Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Biochim Biophys Acta Mol Cell Res ; 1866(12): 118552, 2019 12.
Article in English | MEDLINE | ID: mdl-31487503

ABSTRACT

KIF13B, a kinesin-3 family motor, was originally identified as GAKIN due to its biochemical interaction with human homolog of Drosophila discs-large tumor suppressor (hDLG1). Unlike its homolog KIF13A, KIF13B contains a carboxyl-terminal CAP-Gly domain. To investigate the function of the CAP-Gly domain, we developed a mouse model that expresses a truncated form of KIF13B protein lacking its CAP-Gly domain (KIF13BΔCG), whereas a second mouse model lacks the full-length KIF13A. Here we show that the KIF13BΔCG mice exhibit relatively higher serum cholesterol consistent with the reduced uptake of [3H]CO-LDL in KIF13BΔCG mouse embryo fibroblasts. The plasma level of factor VIII was not significantly elevated in the KIF13BΔCG mice, suggesting that the CAP-Gly domain region of KIF13B selectively regulates LRP1-mediated lipoprotein endocytosis. No elevation of either serum cholesterol or plasma factor VIII was observed in the full length KIF13A null mouse model. The deletion of the CAP-Gly domain region caused subcellular mislocalization of truncated KIF13B concomitant with the mislocalization of LRP1. Mechanistically, the cytoplasmic domain of LRP1 interacts specifically with the alternatively spliced I3 domain of DLG1, which complexes with KIF13B via their GUK-MBS domains, respectively. Importantly, double mutant mice generated by crossing KIF13A null and KIF13BΔCG mice suffer from perinatal lethality showing potential craniofacial defects. Together, this study provides first evidence that the carboxyl-terminal region of KIF13B containing the CAP-Gly domain is important for the LRP1-DLG1-KIF13B complex formation with implications in the regulation of metabolism, cell polarity, and development.


Subject(s)
Discs Large Homolog 1 Protein/metabolism , Kinesins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Membrane Proteins/metabolism , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Circ Res ; 121(4): e9-e19, 2017 Aug 04.
Article in English | MEDLINE | ID: mdl-28684631

ABSTRACT

The biomedical research enterprise depends on the fair and objective peer review of research grants, leading to the distribution of resources through efficient and robust competitive methods. In the United States, federal funding agencies and foundations collectively distribute billions of dollars annually to support biomedical research. For the American Heart Association, a Peer Review Subcommittee is charged with establishing the highest standards for peer review. This scientific statement reviews the current literature on peer review practices, describes the current American Heart Association peer review process and those of other agencies, analyzes the strengths and weaknesses of American Heart Association peer review practices, and recommends best practices for the future.


Subject(s)
American Heart Association , Biomedical Research/standards , Peer Review/standards , Research Support as Topic/standards , Biomedical Research/economics , Biomedical Research/methods , Humans , Peer Review/methods , Research Support as Topic/economics , Research Support as Topic/methods , United States
3.
Hum Mol Genet ; 22(17): 3508-23, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23666527

ABSTRACT

Niemann-Pick disease, type C1 (NPC1), which arises from a mutation in the NPC1 gene, is characterized by abnormal cellular storage and transport of cholesterol and other lipids that leads to hepatic disease and progressive neurological impairment. Oxidative stress has been hypothesized to contribute to the NPC1 disease pathological cascade. To determine whether treatments reducing oxidative stress could alleviate NPC1 disease phenotypes, the in vivo effects of the antioxidant N-acetylcysteine (NAC) on two mouse models for NPC1 disease were studied. NAC was able to partially suppress phenotypes in both antisense-induced (NPC1ASO) and germline (Npc1-/-) knockout genetic mouse models, confirming the presence of an oxidative stress-related mechanism in progression of NPC1 phenotypes and suggesting NAC as a potential molecule for treatment. Gene expression analyses of NAC-treated NPC1ASO mice suggested NAC affects pathways distinct from those initially altered by Npc1 knockdown, data consistent with NAC achieving partial disease phenotype suppression. In a therapeutic trial of short-term NAC administration to NPC1 patients, no significant effects on oxidative stress in these patients were identified other than moderate improvement of the fraction of reduced CoQ10, suggesting limited efficacy of NAC monotherapy. However, the mouse model data suggest that the distinct antioxidant effects of NAC could provide potential treatment of NPC1 disease, possibly in concert with other therapeutic molecules at earlier stages of disease progression. These data also validated the NPC1ASO mouse as an efficient model for candidate NPC1 drug screening, and demonstrated similarities in hepatic phenotypes and genome-wide transcript expression patterns between the NPC1ASO and Npc1-/- models.


