Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Elife ; 132024 Apr 11.
Article in English | MEDLINE | ID: mdl-38602170

ABSTRACT

Stains are known to be anti-inflammatory, but the mechanism remains poorly understood. Here we show that macrophages, either treated with statin in vitro or from statin-treated mice, have reduced cholesterol levels and higher expression of Jmjd3, a H3K27me3 demethylase. We provide evidence that lowering cholesterol levels in macrophages suppresses the ATP synthase in the inner mitochondrial membrane (IMM) and changes the proton gradient in the mitochondria. This activates NFkB and Jmjd3 expression to remove the repressive marker H3K27me3. Accordingly, the epigenome is altered by the cholesterol reduction. When subsequently challenged by the inflammatory stimulus LPS (M1), both macrophages treated with statins in vitro or isolated from statin-treated mice in vivo, express lower levels pro-inflammatory cytokines than controls, while augmenting anti-inflammatory Il10 expression. On the other hand, when macrophages are alternatively activated by IL4 (M2), statins promote the expression of Arg1, Ym1, and Mrc1. The enhanced expression is correlated with the statin-induced removal of H3K27me3 from these genes prior to activation. In addition, Jmjd3 and its demethylase activity are necessary for cholesterol to modulate both M1 and M2 activation. We conclude that upregulation of Jmjd3 is a key event for the anti-inflammatory function of statins on macrophages.

2.
bioRxiv ; 2024 Mar 03.
Article in English | MEDLINE | ID: mdl-36711703

ABSTRACT

Stains are known to be anti-inflammatory, but the mechanism remains poorly understood. Here we show that macrophages, either treated with statin in vitro or from statin-treated mice, have reduced cholesterol levels and higher expression of Jmjd3, a H3K27me3 demethylase. We provide evidence that lowering cholesterol levels in macrophages suppresses the ATP synthase in the inner mitochondrial membrane (IMM) and changes the proton gradient in the mitochondria. This activates NFkB and Jmjd3 expression to remove the repressive marker H3K27me3. Accordingly, the epigenome is altered by the cholesterol reduction. When subsequently challenged by the inflammatory stimulus LPS (M1), both macrophages treated with statins in vitro or isolated from statin-treated mice in vivo, express lower levels pro-inflammatory cytokines than controls, while augmenting anti-inflammatory Il10 expression. On the other hand, when macrophages are alternatively activated by IL4 (M2), statins promote the expression of Arg1, Ym1, and Mrc1. The enhanced expression is correlated with the statin-induced removal of H3K27me3 from these genes prior to activation. In addition, Jmjd3 and its demethylase activity are necessary for cholesterol to modulate both M1 and M2 activation. We conclude that upregulation of Jmjd3 is a key event for the anti-inflammatory function of statins on macrophages.

3.
Neurology ; 100(15): e1565-e1573, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36750387

ABSTRACT

BACKGROUND AND OBJECTIVES: There is significant heterogeneity in the reporting of outcome measures in aneurysmal subarachnoid hemorrhage (aSAH) research. The modified Rankin scale (mRS) is the most commonly reported functional outcome measure. The mRS focuses on physical disability; however, many aSAH survivors experience sequalae in other domains, and the mRS may therefore not capture outcomes important to aSAH survivors. The objective of this study was to assess the clinical relevance of the mRS as a research outcome measure to people with lived aSAH experience. METHODS: We conducted an international cross-sectional survey of 355 aSAH survivors, family members, and caregivers to evaluate patient-perceived outcomes in relation to the mRS. The mRS was assessed using a previously validated web-based tool. RESULTS: Response rate was 60%; respondents from 7 continents were composed of 86% aSAH survivors and 14% family members/caregivers. Agreement between self-assessed outcome and the mRS was poor (Kappa 0.26 [CI 0.14-0.39]). Of the 172 respondents who self-assessed as having had a good aSAH outcome, 122 (71%) had a score of 0-2 on the mRS. Approximately 19% of respondents with a good outcome, based on a measured mRS score of 0-2, self-assessed as having had a poor aSAH outcome. When the mRS score was dichotomized as 0-3 corresponding to a good outcome, agreement between the score and self-assessed outcome remained poor with a Kappa score of 0.40 (CI 0.20-0.60). Approximately 30% of respondents believed that the mRS should not be used as an outcome measure in future aSAH trials. DISCUSSION: The findings suggest that there is poor agreement between aSAH survivors' self-assessed outcome, their actual mRS score, and the dichotomization of the mRS score into good/poor outcomes. Patient-centered and patient-informed outcome measurement tools are needed to guide the aSAH research agenda.


