Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 15(6): 436, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902268

ABSTRACT

Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide, necessitating the identification of novel therapeutic targets. Lysosome Associated Protein Transmembrane 4B (LAPTM4B) is involved in biological processes critical to cancer progression, such as regulation of solute carrier transporter proteins and metabolic pathways, including mTORC1. However, the metabolic processes governed by LAPTM4B and its role in oncogenesis remain unknown. In this study, we conducted unbiased metabolomic screens to uncover the metabolic landscape regulated by LAPTM4B. We observed common metabolic changes in several knockout cell models suggesting of a role for LAPTM4B in suppressing ferroptosis. Through a series of cell-based assays and animal experiments, we demonstrate that LAPTM4B protects tumor cells from erastin-induced ferroptosis both in vitro and in vivo. Mechanistically, LAPTM4B suppresses ferroptosis by inhibiting NEDD4L/ZRANB1 mediated ubiquitination and subsequent proteasomal degradation of the cystine-glutamate antiporter SLC7A11. Furthermore, metabolomic profiling of cancer cells revealed that LAPTM4B knockout leads to a significant enrichment of ferroptosis and associated metabolic alterations. By integrating results from cellular assays, patient tissue samples, an animal model, and cancer databases, this study highlights the clinical relevance of the LAPTM4B-SLC7A11-ferroptosis signaling axis in NSCLC progression and identifies it as a potential target for the development of cancer therapeutics.


Subject(s)
Amino Acid Transport System y+ , Carcinoma, Non-Small-Cell Lung , Ferroptosis , Lung Neoplasms , Proteasome Endopeptidase Complex , Ubiquitin , Ferroptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Animals , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Mice , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Oncogene Proteins/metabolism , Oncogene Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Line, Tumor , Ubiquitination , Mice, Nude , Proteolysis/drug effects
2.
Oncogenesis ; 12(1): 25, 2023 May 06.
Article in English | MEDLINE | ID: mdl-37147294

ABSTRACT

Osteosarcoma (OS) is a rare malignant bone tumor but is one leading cause of cancer mortality in childhood and adolescence. Cancer metastasis accounts for the primary reason for treatment failure in OS patients. The dynamic organization of the cytoskeleton is fundamental for cell motility, migration, and cancer metastasis. Lysosome Associated Protein Transmembrane 4B (LAPTM4B) is an oncogene participating in various biological progress central to cancer biogenesis. However, the potential roles of LAPTM4B in OS and the related mechanisms remain unknown. Here, we established the elevated LAPTM4B expression in OS, and it is essential in regulating stress fiber organization through RhoA-LIMK-cofilin signaling pathway. In terms of mechanism, our data revealed that LAPTM4B promotes RhoA protein stability by suppressing the ubiquitin-mediated proteasome degradation pathway. Moreover, our data show that miR-137, rather than gene copy number and methylation status, contributes to the upregulation of LAPTM4B in OS. We report that miR-137 is capable of regulating stress fiber arrangement, OS cell migration, and metastasis via targeting LAPTM4B. Combining results from cells, patients' tissue samples, the animal model, and cancer databases, this study further suggests that the miR-137-LAPTM4B axis represents a clinically relevant pathway in OS progression and a viable target for novel therapeutics.

3.
J Biol Chem ; 299(4): 104607, 2023 04.
Article in English | MEDLINE | ID: mdl-36924944

ABSTRACT

The glycolipid transfer protein (GLTP) has been linked to many cellular processes aside from its best-known in vitro function as a lipid transport protein. It has been proposed to act as a sensor and regulator of glycosphingolipid homeostasis in cells. Furthermore, through its previously determined interaction with the endoplasmic reticulum membrane protein VAP-A (vesicle-associated membrane protein-associated protein A), GLTP may also be involved in facilitating vesicular transport in cells. In this study, we characterized the phenotype of CRISPR/Cas9-mediated GLTP KO HeLa cells. We showed that motility, three-dimensional growth, and cellular metabolism were all altered by GLTP knockout. Expression of a GLTP mutant incapable of binding VAP disrupted cell spheroid formation, indicating that the GLTP-VAP interaction is linked to cellular adhesion, cohesion, and three-dimensional growth. Most notably, we found evidence that GLTP, through its interaction with VAP-A, affects vesicular trafficking, marking the first cellular process discovered to be directly impacted by a change in GLTP expression.