Subject(s)
Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/genetics , Oxidative Stress/drug effects , Acetylcysteine/administration & dosage , Adolescent , Adult , Animals , Child , Child, Preschool , Cross-Over Studies , Disease Models, Animal , Double-Blind Method , Female , Gene Expression , Humans , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/physiopathology , Oxidative Stress/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism , Young Adult
4.
Arterioscler Thromb Vasc Biol ; 32(11): 2741-50, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22995521

ABSTRACT

OBJECTIVE: Intracellular cholesterol distribution impacts cell function; however, processes influencing endogenous cholesterol trafficking remain largely unknown. Atherosclerosis is associated with vascular inflammation and these studies address the role of inflammatory mediators on smooth muscle cell cholesterol trafficking. METHODS AND RESULTS: Interestingly, in the absence of an exogenous cholesterol source, serum amyloid A increased [(14)C] oleic acid incorporation into cholesteryl ester in rat smooth muscle cells, suggesting endogenous cholesterol trafficking to the endoplasmic reticulum. [(3)H] cholesteryl ester accumulated in cells prelabeled with [(3)H] cholesterol, confirming that serum amyloid A mediated the movement of endogenous cholesterol. Cholesterol movement was dependent upon functional endolysosomes. The cholesterol oxidase-sensitive pool of cholesterol decreased in serum amyloid A-treated cells. Furthermore, the mechanism whereby serum amyloid A induced cholesterol trafficking was determined to be via activation of expression of secretory phospholipase A(2), group IIA (sPLA(2)) and sPLA(2)-dependent activation of sphingomyelinase. Interestingly, although neither tumor necrosis factor-α nor interferon-γ induced cholesterol trafficking, interleukin-1ß induced [(14)C] cholesteryl ester accumulation that was also dependent upon sPLA(2) and sphingomyelinase activities. Serum amyloid A activates smooth muscle cell interleukin-1ß expression, and although the interleukin-1-receptor antagonist inhibited the interleukin-1ß-induced cholesterol trafficking, it had no effect on the movement of cholesterol mediated by serum amyloid A. CONCLUSIONS: These data support a role for inflammation in endogenous smooth muscle cell cholesterol trafficking from the plasma membrane to the endoplasmic reticulum.


Subject(s)
Cholesterol/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Serum Amyloid A Protein/metabolism , Animals , Animals, Newborn , Biological Transport , Cells, Cultured , Cholesterol Esters/metabolism , Cholesterol Oxidase/metabolism , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Interferon-gamma/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , Lipoproteins, IDL/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oleic Acid/metabolism , Phospholipases A2, Secretory/antagonists & inhibitors , Phospholipases A2, Secretory/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
5.
PLoS One ; 5(9): e12941, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20886067