Subject(s)
Subarachnoid Hemorrhage , Humans , Subarachnoid Hemorrhage/diagnosis , Subarachnoid Hemorrhage/therapy , Cross-Sectional Studies , Outcome Assessment, Health Care , Patients , Retrospective Studies
4.
Science ; 377(6606): 666-669, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35926054

ABSTRACT

Muscle stem cells (MuSCs) reside in a specialized niche that ensures their regenerative capacity. Although we know that innate immune cells infiltrate the niche in response to injury, it remains unclear how MuSCs adapt to this altered environment for initiating repair. Here, we demonstrate that inflammatory cytokine signaling from the regenerative niche impairs the ability of quiescent MuSCs to reenter the cell cycle. The histone H3 lysine 27 (H3K27) demethylase JMJD3, but not UTX, allowed MuSCs to overcome inhibitory inflammation signaling by removing trimethylated H3K27 (H3K27me3) marks at the Has2 locus to initiate production of hyaluronic acid, which in turn established an extracellular matrix competent for integrating signals that direct MuSCs to exit quiescence. Thus, JMJD3-driven hyaluronic acid synthesis plays a proregenerative role that allows MuSC adaptation to inflammation and the initiation of muscle repair.


Subject(s)
Hyaluronic Acid , Inflammation , Jumonji Domain-Containing Histone Demethylases , Muscle, Skeletal , Myoblasts, Skeletal , Regeneration , Stem Cell Niche , Animals , Cell Cycle , Histones , Humans , Hyaluronic Acid/biosynthesis , Inflammation/metabolism , Interferon-gamma/metabolism , Interleukin-6 , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Myoblasts, Skeletal/metabolism
5.
Arterioscler Thromb Vasc Biol ; 39(2): 137-149, 2019 02.
Article in English | MEDLINE | ID: mdl-30580573

ABSTRACT

Objective- During atherosclerosis, LDLs (low-density lipoproteins) accumulate in the arteries, where they become modified, aggregated, and retained. Such deposits of aggregated LDL (agLDL) can be recognized by macrophages, which attempt to digest and clear them. AgLDL catabolism promotes internalization of cholesterol and foam cell formation, which leads to the progression of atherosclerosis. Therapeutic blockade of this process may delay disease progression. When macrophages interact with agLDL in vitro, they form a novel extracellular, hydrolytic compartment-the lysosomal synapse (LS)-aided by local actin polymerization to digest agLDL. Here, we investigated the specific regulators involved in actin polymerization during the formation of the LS. Approach and Results- We demonstrate in vivo that atherosclerotic plaque macrophages contacting agLDL deposits polymerize actin and form a compartment strikingly similar to those made in vitro. Live cell imaging revealed that macrophage cortical F-actin depolymerization is required for actin polymerization to support the formation of the LS. This depolymerization is cofilin-1 dependent. Using siRNA-mediated silencing, pharmacological inhibition, genetic knockout, and stable overexpression, we elucidate key roles for Cdc42 Rho GTPase and GEF (guanine nucleotide exchange factor) Vav in promoting actin polymerization during the formation of the LS and exclude a role for Rac1. Conclusions- These results highlight critical roles for dynamic macrophage F-actin rearrangement and polymerization via cofilin-1, Vav, and Cdc42 in LS formation, catabolism of agLDL, and foam cell formation. These proteins might represent therapeutic targets to treat atherosclerotic disease.


Subject(s)
Actins/chemistry , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Protein Aggregates , Proto-Oncogene Proteins c-vav/physiology , cdc42 GTP-Binding Protein/physiology , Animals , Lipoproteins, LDL/chemistry , Mice , Polymerization , RAW 264.7 Cells
6.
Elife ; 72018 11 13.
Article in English | MEDLINE | ID: mdl-30422112

ABSTRACT

The plasma membrane in mammalian cells is rich in cholesterol, but how the cholesterol is partitioned between the two leaflets of the plasma membrane remains a matter of debate. Recently, Liu et al. used domain 4 (D4) of perfringolysin O as a cholesterol sensor to argue that cholesterol is mostly in the exofacial leaflet (Liu et al., 2017). This conclusion was made by interpreting D4 binding in live cells using in vitro calibrations with liposomes. However, liposomes may be unfaithful in mimicking the plasma membrane, as we demonstrate here. Also, D4 binding is highly sensitive to the presence of cytosolic proteins. In addition, we find that a D4 variant, which requires >35 mol% cholesterol to bind to liposomes in vitro, does in fact bind to the cytoplasmic leaflet of the plasma membrane in a cholesterol-dependent manner. Thus, we believe, based on the current evidence, that it is unlikely that there is a significantly higher proportion of cholesterol in the exofacial leaflet of the plasma membrane compared to the cytosolic leaflet.