Subject(s)
Biological Transport , Carrier Proteins , Cell Membrane , Humans , Biological Transport/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , HeLa Cells , Gene Knockout Techniques , Protein Binding/genetics , Gene Expression Regulation/genetics , Cytosol/metabolism , Cell Movement/genetics
4.
Article in English | MEDLINE | ID: mdl-33181324

ABSTRACT

Lysosome Associated Protein Transmembrane 4B (LAPTM4B) is a four-membrane spanning ceramide interacting protein that regulates mTORC1 signaling. Here, we show that LAPTM4B is sorted into intraluminal vesicles (ILVs) of multivesicular endosomes (MVEs) and released in small extracellular vesicles (sEVs) into conditioned cell culture medium and human urine. Efficient sorting of LAPTM4B into ILV membranes depends on its third transmembrane domain containing a sphingolipid interaction motif (SLim). Unbiased lipidomic analysis reveals a strong enrichment of glycosphingolipids in sEVs secreted from LAPTM4B knockout cells and from cells expressing a SLim-deficient LAPTM4B mutant. The altered sphingolipid profile is accompanied by a distinct SLim-dependent co-modulation of ether lipid species. The changes in the lipid composition of sEVs derived from LAPTM4B knockout cells is reflected by an increased stability of membrane nanodomains of sEVs. These results identify LAPTM4B as a determinant of the glycosphingolipid profile and membrane properties of sEVs.


Subject(s)
Exosomes/metabolism , Glycosphingolipids/metabolism , Membrane Proteins/metabolism , Oncogene Proteins/metabolism , Cell Line, Tumor , Endosomes/metabolism , Gene Knockout Techniques , Humans , Lipid Metabolism , Lipidomics , Membrane Proteins/genetics , Oncogene Proteins/genetics
5.
Am J Pathol ; 190(10): 2018-2028, 2020 10.
Article in English | MEDLINE | ID: mdl-32679228

ABSTRACT

Studies of lysosome associated protein transmembrane 4B (LAPTM4B) have mainly focused on the 35-kDa isoform and its association with poor prognosis in cancers. Here, by employing a novel monoclonal antibody, the authors found that the 24-kDa LAPTM4B isoform predominated in most, both healthy and malignant, human cells and tissues studied. LAPTM4B-24 lacks the extreme N-terminus and, contrary to LAPTM4B-35, failed to promote cell migration. The endogenous LAPTM4B-24 protein was subject to rapid turnover with a t1/2 of approximately 1 hour. The protein was degraded by both lysosomal and proteasomal pathways, and its levels were increased by the availability of nutrients and lysosomal ceramide. These findings underscore the pathophysiological relevance of the LAPTM4B-24 isoform and identify it as a dynamically regulated effector in lysosomal nutrient signaling.


Subject(s)
Cell Movement/physiology , Ceramides/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Oncogene Proteins/metabolism , Humans , Protein Isoforms/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
6.
ACS Cent Sci ; 4(5): 548-558, 2018 May 23.
Article in English | MEDLINE | ID: mdl-29806001

ABSTRACT

Membrane proteins are functionally regulated by the composition of the surrounding lipid bilayer. The late endosomal compartment is a central site for the generation of ceramide, a bioactive sphingolipid, which regulates responses to cell stress. The molecular interactions between ceramide and late endosomal transmembrane proteins are unknown. Here, we uncover in atomistic detail the ceramide interaction of Lysosome Associated Protein Transmembrane 4B (LAPTM4B), implicated in ceramide-dependent cell death and autophagy, and its functional relevance in lysosomal nutrient signaling. The ceramide-mediated regulation of LAPTM4B depends on a sphingolipid interaction motif and an adjacent aspartate residue in the protein's third transmembrane (TM3) helix. The interaction motif provides the preferred contact points for ceramide while the neighboring membrane-embedded acidic residue confers flexibility that is subject to ceramide-induced conformational changes, reducing TM3 bending. This facilitates the interaction between LAPTM4B and the amino acid transporter heavy chain 4F2hc, thereby controlling mTORC signaling. These findings provide mechanistic insights into how transmembrane proteins sense and respond to ceramide.