ABSTRACT

BACKGROUND: Niemann-Pick type C (NPC) disease is a lysosomal storage disease characterized by the accumulation of cholesterol and glycosphingolipids. The majority of NPC patients die in their teen years due to progressive neurodegeneration; however, half of NPC patients also suffer from cholestasis, prolonged jaundice, and hepatosplenomegaly. We previously showed that a key mediator of NPC liver disease is tumor necrosis factor (TNF) α, which is involved in both proinflammatory and apoptotic signaling cascades. In this study, we tested the hypothesis that blocking TNF action with an anti-TNF monoclonal antibody (CNTO5048) will slow the progression of NPC liver disease. METHODOLOGY/PRINCIPAL FINDINGS: Treatment of wild-type C57BL/6 mice with NPC1-specific antisense oligonucleotides led to knockdown of NPC1 protein expression in the liver. This caused classical symptoms of NPC liver disease, including hepatic cholesterol accumulation, hepatomegaly, elevated serum liver enzymes, and lipid laden macrophage accumulation. In addition, there was a significant increase in the number of apoptotic cells and a proliferation of stellate cells. Concurrent treatment of NPC1 knockdown mice with anti-TNF had no effect on the primary lipid storage or accumulation of lipid-laden macrophages. However, anti-TNF treatment slightly blunted the increase in hepatic apoptosis and stellate cell activation that was seen with NPC1 knockdown. CONCLUSIONS/SIGNIFICANCE: Current therapeutic options for NPC disease are limited. Our results provide proof of principle that pharmacologically blocking the TNF-α inflammatory cascade can slightly reduce certain markers of NPC disease. Small molecule inhibitors of TNF that penetrate tissues and cross the blood-brain barrier may prove even more beneficial.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Mice , Niemann-Pick Disease, Type C/drug therapy , Tumor Necrosis Factor-alpha/immunology , Animals , Apoptosis/drug effects , Cholesterol/metabolism , Drug Evaluation, Preclinical , Female , Humans , Intracellular Signaling Peptides and Proteins , Lipid Metabolism/drug effects , Liver/cytology , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Proteins/genetics , Proteins/metabolism
6.
J Lipid Res ; 51(8): 2372-83, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20418540

ABSTRACT

Loss of function of Niemann-Pick C1 (NPC1) leads to lysosomal free cholesterol storage, resulting in the neurodegenerative disease Niemann-Pick disease type C (NPC). Significant numbers of patients with NPC also suffer from liver disease. Currently, no treatments exist that alter patient outcome, and it is unknown if recovery from tissue damage can occur even if a treatment were found. Our laboratory developed a strategy to test whether mice can recover from NPC liver disease. We used antisense oligonucleotides to knock down hepatic expression of NPC1 in BALB/C mice for either 9 or 15 weeks. This recapitulated liver disease with hepatomegaly, cell death, and fibrosis. Then, antisense oligonucleotide treatment was halted for an additional 4, 9, or 15 weeks. We report that significant liver recovery occurred even when NPC1 protein expression only partially returned to normal. Several pathological phenotypes were alleviated, including hepatomegaly, cholesterol storage, and liver cell death. Histological examination revealed that foamy cell accumulation was relieved; however, liver fibrosis increased. Additionally, resolution of cholesterol storage and liver cell death took longer in mice with long-term knockdown. Finally, we found that transcription of cholesterol homeostatic genes was significantly disrupted during the recovery phase after long-term knockdown.


Subject(s)
Liver Diseases/genetics , Liver Diseases/therapy , Niemann-Pick Disease, Type C/complications , Animals , Apoptosis/genetics , Base Sequence , Cell Proliferation , Cholesterol/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Hepatomegaly/genetics , Homeostasis/genetics , Intracellular Signaling Peptides and Proteins , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Diseases/complications , Liver Diseases/pathology , Macrophages/metabolism , Mice , Niemann-Pick C1 Protein , Oligonucleotides, Antisense/genetics , Phenotype , Proteins/genetics , Proteins/metabolism , Time Factors
7.
J Lipid Res ; 50(2): 327-33, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18815434

