Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Acta Paediatr Suppl ; 92(443): 63-73; discussion 45, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14989468

ABSTRACT

UNLABELLED: The cellular location of Niemann-Pick C2 protein (NPC2) in cultured human fibroblasts and Chinese hamster ovary cells was examined immunocytochemically and in living cells by expression of a functional red fluorescent protein chimeric analogue. RESULTS: NPC2 is present in the lysosomes of both cholesterol-depleted and -replenished cells, unlike Niemann-Pick C1 protein (NPC1) which is recruited to late endosomes only upon uptake of low-density lipoprotein. With mobilization of cholesterol from lysosomes, immunocytochemical detection of NPC2 in lysosomes is greatly diminished, whereas NPC1 remains in the late endosomal compartment. We found a partial overlap in the trafficking and organellar sites of accumulation of NPC2 and NPC1. In living cells, NPC2 traffics with NPC1 in late endosomal tubules. However, in contrast to NPC1, which remains either in late endosomal vesicles and tubules or at the peripheries of cholesterol-laden lysosomes, NPC2 moves into the central core of lysosomes. Glycolipid analysis reveals that, in contrast to null mutant NPC1 cells, which accumulate GM2 ganglioside only at the plasma membrane, with no endocytic storage, absence of NPC2 protein in null mutant NPC2 cells does not block internalization of GM2 into endocytic vesicles. This difference in the cellular distribution of GM2 in NPC1 and NPC2 null mutants is the first report of a variation in the phenotypic expression of these genotypically distinct lesions. CONCLUSION: We speculate that while NPC1 may play a major role in the sorting of glycolipids as well as cholesterol within the late endosomes, NPC2 primarily plays a role in the egress of cholesterol and, potentially, glycolipids from lysosomes. These proteins appear not to be integrated into a tightly bound biological complex, but rather represent separate functional entities that complement each other.


Subject(s)
Carrier Proteins/metabolism , Endosomes/metabolism , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Niemann-Pick Diseases/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Histocytochemistry , Intracellular Signaling Peptides and Proteins , Luminescent Proteins , Lysosomes , Microscopy, Confocal , Niemann-Pick C1 Protein , Polymerase Chain Reaction , Protein Transport/physiology , Transfection , Vesicular Transport Proteins , Red Fluorescent Protein
2.
J Biol Chem ; 276(29): 27584-90, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11349133

ABSTRACT

ABCA1, the ATP-binding cassette protein mutated in Tangier disease, mediates the efflux of excess cellular sterol to apoA-I and thereby the formation of high density lipoprotein. The intracellular localization and trafficking of ABCA1 was examined in stably and transiently transfected HeLa cells expressing a functional human ABCA1-green fluorescent protein (GFP) fusion protein. The fluorescent chimeric ABCA1 transporter was found to reside on the cell surface and on intracellular vesicles that include a novel subset of early endosomes, as well as late endosomes and lysosomes. Studies of the localization and trafficking of ABCA1-GFP in the presence of brefeldin A or monensin, agents known to block intracellular vesicular trafficking, as well as apoA-I-mediated cellular lipid efflux, showed that: (i) ABCA1 functions in lipid efflux at the cell surface, and (ii) delivery of ABCA1 to lysosomes for degradation may serve as a mechanism to modulate its surface expression. Time-lapse fluorescence microscopy revealed that ABCA1-GFP-containing early endosomes undergo fusion, fission, and tubulation and transiently interact with one another, late endocytic vesicles, and the cell surface. These studies establish a complex intracellular trafficking pathway for human ABCA1 that may play important roles in modulating ABCA1 transporter activity and cellular cholesterol homeostasis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Apolipoprotein A-I/metabolism , Biological Transport , CHO Cells , Cell Compartmentation , Cricetinae , Cycloheximide/pharmacology , Endocytosis , Green Fluorescent Proteins , HeLa Cells , Humans , Immunohistochemistry , Luminescent Proteins/genetics , Monensin/pharmacology , Recombinant Fusion Proteins/genetics , Sterols/metabolism , Transfection
3.
Proc Natl Acad Sci U S A ; 98(8): 4466-71, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11296289

ABSTRACT

Niemann-Pick type C1 (NPC1) disease results from a defect in the NPC1 protein and is characterized by a pathological accumulation of cholesterol and glycolipids in endocytic organelles. We followed the biosynthesis and trafficking of NPC1 with the use of a functional green fluorescent protein-fused NPC1. Newly synthesized NPC1 is exported from the endoplasmic reticulum and requires transit through the Golgi before it is targeted to late endosomes. NPC1-containing late endosomes then move by a dynamic process involving tubulation and fission, followed by rapid retrograde and anterograde migration along microtubules. Cell fusion studies with normal and mutant NPC1 cells show that exchange of contents between late endosomes and lysosomes depends upon ongoing tubulovesicular late endocytic trafficking. In turn, rapid endosomal tubular movement requires an intact NPC1 sterol-sensing domain and is retarded by an elevated endosomal cholesterol content. We conclude that the neuropathology and cellular lysosomal lipid accumulation in NPC1 disease results, at least in part, from striking defects in late endosomal tubulovesicular trafficking.


