Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
iScience ; 25(9): 104941, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36065186

ABSTRACT

Numerous studies have established the involvement of lysosomal and mitochondrial dysfunction in the pathogenesis of neurodegenerative disorders such as Alzheimer's and Parkinson diseases. Building on our previous studies of the neurodegenerative lysosomal lipidosis Niemann-Pick C1 (NPC1), we have unexpectedly discovered that activation of the mitochondrial chaperone tumor necrosis factor receptor-associated protein 1 (TRAP1) leads to the correction of the lysosomal storage phenotype in patient cells from multiple lysosomal storage disorders including NPC1. Using small compound activators specific for TRAP1, we find that activation of this chaperone leads to a generalized restoration of lysosomal and mitochondrial health. Mechanistically, we show that this process includes inhibition of oxidative phosphorylation and reduction of oxidative stress, which results in activation of AMPK and ultimately stimulates lysosome recycling. Thus, TRAP1 participates in lysosomal-mitochondrial crosstalk to maintain cellular homeostasis and could represent a potential therapeutic target for multiple disorders.

2.
J Clin Invest ; 127(6): 2081-2090, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28504649

ABSTRACT

Targeted cancer therapies, which act on specific cancer-associated molecular targets, are predominantly inhibitors of oncogenic kinases. While these drugs have achieved some clinical success, the inactivation of kinase signaling via stimulation of endogenous phosphatases has received minimal attention as an alternative targeted approach. Here, we have demonstrated that activation of the tumor suppressor protein phosphatase 2A (PP2A), a negative regulator of multiple oncogenic signaling proteins, is a promising therapeutic approach for the treatment of cancers. Our group previously developed a series of orally bioavailable small molecule activators of PP2A, termed SMAPs. We now report that SMAP treatment inhibited the growth of KRAS-mutant lung cancers in mouse xenografts and transgenic models. Mechanistically, we found that SMAPs act by binding to the PP2A Aα scaffold subunit to drive conformational changes in PP2A. These results show that PP2A can be activated in cancer cells to inhibit proliferation. Our strategy of reactivating endogenous PP2A may be applicable to the treatment of other diseases and represents an advancement toward the development of small molecule activators of tumor suppressor proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Activators/pharmacology , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Enzyme Activation , Enzyme Activators/chemistry , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Protein Binding , Protein Phosphatase 2/chemistry , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays
3.
J Biol Chem ; 292(11): 4395-4410, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28031458

ABSTRACT

Niemann-Pick type C (NP-C) disease is a fatal genetic lipidosis for which there is no Food and Drug Administration (FDA)-approved therapy. Vorinostat, an FDA-approved inhibitor of histone deacetylases, ameliorates lysosomal lipid accumulation in cultured NP-C patient fibroblasts. To assess the therapeutic potential of histone deacetylase inhibition, we pursued these in vitro observations in two murine models of NP-C disease. Npc1nmf164 mice, which express a missense mutation in the Npc1 gene, were treated intraperitoneally, from weaning, with the maximum tolerated dose of vorinostat (150 mg/kg, 5 days/week). Disease progression was measured via gene expression, liver function and pathology, serum and tissue lipid levels, body weight, and life span. Transcriptome analyses of treated livers indicated multiple changes consistent with reversal of liver dysfunction that typifies NP-C disease. Significant improvements in liver pathology and function were achieved by this treatment regimen; however, NPC1 protein maturation and levels, disease progression, weight loss, and animal morbidity were not detectably altered. Vorinostat concentrations were >200 µm in the plasma compartment of treated animals but were almost 100-fold lower in brain tissue. Apolipoprotein B metabolism and the expression of key components of lipid homeostasis in primary hepatocytes from null (Npc1-/-) and missense (Npc1nmf164 ) mutant mice were altered by vorinostat treatment, consistent with a response by these cells independent of the status of the Npc1 locus. These results suggest that HDAC inhibitors have utility to treat visceral NP-C disease. However, it is clear that improved blood-brain barrier penetration will be required to alleviate the neurological symptoms of human NP-C disease.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Liver/drug effects , Liver/physiopathology , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/genetics , Proteins/genetics , Animals , Apolipoproteins B/metabolism , Cells, Cultured , Cholesterol/genetics , Cholesterol/metabolism , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacokinetics , Homeostasis/drug effects , Humans , Hydroxamic Acids/pharmacokinetics , Intracellular Signaling Peptides and Proteins , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mutation, Missense , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/pathology , Niemann-Pick Disease, Type C/physiopathology , Proteins/metabolism , Transcriptome/drug effects , Vorinostat
4.
PLoS One ; 8(8): e74169, 2013.
Article in English | MEDLINE | ID: mdl-23977398