ABSTRACT

Niemann-Pick type C (NPC) is a fatal autosomal recessive lysosomal storage disease clinically characterized by neurodegeneration and liver disease. Heterogeneous mutations in the NPC1 and NPC2 genes cause impaired egress of free cholesterol from lysosomes, leading to accumulation of cholesterol and glycosphingolipids. Key features of NPC liver disease include hepatic apoptosis, inflammation, and fibrosis. It is unclear what signaling events regulate these disease processes in NPC. We hypothesize that tumor necrosis factor alpha (TNF-alpha), which is involved in both proinflammatory and apoptotic signaling cascades, is a key mediator of inflammation, apoptosis, and fibrosis in NPC liver disease. In this study, we evaluated the role of TNF-alpha signaling in NPC liver disease by utilizing NPC1-specific antisense oligonucleotides to knock down NPC1 expression in control and TNF-alpha knockout mice. In the absence of TNF-alpha, NPC1 knockdown produced liver disease with significantly less inflammation, apoptosis, and fibrosis.


Subject(s)
Apoptosis , Hepatocytes/pathology , Niemann-Pick Diseases/etiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis/genetics , Cell Proliferation , Female , Fibrosis/metabolism , Hepatocytes/metabolism , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Niemann-Pick Diseases/metabolism , Niemann-Pick Diseases/pathology , Tumor Necrosis Factor-alpha/genetics
8.
Hepatology ; 47(5): 1504-12, 2008 May.
Article in English | MEDLINE | ID: mdl-18438776

ABSTRACT

UNLABELLED: Niemann-Pick type C (NPC) is a fatal autosomal recessive lipidosis that is characterized by lysosomal storage of cholesterol and glycosphingolipids. Patients exhibit prolonged neonatal jaundice, hepatosplenomegaly, and progressive neurodegeneration that generally result in death by the teen years. Most clinical cases are caused by mutations in the NPC1 gene. Current mouse models of NPC are not well suited for studying the liver disease due to the rapidly progressing neurological disease. To facilitate study of NPC-associated liver dysfunction, we have developed a novel mouse model using antisense oligonucleotides to ablate NPC1 expression primarily in the liver. Here, we show that the NPC1 knockdown leads to a liver disease phenotype similar to that of patients with NPC and the NPC(nih) mouse model. Key features include hepatomegaly, lipid storage, elevated serum liver enzymes, and increased apoptosis. CONCLUSION: This novel NPC1 antisense mouse model will allow delineation of the mechanism by which NPC1 dysfunction leads to liver cell death.


Subject(s)
Niemann-Pick Disease, Type C/genetics , Oligonucleotides, Antisense/pharmacology , Proteins/genetics , Animals , Apoptosis , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Death/drug effects , Cell Division , Cholesterol/metabolism , Female , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Liver/pathology , Mice , Mice, Inbred BALB C , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/pathology , Polymerase Chain Reaction , RNA, Messenger/genetics , Viscera/drug effects , Viscera/metabolism , Viscera/pathology
9.
Biochem J ; 408(1): e1-3, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17956226

ABSTRACT

Dietary and biliary cholesterol are taken up by intestinal epithelial cells and transported to the endoplasmic reticulum. At the endoplasmic reticulum, cholesterol is esterified, packaged into chylomicrons and secreted into the lymph for delivery to the bloodstream. NPC1L1 (Niemann-Pick C1-like 1) is a protein on the enterocyte brush-border membrane that facilitates cholesterol absorption. Cholesterol's itinerary as it moves to the endoplasmic reticulum is unknown, as is the identity of any cellular proteins that facilitate the movement. Two proteins that play an important role in intracellular cholesterol transport and could potentially influence NPC1L1-mediated cholesterol uptake are NPC1 and NPC2 (Niemann-Pick type C disease proteins 1 and 2). In this issue of the Biochemical Journal, Dixit and colleagues show that the absence or presence of NPC1 and NPC2 has no effect on intestinal cholesterol absorption in the mouse. Thus neither protein fills the gap in our knowledge of intra-enterocyte cholesterol transport. Furthermore, the NPC1/NPC2 pathway would not be a good target for limiting the uptake of dietary cholesterol.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Intestinal Absorption , Membrane Glycoproteins/metabolism , Niemann-Pick Diseases/metabolism , Animals , Biomarkers
10.
J Biol Chem ; 280(11): 10333-9, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15632139