Subject(s)
Endosomes/metabolism , Niemann-Pick Diseases/metabolism , Animals , Blotting, Western , CHO Cells , Carrier Proteins/metabolism , Cell Compartmentation , Cholesterol/metabolism , Cricetinae , Endocytosis , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Green Fluorescent Proteins , Humans , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/metabolism , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Niemann-Pick C1 Protein
4.
J Biol Chem ; 276(5): 3417-25, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11032830

ABSTRACT

The Niemann-Pick C1 (NPC1) protein and endocytosed low density lipoprotein (LDL)-derived cholesterol were shown to enrich separate subsets of vesicles containing lysosomal associated membrane protein 2. Localization of Rab7 in the NPC1-containing vesicles and enrichment of lysosomal hydrolases in the cholesterol-containing vesicles confirmed that these organelles were late endosomes and lysosomes, respectively. Lysobisphosphatidic acid, a lipid marker of the late endosomal pathway, was found in the cholesterol-enriched lysosomes. Recruitment of NPC1 to Rab7 compartments was stimulated by cellular uptake of cholesterol. The NPC1 compartment was shown to be enriched in glycolipids, and internalization of GalNAcbeta1-4[NeuAcalpha2-3]Galbeta1-4Glcbeta1-1'-ceramide (G(M2)) into endocytic vesicles depends on the presence of NPC1 protein. The glycolipid profiles of the NPC1 compartment could be modulated by LDL uptake and accumulation of lysosomal cholesterol. Expression in cells of biologically active NPC1 protein fused to green fluorescent protein revealed rapidly moving and flexible tubular extensions emanating from the NPC1-containing vesicles. We conclude that the NPC1 compartment is a dynamic, sterol-modulated sorting organelle involved in the trafficking of plasma membrane-derived glycolipids as well as plasma membrane and endocytosed LDL cholesterol.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Endosomes/metabolism , Glycolipids/metabolism , Membrane Glycoproteins/metabolism , Animals , Biological Transport , CHO Cells , Carrier Proteins/genetics , Cell Compartmentation , Cells, Cultured , Cricetinae , DNA, Complementary/genetics , Histocytochemistry , Humans , Intracellular Membranes/metabolism , Intracellular Signaling Peptides and Proteins , Lipoproteins, LDL , Lysosomes/metabolism , Membrane Glycoproteins/genetics , Niemann-Pick C1 Protein , Protein Transport/physiology , Subcellular Fractions , Transfection
5.
Exp Cell Res ; 259(1): 247-56, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10942596

ABSTRACT

Mutations in the NPC1 gene cause Niemann-Pick type C disease, which is characterized by the accumulation of free cholesterol and other lipids in lysosomes. The NPC1 glycoprotein is located in a late endosomal compartment that transiently interacts with lysosomes. To identify factors regulating NPC1 expression and action, we analyzed the function of the human NPC1 promoter in human-derived ovarian, hepatic, and neuronal cells. A fragment containing the first 208 base pairs upstream from the major transcription initiation site was sufficient to drive near maximal NPC1 promoter activity. Deletion analysis revealed that sequences between base pairs -111 and -37 play an important role in controlling NPC1 transcription. Treatment of proliferating granulosa cells with 30 microM progesterone, which induces a reversible phenocopy of the cholesterol trafficking defect of Niemann-Pick type C disease, increased NPC1 mRNA levels threefold. The protein synthesis inhibitor, cycloheximide, also increased NPC1 mRNA levels, augmenting the progesterone-induced increase in NPC1 mRNA abundance. Progesterone treatment was shown to increase the mRNA half-life, but did not affect NPC1 promoter activity. Cysteine residues in a "cysteine-rich" loop predicted to reside in the intralumenal compartment of vesicles containing NPC1 were mutated, resulting in proteins that were incapable of correcting the cholesterol trafficking defect in CT60 cells, a Chinese hamster cell line in which the endogenous NPC1 gene is inactivated. Converting isoleucine 1061, also predicted to lie within the cysteine-rich loop, to a threonine residue inactivated the protein as well. The I1061T mutation is one of the most common mutations in Niemann-Pick type C disease. All of the cysteine-rich loop mutants were localized to cholesterol-engorged lysosomes in a pattern mimicking the distribution of NPC1 in progesterone-treated cells. A recombinant protein representing the cysteine-rich loop was shown to bind to a zinc-NTA agarose column. We conclude: (1) that cis elements residing in the first 111 base pairs upstream from the transcription start site are critical for transcription of the NPC1 gene; (2) that NPC1 expression is subject to posttranscriptional regulation in response to treatments that disrupt NPC1 function; and (3) that an intralumenal cysteine-rich loop with zinc-binding activity is critical to NPC1's ability to unload lysosomal cargo.