Subject(s)
Cell Membrane/chemistry , Cholesterol/analysis , Animals , Lipid Bilayers/analysis , Lipids/analysis , Liposomes/analysis , Membranes
7.
Proc Natl Acad Sci U S A ; 114(30): 7999-8004, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696297

ABSTRACT

mTORC1 is known to activate sterol regulatory element-binding proteins (SREBPs) including SREBP-2, a master regulator of cholesterol synthesis. Through incompletely understood mechanisms, activated mTORC1 triggers translocation of SREBP-2, an endoplasmic reticulum (ER) resident protein, to the Golgi where SREBP-2 is cleaved to translocate to the nucleus and activate gene expression for cholesterol synthesis. Low ER cholesterol is a well-established trigger for SREBP-2 activation. We thus investigated whether mTORC1 activates SREBP-2 by reducing cholesterol delivery to the ER. We report here that mTORC1 activation is accompanied by low ER cholesterol and an increase of SREBP-2 activation. Conversely, a decrease in mTORC1 activity coincides with a rise in ER cholesterol and a decrease in SERBP-2 activity. This rise in ER cholesterol is of lysosomal origin: blocking the exit of cholesterol from lysosomes by U18666A or NPC1 siRNA prevents ER cholesterol from increasing and, consequently, SREBP-2 is activated without mTORC1 activation. Furthermore, when mTORC1 activity is low, cholesterol is delivered to lysosomes through two membrane trafficking pathways: autophagy and rerouting of endosomes to lysosomes. Indeed, with dual blockade of both pathways by Atg5-/- and dominant-negative rab5, ER cholesterol fails to increase when mTORC1 activity is low, and SREBP-2 is activated. Conversely, overexpressing constitutively active Atg7, which forces autophagy and raises ER cholesterol even when mTORC1 activity is high, suppresses SREBP-2 activation. We conclude that mTORC1 actively suppresses autophagy and maintains endosomal recycling, thereby preventing endosomes and autophagosomes from reaching lysosomes. This results in a reduction of cholesterol in the ER and activation of SREBP-2.


Subject(s)
Autophagosomes/physiology , Cholesterol/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , HEK293 Cells , Humans
8.
J Biol Chem ; 292(14): 5737-5747, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28196862

ABSTRACT

The plasma membrane of mammalian cells undergoes constitutive endocytosis, endocytic sorting, and recycling, which delivers nutrients to the lysosomes. The receptors, along with membrane lipids, are normally returned to the plasma membrane to sustain this action. It is not known, however, whether this process is influenced by metabolic conditions. Here we report that endocytic recycling requires active mechanistic target of rapamycin (aka mammalian target of rapamycin) (mTORC1), a master metabolic sensor. Upon mTORC1 inactivation, either by starvation or by inhibitor, recycling receptors and plasma membrane lipids, such as transferrin receptors and sphingomyelin, are delivered to the lysosomes. This lysosomal targeting is independent of canonical autophagy: both WT and Atg5-/- mouse embryonic fibroblasts responded similarly. Furthermore, we identify hepatocyte growth factor-regulated tyrosine kinase substrate (Hrs), an endosomal sorting complexes required for transport (ESCORT-0) component, as a downstream target of mTORC1. Hrs requires mTORC1 activity to maintain its protein expression level. Silencing Hrs without decreasing mTORC1 activity is sufficient to target transferrin and sphingomyelin to the lysosomes. It is thus evident that the canonical recycling pathway is under the regulation of mTORC1 and likely most predominant in proliferating cells where mTORC1 is highly active.


Subject(s)
Embryo, Mammalian/metabolism , Endocytosis/physiology , Fibroblasts/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , Sphingomyelins/metabolism , TOR Serine-Threonine Kinases/metabolism , Transferrin/metabolism , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Biological Transport, Active/physiology , Cell Proliferation/physiology , Cells, Cultured , Embryo, Mammalian/cytology , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Fibroblasts/cytology , Lysosomes/genetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Sphingomyelins/genetics , TOR Serine-Threonine Kinases/genetics , Transferrin/genetics
9.
Autophagy ; 11(7): 1025-36, 2015.
Article in English | MEDLINE | ID: mdl-26043942