ABSTRACT

Niemann-Pick type C (NPC) is an autosomal recessive lipid storage disorder characterized by lysosomal accumulation of cholesterol and gangliosides resulting from a defect in intracellular lipid trafficking. The NPC1 gene encodes a 1278-amino acid integral membrane protein involved in the sub-cellular trafficking of lipids. The exact biological function of NPC1 remains unclear. Recent evidence suggests that NPC1 is a eukaryotic member of the RND permease family of transport proteins, which when expressed in bacteria is capable of transporting fatty acids. The goal of this project was to assess the role of NPC1 in the transport of fatty acids in primary human fibroblasts using normal fibroblasts and fibroblasts from patients with three lysosomal storage diseases: NPC, mucolipidosis IV, and Sandhoff disease. If NPC1 is a fatty acid transporter, we expect to find fatty acid accumulation only in NPC fibroblasts. We used three experimental approaches to assess the role of NPC1 as a fatty acid transporter. First, we evaluated the accumulation versus metabolism of low density lipoprotein-derived oleic acid. Second, we assessed the amount of free fatty acid present after growth in lipoprotein-containing media. Third, we assessed the cellular accumulation of acriflavine, a fluorescent substrate for a number of resistance-nodulation-cell division permease transporters. Our results indicate that fatty acid flux through NPC1-deficient lysosomes is normal.


Subject(s)
Carrier Proteins/metabolism , Fatty Acids/metabolism , Lysosomes/metabolism , Membrane Glycoproteins/metabolism , Acriflavine/metabolism , Acriflavine/pharmacology , Cells, Cultured , Cholesterol/metabolism , Chromatography, Gas , Dextrans/chemistry , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Lipid Metabolism , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/metabolism , Niemann-Pick C1 Protein , Niemann-Pick Diseases/metabolism , Oleic Acid/metabolism , Protein Transport
11.
Biochim Biophys Acta ; 1685(1-3): 22-7, 2004 Oct 11.
Article in English | MEDLINE | ID: mdl-15465423

ABSTRACT

Niemann-Pick C 1 (NPC1) is a large integral membrane glycoprotein that resides in late endosomes, whereas NPC2 is a small soluble protein found in the lumen of lysosomes. Mutations in either NPC1 or NPC2 result in aberrant lipid transport from endocytic compartments, which results in lysosomal storage of a complex mixture of lipids, primarily cholesterol and glycosphingolipids. What are the biological functions of the NPC1 and NPC2 proteins? Here we review what is known about the intracellular itinerary of these two proteins as they facilitate lipid transport. We propose that the intracellular trafficking patterns of these proteins will provide clues about their function.


Subject(s)
Carrier Proteins/metabolism , Glycoproteins/metabolism , Lipid Metabolism , Membrane Glycoproteins/metabolism , Niemann-Pick Diseases/metabolism , Animals , Endosomes/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Models, Biological , Niemann-Pick C1 Protein , Protein Transport , Vesicular Transport Proteins
12.
Biochim Biophys Acta ; 1685(1-3): 83-7, 2004 Oct 11.
Article in English | MEDLINE | ID: mdl-15465429

ABSTRACT

The molecular isolation of NPC1 and NPC2, the genes defective in patients with Niemann-Pick disease type C (NP-C), has heralded in an exponential increase in our understanding of this syndrome and thus of human intracellular sterol transport. Despite this, neither the mechanisms of action nor the substrates for these putative transporters have been defined. In this overview, we describe our perspectives on the current awareness of the genetic determination and cellular biology of this syndrome, with emphasis on the underlying events that lead to neurodegeneration and the manner in which they might eventually be treated.