7.
Int J Mol Sci ; 18(5)2017 May 01.
Article in English | MEDLINE | ID: mdl-28468312

ABSTRACT

Neuronal ceroid lipofuscinoses (NCLs) are autosomal recessive progressive encephalopathies caused by mutations in at least 14 different genes. Despite extensive studies performed in different NCL animal models, the molecular mechanisms underlying neurodegeneration in NCLs remain poorly understood. To model NCL in human cells, we generated induced pluripotent stem cells (iPSCs) by reprogramming skin fibroblasts from a patient with CLN5 (ceroid lipofuscinosis, neuronal, 5) disease, the late infantile variant form of NCL. These CLN5 patient-derived iPSCs (CLN5Y392X iPSCs) harbouring the most common CLN5 mutation, c.1175_1176delAT (p.Tyr392X), were further differentiated into neural lineage cells, the most affected cell type in NCLs. The CLN5Y392X iPSC-derived neural lineage cells showed accumulation of autofluorescent storage material and subunit C of the mitochondrial ATP synthase, both representing the hallmarks of many forms of NCLs, including CLN5 disease. In addition, we detected abnormalities in the intracellular organelles and aberrations in neuronal sphingolipid transportation, verifying the previous findings obtained from Cln5-deficient mouse macrophages. Therefore, patient-derived iPSCs provide a suitable model to study the mechanisms of NCL diseases.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Phenotype , Cell Differentiation , Cell Lineage , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Lysosomal Membrane Proteins , Mutation , Neuronal Ceroid-Lipofuscinoses/pathology
8.
Chem Phys Lipids ; 194: 29-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26343174

ABSTRACT

Lipids are often introduced into cell membranes directly from solvent or from lipophilic artificial carriers, such as cyclodextrins. A physiological lipid entry route into mammalian cells is via lipoprotein mediated uptake. In this review, we discuss the introduction of BODIPY-labeled sterol and sphingolipid analogs into mammalian cells via high- or low-density lipoproteins, and the novel findings made by using this strategy. Lipoprotein mediated delivery favors endocytic uptake and initial incorporation of the lipid into membranes of the endosomal compartments. This routing can therefore highlight physiological mechanisms of lipid entry into and exit from the endo-lysosomal membrane system. The underlying principles are of key importance for instance in controlling plasma cholesterol levels and in the development and regression of lysosomal lipid storage diseases. A common denominator for the BODIPY-labeled lipid analogs discussed in this review is that they were synthesized by late Robert Bittman, whose scientific impact radiates far beyond his lifework in organic chemistry.


Subject(s)
Boron Compounds/metabolism , Fluorescent Dyes/metabolism , Lipoproteins/metabolism , Sphingolipids/metabolism , Sterols/metabolism , Animals , Biological Transport , Boron Compounds/chemistry , Cell Membrane/chemistry , Cell Membrane/metabolism , Endosomes/chemistry , Endosomes/metabolism , Fluorescent Dyes/chemistry , Humans , Lipoproteins/chemistry , Sphingolipids/chemistry , Sterols/chemistry
9.
Lipid Insights ; 8(Suppl 1): 11-20, 2015.
Article in English | MEDLINE | ID: mdl-26715852

ABSTRACT

Ceramide and sphingosine and their phosphorylated counterparts are recognized as "bioactive sphingolipids" and modulate membrane integrity, the activity of enzymes, or act as ligands of G protein-coupled receptors. The subcellular distribution of the bioactive sphingolipids is central to their function as the same lipid can mediate diametrically opposite effects depending on its location. To ensure that these lipids are present in the right amount and in the appropriate organelles, cells employ selective lipid transport and compartmentalize sphingolipid-metabolizing enzymes to characteristic subcellular sites. Our knowledge of key mechanisms involved in sphingolipid signaling and trafficking has increased substantially in the past decades-thanks to advances in biochemical and cell biological methods. In this review, we focus on the bioactive sphingolipids and discuss how the combination of studies in cells and in model membranes have contributed to our understanding of how they behave and function in living organisms.