ABSTRACT

A homozygous mutation in the DST (dystonin) gene causes a newly identified lethal form of hereditary sensory and autonomic neuropathy in humans (HSAN-VI). DST loss of function similarly leads to sensory neuron degeneration and severe ataxia in dystonia musculorum (Dst(dt)) mice. DST is involved in maintaining cytoskeletal integrity and intracellular transport. As autophagy is highly reliant upon stable microtubules and motor proteins, we assessed the influence of DST loss of function on autophagy using the Dst(dt-Tg4) mouse model. Electron microscopy (EM) revealed an accumulation of autophagosomes in sensory neurons from these mice. Furthermore, we demonstrated that the autophagic flux was impaired. Levels of LC3-II, a marker of autophagosomes, were elevated. Consequently, Dst(dt-Tg4) sensory neurons displayed impaired protein turnover of autophagosome substrate SQTSM1/p62 and of polyubiquitinated proteins. Interestingly, in a previously described Dst(dt-Tg4) mouse model that is partially rescued by neuronal specific expression of the DST-A2 isoform, autophagosomes, autolysosomes, and damaged organelles were reduced when compared to Dst(dt-Tg4) mutant mice. LC3-II, SQTSM1, polyubiquitinated proteins and autophagic flux were also restored to wild-type levels in the rescued mice. Finally, a significant decrease in DNAIC1 (dynein, axonemal, intermediate chain 1; the mouse ortholog of human DNAI1), a member of the DMC (dynein/dynactin motor complex), was noted in Dst(dt-Tg4) dorsal root ganglia and sensory neurons. Thus, DST-A2 loss of function perturbs late stages of autophagy, and dysfunctional autophagy at least partially underlies Dst(dt) pathogenesis. We therefore conclude that the DST-A2 isoform normally facilitates autophagy within sensory neurons to maintain cellular homeostasis.


Subject(s)
Autophagy , Dystonia/pathology , Sensory Receptor Cells/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/genetics , Cytoskeletal Proteins/genetics , Dynactin Complex , Dystonia/metabolism , Dystonin , Heat-Shock Proteins/metabolism , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/genetics , Phagosomes/metabolism , Phagosomes/ultrastructure , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/ultrastructure , Sequestosome-1 Protein
10.
PLoS One ; 9(11): e113789, 2014.
Article in English | MEDLINE | ID: mdl-25415591

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) plays an essential role in mediating cholesterol efflux to apolipoprotein A-I (apoA-I), a major housekeeping mechanism for cellular cholesterol homeostasis. After initial engagement with ABCA1, apoA-I directly interacts with the plasma membrane to acquire cholesterol. This apoA-I lipidation process is also known to require cellular signaling processes, presumably to support cholesterol trafficking to the plasma membrane. We report here that one of major signaling pathways in mammalian cells, Akt, is also involved. In several cell models that express ABCA1 including macrophages, pancreatic beta cells and hepatocytes, inhibition of Akt increases cholesterol efflux to apoA-I. Importantly, Akt inhibition has little effect on cells expressing non-functional mutant of ABCA1, implicating a specific role of Akt in ABCA1 function. Furthermore, we provide evidence that mTORC1, a major downstream target of Akt, is also a negative regulator of cholesterol efflux. In cells where mTORC1 is constitutively activated due to tuberous sclerosis complex 2 deletion, cholesterol efflux to apoA-I is no longer sensitive to Akt activity. This suggests that Akt suppresses cholesterol efflux through mTORC1 activation. Indeed, inhibition of mTORC1 by rapamycin or Torin-1 promotes cholesterol efflux. On the other hand, autophagy, one of the major pathways of cholesterol trafficking, is increased upon Akt inhibition. Furthermore, Akt inhibition disrupts lipid rafts, which is known to promote cholesterol efflux to apoA-I. We therefore conclude that Akt, through its downstream targets, mTORC1 and hence autophagy, negatively regulates cholesterol efflux to apoA-I.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Apolipoprotein A-I/metabolism , Cholesterol/metabolism , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , ATP Binding Cassette Transporter 1/genetics , Animals , Apolipoprotein A-I/genetics , Biological Transport, Active/genetics , Cell Line , Cholesterol/genetics , Cricetinae , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics
11.
Arterioscler Thromb Vasc Biol ; 33(2): 224-31, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23241412

ABSTRACT

OBJECTIVE: Apolipoprotein (apo) A-II is the second major apo of high-density lipoproteins, yet its pathophysiological roles in the development of atherosclerosis remain unknown. We aimed to examine whether apo A-II plays any role in atherogenesis and, if so, to elucidate the mechanism involved. METHODS AND RESULTS: We compared the susceptibility of human apo A-II transgenic (Tg) rabbits to cholesterol diet-induced atherosclerosis with non-Tg littermate rabbits. Tg rabbits developed significantly less aortic and coronary atherosclerosis than their non-Tg littermates, while total plasma cholesterol levels were similar. Atherosclerotic lesions of Tg rabbits were characterized by reduced macrophages and smooth muscle cells, and apo A-II immunoreactive proteins were frequently detected in the lesions. Tg rabbits exhibited low levels of plasma C-reactive protein and blood leukocytes compared with non-Tg rabbits, and high-density lipoproteins of Tg rabbit plasma exerted stronger cholesterol efflux activity and inhibitory effects on the inflammatory cytokine expression by macrophages in vitro than high-density lipoproteins isolated from non-Tg rabbits. In addition, ß-very-low-density lipoproteins of Tg rabbits were less sensitive to copper-induced oxidation than ß-very-low-density lipoproteins of non-Tg rabbits. CONCLUSIONS: These results suggest that enrichment of apo A-II in high-density lipoprotein particles has atheroprotective effects and apo A-II may become a target for the treatment of atherosclerosis.