Subject(s)
Niemann-Pick Diseases/metabolism , Niemann-Pick Diseases/physiopathology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cholesterol/genetics , Cholesterol/metabolism , Forecasting , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Niemann-Pick C1 Protein , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/therapy , Sphingolipids/genetics , Sphingolipids/metabolism , Vesicular Transport Proteins
13.
J Biol Chem ; 278(17): 14850-6, 2003 Apr 25.
Article in English | MEDLINE | ID: mdl-12591922

ABSTRACT

Niemann-Pick disease type C (NPC) is characterized by lysosomal storage of cholesterol and gangliosides, which results from defects in intracellular lipid trafficking. Most studies of NPC1 have focused on its role in intracellular cholesterol movement. Our hypothesis is that NPC1 facilitates the egress of cholesterol from late endosomes, which are where active NPC1 is located. When NPC1 is defective, cholesterol does not exit late endosomes; instead, it is carried along to lysosomal storage bodies, where it accumulates. In this study, we addressed whether cholesterol is transported from endosomes to the plasma membrane before reaching NPC1-containing late endosomes. Our study was conducted in Chinese hamster ovary cell lines that display the classical NPC biochemical phenotype and belong to the NPC1 complementation group. We used three approaches to test whether low density lipoprotein (LDL)-derived cholesterol en route to NPC1-containing organelles passes through the plasma membrane. First, we used cyclodextrins to measure the arrival of LDL cholesterol at the plasma membrane and found that the arrival of LDL cholesterol in a cyclodextrin-accessible pool was significantly delayed in NPC1 cells. Second, the movement of LDL cholesterol to NPC1-containing late endosomes was assessed and found to be normal in Chinese hamster ovary mutant 3-6, which exhibits defective movement of plasma membrane cholesterol to intracellular membranes. Third, we examined the movement of plasma membrane cholesterol to the endoplasmic reticulum and found that this pathway is intact in NPC1 cells, i.e. it does not pass through NPC1-containing late endosomes. Our data suggest that in NPC1 cells LDL cholesterol traffics directly through endosomes to lysosomes, bypassing the plasma membrane, and is trapped there because of dysfunctional NPC1.


Subject(s)
Carrier Proteins/physiology , Cell Membrane/metabolism , Cholesterol, LDL/metabolism , Animals , Biological Transport , CHO Cells , Carrier Proteins/genetics , Cell Compartmentation , Cricetinae , Cyclodextrins , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Niemann-Pick Diseases/pathology , Time Factors
14.
J Lipid Res ; 44(1): 182-92, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12518037

ABSTRACT

Plasmalogens are a major sub-class of ethanolamine and choline phospholipids in which the sn-1 position has a long chain fatty alcohol attached through a vinyl ether bond. These phospholipids are proposed to play a role in membrane fusion-mediated events. In this study, we investigated the role of the ethanolamine plasmalogen plasmenylethanolamine (PlsEtn) in intracellular cholesterol transport in Chinese hamster ovary cell mutants NRel-4 and NZel-1, which have single gene defects in PlsEtn biosynthesis. We found that PlsEtn was essential for specific cholesterol transport pathways, those from the cell surface or endocytic compartments to acyl-CoA/cholesterol acyltransferase in the endoplasmic reticulum. The movement of cholesterol from the endoplasmic reticulum or endocytic compartments to the cell surface was normal in PlsEtn-deficient cells. Also, vesicle trafficking was normal in PlsEtn-deficient cells, as measured by fluid phase endocytosis and exocytosis, as was the movement of newly-synthesized proteins to the cell surface. The mutant cholesterol transport phenotype was due to the lack of PlsEtn, since it was corrected when NRel-4 cells were transfected with a cDNA encoding the missing enzyme or supplied with a metabolic intermediate that enters the PlsEtn biosynthetic pathway downstream of the defect. Future work must determine the precise role that plasmalogens have on cholesterol transport to the endoplasmic reticulum.