10.
Nat Chem Biol ; 11(10): 799-806, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26280656

ABSTRACT

Lysosome-associated protein transmembrane-4b (LAPTM4B) associates with poor prognosis in several cancers, but its physiological function is not well understood. Here we use novel ceramide probes to provide evidence that LAPTM4B interacts with ceramide and facilitates its removal from late endosomal organelles (LEs). This lowers LE ceramide in parallel with and independent of acid ceramidase-dependent catabolism. In LAPTM4B-silenced cells, LE sphingolipid accumulation is accompanied by lysosomal membrane destabilization. However, these cells resist ceramide-driven caspase-3 activation and apoptosis induced by chemotherapeutic agents or gene silencing. Conversely, LAPTM4B overexpression reduces LE ceramide and stabilizes lysosomes but sensitizes to drug-induced caspase-3 activation. Together, these data uncover a cellular ceramide export route from LEs and identify LAPTM4B as its regulator. By compartmentalizing ceramide, LAPTM4B controls key sphingolipid-mediated cell death mechanisms and emerges as a candidate for sphingolipid-targeting cancer therapies.


Subject(s)
Apoptosis/physiology , Ceramides/metabolism , Endosomes/metabolism , Membrane Proteins/metabolism , Oncogene Proteins/metabolism , Anthracyclines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biological Transport , Cell Line, Tumor , Gene Silencing , Humans , Intracellular Membranes/metabolism , Membrane Proteins/genetics , Oncogene Proteins/genetics , Paclitaxel/pharmacology , Protein Binding , RNA, Small Interfering/genetics , Sphingomyelins/metabolism
11.
Biochim Biophys Acta ; 1853(9): 2173-82, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25892494

ABSTRACT

Caveolae are plasma membrane invaginations enriched in sterols and sphingolipids. Sphingosine kinase 1 (SK1) is an oncogenic protein that converts sphingosine to sphingosine 1-phosphate (S1P), which is a messenger molecule involved in calcium signaling. Caveolae contain calcium responsive proteins, but the effects of SK1 or S1P on caveolar calcium signaling have not been investigated. We generated a Caveolin-1-Aequorin fusion protein (Cav1-Aeq) that can be employed for monitoring the local calcium concentration at the caveolae ([Ca²âº]cav). In HeLa cells, Cav1-Aeq reported different [Ca²âº] as compared to the plasma membrane [Ca²âº] in general (reported by SNAP25-Aeq) or as compared to the cytosolic [Ca²âº] (reported by cyt-Aeq). The Ca²âº signals detected by Cav1-Aeq were significantly attenuated when the caveolar structures were disrupted by methyl-ß-cyclodextrin, suggesting that the caveolae are specific targets for Ca²âº signaling. HeLa cells overexpressing SK1 showed increased [Ca²âº]cav during histamine-induced Ca²âº mobilization in the absence of extracellular Ca²âº as well as during receptor-operated Ca²âº entry (ROCE). The SK1-induced increase in [Ca²âº]cav during ROCE was reverted by S1P receptor antagonists. In accordance, pharmacologic inhibition of SK1 reduced the [Ca²âº]cav during ROCE. S1P treatment stimulated the [Ca²âº]cav upon ROCE. The Ca²âº responses at the plasma membrane in general were not affected by SK1 expression. In summary, our results show that SK1/S1P-signaling regulates Ca²âº signals at the caveolae. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.


Subject(s)
Aequorin/biosynthesis , Calcium Signaling/physiology , Caveolae/metabolism , Caveolin 1/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Recombinant Fusion Proteins/biosynthesis , Aequorin/genetics , Calcium/metabolism , Calcium Signaling/drug effects , Caveolin 1/genetics , HeLa Cells , Humans , Lysophospholipids/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Recombinant Fusion Proteins/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
12.
Dev Cell ; 27(3): 249-62, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24209575