Subject(s)
Carrier Proteins , Membrane Glycoproteins , Niemann-Pick Diseases/genetics , Promoter Regions, Genetic/genetics , Proteins , Animals , CHO Cells , Carcinoma, Hepatocellular , Cricetinae , Cysteine , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/physiology , Niemann-Pick C1 Protein , Plasmids , Progesterone/pharmacology , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , RNA Processing, Post-Transcriptional/drug effects , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , Transfection , Tumor Cells, Cultured , Zinc/metabolism
6.
Exp Cell Res ; 255(1): 56-66, 2000 Feb 25.
Article in English | MEDLINE | ID: mdl-10666334

ABSTRACT

Steroidogenic cells represent unique systems for the exploration of intracellular cholesterol trafficking. We employed cytochemical and biochemical methods to explore the expression, regulation, and function of the Niemann-Pick C1 protein (NPC1) in human granulosa-lutein cells. NPC1 was localized in a subset of lysosome-associated membrane glycoprotein 2 (LAMP-2)-positive vesicles. By analyzing the sensitivity of NPC1 N-linked oligosaccharide chains to glycosidases and neuraminidase, evidence was obtained for movement of nascent NPC1 from the endoplasmic reticulum through the medial and trans compartments of the Golgi apparatus prior to its appearance in cytoplasmic vesicles. NPC1 protein content and the morphology and cellular distribution of NPC1-containing vesicles were not affected by treatment of the granulosa-lutein cells with 8-Br-cAMP, which stimulates cholesterol metabolism into progesterone. In contrast, steroidogenic acute regulatory (StAR) protein levels were increased by 8-Br-cAMP. Incubation of granulosa-lutein cells with low-density lipoprotein (LDL) in the presence of the hydrophobic amine, U18666A, caused accumulation of free cholesterol in granules, identified by filipin staining, that contained LAMP-2 and NPC1. These granules also stained for neutral lipid with Nile red, reflecting accumulation of LDL-derived cholesterol esters. LDL-stimulated progesterone synthesis was completely blocked by U18666A, leaving steroid output at levels similar to those of cells incubated in the absence of LDL. The hydrophobic amine also blocked the LDL augmentation of 8-Br-cAMP-stimulated progesterone synthesis, reducing steroid production to levels seen in cells stimulated with 8-Br-cAMP in the absence of LDL. Steroidogenesis recovered after U18666A was removed from the culture medium. U18666A treatment caused a 2-fold or more increase in NPC1 protein and mRNA levels, suggesting that disruption of NPC1's function activates a compensatory mechanism resulting in increased NPC1 synthesis. We conclude that the NPC1 compartment plays an important role in the trafficking of LDL-derived substrate in steroidogenic cells; that NPC1 expression is up-regulated when NPC1 action is blocked; and that the NPC1 compartment can be functionally separated from other intracellular pathways contributing substrate for steroidogenesis.


Subject(s)
Carrier Proteins , Cholesterol/metabolism , Granulosa Cells/metabolism , Luteal Cells/metabolism , Membrane Glycoproteins , Proteins/metabolism , Steroids/biosynthesis , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Androstenes/pharmacology , Animals , Anticholesteremic Agents/pharmacology , Biological Transport , CHO Cells , Cells, Cultured , Cricetinae , Female , Gene Expression/drug effects , Granulosa Cells/cytology , Granulosa Cells/drug effects , Humans , Intracellular Fluid/metabolism , Intracellular Signaling Peptides and Proteins , Lipoproteins, LDL/pharmacology , Luteal Cells/cytology , Luteal Cells/drug effects , Lysosomes/metabolism , Niemann-Pick C1 Protein , Phosphoproteins/biosynthesis , Progesterone/biosynthesis , Progestins/biosynthesis , Proteins/genetics
7.
J Biol Chem ; 274(31): 21861-6, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419504

ABSTRACT

Niemann-Pick type C (NPC) disease, characterized by accumulation of low density lipoprotein-derived free cholesterol in lysosomes, is caused by mutations in the NPC1 gene. We examined the ability of wild-type NPC1 and NPC1 mutants to correct the NPC sterol trafficking defect and their subcellular localization in CT60 cells. Cells transfected with wild-type NPC1 expressed 170- and 190-kDa proteins. Tunicamycin treatment resulted in a 140-kDa protein, the deduced size of NPC1, suggesting that NPC1 is N-glycosylated. Mutation of all four asparagines in potential N-terminal N-glycosylation sites to glutamines resulted in a 20-kDa reduction of the expressed protein. Proteins with a single N-glycosylation site mutation localized to late endosome/lysosomal compartments, as did wild-type NPC1, and each corrected the cholesterol trafficking defect. However, mutation of all four potential N-glycosylation sites reduced ability to correct the NPC phenotype commensurate with reduced expression of the protein. Mutations in the putative sterol-sensing domain resulted in inactive proteins targeted to lysosomal membranes encircling cholesterol-laden cores. N-terminal leucine zipper motif mutants could not correct the NPC defect, although they accumulated in lysosomal membranes. We conclude that NPC1 is a glycoprotein that must have an intact sterol-sensing domain and leucine zipper motif for cholesterol-mobilizing activity.