ABSTRACT

Activation of protein kinase C (PKC) has previously been shown to ameliorate the cholesterol transport defect in Niemann Pick Type C1 (NPC1) cells, presumably by increasing the soluble levels of one of its substrates, vimentin. This activity would then restore the vimentin cycle in these cells and allow vimentin-dependent retrograde transport to proceed. Here, we further investigate the effects of PKC activation in NPC1 cells by evaluating different isoforms for their ability to solubilize vimentin and correct the NPC1 cholesterol storage phenotype. We also examine the effects of PKC activators, including free fatty acids and the PKC-specific activator diazoxide, on the NPC1 disease phenotype. Our results indicate that PKC isoforms α, ßII, and ε have the greatest effects on vimentin solubilization. Furthermore, expression or activation of PKCε in NPC1 cells dramatically reduces the amount of stored cholesterol and restores cholesterol transport out of endocytic vesicles. These results provide further support for the contribution of PKCs in NPC1 disease pathogenesis and suggest that PKCs may be targeted in future efforts to develop therapeutics for NPC1 disease.


Subject(s)
Cholesterol/metabolism , Protein Kinase C/metabolism , Animals , Biological Transport/drug effects , CHO Cells , Cell Extracts , Cricetinae , Cricetulus , Enzyme Activation/drug effects , Esterification/drug effects , Fatty Acids/pharmacology , Niemann-Pick Disease, Type C/enzymology , Phenotype , Solubility , Sphingolipids/metabolism , Vimentin/metabolism , rab GTP-Binding Proteins/metabolism
5.
PLoS One ; 7(4): e34424, 2012.
Article in English | MEDLINE | ID: mdl-22514632

ABSTRACT

MLN64 is an integral membrane protein localized to the late endosome and plasma membrane that is thought to function as a mediator of cholesterol transport from endosomal membranes to the plasma membrane and/or mitochondria. The protein consists of two distinct domains: an N-terminal membrane-spanning domain that shares homology with the MENTHO protein and a C-terminal steroidogenic acute regulatory protein (StAR)-related lipid transfer (START) domain that binds cholesterol. To further characterize the MLN64 protein, full-length and truncated proteins were overexpressed in cells and the effects on MLN64 trafficking and endosomal morphology were observed. To gain insight into MLN64 function, affinity chromatography and mass spectrometric techniques were used to identify potential MLN64 interacting partners. Of the 15 candidate proteins identified, 14-3-3 was chosen for further characterization. We show that MLN64 interacts with 14-3-3 in vitro as well as in vivo and that the strength of the interaction is dependent on the 14-3-3 isoform. Furthermore, blocking the interaction through the use of a 14-3-3 antagonist or MLN64 mutagenesis delays the trafficking of MLN64 to the late endosome and also results in the dispersal of endocytic vesicles to the cell periphery. Taken together, these studies have determined that MLN64 is a novel 14-3-3 binding protein and indicate that 14-3-3 plays a role in the endosomal trafficking of MLN64. Furthermore, these studies suggest that 14-3-3 may be the link by which MLN64 exerts its effects on the actin-mediated endosome dynamics.


Subject(s)
14-3-3 Proteins/metabolism , Carrier Proteins/metabolism , Endosomes/metabolism , Membrane Proteins/metabolism , 14-3-3 Proteins/genetics , Animals , Binding Sites , COS Cells , Carrier Proteins/genetics , Cell Line, Tumor , Chlorocebus aethiops , Humans , Mass Spectrometry , Membrane Proteins/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding/genetics , Protein Binding/physiology
6.
J Biol Chem ; 286(27): 23842-51, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21489983