Subject(s)
Aorta/metabolism , Aortic Diseases/prevention & control , Apolipoprotein A-II/metabolism , Atherosclerosis/prevention & control , Coronary Artery Disease/prevention & control , Coronary Vessels/metabolism , Animals , Animals, Genetically Modified , Aorta/immunology , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Apolipoprotein A-II/blood , Apolipoprotein A-II/genetics , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cholesterol, Dietary/blood , Coronary Artery Disease/blood , Coronary Artery Disease/etiology , Coronary Artery Disease/genetics , Coronary Artery Disease/immunology , Coronary Artery Disease/pathology , Coronary Vessels/immunology , Coronary Vessels/pathology , Cytokines/blood , Disease Models, Animal , Female , Humans , Inflammation Mediators/blood , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Lipoproteins, VLDL/blood , Male , Oxidation-Reduction , Plaque, Atherosclerotic , Rabbits , Time Factors
12.
J Biol Chem ; 287(48): 40502-12, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23055522

ABSTRACT

BACKGROUND: ABCA1 is known to suppress proinflammatory cytokines. RESULTS: ABCA1 activates PKA and up-regulates anti-inflammatory cytokine IL-10. Elevated PKA transforms macrophages to M2-like phenotype. Disrupting lipid rafts by statins MCD, and filipin recuperates ABCA1 phenotype and likely functions downstream of ABCA1. CONCLUSION: By modulating cholesterol, ABCA1 activates PKA. This generates M2-like macrophages. SIGNIFICANCE: ABCA1 does not simply suppress inflammatory response. It promotes M2-like activation and facilitates resolution. Nonresolving inflammatory response from macrophages is a major characteristic of atherosclerosis. Macrophage ABCA1 has been previously shown to suppress the secretion of proinflammatory cytokine. In the present study, we demonstrate that ABCA1 also promotes the secretion of IL-10, an anti-inflammatory cytokine critical for inflammation resolution. ABCA1(+/+) bone marrow-derived macrophages secrete more IL-10 but less proinflammatory cytokines than ABCA1(-/-) bone marrow-derived macrophages, similar to alternatively activated (M2) macrophages. We present evidence that ABCA1 activates PKA and that this elevated PKA activity contributes to M2-like inflammatory response from ABCA1(+/+) bone marrow-derived macrophages. Furthermore, cholesterol lowering by statins, methyl-ß-cyclodextrin, or filipin also activates PKA and, consequently, transforms macrophages toward M2-like phenotype. Conversely, cholesterol enrichment suppresses PKA activity and promotes M1-like inflammatory response. As the primary function of ABCA1 is cholesterol removal, our results suggest that ABCA1 activates PKA by regulating cholesterol. Indeed, forced cholesterol enrichment in ABCA1-expressing macrophages suppresses PKA activation and elicits M1-like response. Collectively, these findings reveal a novel protective process by ABCA1-activated PKA in macrophages. They also suggest cholesterol lowering in extra-hepatic tissues by statins as an anti-inflammation strategy.


Subject(s)
ATP-Binding Cassette Transporters/immunology , Cyclic AMP-Dependent Protein Kinases/immunology , Interleukin-10/immunology , Macrophages/immunology , Toll-Like Receptor 4/immunology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/immunology , Cell Line , Cholesterol/immunology , Cricetinae , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation , Humans , Macrophages/enzymology , Mice , Mice, Knockout , Protein Structure, Tertiary , Toll-Like Receptor 4/genetics
13.
J Mol Cell Biol ; 4(6): 386-97, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22847234

ABSTRACT

Skeletal muscle differentiation is mediated by a complex gene expression program requiring both the muscle-specific transcription factor Myogenin (Myog) and p38α MAPK (p38α) signaling. However, the relative contribution of Myog and p38α to the formation of mature myotubes remains unknown. Here, we have uncoupled the activity of Myog from that of p38α to gain insight into the individual roles of these proteins in myogenesis. Comparative expression profiling confirmed that Myog activates the expression of genes involved in muscle function. Furthermore, we found that in the absence of p38α signaling, Myog expression leads to the down-regulation of genes involved in cell cycle progression. Consistent with this, the expression of Myog is sufficient to induce cell cycle exit. Interestingly, p38α-defective, Myog-expressing myoblasts fail to form multinucleated myotubes, suggesting an important role for p38α in cell fusion. Through the analysis of p38α up-regulated genes, the tetraspanin CD53 was identified as a candidate fusion protein, a role confirmed both ex vivo in primary myoblasts, and in vivo during myofiber regeneration in mice. Thus, our study has revealed an unexpected role for Myog in mediating cell cycle exit and has identified an essential role for p38α in cell fusion through the up-regulation of CD53.