Subject(s)
Carrier Proteins , Membrane Glycoproteins , Mutagenesis, Site-Directed , Niemann-Pick Diseases/genetics , Proteins/genetics , Proteins/metabolism , Sterols/metabolism , Amino Acid Substitution , Animals , Asparagine , Binding Sites , CHO Cells , Cholesterol/metabolism , Cricetinae , Endosomes/metabolism , Glutamine , Glycosylation , Humans , Intracellular Membranes/metabolism , Intracellular Signaling Peptides and Proteins , Leucine Zippers , Lysosomes/metabolism , Molecular Weight , Niemann-Pick C1 Protein , Phenotype , Point Mutation , Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
8.
J Biol Chem ; 274(14): 9627-35, 1999 Apr 02.
Article in English | MEDLINE | ID: mdl-10092649

ABSTRACT

Niemann-Pick C disease (NP-C) is a neurovisceral lysosomal storage disorder. A variety of studies have highlighted defective sterol trafficking from lysosomes in NP-C cells. However, the heterogeneous nature of additional accumulating metabolites suggests that the cellular lesion may involve a more generalized block in retrograde lysosomal trafficking. Immunocytochemical studies in fibroblasts reveal that the NPC1 gene product resides in a novel set of lysosome-associated membrane protein-2 (LAMP2)(+)/mannose 6-phosphate receptor(-) vesicles that can be distinguished from cholesterol-enriched LAMP2(+) lysosomes. Drugs that block sterol transport out of lysosomes also redistribute NPC1 to cholesterol-laden lysosomes. Sterol relocation from lysosomes in cultured human fibroblasts can be blocked at 21 degrees C, consistent with vesicle-mediated transfer. These findings suggest that NPC1(+) vesicles may transiently interact with lysosomes to facilitate sterol relocation. Independent of defective sterol trafficking, NP-C fibroblasts are also deficient in vesicle-mediated clearance of endocytosed [14C]sucrose. Compartmental modeling of the observed [14C]sucrose clearance data targets the trafficking defect caused by mutations in NPC1 to an endocytic compartment proximal to lysosomes. Low density lipoprotein uptake by normal cells retards retrograde transport of [14C]sucrose through this same kinetic compartment, further suggesting that it may contain the sterol-sensing NPC1 protein. We conclude that a distinctive organelle containing NPC1 mediates retrograde lysosomal transport of endocytosed cargo that is not restricted to sterol.


Subject(s)
Carrier Proteins , Lysosomes/metabolism , Niemann-Pick Diseases/metabolism , Proteins/metabolism , Amino Acid Sequence , Antibodies , Antigens, CD/metabolism , Biological Transport , Cell Compartmentation , Cholesterol/metabolism , Endocytosis , Humans , Intracellular Signaling Peptides and Proteins , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Niemann-Pick C1 Protein , Niemann-Pick Diseases/genetics , Proteins/genetics , Receptor, IGF Type 2/metabolism , Structure-Activity Relationship , Sucrose/metabolism
9.
Proc Natl Acad Sci U S A ; 96(3): 805-10, 1999 Feb 02.
Article in English | MEDLINE | ID: mdl-9927649

ABSTRACT

Niemann-Pick type C (NPC) disease is an inherited lipid storage disorder that affects the viscera and central nervous system. A characteristic feature of NPC cells is the lysosomal accumulation of low density lipoprotein-derived cholesterol. To elucidate important structural features of the recently identified NPC1 gene product defective in NPC disease, we examined the ability of wild-type NPC1 and NPC1 mutants to correct the excessive lysosomal storage of low density lipoprotein-derived cholesterol in a model cell line displaying the NPC cholesterol-trafficking defect (CT60 Chinese hamster ovary cells). CT60 cells transfected with human wild-type NPC1 contained immunoreactive proteins of 170 and 190 kDa localized to the lysosomal/endosomal compartment. Wild-type NPC1 protein corrected the NPC cholesterol-trafficking defect in the CT60 cells. Mutation of conserved cysteine residues in the NPC1 N terminus to serine residues resulted in proteins targeted to lysosomal membranes encircling cholesterol-laden cores, whereas deletion of the C-terminal 4-aa residues containing the LLNF lysosome-targeting motif resulted in the expression of protein localized to the endoplasmic reticulum. None of these mutant NPC1 proteins corrected the NPC cholesterol-trafficking defect in CT60 cells. We conclude that transport of the NPC1 protein to the cholesterol-laden lysosomal compartment is essential for expression of its biological activity and that domains in the N terminus of the NPC1 protein are critical for mobilization of cholesterol from lysosomes.