Subject(s)
Cholesterol/metabolism , Plasmalogens/metabolism , Biological Transport , Cell Membrane/metabolism , Cells, Cultured , Endocytosis , Endoplasmic Reticulum/metabolism , Esterification , Exocytosis , Filipin , Lysosomes/metabolism , Microscopy, Fluorescence , Molecular Structure , Plasmalogens/biosynthesis
15.
J Lipid Res ; 43(10): 1708-17, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12364555

ABSTRACT

Niemann-Pick C (NPC) is an autosomal recessive lysosomal lipid storage disease characterized by progressive central nervous system degeneration. In cultured human NPC fibroblasts, LDL-derived cholesterol accumulates in lysosomes and endosomes, LDL-cholesterol transport from endocytic compartments to other cellular compartments is delayed, and LDL does not elicit normal homeostatic responses. Currently, there is no therapy that delays the onset of neurological symptoms or prolongs the life span of NPC children. We have developed and implemented an amphotericin B-mediated cytotoxicity assay to screen for potential therapeutic drugs that induce cholesterol movement in cultured NPC cells. NPC cells are relatively resistant to amphotericin B killing due to intracellular sequestration of cellular cholesterol. The screen was carried out using simian virus 40-transformed ovarian granulosa cells from the npc (nih) mouse model of NPC disease. A library of 44240 compounds was screened and 55 compounds were identified that promote amphotericin B-mediated killing of NPC cells. One compound, NP-27, corrected the NPC phenotype by four different measures of cholesterol homeostasis. In addition to making NPC cells more sensitive to amphotericin B, NP-27 stimulated two separate cholesterol transport pathways and restored LDL stimulation of cholesterol esterification to near normal levels.


Subject(s)
Niemann-Pick Diseases/drug therapy , Nitrovin/chemistry , Nitrovin/pharmacology , Amphotericin B/toxicity , Animals , Biological Transport/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Cholesterol/biosynthesis , Cholesterol/metabolism , Cholesterol, LDL/biosynthesis , Drug Evaluation, Preclinical , Female , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Lysosomes/metabolism , Mice , Niemann-Pick Diseases/metabolism , Phenotype , Tritium
17.
J Biol Chem ; 277(10): 8267-72, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11751901

ABSTRACT

Perilipin (Peri) A is a phosphoprotein located at the surface of intracellular lipid droplets in adipocytes. Activation of cyclic AMP-dependent protein kinase (PKA) results in the phosphorylation of Peri A and hormone-sensitive lipase (HSL), the predominant lipase in adipocytes, with concurrent stimulation of adipocyte lipolysis. To investigate the relative contributions of Peri A and HSL in basal and PKA-mediated lipolysis, we utilized NIH 3T3 fibroblasts lacking Peri A and HSL but stably overexpressing acyl-CoA synthetase 1 (ACS1) and fatty acid transport protein 1 (FATP1). When incubated with exogenous fatty acids, ACS1/FATP1 cells accumulated 5 times more triacylglycerol (TG) as compared with NIH 3T3 fibroblasts. Adenoviral-mediated expression of Peri A in ACS1/FATP1 cells enhanced TG accumulation and inhibited lipolysis, whereas expression of HSL fused to green fluorescent protein (GFPHSL) reduced TG accumulation and enhanced lipolysis. Forskolin treatment induced Peri A hyperphosphorylation and abrogated the inhibitory effect of Peri A on lipolysis. Expression of a mutated Peri A Delta 3 (Ser to Ala substitutions at PKA consensus sites Ser-81, Ser-222, and Ser-276) reduced Peri A hyperphosphorylation and blocked constitutive and forskolin-stimulated lipolysis. Thus, perilipin expression and phosphorylation state are critical regulators of lipid storage and hydrolysis in ACS1/FATP1 cells.


Subject(s)
Adenoviridae/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphoproteins/metabolism , Sterol Esterase/metabolism , 3T3 Cells , Alanine/chemistry , Animals , Bacterial Outer Membrane Proteins/metabolism , Blotting, Western , Carrier Proteins , Cell Line , Colforsin/pharmacology , Hydrolysis , Lipid Metabolism , Mice , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Perilipin-1 , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Conformation , Serine/chemistry , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...