ABSTRACT

Niemann-Pick type C (NP-C) disease is a fatal lysosomal lipid storage disorder for which no effective therapy exists. A genome-wide, conditional synthetic lethality screen was performed using the yeast model of NP-C disease during anaerobiosis, an auxotrophic condition that requires yeast to utilize exogenous sterol. We identified 12 pathways and 13 genes as modifiers of the absence of the yeast NPC1 ortholog (NCR1) and quantified the impact of loss of these genes on sterol metabolism in ncr1Δ strains grown under viable aerobic conditions. Deletion of components of the yeast NuA4 histone acetyltransferase complex in ncr1Δ strains conferred anaerobic inviability and accumulation of multiple sterol intermediates. Thus, we hypothesize an imbalance in histone acetylation in human NP-C disease. Accordingly, we show that the majority of the 11 histone deacetylase (HDAC) genes are transcriptionally up-regulated in three genetically distinct fibroblast lines derived from patients with NP-C disease. A clinically approved HDAC inhibitor (suberoylanilide hydroxamic acid) reverses the dysregulation of the majority of the HDAC genes. Consequently, three key cellular diagnostic criteria of NP-C disease are dramatically ameliorated as follows: lysosomal accumulation of both cholesterol and sphingolipids and defective esterification of LDL-derived cholesterol. These data suggest HDAC inhibition as a candidate therapy for NP-C disease. We conclude that pathways that exacerbate lethality in a model organism can be reversed in human cells as a novel therapeutic strategy. This "exacerbate-reverse" approach can potentially be utilized in any model organism for any disease.


Subject(s)
Cholesterol/metabolism , Lysosomes/metabolism , Niemann-Pick Disease, Type C/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Sphingolipids/metabolism , Anaerobiosis/drug effects , Biological Transport/drug effects , Biological Transport/genetics , Cell Line , Cholesterol/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Lysosomes/genetics , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sphingolipids/genetics
7.
PLoS One ; 5(11): e15054, 2010 Nov 29.
Article in English | MEDLINE | ID: mdl-21124786

ABSTRACT

Cyclodextrins (CDs) have long been used to manipulate cellular cholesterol levels both in vitro and in vivo, but their direct effects at a cellular level are not well characterized. Recently, CDs have garnered much interest because of their ability to clear stored cholesterol from Niemann Pick Type C (NPC) cells and markedly prolong the life of NPC1 disease mice. Here, we investigate the hypothesis that treatment with 2-hydroxypropyl- ß-cyclodextrin (HPB-CD) stimulates lysosomal exocytosis in a calcium-enhanced manner. We propose that this exocytosis is the mechanism by which HPB-CD ameliorates the endolysosomal cholesterol storage phenotype in NPC cells. These findings have significant implications for the use of HPB-CD in biochemical assays and data interpretation as well as for their use for the treatment for NPC and other disorders.


Subject(s)
Calcium/metabolism , Exocytosis/drug effects , Lysosomes/metabolism , beta-Cyclodextrins/pharmacology , 2-Hydroxypropyl-beta-cyclodextrin , Animals , Blotting, Western , Cell Line , Cell Survival/drug effects , Cholesterol/metabolism , Excipients/pharmacology , Exosomes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Niemann-Pick C1 Protein , Proteins/genetics , Proteins/metabolism , beta-N-Acetylhexosaminidases/metabolism
8.
J Lipid Res ; 50(5): 915-23, 2009 May.
Article in English | MEDLINE | ID: mdl-19141868

ABSTRACT

Diosgenin exists in some food supplements and herbal medicines and lowers plasma cholesterol by increasing fecal cholesterol excretion. It is believed that diosgenin promotes fecal cholesterol excretion by stimulating biliary cholesterol secretion and decreasing intestinal cholesterol absorption. Niemann-Pick C1-like 1 (NPC1L1) was recently identified as an essential protein for intestinal cholesterol absorption. To determine the relative contribution of biliary secretion and intestinal absorption of cholesterol in diosgenin-stimulated fecal cholesterol excretion, wild-type (WT) and NPC1L1-knockout (L1KO) mice were fed a diet with or without 1% diosgenin. Fecal cholesterol excretion (mumol/day/100 g body weight) increased in diosgenin-fed WT and L1KO mice from 4.2 to 52 and from 63 to 140, respectively. Surprisingly, this increase in diosgenin-treated versus untreated L1KO mice (77) was even greater than that seen in diosgenin-treated versus untreated WT mice (47.8). Additionally, WT and L1KO mice fed the diosgenin diet had similar increases in biliary cholesterol concentration, despite unaltered hepatic expression of the hepatobiliary cholesterol transporter, ATP binding cassette transporters G5 and G8. Facilitated cholesterol excretion in diosgenin-treated WT and L1KO mice was associated with decreased hepatic and plasma cholesterol and increased liver expression of cholesterol synthetic genes. In contrast, diosgenin had no effect on the intestinal expression of NPC1L1 and cholesterol synthetic genes. In an in vitro assay, diosgenin was unable to block NPC1L1-dependent cholesterol uptake. In conclusion, diosgenin stimulation of fecal cholesterol excretion is independent of NPC1L1-mediated cholesterol absorption.