Subject(s)
Carrier Proteins , Cholesterol/metabolism , Lysosomes/metabolism , Membrane Glycoproteins , Proteins/metabolism , Amino Acid Substitution , Animals , CHO Cells , Cricetinae , Endoplasmic Reticulum/metabolism , Filipin/analysis , Green Fluorescent Proteins , Humans , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/metabolism , Mutagenesis, Site-Directed , Niemann-Pick C1 Protein , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/metabolism , Proteins/chemistry , Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Transfection
10.
J Lab Clin Med ; 131(3): 215-21, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9523844

ABSTRACT

Glycosylphosphatidylinositol-anchored proteins (GPI-proteins) expressed on the outer leaflet of cell membranes are involved in diverse physiologic as well as pathologic processes in humans. Previously, we demonstrated the intercellular transfer of overexpressed CD4-GPI in vitro from transduced HeLa cells to their parental cell line. In this report we present further information on the transfer process and the nature of the transferred GPI-proteins. In mixed-cell populations, the transfer of CD4-GPI was detectable within minutes at levels proportional to the ratio of donor and recipient cells. The amount of CD4-GPI detected with flow cytometry on the surface of the recipient cells varied according to cell type. Microscopy of mixed cell populations revealed discrete CD4-GPI containing aggregates on the target cells, whereas colocalized transfer of cytoplasm was not detected. Separation of cocultivated cells by semipermeable membranes largely prevented CD4-GPI transfer, but aggregates containing CD4-GPI were demonstrated by electron microscopy in supernatants passed through filters of 0.4-mm pore size.


Subject(s)
CD4 Antigens/metabolism , Cell Communication/physiology , Glycosylphosphatidylinositols/metabolism , Animals , Cell Count , Cell Line/metabolism , Cells, Cultured , Coculture Techniques , Flow Cytometry/methods , Humans , Microscopy, Fluorescence/methods , Recombinant Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 95(3): 1207-12, 1998 Feb 03.
Article in English | MEDLINE | ID: mdl-9448310

ABSTRACT

Mucolipidosis type IV is an autosomal recessive lysosomal storage disease of unknown etiology that causes severe neurological and ophthalmological abnormalities. In an attempt to obtain insight into the nature of the metabolic abnormality in this disorder, we prospectively evaluated 15 consecutive patients, aged 2 to 23 years, over a period of 22 months. The finding of iron deficiency in some of the patients led us to the discovery that all patients but one had markedly elevated blood gastrin levels. None had vitamin B12 deficiency. Gastroscopy in three patients showed normal gross appearance of the mucosa in two patients, 4 and 7 years old, and mucosal atrophy in a 22-year-old. Parietal cells were present in normal numbers and contained large cytoplasmic inclusions that were confirmed immunohistochemically to be lysosomal in nature. Other gastric epithelial cells appeared normal. Parietal cells contained very few tubulovesicular membranes, suggesting cellular activation, whereas apical canaliculi appeared relatively nonactivated. Both subunits of the parietal cell H+/K+-ATPase were present, and both partially colocalized with f-actin at the apical membrane. We conclude that patients with mucolipidosis type IV are constitutively achlorhydric and have partially activated parietal cells. We hypothesize that the defective protein in this disease is closely associated with the final stages of parietal cell activation and is critical for a specific type of cellular vacuolar trafficking between the cytoplasm and the apical membrane domain.


Subject(s)
Achlorhydria/complications , Mucolipidoses/complications , Achlorhydria/pathology , Actins/metabolism , Adolescent , Adult , Child , Child, Preschool , Gastric Acid/chemistry , Gastroscopy , H(+)-K(+)-Exchanging ATPase/metabolism , Humans , Microscopy, Confocal , Mucolipidoses/pathology , Mucolipidoses/physiopathology , Parietal Cells, Gastric/pathology
12.
J Histochem Cytochem ; 45(8): 1083-96, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9267469

ABSTRACT

Insulin stimulates glucose transport in rat adipose cells through the translocation of GLUT4 from a poorly defined intracellular compartment to the cell surface. We employed confocal microscopy to determine the in situ localization of GLUT4 relative to vesicle, Golgi, and endosomal proteins in these physiological insulin target cells. Three-dimensional analyses of GLUT4 immunostaining in basal cells revealed an intracellular punctate, patchy distribution both in the perinuclear region and scattered throughout the cytoplasm. VAMP2 closely associates with GLUT4 in many punctate vesicle-like structures. A small fraction of GLUT4 overlaps with TGN38-mannosidase II, gamma-adaptin, and mannose-6-phosphate receptors in the perinuclear region, presumably corresponding to late endosome and trans-Golgi network structures. GLUT4 does not co-localize with transferrin receptors, clathrin, and Igp-120. After insulin treatment, GLUT4 partially redistributes to the cell surface and decreases in the perinuclear area. However, GLUT4 remains co-localized with TGN38-mannosidase II and gamma-adaptin. Therefore, the basal compartment from which GLUT4 is translocated in response to insulin comprises specialized post-endosomal VAMP2-positive vesicles, distinct from the constitutively recycling endosomes. These results are consistent with a kinetic model in which GLUT4 is sequestered through two or more intracellular pools in series.