ABSTRACT

Mammalian cells acquire cholesterol, a major membrane constituent, via low-density lipoprotein (LDL) uptake. However, the mechanisms by which LDL cholesterol reaches the plasma membrane (PM) have remained obscure. Here, we applied LDL labeled with BODIPY cholesteryl linoleate to identify this pathway in living cells. The egress of BODIPY cholesterol (BC) from late endosomal (LE) organelles was dependent on acid lipase and Niemann-Pick C1 (NPC1) protein, as for natural cholesterol. We show that NPC1 was needed to recruit Rab8a to BC-containing LEs, and Rab8a enhanced the motility and segregation of BC- and CD63-positive organelles from lysosomes. The BC carriers docked to the cortical actin by a Rab8a- and Myosin5b (Myo5b)-dependent mechanism, typically in the proximity of focal adhesions (FAs). LDL increased the number and dynamics of FAs and stimulated cell migration in an acid lipase, NPC1, and Rab8a-dependent fashion, providing evidence that this cholesterol delivery route to the PM is important for cell movement.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Cholesterol, LDL/metabolism , Myosins/metabolism , rab GTP-Binding Proteins/metabolism , Biological Transport , Carrier Proteins/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Endosomes/drug effects , Endosomes/metabolism , Fluorescent Antibody Technique , Focal Adhesions/physiology , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Microscopy, Immunoelectron , Niemann-Pick C1 Protein , Porphobilinogen/analogs & derivatives , Porphobilinogen/pharmacology , Tetraspanin 30/metabolism , Wound Healing/drug effects
13.
J Cell Sci ; 126(Pt 17): 3961-71, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23813961

ABSTRACT

N-myc downstream-regulated gene 1 (NDRG1) mutations cause Charcot-Marie-Tooth disease type 4D (CMT4D). However, the cellular function of NDRG1 and how it causes CMT4D are poorly understood. We report that NDRG1 silencing in epithelial cells results in decreased uptake of low-density lipoprotein (LDL) due to reduced LDL receptor (LDLR) abundance at the plasma membrane. This is accompanied by the accumulation of LDLR in enlarged EEA1-positive endosomes that contain numerous intraluminal vesicles and sequester ceramide. Concomitantly, LDLR ubiquitylation is increased but its degradation is reduced and ESCRT (endosomal sorting complex required for transport) proteins are downregulated. Co-depletion of IDOL (inducible degrader of the LDLR), which ubiquitylates the LDLR and promotes its degradation, rescues plasma membrane LDLR levels and LDL uptake. In murine oligodendrocytes, Ndrg1 silencing not only results in reduced LDL uptake but also in downregulation of the oligodendrocyte differentiation factor Olig2. Both phenotypes are rescued by co-silencing of Idol, suggesting that ligand uptake through LDLR family members controls oligodendrocyte differentiation. These findings identify NDRG1 as a novel regulator of multivesicular body formation and endosomal LDLR trafficking. The deficiency of functional NDRG1 in CMT4D might impair lipid processing and differentiation of myelinating cells.


Subject(s)
Cell Cycle Proteins/metabolism , Charcot-Marie-Tooth Disease/metabolism , Endosomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Receptors, LDL/metabolism , Refsum Disease/metabolism , Androstenes/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Line, Tumor , Cell Membrane/metabolism , Charcot-Marie-Tooth Disease/genetics , Down-Regulation , Endocytosis/genetics , Endosomal Sorting Complexes Required for Transport/biosynthesis , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lipoproteins, LDL/metabolism , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Protein Transport/genetics , RNA Interference , RNA, Small Interfering , Refsum Disease/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
15.
Traffic ; 13(9): 1234-43, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22607065

ABSTRACT

The late endosomal/lysosomal compartment (LE/LY) plays a key role in sphingolipid breakdown, with the last degradative step catalyzed by acid ceramidase. The released sphingosine can be converted to ceramide in the ER and transported by ceramide transfer protein (CERT) to the Golgi for conversion to sphingomyelin. The mechanism by which sphingosine exits LE/LY is unknown but Niemann-Pick C1 protein (NPC1) has been suggested to be involved. Here, we used sphingomyelin, ceramide and sphingosine labeled with [(3)H] in carbon-3 of the sphingosine backbone and targeted them to LE/LY in low-density lipoprotein (LDL) particles. These probes traced LE/LY sphingolipid degradation and recycling as suggested by (1) accumulation of [(3)H]-sphingomyelin-derived [(3)H]-ceramide and depletion of [(3)H]-sphingosine upon acid ceramidase depletion, and (2) accumulation of [(3)H]-sphingosine-derived [(3)H]-ceramide and attenuation of [(3)H]-sphingomyelin synthesis upon CERT depletion. NPC1 silencing did not result in the accumulation of [(3)H]-sphingosine derived from [(3)H]-sphingomyelin/LDL or [(3)H]-ceramide/LDL. Additional evidence against NPC1 playing a significant role in LE/LY sphingosine export was obtained in experiments using the [(3)H]-sphingolipids or a fluorescent sphingosine derivative in NPC1 knock-out cells. Instead, NPC1-deficient cells displayed an increased affinity for sphingosine independently of protein-mediated lipid transport. This likely contributes to the increased sphingosine content of NPC1 cells.