Subject(s)
Cholesterol/metabolism , Diosgenin/metabolism , Feces/chemistry , Membrane Transport Proteins/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Cell Line , Diet , Diosgenin/administration & dosage , Humans , Intestinal Mucosa/metabolism , Lipoproteins/metabolism , Liver/metabolism , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats
9.
J Lipid Res ; 50(2): 293-300, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18796403

ABSTRACT

Mice lacking Niemann-Pick C1-Like 1 (NPC1L1) (NPC1L1(-/-)mice) exhibit a defect in intestinal absorption of cholesterol and phytosterols. However, wild-type (WT) mice do not efficiently absorb and accumulate phytosterols either. Cell-based studies show that NPC1L1 is a much weaker transporter for phytosterols than cholesterol. In this study, we examined the role of NPC1L1 in phytosterol and cholesterol trafficking in mice lacking ATP-binding cassette (ABC) transporters G5 and G8 (G5/G8(-/-) mice). G5/G8(-/-) mice develop sitosterolemia, a genetic disorder characterized by the accumulation of phytosterols in blood and tissues. We found that mice lacking ABCG5/G8 and NPC1L1 [triple knockout (TKO) mice] did not accumulate phytosterols in plasma and the liver. TKO mice, like G5/G8(-/-) mice, still had a defect in hepatobiliary cholesterol secretion, which was consistent with TKO versus NPC1L1(-/-) mice exhibiting a 52% reduction in fecal cholesterol excretion. Because fractional cholesterol absorption was reduced similarly in NPC1L1(-/-) and TKO mice, by subtracting fecal cholesterol excretion in TKO mice from NPC1L1(-/-) mice, we estimated that a 25g NPC1L1(-/-) mouse may secrete about 4 mumol of cholesterol daily via the G5/G8 pathway. In conclusion, NPC1L1 is essential for phytosterols to enter the body in mice.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Lipoproteins/genetics , Membrane Transport Proteins/genetics , Sitosterols/blood , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/metabolism , Animals , Cholesterol/metabolism , Lipoproteins/metabolism , Liver/metabolism , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Phytosterols/blood , Phytosterols/metabolism , Sitosterols/metabolism
10.
Biol Cell ; 101(3): 141-52, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18681838

ABSTRACT

BACKGROUND INFORMATION: Within the group of lysosomal storage diseases, NPC1 [NPC (Niemann-Pick type C) 1] disease is a lipidosis characterized by excessive accumulation of free cholesterol as well as gangliosides, glycosphingolipids and fatty acids in the late E/L (endosomal/lysosomal) system (Chen et al., 2005) due to a defect in late endosome lipid egress. We have previously demonstrated that expression of the small GTPase Rab9 in NPC1 cells can rescue the lipid transport block phenotype (Walter et al., 2003), albeit by an undefined mechanism. RESULTS: To investigate further the mechanism by which Rab9 facilitates lipid movement from late endosomes we sought to identify novel Rab9 binding/interacting proteins. In the present study, we report that Rab9 interacts with the intermediate filament phosphoprotein vimentin and this interaction is altered by lipid accumulation in late endosomes, which results in inhibition of PKC (protein kinase C) and hypophosphorylation of vimentin, leading to late endosome dysfunction. Intermediate filament hypophosphorylation, aggregation and entrapment of Rab9 ultimately leads to transport defects and inhibition of lipid egress from late endosomes. CONCLUSIONS: These results reveal a previously unappreciated interaction between Rab proteins and intermediate filaments in regulating intracellular lipid transport.