Subject(s)
Adipocytes/chemistry , Endosomes/chemistry , Insulin/pharmacology , Membrane Proteins/analysis , Monosaccharide Transport Proteins/analysis , Muscle Proteins , Adipocytes/drug effects , Adipocytes/ultrastructure , Animals , Cell Compartmentation , Cell Membrane/chemistry , Endosomes/drug effects , Fluorescent Antibody Technique, Indirect , Glucose Transporter Type 4 , Image Processing, Computer-Assisted , Male , Microscopy, Confocal , R-SNARE Proteins , Rats , Rats, Sprague-Dawley
13.
Science ; 277(5323): 228-31, 1997 Jul 11.
Article in English | MEDLINE | ID: mdl-9211849

ABSTRACT

Niemann-Pick type C (NP-C) disease, a fatal neurovisceral disorder, is characterized by lysosomal accumulation of low density lipoprotein (LDL)-derived cholesterol. By positional cloning methods, a gene (NPC1) with insertion, deletion, and missense mutations has been identified in NP-C patients. Transfection of NP-C fibroblasts with wild-type NPC1 cDNA resulted in correction of their excessive lysosomal storage of LDL cholesterol, thereby defining the critical role of NPC1 in regulation of intracellular cholesterol trafficking. The 1278-amino acid NPC1 protein has sequence similarity to the morphogen receptor PATCHED and the putative sterol-sensing regions of SREBP cleavage-activating protein (SCAP) and 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase.


Subject(s)
Carrier Proteins , Cholesterol/metabolism , Drosophila Proteins , Membrane Glycoproteins , Niemann-Pick Diseases/genetics , Proteins/genetics , Amino Acid Sequence , Cholesterol, LDL/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 18 , Cloning, Molecular , Homeostasis , Humans , Hydroxymethylglutaryl CoA Reductases/chemistry , Insect Proteins/chemistry , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Membrane Proteins/chemistry , Molecular Sequence Data , Mutation , Niemann-Pick C1 Protein , Niemann-Pick Diseases/metabolism , Polymorphism, Single-Stranded Conformational , Proteins/chemistry , Proteins/physiology , Receptors, Cell Surface/chemistry , Sequence Homology, Amino Acid , Transfection
14.
Pediatr Res ; 40(3): 429-37, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8865280

ABSTRACT

The effect of diet, human milk or formula, on gastric function (lipase and pepsin activity, pH, and volume) and intragastric digestion of fat was assessed in 28 appropriate for gestational age preterm infants (gestational age, 28.9 +/- 1.4, 29.1 +/- 0.9, 29.5 +/- 0.6 wk; birth weight, 1.00 +/- 0.14 to 1.18 +/- 0.07 kg). The infants were fed either human milk (n = 11), SMA Super Preemie formula (n = 9), or Similac, Special Care formula (n = 8). Fasting and postprandial activity of digestive enzymes, pH, and gastric volume (measured before or during 50 min after gavage feeding) did not differ as a function of diet among the three groups of infants. Gastric lipase output, 23.1 +/- 5.1, 28.3 +/- 6.6, and 22.5 +/- 6.4 (U/kg of body weight) in human milk-, SMA SP-, or Similac SC-fed infants was comparable to the gastric lipase output of healthy adults fed a high fat diet (22.6 +/- 3.0). Pepsin output was, however, significantly lower (597 +/- 77, 743 +/- 97, and 639 +/- 142 U/kg of body weight) in human milk-, SMA SP-, and Similac SC-fed infants) than in healthy adults (3352 +/- 753 U/kg). The hydrolysis of dietary fat was 1.7-2.5-fold higher (p < 0.01) in human milk-fed infants than in infants fed either formula. We conclude that differences in type of feeding, i.e. different fatty acid profiles (long chain or medium chain triglycerides), different emulsions (natural or artificial), and different fat particle sizes do not affect the level of activity of gastric enzymes. However, the triglyceride within milk fat globules appears to be more accessible to gastric lipase than that within formula fat particles. We suggest that the contribution of gastric lipase to overall fat digestion might be greater in the newborn (a period of pancreatic insufficiency) than in the adult.


