus (Mitf) affect the development of different cell types, including melanocytes, retinal pigment epithelial cells of the eye, and osteoclasts. The MITF protein is a member of the MYC supergene family of basic-helix-loop-helix-leucine-zipper (bHLHZip) transcription factors and is known to regulate the expression of cell-specific target genes by binding DNA as homodimer or as heterodimer with related proteins. The many mutations isolated at the locus have different effects on the phenotype and can be arranged in an allelic series in which the phenotypes range from near normal to white microphthalmic animals with osteopetrosis. Previous investigations have shown that certain combinations of Mitf alleles complement each other, resulting in a phenotype more normal than that of each homozygote alone. Here we analyze this interallelic complementation in detail and show that it is limited to one particular allele, Mitf(Mi-white) (Mitf(Mi-wh)), a mutation affecting the DNA-binding domain. Both loss- and gain-of-function mutations are complemented, as are other Mitf mutations affecting the DNA-binding domain. Furthermore, this behavior is not restricted to particular cell types: Both eye development and coat color phenotypes are complemented. Our analysis suggests that Mitf(Mi-wh)-associated interallelic complementation is due to the unique biochemical nature of this mutation.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Complementation Test , Transcription Factors/genetics , Animals , Eye Abnormalities , Heterozygote , Mice , Microphthalmia-Associated Transcription Factor , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...