Subject(s)
Endosomes/metabolism , Niemann-Pick Disease, Type C/metabolism , Protein Kinase C/metabolism , Vimentin/metabolism , rab GTP-Binding Proteins/metabolism , Cell Line , Cholesterol/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Humans , Intermediate Filaments/metabolism , Lipid Metabolism , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Phosphorylation , Protein Binding , Sphingosine/pharmacology , rab GTP-Binding Proteins/genetics
11.
Am J Physiol Gastrointest Liver Physiol ; 295(4): G776-83, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18718999

ABSTRACT

The impact of NPC1L1 and ezetimibe on cholesterol absorption are well documented. However, their potential consequences relative to absorption and metabolism of other nutrients have been only minimally investigated. Thus studies were undertaken to investigate the possible effects of this protein and drug on fat absorption, weight gain, and glucose metabolism by using Npc1l1(-/-) and ezetimibe-treated mice fed control and high-fat, high-sucrose diets. Results show that lack of NPC1L1 or treatment with ezetimibe reduces weight gain when animals are fed a diabetogenic diet. This resistance to diet-induced obesity results, at least in part, from significantly reduced absorption of dietary saturated fatty acids, particularly stearate and palmitate, since food intake did not differ between groups. Expression analysis showed less fatty acid transport protein 4 (FATP4) in intestinal scrapings of Npc1l1(-/-) and ezetimibe-treated mice, suggesting an important role for FATP4 in intestinal absorption of long-chain fatty acids. Concomitant with resistance to weight gain, lack of NPC1L1 or treatment with ezetimibe also conferred protection against diet-induced hyperglycemia and insulin resistance. These unexpected beneficial results may be clinically important, given the focus on NPC1L1 as a target for the treatment of hypercholesterolemia.


Subject(s)
Azetidines/pharmacology , Diabetes Mellitus/etiology , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Fatty Acids/metabolism , Intestinal Absorption/physiology , Membrane Transport Proteins/deficiency , Obesity/prevention & control , Animals , Diabetes Mellitus/prevention & control , Ezetimibe , Fatty Acid Transport Proteins/biosynthesis , Female , Hyperglycemia/prevention & control , Male , Membrane Transport Proteins/physiology , Mice
12.
Arterioscler Thromb Vasc Biol ; 28(3): 448-54, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18187667

ABSTRACT

OBJECTIVE: Activation of liver x receptor (LXR) raises plasma HDL-cholesterol (HDL-C) in mice. Interestingly, the LXR agonist GW3965 fails to raise plasma HDL-C in mice lacking intestinal ABCA1, indicating that intestinal ABCA1 plays a predominant role in GW3965-mediated HDL production. How this is coupled to intestinal function remains elusive. Because cholesterol is essential for HDL assembly and directly regulates intestinal ABCA1 expression via activating LXR, we hypothesized that cholesterol absorption, a major function of intestine, modulates LXR-dependent HDL formation. METHODS AND RESULTS: Mice lacking Niemann-Pick C1-Like 1 (NPC1L1) (L1-KO mice), a gene that is essential for cholesterol absorption, were treated with LXR agonist T0901317 for 7 days. Intriguingly, this treatment failed to significantly raise plasma HDL-C but caused a much greater fecal cholesterol excretion in L1-KO mice. The intestinal ABCA1 mRNA level was about 4-fold lower in L1-KO versus wild-type mice, and increased 3.9-fold and 8.8-fold after T0901317 treatment in wild-type and L1-KO mice, respectively. Hepatic ABCA1 failed to respond to T0901317 in mice of both genotypes, although hepatic mRNAs for many LXR target genes were higher in the T0901317-treated versus untreated wild-type animals. CONCLUSIONS: NPC1L1 is required for an LXR agonist to increase plasma HDL-C in mice.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Benzoates/pharmacology , Benzylamines/pharmacology , Cholesterol, HDL/drug effects , Cholesterol, HDL/metabolism , Niemann-Pick Disease, Type C/drug therapy , ATP-Binding Cassette Transporters/genetics , Analysis of Variance , Animals , Blood Chemical Analysis , Cholesterol, HDL/blood , DNA-Binding Proteins , Disease Models, Animal , Injections, Intraperitoneal , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Niemann-Pick Disease, Type C/blood , Niemann-Pick Disease, Type C/genetics , Orphan Nuclear Receptors , Probability , Random Allocation , Receptors, Cytoplasmic and Nuclear , Sensitivity and Specificity
13.
Biochim Biophys Acta ; 1771(9): 1140-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17689140