Subject(s)
Membrane Glycoproteins/deficiency , Sphingosine/metabolism , Animals , CHO Cells , Carrier Proteins , Cell Line, Tumor , Ceramides/metabolism , Cricetinae , Cricetulus , Endosomes/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Lipoproteins, LDL/metabolism , Lysosomes/metabolism , Niemann-Pick C1 Protein , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Sphingolipidoses/metabolism , Sphingomyelins/metabolism
16.
Neurobiol Dis ; 46(1): 19-29, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22182690

ABSTRACT

CLN5 disease, late infantile variant phenotype neuronal ceroid lipofuscinosis, is a severe neurodegenerative disease caused by mutations in the CLN5 gene, which encodes a lysosomal protein of unknown function. Cln5-deficiency in mice leads to loss of thalamocortical neurons, and glial activation, but the underlying mechanisms are poorly understood. We have now studied the gene expression of Cln5 in the mouse brain and show that it increases gradually with age and differs between neurons and glia, with the highest expression in microglia. In Cln5(-/-) mice, we documented early and significant microglial activation that was already evident at 3 months of age. Loss of Cln5 also leads to defective myelination in vitro and in the developing mouse brain. This was accompanied by early alterations in serum lipid profiles, dysfunctional cellular metabolism and lipid transport in Cln5(-/-) mice. Taken together, these data provide significant new information about events associated with Cln5-deficiency, revealing altered myelination and disturbances in lipid metabolism, together with an early neuroimmune response.


Subject(s)
Demyelinating Diseases/physiopathology , Lipid Metabolism/physiology , Membrane Glycoproteins/deficiency , Microglia/metabolism , Animals , Cells, Cultured , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Disease Models, Animal , Lipid Metabolism/genetics , Lipid Metabolism Disorders/genetics , Lipid Metabolism Disorders/metabolism , Lipid Metabolism Disorders/pathology , Lysosomal Membrane Proteins , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/metabolism , Neurons/pathology
17.
Cold Spring Harb Perspect Biol ; 3(8): a004713, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21482741

ABSTRACT

Eukaryotic cells can synthesize thousands of different lipid molecules that are incorporated into their membranes. This involves the activity of hundreds of enzymes with the task of creating lipid diversity. In addition, there are several, typically redundant, mechanisms to transport lipids from their site of synthesis to other cellular membranes. Biosynthetic lipid transport helps to ensure that each cellular compartment will have its characteristic lipid composition that supports the functions of the associated proteins. In this article, we provide an overview of the biosynthesis of the major lipid constituents of cell membranes, that is, glycerophospholipids, sphingolipids, and sterols, and discuss the mechanisms by which these newly synthesized lipids are delivered to their target membranes.


Subject(s)
Cell Membrane/metabolism , Lipid Metabolism , Membrane Lipids/biosynthesis , Animals , Humans
18.
BMC Cell Biol ; 11: 45, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20573281

ABSTRACT

BACKGROUND: Sphingosine-1-phosphate (S1P) is a bioactive lipid that regulates a multitude of cellular functions, including cell proliferation, survival, migration and angiogenesis. S1P mediates its effects either by signaling through G protein-coupled receptors (GPCRs) or through an intracellular mode of action. In this study, we have investigated the mechanism behind S1P-induced survival signalling. RESULTS: We found that S1P protected cells from FasL-induced cell death in an NF-kappaB dependent manner. NF-kappaB was activated by extracellular S1P via S1P2 receptors and Gi protein signaling. Our study also demonstrates that extracellular S1P stimulates cells to rapidly produce and secrete additional S1P, which can further amplify the NF-kappaB activation. CONCLUSIONS: We propose a self-amplifying loop of autocrine S1P with capacity to enhance cell survival. The mechanism provides increased understanding of the multifaceted roles of S1P in regulating cell fate during normal development and carcinogenesis.