Subject(s)
Dietary Fats/metabolism , Digestion/physiology , Infant Food , Infant, Premature/physiology , Milk, Human/physiology , Stomach/physiology , Evaluation Studies as Topic , Female , Humans , Hydrogen-Ion Concentration , Infant, Newborn , Intestinal Absorption/physiology , Lipase/metabolism , Lipolysis/physiology , Male , Pepsin A/metabolism , Stomach/anatomy & histology
15.
J Biol Chem ; 271(35): 21604-13, 1996 Aug 30.
Article in English | MEDLINE | ID: mdl-8702948

ABSTRACT

The sterol binding agent 2-hydroxypropyl-beta-cyclodextrin is shown to be a convenient and useful experimental tool to probe intracellular pathways of cholesterol transport. Biochemical and cytochemical studies reveal that cyclodextrin specifically removes plasma membrane cholesterol. Depletion of plasma membrane sphingomyelin greatly accelerated cyclodextrin-mediated cholesterol removal. Cholesterol arriving at the plasma membrane from lysosomes and the endoplasmic reticulum was also removed by cyclodextrin. Cellular cholesterol esterification linked to the mobilization of cholesterol from lysosomes was strongly attenuated by cyclodextrin, suggesting that the major portion of endocytosed cholesterol is delivered from lysosomes to the endoplasmic reticulum via the plasma membrane. Evidence for translocation of lysosomal cholesterol to the endoplasmic reticulum by a plasma membrane-independent pathway is provided by the finding that cyclodextrin loses its ability to suppress esterification when plasma membrane sphingomyelin is depleted. The Golgi apparatus appears to play an active role in directing the relocation of lysosomal cholesterol to the plasma membrane since brefeldin A also abrogated cyclodextrin-mediated suppression of cholesterol esterification. Using cyclodextrin we further show that attenuated esterification of lysosomal cholesterol in Niemann-Pick C cells reflects defective translocation of cholesterol to the plasma membrane that may be linked to abnormal Golgi trafficking.


Subject(s)
Cholesterol, LDL/metabolism , Cyclodextrins/metabolism , beta-Cyclodextrins , 2-Hydroxypropyl-beta-cyclodextrin , Biological Transport , Cell Compartmentation , Cell Membrane/metabolism , Culture Techniques , Cyclodextrins/chemistry , Golgi Apparatus/metabolism , Humans , Lysosomes/metabolism , Microscopy, Fluorescence , Molecular Probes , Sphingomyelins/metabolism
16.
Int J Obes Relat Metab Disord ; 20 Suppl 3: S97-101, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8680486

ABSTRACT

Perilipins are a family of unique proteins intimately associated with the limiting surface of neutral lipid storage droplets in adipocytes and in steroidogenic cells. Lipid hydrolysis in these cells is initiated by cAMP, which leads to phosphorylation of hormone-sensitive lipase in adipocytes and cholesteryl esterase in steroidogenic cells by protein kinase A. Although the concurrent phosphorylation of perilipin by this kinase suggests a role for these proteins in lipid breakdown, a role for these proteins in lipid packaging or in maintaining the lipid droplet structure cannot be excluded.


Subject(s)
Adipocytes/metabolism , Lipid Metabolism , Lipids/chemistry , Phosphoproteins/physiology , Steroids/biosynthesis , Animals , Carrier Proteins , Lipolysis/physiology , Perilipin-1 , Phosphorylation , Sterol Esterase/metabolism
17.
J Lipid Res ; 36(6): 1211-26, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7665999

ABSTRACT

Immunocytochemistry was used to determine the intracellular location of perilipins in adipocytes and the occurrence of these proteins in tissues involved in triacylglycerol metabolism. Confocal microscopy and 3-dimensional analysis of 3T3-L1 adipocytes showed that perilipin immunofluorescence, present on the surfaces of all sized lipid droplets, appeared unevenly dispersed on the surfaces of many large lipid droplets. Electron microscopy revealed that immunogold staining for perilipin was located directly on the surface layer apposed to and surrounding the core triacylglycerol of intracellular lipid droplets of adipocytes in culture or from white and brown adipose tissue. Freeze-fracture electron microscopy indicated that the hydrophobic face of this surface monolayer contained particles identical in size and distribution to intramembranous particles (IMPs), which are unique structural features of the hydrophobic faces of bilayered membranes. Also, freeze-fracture replicas revealed areas of continuity between the surface layer of lipid droplets and the membrane leaflets of endoplasmic reticulum, suggesting that the droplet monolayer surface is an area of endoplasmic reticulum membrane leaflet modified by its unique content of perilipin. Microperoxisomes, identified by immunostaining for catalase, were found closely associated with lipid droplets, but external to and not in contact with the lipid droplet surface layer. Vimentin, identified by immunofluorescence, was present around the periphery of most lipid droplets in 3T3-L1 cells during early stages of adipocyte development but, in contrast to perilipins, vimentin was not around the periphery of many large lipid droplets in mature cells. Although perilipin was at the surface of lipid droplets in adipocytes of lactating mammary gland, none was found to be associated with the milk lipid droplets in alveolar epithelial cells, nor was the protein found on the surfaces of lipid droplets in hepatocytes. Studies in mammary gland show that perilipin immunostaining will be a valuable tool for the identification of tissue adipocytes severely depleted of their triacylglycerol stores and thus without their characteristic spherical shape. Perilipin's singular location on the surface monolayer of intracellular lipid droplets supports an intimate role for the protein in the triacylglycerol metabolic functions of adipocytes.