ABSTRACT

We compared cholesterol uptake into brush border membrane vesicles (BBMV) made from the small intestines of either wild-type or Niemann-Pick C1-like 1 (NPC1L1) knockout mice to elucidate the contribution of NPC1L1 to facilitated uptake; this uptake involves cholesterol transport from lipid donor particles into the BBM of enterocytes. The lack of NPC1L1 in the BBM of the knockout mice had no effect on the rate of cholesterol uptake. It follows that NPC1L1 cannot be the putative high-affinity, ezetimibe-sensitive cholesterol transporter in the brush border membrane (BBM) as has been proposed by others. The following findings substantiate this conclusion: (I) NPC1L1 is not a brush border membrane protein but very likely localized to intracellular membranes; (II) the cholesterol absorption inhibitor ezetimibe and its analogues reduce cholesterol uptake to the same extent in wild-type and NPC1L1 knockout mouse BBMV. These findings indicate that the prevailing belief that NPC1L1 facilitates intestinal cholesterol uptake into the BBM and its interaction with ezetimibe is responsible for the inhibition of this process can no longer be sustained.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Cholesterol/metabolism , Intestinal Mucosa , Intestines , Membrane Transport Proteins/metabolism , Microvilli , Animals , Anticholesteremic Agents/chemistry , Azetidines/chemistry , Cell Membrane/metabolism , Ezetimibe , Intestinal Mucosa/metabolism , Intestines/anatomy & histology , Intestines/drug effects , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvilli/drug effects , Microvilli/metabolism , Transport Vesicles/metabolism
14.
J Clin Invest ; 117(7): 1968-78, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17571164

ABSTRACT

Niemann-Pick C1-like 1 (NPC1L1) is required for cholesterol absorption. Intestinal NPC1L1 appears to be a target of ezetimibe, a cholesterol absorption inhibitor that effectively lowers plasma LDL-cholesterol in humans. However, human liver also expresses NPC1L1. Hepatic function of NPC1L1 was previously unknown, but we recently discovered that NPC1L1 localizes to the canalicular membrane of primate hepatocytes and that NPC1L1 facilitates cholesterol uptake in hepatoma cells. Based upon these findings, we hypothesized that hepatic NPC1L1 allows the retention of biliary cholesterol by hepatocytes and that ezetimibe disrupts hepatic function of NPC1L1. To test this hypothesis, transgenic mice expressing human NPC1L1 in hepatocytes (L1-Tg mice) were created. Hepatic overexpression of NPC1L1 resulted in a 10- to 20-fold decrease in biliary cholesterol concentration, but not phospholipid and bile acid concentrations. This decrease was associated with a 30%-60% increase in plasma cholesterol, mainly because of the accumulation of apoE-rich HDL. Biliary and plasma cholesterol concentrations in these animals were virtually returned to normal with ezetimibe treatment. These findings suggest that in humans, ezetimibe may reduce plasma cholesterol by inhibiting NPC1L1 function in both intestine and liver, and hepatic NPC1L1 may have evolved to protect the body from excessive biliary loss of cholesterol.


Subject(s)
Azetidines/pharmacology , Bile/drug effects , Bile/metabolism , Cholesterol/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoproteins E/blood , Bile Acids and Salts/metabolism , Cell Membrane/metabolism , Cholesterol/blood , Ezetimibe , Humans , Liver/metabolism , Male , Membrane Proteins/genetics , Membrane Transport Proteins , Mice , Mice, Transgenic , Phospholipids/metabolism
15.
Biochim Biophys Acta ; 1771(9): 1132-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17442616