Subject(s)
Lysophospholipids/biosynthesis , NF-kappa B/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Autocrine Communication , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fas Ligand Protein/metabolism , Feedback, Physiological , Flavonoids/pharmacology , Humans , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , Receptors, Lysosphingolipid/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingosine/antagonists & inhibitors , Sphingosine/biosynthesis , Sphingosine/genetics , Transgenes/genetics
19.
Circ Res ; 106(4): 720-9, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20056921

ABSTRACT

RATIONALE: The synthetic sphingosine analog FTY720 is undergoing clinical trials as an immunomodulatory compound, acting primarily via sphingosine 1-phosphate receptor activation. Sphingolipid and cholesterol homeostasis are closely connected but whether FTY720 affects atherogenesis in humans is not known. OBJECTIVE: We examined the effects of FTY720 on the processing of scavenged lipoprotein cholesterol in human primary monocyte-derived macrophages. METHODS AND RESULTS: FTY720 did not affect cholesterol uptake but inhibited its delivery to the endoplasmic reticulum, reducing cellular free cholesterol cytotoxicity. This was accompanied by increased levels of Niemann-Pick C1 protein (NPC1) and ATP-binding cassette transporter (ABC)A1 proteins and increased efflux of endosomal cholesterol to apolipoprotein A-I. These effects were not dependent on sphingosine 1-phosphate receptor activation. Instead, FTY720 stimulated the production of 27-hydroxycholesterol, an endogenous ligand of the liver X receptor, leading to liver X receptor-induced upregulation of ABCA1. Fluorescently labeled FTY720 was targeted to late endosomes, and the FTY720-induced upregulation of ABCA1 was NPC1-dependent, but the endosomal exit of FTY720 itself was not. CONCLUSIONS: We conclude that FTY720 decreases cholesterol toxicity in primary human macrophages by reducing the delivery of scavenged lipoprotein cholesterol to the endoplasmic reticulum and facilitating its release to physiological extracellular acceptors. Furthermore, FTY720 stimulates 27-hydroxycholesterol production, providing an explanation for the atheroprotective effects and identifying a novel mechanism by which FTY720 modulates signaling.


Subject(s)
Atherosclerosis/prevention & control , Cholesterol/metabolism , Hydroxycholesterols/metabolism , Macrophages/drug effects , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/metabolism , Atherosclerosis/metabolism , Biological Transport , Carrier Proteins/metabolism , Cell Culture Techniques , Cell Death , Cell Survival , Cells, Cultured , Cholesterol Esters/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endosomes/drug effects , Endosomes/metabolism , Fingolimod Hydrochloride , Humans , Intracellular Signaling Peptides and Proteins , Lipoproteins, LDL/metabolism , Liver X Receptors , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Niemann-Pick C1 Protein , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/metabolism , Receptors, Scavenger/drug effects , Receptors, Scavenger/metabolism , Signal Transduction/drug effects , Sphingosine/pharmacology , Time Factors
20.
J Cell Physiol ; 216(1): 245-52, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18431718

ABSTRACT

The mammalian canonical transient receptor channels (TRPCs) are considered to be candidates for store-operated calcium channels (SOCCs). Many studies have addressed how TRPC3 channels are affected by depletion of intracellular calcium stores. Conflicting results have been shown for TRPC3 regarding its function, and this has been linked to its level of expression in various systems. In the present study, we have investigated how overexpression of TRPC3 interferes with the regulation of intracellular calcium stores. We demonstrate that overexpression of TRPC3 reduces the mobilization of calcium in response to stimulation of the cells with thapsigargin (TG) or the G-protein coupled receptor agonist sphingosine-1-phosphate (S1P). Our results indicate that this is the result of the expression of TRPC3 channels in the endoplasmic reticulum (ER), thus depleting ER calcium stores. OAG evoked calcium entry in cells overexpressing TRPC3, indicating that functional TRPC3 channels were also expressed in the plasma membrane. Taken together, our results show that overexpression of the putative SOCC, TRPC3, actually reduces the calcium content of intracellular stores, but does not enhance agonist-evoked or store-dependent calcium entry. Our results may, in part, explain the conflicting results obtained in previous studies on the actions of TRPC3 channels.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , TRPC Cation Channels/metabolism , Animals , Calcium Channels/genetics , Calnexin/metabolism , Cell Line , Diglycerides/metabolism , Endoplasmic Reticulum/metabolism , Humans , Ion Channel Gating , Lysophospholipids/metabolism , Patch-Clamp Techniques , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , TRPC Cation Channels/genetics , Thapsigargin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...