Subject(s)
Adipocytes/chemistry , Adipocytes/ultrastructure , Lipids/analysis , Phosphoproteins/analysis , 3T3 Cells , Adipose Tissue, Brown/ultrastructure , Animals , Carrier Proteins , Cells, Cultured , Endoplasmic Reticulum/chemistry , Epididymis/ultrastructure , Female , Fluorescent Antibody Technique , Freeze Fracturing , Humans , Lactation , Liver/chemistry , Liver/ultrastructure , Male , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/ultrastructure , Mice , Microbodies/chemistry , Microscopy, Confocal , Microscopy, Electron , Perilipin-1 , Rats , Rats, Sprague-Dawley , Vimentin/analysis
18.
Pediatr Res ; 37(6): 687-92, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7651750

ABSTRACT

Mucolipidosis 4 (ML4) is an autosomal recessive disorder with both lipid and mucopolysaccharide storage. The disease is characterized by severe visual impairment and psychomotor retardation. In our effort to find a phenotypic marker for ML4 fibroblasts, living cells were stained with fluorescent compounds. The staining pattern in cells was complicated by autofluorescence. A careful study revealed that auto-fluorescence by itself was a sufficient marker for viable ML4 fibroblasts. ML4 cells in cultures obtained from four unrelated patients contain auto-fluorescent material. Auto-fluorescence was noted over a wide range of excitation wavelengths from approximately 365 to approximately 546 nm. The most intense fluorescence was observed in the lower wave-length range. Cultured fibroblasts from normal individuals or obligate ML4 heterozygotes did not fluoresce under adequately controlled culture conditions. High passage number of inadequate feeding caused a small proportion of fibroblasts obtained from normal individuals to auto-fluoresce. The auto-fluorescent material co-localized with phase-dense inclusion bodies, shown to be lysosomes by staining with LAMP-ab. These findings imply that fluorescence may relate to the specific compound(s) stored in the lysosomes. In a comparative study, neuronal ceroid lipofuscinosis fibroblasts were also fluorescent. Fibroblasts from other diseases such as Gaucher disease and glycogenosis type 2 did not show any fluorescence. These findings are currently used in our functional cloning strategy for determining the gene involved in ML4.


Subject(s)
Mucolipidoses/pathology , Skin/pathology , Biomarkers/chemistry , Cells, Cultured , Fibroblasts/chemistry , Fibroblasts/pathology , Fluorescence , Humans , Microscopy, Fluorescence , Mucolipidoses/genetics , Mutation , Skin/chemistry
19.
J Steroid Biochem Mol Biol ; 51(3-4): 143-8, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7981123

ABSTRACT

Steroid-free glucocorticoid receptors are generally considered to reside in the cytoplasm of cells. After the binding of steroids, the receptors translocate into the nucleus in a manner that has been proposed to involve microtubules. However, some results with inhibitors of microtubule assembly argue to the contrary. In all of these studies, only the whole cell localization of receptors has been examined; the biological activity of these receptors has not been determined. We now report that steroid-induced gene expression is maintained in the absence of intact microtubules. This argues that microtubules are not required for either the nuclear translocation or biological activity of glucocorticoid receptors.


Subject(s)
Gene Expression Regulation , Microtubules/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cell Nucleus/metabolism , Colchicine/pharmacology , Cytoplasm/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , HeLa Cells , Humans , Microtubules/drug effects , Microtubules/ultrastructure , Rats , Steroids/pharmacology , Transcriptional Activation , Transfection
20.
Biochem Biophys Res Commun ; 171(1): 38-45, 1990 Aug 31.
Article in English | MEDLINE | ID: mdl-2393397

ABSTRACT

Type C Niemann-Pick disease (NPC) is an autosomal recessive neurovisceral storage disorder in which defective intracellular cholesterol processing has been demonstrated in fibroblasts from NPC patients and obligate heterozygotes. In the present paper, the ability to esterify LDL-cholesterol was examined in cultured lymphocytes from 8 NPC patients, 8 obligate heterozygotes and 8 controls. Cholesteryl ester synthesis was 8% (+/- 5%) and 45% (+/- 16%) of controls in homozygous and heterozygous cell lines, respectively. Histochemical and electron microscopic examinations confirmed that this biochemical lesion was associated with abnormal intracellular accumulation of unesterified cholesterol in mutant lymphocytes. These results demonstrate that measurement of cholesterol esterification in cultured lymphocytes offers a quick and reliable means of confirming the diagnosis of NPC and that these cells may be useful for probing the primary molecular lesion of NPC.


Subject(s)
Cholesterol, LDL/metabolism , Lipoproteins, LDL/metabolism , Lymphocytes/metabolism , Niemann-Pick Diseases/metabolism , Cell Compartmentation , Cholesterol Esters/metabolism , Filipin/analysis , Heterozygote , Homozygote , Humans , In Vitro Techniques , Intracellular Membranes/metabolism , Microscopy, Electron , Microscopy, Fluorescence , Niemann-Pick Diseases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...