ABSTRACT

Recent studies have documented the importance of Niemann-Pick C1-like 1 protein (NPC1L1), a putative physiological target of the drug ezetimibe, in mediating intestinal cholesterol absorption. However, whether NPC1L1 is the high affinity cholesterol binding protein on intestinal brush border membranes is still controversial. In this study, brush border membrane vesicles (BBMV) from wild type and NPC1L1-/- mice were isolated and assayed for micellar cholesterol binding in the presence or absence of ezetimibe. Results confirmed the loss of the high affinity component of cholesterol binding when wild type BBMV preparations were incubated with antiserum against the class B type 1 scavenger receptor (SR-BI) in the reaction mixture similar to previous studies. Subsequently, second order binding of cholesterol was observed with BBMV from wild type and NPC1L1-/- mice. The inclusion of ezetimibe in these in vitro reaction assays resulted in the loss of the high affinity component of cholesterol interaction. Surprisingly, BBMVs from NPC1L1-/- mice maintained active binding of cholesterol. These results documented that SR-BI, not NPC1L1, is the major protein responsible for the initial high affinity cholesterol ligand binding process in the cholesterol absorption pathway. Additionally, ezetimibe may inhibit BBM cholesterol binding through targets such as SR-BI in addition to its inhibition of NPC1L1.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Cholesterol/metabolism , Intestinal Mucosa/metabolism , Membrane Transport Proteins/metabolism , Scavenger Receptors, Class B/metabolism , Transport Vesicles/metabolism , Animals , Anticholesteremic Agents/chemistry , Apolipoprotein A-I/pharmacology , Azetidines/chemistry , Dose-Response Relationship, Drug , Ezetimibe , Female , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/ultrastructure , Male , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Micelles , Microvilli/drug effects , Microvilli/metabolism , Microvilli/ultrastructure , Scavenger Receptors, Class B/genetics
16.
Dev Cell ; 12(4): 481-3, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17419988

ABSTRACT

The Niemann-Pick C proteins have slowly emerged as regulators of subcellular lipid transport and sterol absorption at the small intestine. A recent article in Cell Metabolism suggests that in addition to their significant structural and sequence homology, these proteins may orchestrate their functions in a previously unappreciated fashion (Voght et al., 2007).


Subject(s)
Biological Transport , Drosophila melanogaster/metabolism , Niemann-Pick Diseases/metabolism , Sterols/pharmacokinetics , Absorption , Animals , Cholesterol/pharmacokinetics , Mice , Models, Biological
17.
J Neurosci ; 27(8): 1879-91, 2007 Feb 21.
Article in English | MEDLINE | ID: mdl-17314284

ABSTRACT

Niemann-Pick disease type C (NPC) is an inherited lipid storage disorder caused by mutations in NPC1 or NPC2 genes. Loss of function of either protein results in the endosomal accumulation of cholesterol and other lipids, progressive neurodegeneration, and robust glial cell activation. Here, we report that cultured human NPC fibroblasts secrete interferon-beta, interleukin-6 (IL-6), and IL-8, and contain increased levels of signal transducers and activators of transcription (STATs). These cells also contained increased levels of Toll-like receptor 4 (TLR4) that accumulated in cholesterol-enriched endosomes/lysosomes, and small interfering RNA knockdown of this receptor reduced cytokine secretion. In the NPC1-/- mouse brain, glial cells expressed TLR4 and IL-6, whereas both glial and neuronal cells expressed STATs. Genetic deletion of TLR4 in NPC1-/- mice reduced IL-6 secretion by cultured fibroblasts but failed to alter STAT levels or glial cell activation in the brain. In contrast, genetic deletion of IL-6 normalized STAT levels and suppressed glial cell activation. These findings indicate that constitutive cytokine secretion leads to activation of STATs in NPC fibroblasts and that this secretion is partly caused by an endosomal accumulation of TLR4. These results also suggest that similar signaling events may underlie glial cell activation in the NPC1-/- mouse brain.


Subject(s)
Cytokines/metabolism , Endosomes/metabolism , Fibroblasts/metabolism , Niemann-Pick Disease, Type C/metabolism , STAT Transcription Factors/metabolism , Toll-Like Receptor 4/metabolism , Animals , Brain/pathology , Brain/physiopathology , Cells, Cultured , Culture Media/pharmacology , Fibroblasts/drug effects , Humans , Interferon-beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Longevity , Mice , Mice, Knockout , Neuroglia , Niemann-Pick Disease, Type C/pathology , Niemann-Pick Disease, Type C/physiopathology
18.
J Cell Sci ; 119(Pt 13): 2643-53, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16757520