Subject(s)
Gene Expression/genetics , Mitogen-Activated Protein Kinase 14/genetics , Muscle Development/genetics , Muscle Development/physiology , Myoblasts, Skeletal/physiology , Myogenin/genetics , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Fusion , Cell Line , Cell Proliferation , Down-Regulation/genetics , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Myoblasts, Skeletal/metabolism , Myogenin/metabolism , Regeneration/genetics , Regeneration/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Tetraspanin 25/genetics , Tetraspanin 25/metabolism , Up-Regulation/genetics
14.
Acta Biochim Biophys Sin (Shanghai) ; 44(2): 119-28, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22232299

ABSTRACT

The Niemann-Pick type C1 (NPC1) is located mainly in the membranes of the late endosome/lysosome and controls the intracellular cholesterol trafficking from the late endosome/lysosome to the plasma membrane. It has been reported that oxidized low-density lipoprotein (oxLDL) can up-regulate NPC1 expression. However, the detailed mechanisms are not fully understood. In this study, we investigated the effect of oxLDL stimulation on NPC1 expression in THP-1 macrophages. Our results showed that oxLDL up-regulated NPC1 expression at both mRNA and protein levels in a dose-dependent and time-dependent manner. In addition, oxLDL also induced the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). Treatment with oxLDL significantly increased cyclooxygenase-2 (COX-2) mRNA and protein expression in the macrophages, and these increases were suppressed by the ERK1/2 inhibitor PD98059 or ERK1/2 small interfering RNA (siRNA) treatment. OxLDL up-regulated the expression of peroxisome proliferator-activated receptor α (PPARα) at the mRNA and protein levels, which could be abolished by COX-2 siRNA or COX-2 inhibitor NS398 treatment in these macrophages. OxLDL dramatically elevated cellular cholesterol efflux, which was abrogated by inhibiting ERK1/2 and/or COX-2. In addition, oxLDL-induced NPC1 expression and cellular cholesterol efflux were reversed by PPARα siRNA or GW6471, an antagonist of PPARα. Taken together, these results provide the evidence that oxLDL can up-regulate the expression of the NPC1 through ERK1/2/COX-2/PPARα-signaling pathway in macrophages.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Lipoproteins, LDL/pharmacology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Base Sequence , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Intracellular Signaling Peptides and Proteins , Lipoproteins, LDL/metabolism , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/metabolism , Models, Biological , Niemann-Pick C1 Protein , Oxazoles/pharmacology , PPAR alpha/antagonists & inhibitors , PPAR alpha/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Tyrosine/analogs & derivatives , Tyrosine/pharmacology , Up-Regulation/drug effects
15.
Lipids ; 46(9): 795-804, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21710238

ABSTRACT

ATP binding cassette A1 (ABCA1) transports cholesterol, phospholipids and lipophilic molecules to and across cellular membranes. We examined if ABCA1 expression altered cellular de novo glycerolipid biosynthesis in growing Baby hamster kidney (BHK) cells. Mock BHK cells or cells expressing a mifepristone-inducible ABCA1 (ABCA1) were incubated plus or minus mifepristone and then with [(3)H]serine or [(3)H]inositol or [(3)H]ethanolamine or [methyl-(3)H]choline or [(3)H]glycerol or [(14)C]oleate and radioactivity incorporated into glycerolipids determined. Mifepristone did not affect [1,3-(3)H]glycerol or [(14)C]oleate or [(3)H]ethanolamine or [methyl-(3)H]choline uptake in BHK cells. In contrast, [(3)H]glycerol and [(14)C]oleate incorporated into phosphatidylserine (PtdSer) were elevated 2.4-fold (p < 0.05) and 54% (p < 0.05), respectively, upon ABCA1 induction confirming increased PtdSer biosynthesis from these precursors. However, mifepristone inhibited [(3)H]serine uptake and incorporation into PtdSer indicating that PtdSer synthesis from serine in BHK cells is dependent on serine uptake. Mifepristone stimulated [(3)H]inositol uptake in mock and ABCA1 cells but not its incorporation into phosphatidylinositol indicating that its synthesis from inositol is independent of inositol uptake in BHK cells. [(3)H]glycerol and [(14)C]oleate incorporated into triacylglycerol were reduced and into diacylglycerol elevated only in mifepristone-induced ABCA1 expressing cells due to a decrease in diacylglycerol acyltransferase-1 (DGAT-1) activity. The presence of trichostatin A, a class I and II histone deacetylase inhibitor, reversed the ABCA1-mediated reduction in DGAT-1 activity but did not affect DGAT-1 mRNA expression. Thus, mifepristone has diverse effects on de novo glycerolipid synthesis. We suggest that caution should be exercised when using mifepristone-inducible systems for studies of glycerolipid metabolism in cells expressing glucocorticoid responsive receptors.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Gene Expression/drug effects , Glycerides/biosynthesis , Mifepristone/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Cell Line , Cricetinae , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Enzyme Assays , Glycerol/metabolism , Glycerophospholipids/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Inositol/metabolism , Oleic Acid/metabolism , Phosphatidylserines/biosynthesis , Serine/metabolism
16.
Am J Physiol Cell Physiol ; 301(4): C886-94, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21697542

ABSTRACT

ATP-binding cassette protein A1 (ABCA1) is a key plasma membrane protein required for the efflux of cellular cholesterol to extracellular acceptors, particularly to apolipoprotein A-I (apoA-I). This process is essential to maintain cholesterol homeostasis in the body. The detailed molecular mechanisms, however, are still insufficiently understood. Also, the molecular identity of ABCA1, i.e., channel, pump, or flippase, remains unknown. In this study we analyzed extracellular ATP levels in the medium of ABCA1-expressing BHK cells and RAW macrophages and compared them to the medium of nonexpressing cells. We found that extracellular ATP concentrations are significantly elevated when cells express ABCA1. Importantly, a dysfunctional ABCA1 mutant (A937V), when expressed similarly as wild-type ABCA1, is unable to raise extracellular ATP concentration, which suggests a casual relationship between functional ABCA1 and elevated extracellular ATP. To explore the physiological role of extracellular ATP, we analyzed ABCA1-mediated cholesterol efflux under conditions where extracellular ATP levels were modulated. We found that increasing extracellular ATP within the physiological range, i.e., <µM, promotes cholesterol efflux to apoA-I. On the other hand, removing extracellular ATP, either by adding apyrase to the medium or by expressing a plasma membrane-bound ectonucleotidase, CD39, abolishes cholesterol efflux to apoA-I. On the basis of these results, we conclude that, through direct or indirect mechanisms, ABCA1 functions to raise ATP levels in the medium. This elevated extracellular ATP is required for ABCA1-mediated cholesterol efflux to apoA-I.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Apolipoprotein A-I/metabolism , Cholesterol/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apyrase/genetics , Apyrase/metabolism , Cell Line , Cricetinae , Gene Expression Regulation , Macrophages/drug effects , Macrophages/metabolism , Mice , Mifepristone/pharmacology , Mutation
17.
J Biol Chem ; 286(5): 3370-8, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21106522

ABSTRACT

Macrophage foam cell is the predominant cell type in atherosclerotic lesions. Removal of excess cholesterol from macrophages thus offers effective protection against atherosclerosis. Here we report that a protein kinase A (PKA)-anchoring inhibitor, st-Ht31, induces robust cholesterol/phospholipid efflux, and ATP-binding cassette transporter A1 (ABCA1) greatly facilitates this process. Remarkably, we found that st-Ht31 completely reverses foam cell formation, and this process is ABCA1-dependent. The reversal is also accompanied by the restoration of well modulated inflammatory response to LPS. There is no detectable toxicity associated with st-Ht31, even when cells export up to 20% cellular cholesterol per hour. Using FRET-based PKA biosensors in live cells, we provide evidence that st-Ht31 drives cholesterol efflux by elevating PKA activity specifically in the cytoplasm. Furthermore, ABCA1 facilitates st-Ht31 uptake. This allows st-Ht31 to effectively remove cholesterol from ABCA1-expressing cells. We speculate that de-anchoring of PKA offers a novel therapeutic strategy to remove excess cholesterol from lipid-laden lesion macrophages.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cholesterol/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Foam Cells/drug effects , Proteins/pharmacology , ATP Binding Cassette Transporter 1 , Animals , Biological Transport/drug effects , Cell Line , Foam Cells/cytology , Macrophages/cytology , Mice
18.
J Lipid Res ; 51(5): 1144-56, 2010 May.
Article in English | MEDLINE | ID: mdl-19965585

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) is required for the lipidation of apolipoprotein A-I (apoA-I), although molecular mechanisms supporting this process remain poorly defined. In this study, we focused on the role of cytosolic Ca(2+) and its signaling and found that cytosolic Ca(2+) was required for cholesterol efflux to apoA-I. Removing extracellular Ca(2+) or chelating cytosolic Ca(2+) were equally inhibitory for apoA-I lipidation. We provide evidence that apoA-I induced Ca(2+) influx from the medium. We further demonstrate that calcineurin activity, the downstream target of Ca(2+) influx, was essential; inhibition of calcineurin activity by cyclosporine A or FK506 completely abolished apoA-I lipidation. Furthermore, calcineurin inhibition abolished apoA-I binding and diminished JAK2 phosphorylation, an established signaling event for cholesterol efflux to apoA-I. Finally, we demonstrate that neither Ca(2+) manipulation nor calcineurin inhibition influenced ABCA1's capacity to release microparticles or to remodel the plasma membrane. We conclude that this Ca(2+)-dependent calcineurin/JAK2 pathway is specifically responsible for apoA-I lipidation without directly modifying ABCA1 activity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/metabolism , Calcineurin/metabolism , Calcium/metabolism , Cholesterol/metabolism , Gene Expression Regulation , Signal Transduction , ATP Binding Cassette Transporter 1 , Animals , Calcineurin Inhibitors , Calcium/pharmacology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cytosol/drug effects , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Space/drug effects , Extracellular Space/metabolism , Humans , Janus Kinase 2/metabolism , Mice , Phosphorylation/drug effects , Signal Transduction/drug effects
19.
J Lipid Res ; 50(3): 456-466, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18941142

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) mediates cholesterol efflux to lipid-poor apolipoprotein A-I (apoA-I) and generates HDL. Here, we demonstrate that ABCA1 also directly mediates the production of apoA-I free microparticles. In baby hamster kidney (BHK) cells and RAW macrophages, ABCA1 expression led to lipid efflux in the absence of apoA-I and released large microparticles devoid of apoB and apoE. We provide evidence that these microparticles are an integral component of the classical cholesterol efflux pathway when apoA-I is present and accounted for approximately 30% of the total cholesterol released to the medium. Furthermore, microparticle release required similar ABCA1 activities as was required for HDL production. For instance, a nucleotide binding domain mutation in ABCA1 (A937V) that impaired HDL generation also abolished microparticle release. Similarly, inhibition of protein kinase A (PKA) prevented the release of both types of particles. Interestingly, physical modulation of membrane dynamics affected HDL and microparticle production, rigidifying the plasma membrane with wheat germ agglutinin inhibited HDL and microparticle release, whereas increasing the fluidity promoted the production of these particles. Given the established role of ABCA1 in expending nonraft or more fluid-like membrane domains, our results suggest that both HDL and microparticle release is favored by a more fluid plasma membrane. We speculate that ABCA1 enhances the dynamic movement of the plasma membrane, which is required for apoA-I lipidation and microparticle formation.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cell-Derived Microparticles/metabolism , Cholesterol/metabolism , Lipoproteins, HDL/metabolism , ATP Binding Cassette Transporter 1 , Animals , Apolipoprotein A-I/metabolism , Biological Transport, Active/drug effects , Cell Line , Cell-Derived Microparticles/drug effects , Cricetinae , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Membrane Fluidity/drug effects , Membrane Fluidity/physiology , Mice , Mifepristone/pharmacology , Recombinant Proteins/metabolism , Wheat Germ Agglutinins/pharmacology
20.
J Biol Chem ; 283(23): 16178-86, 2008 Jun 06.
Article in English | MEDLINE | ID: mdl-18385134

ABSTRACT

ATP-binding cassette transporter (ABC) A1 is required for the lipidation of apolipoprotein A-I to generate high density lipoprotein (HDL). This process is proposed to occur through a retro-endocytosis pathway in which apoA-I internalizes with ABCA1 and generates HDL from the endosomal compartments before resecretion. The aim of this study was to determine the route of apoA-I endocytosis and whether endocytosis contributes to HDL biogenesis. Using confocal microscopy, we found that internalized apoA-I only transiently colocalized with transferrin, a retro-endocytosis marker. Instead, apoA-I perfectly colocalized with a bulk phase uptake marker (fluorescein isothiocyanate-dextran) and, at later time points, with LysoTracker in several cell models including macrophages, fibroblasts, and baby hamster kidney cells. ABCA1 colocalized poorly with internalized apoA-I. To determine the contribution of internalized apoA-I to HDL biogenesis, we specifically removed apoA-I from the cell surface and analyzed the fate of internalized apoA-I. We found that 23% of cell-associated apoA-I was internalized at steady state. Of internalized apoA-I, only 20% was converted to HDL, and the rest was degraded, consistent with a lysosomal destination. We also found that apoA-I was released approximately five times faster from the plasma membrane than from the intracellular compartments. From these kinetic parameters, we estimated that approximately 5.6% of apoA-I that interacts with cells is degraded and that internalized apoA-I contributes to approximately 1.4% of total HDL production. We also found that blocking endocytosis with sucrose or cytochalasin D did not decrease cholesterol efflux or HDL biogenesis. We therefore conclude that the plasma membrane is the main platform where ABCA1-mediated lipidation of apoA-I occurs.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , Lipoproteins, HDL/metabolism , Protein Processing, Post-Translational/physiology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Apolipoprotein A-I/genetics , Cell Line , Cell Membrane/genetics , Cricetinae , Endocytosis/physiology , Endosomes/genetics , Humans , Kinetics , Lipoproteins, HDL/genetics , Mice , Protein Transport/physiology , Transferrin/genetics , Transferrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...