ABSTRACT

Niemann-Pick disease type C (NPC) is an inherited lipid storage disorder caused by mutations in NPC1 or NPC2. NPC1 is a polytopic glycoprotein that contains a sterol-sensing domain, whereas NPC2 is a soluble protein that contains an MD-2-like lipid-recognition domain. In the current study, we addressed the hypothesis that ubiquitylation of NPC1 might be regulated by cholesterol. We found that depletion of cellular cholesterol facilitated ubiquitylation of NPC1 expressed in COS cells. A loss-of-function mutant, NPC1(P691S), which contains an amino acid substitution in the sterol-sensing domain, failed to respond to cholesterol depletion. Another mutant, NPC1(deltaLLNF), which lacks the endosomal-targeting motif, also failed to respond. SKD1(E235Q), a dominant-negative mutant of SKD1/Vps4 that inhibits disassembly of the endosomal sorting complex required for transport (ESCRT), caused an accumulation of ubiquitylated NPC1. SKD1(E235Q) associated with NPC1 on the endosomal membrane, whereas wild-type SKD1 associated with NPC1 only when cells were depleted of cholesterol. Similarly, in control human skin fibroblasts, cholesterol depletion facilitated ubiquitylation of endogenous NPC1. In patient cells that lack NPC2 function, NPC1 was ubiquitylated regardless of cellular cholesterol levels, suggesting that NPC2 is required to prevent NPC1 ubiquitylation under cholesterol-rich conditions. These results suggest that ubiquitylation of NPC1 and its association with the ESCRT complex are controlled by endosomal cholesterol levels utilizing a mechanism that involves NPC2.


Subject(s)
Adenosine Triphosphatases/metabolism , Carrier Proteins/metabolism , Cholesterol/pharmacology , Membrane Glycoproteins/metabolism , Protein Processing, Post-Translational/drug effects , Repressor Proteins/metabolism , Ubiquitin/metabolism , ATPases Associated with Diverse Cellular Activities , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Endosomal Sorting Complexes Required for Transport , Endosomes/metabolism , Fibroblasts/drug effects , Glycoproteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Leupeptins/pharmacology , Mutant Proteins/metabolism , Niemann-Pick C1 Protein , Protein Binding , Skin/cytology , Skin/drug effects , Vesicular Transport Proteins
19.
Curr Opin Lipidol ; 17(3): 221-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16680025

ABSTRACT

PURPOSE OF REVIEW: In the past 2 years the Niemann-Pick C1-like 1 protein has rapidly emerged as a key regulator of intestinal cholesterol absorption. This review covers the limited number of published reports to develop a hypothesis of the potential function of Niemann-Pick C1-like 1 protein and outlines questions that should be addressed in future studies. RECENT FINDINGS: Considerable disagreement regarding the potential function and subcellular location of Niemann-Pick C1-like 1 protein has complicated interpretation of published results and evaluation of their biologic significance. Recent reports suggest, however, that Niemann-Pick C1-like 1 protein plays a key role in modulating the subcellular fate of multiple lipids including cholesterol and sphingolipids. In addition, this function may require Niemann-Pick C1-like 1 protein to move between different cellular locations. SUMMARY: This paper proposes a model of Niemann-Pick C1-like 1 protein function based on available data and on knowledge of its close relative and homologue the Niemann-Pick C1 protein, whose precise function, albeit still elusive, is more extensively characterized. It also raises new questions with respect to Niemann-Pick C1-like 1 protein function and discusses potential future directions.


Subject(s)
Homeostasis , Lipid Metabolism , Membrane Transport Proteins/metabolism , Niemann-Pick Diseases/metabolism , Animals , Azetidines/pharmacology , Biological Transport , Ezetimibe , Humans
20.
Trends Biochem Sci ; 30(9): 498-505, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16054367

ABSTRACT

Cholesterol accumulation in the endosomes and lysosomes of Niemann-Pick C (NPC) cells is considered to be the hallmark of this disorder, so the main focus of NPC research has revolved around cholesterol and its role in disease pathogenesis. However, recent data indicate that cholesterol is not the primary culprit in this human lipidosis. I propose a new hypothesis for the potential action or function of the NPC1 protein in the endosome. In this context, the relationship of NPC2 and NPC1 is also discussed.


Subject(s)
Carrier Proteins/physiology , Cholesterol/pharmacokinetics , Membrane Glycoproteins/physiology , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/physiopathology , Cholesterol/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Models, Chemical , Niemann-Pick C1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL