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











Publication year range
1.
Mol Cell Biol ; 38(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29555727

ABSTRACT

Retrograde transport (RT) allows cells to retrieve receptors and other cellular cargoes for delivery to the Golgi apparatus, contributing to the maintenance of cellular homeostasis. This transport route is also commonly used by several bacterial toxins to exert their deleterious actions on eukaryotic cells. While the retrograde transport process has been well characterized, the contribution of microRNAs (miRNAs) in regulating this cellular transport mechanism remains unknown. Here, we determined that mir-199a and mir-199b, members of the intronic miRNA family, coordinate genes regulating RT and endosome trafficking. We demonstrate that miR-199a-5p attenuates the expression of Vps26A, Rab9B, and M6PR, thereby controlling RT from endosomes to the trans-Golgi network (TGN). Importantly, we found that overexpression of a Vps26A construct resistant to the inhibitory action of miR-199a-5p abrogates the effect of miR-199a-5p on RT. Finally, we demonstrate that miR-199-5p overexpression attenuates Shiga toxin type 1 (Stx1)-mediated inhibition of protein biosynthesis. In summary, our work identifies the first noncoding RNA that influences RT and reduces the inhibition of protein biosynthesis caused by bacterial toxins.


Subject(s)
Biological Transport/drug effects , Cell Differentiation/drug effects , Endosomes/drug effects , MicroRNAs/pharmacology , Protein Biosynthesis/drug effects , Biological Transport/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Movement/drug effects , Cell Movement/genetics , Endosomes/metabolism , Humans , MicroRNAs/genetics , Protein Biosynthesis/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
2.
Atherosclerosis ; 243(2): 499-509, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26520906

ABSTRACT

RATIONALE: Recently, there has been significant interest in the therapeutic administration of miRNA mimics and inhibitors to treat cardiovascular disease. In particular, miR-27b has emerged as a regulatory hub in cholesterol and lipid metabolism and potential therapeutic target for treating atherosclerosis. Despite this, the impact of miR-27b on lipid levels in vivo remains to be determined. As such, here we set out to further characterize the role of miR-27b in regulating cholesterol metabolism in vitro and to determine the effect of miR-27b overexpression and inhibition on circulating and hepatic lipids in mice. METHODS AND RESULTS: Our results identify miR-27b as an important regulator of LDLR activity in human and mouse hepatic cells through direct targeting of LDLR and LDLRAP1. In addition, we report that modulation of miR-27b expression affects ABCA1 protein levels and cellular cholesterol efflux to ApoA1 in human hepatic Huh7 cells. Overexpression of pre-miR-27b in the livers of wild-type mice using AAV8 vectors increased pre-miR-27b levels 50-fold and reduced hepatic ABCA1 and LDLR expression by 50% and 20%, respectively, without changing circulating and hepatic cholesterol and triglycerides. To determine the effect of endogenous miR-27b on circulating lipids, wild-type mice were fed a Western diet for one month and injected with 5 mg/kg of LNA control or LNA anti-miR-27b oligonucleotides. Following two weeks of treatment, the expression of ABCA1 and LDLR were increased by 10-20% in the liver, demonstrating effective inhibition of miR-27b function. Intriguingly, no differences in circulating and hepatic lipids were observed between treatment groups. CONCLUSIONS: The results presented here provide evidence that short-term modulation of miR-27b expression in wild-type mice regulates hepatic LDLR and ABCA1 expression but does not influence plasma and hepatic lipid levels.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol/blood , Diet, High-Fat , Liver/metabolism , MicroRNAs/metabolism , Receptors, LDL/metabolism , 3' Untranslated Regions , ATP Binding Cassette Transporter 1/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biomarkers/blood , COS Cells , Chlorocebus aethiops , Computational Biology , Databases, Genetic , Gene Expression Regulation , Gene Regulatory Networks , Hep G2 Cells , Humans , Macaca mulatta , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Receptors, LDL/genetics , Time Factors , Transfection , Triglycerides/blood
3.
Nat Med ; 21(11): 1280-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26437365

ABSTRACT

The hepatic low-density lipoprotein receptor (LDLR) pathway is essential for clearing circulating LDL cholesterol (LDL-C). Whereas the transcriptional regulation of LDLR is well characterized, the post-transcriptional mechanisms that govern LDLR expression are just beginning to emerge. Here we develop a high-throughput genome-wide screening assay to systematically identify microRNAs (miRNAs) that regulate LDLR activity in human hepatic cells. From this screen we identified and characterized miR-148a as a negative regulator of LDLR expression and activity and defined a sterol regulatory element-binding protein 1 (SREBP1)-mediated pathway through which miR-148a regulates LDL-C uptake. In mice, inhibition of miR-148a increased hepatic LDLR expression and decreased plasma LDL-C. Moreover, we found that miR-148a regulates hepatic expression of ATP-binding cassette, subfamily A, member 1 (ABCA1) and circulating high-density lipoprotein cholesterol (HDL-C) levels in vivo. These studies uncover a role for miR-148a as a key regulator of hepatic LDL-C clearance through direct modulation of LDLR expression and demonstrate the therapeutic potential of inhibiting miR-148a to ameliorate an elevated LDL-C/HDL-C ratio, a prominent risk factor for cardiovascular disease.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Hepatocytes/metabolism , Liver/metabolism , MicroRNAs/genetics , Receptors, LDL/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Gene Expression Regulation , Hep G2 Cells , High-Throughput Screening Assays , Humans , Mice , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Receptors, LDL/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
4.
J Cell Sci ; 128(17): 3197-209, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26163491

ABSTRACT

Small non-coding RNAs (microRNAs) are important regulators of gene expression that modulate many physiological processes; however, their role in regulating intracellular transport remains largely unknown. Intriguingly, we found that the dynamin (DNM) genes, a GTPase family of proteins responsible for endocytosis in eukaryotic cells, encode the conserved miR-199a and miR-199b family of miRNAs within their intronic sequences. Here, we demonstrate that miR-199a and miR-199b regulate endocytic transport by controlling the expression of important mediators of endocytosis such as clathrin heavy chain (CLTC), Rab5A, low-density lipoprotein receptor (LDLR) and caveolin-1 (Cav-1). Importantly, miR-199a-5p and miR-199b-5p overexpression markedly inhibits CLTC, Rab5A, LDLR and Cav-1 expression, thus preventing receptor-mediated endocytosis in human cell lines (Huh7 and HeLa). Of note, miR-199a-5p inhibition increases target gene expression and receptor-mediated endocytosis. Taken together, our work identifies a new mechanism by which microRNAs regulate intracellular trafficking. In particular, we demonstrate that the DNM, miR-199a-5p and miR-199b-5p genes act as a bifunctional locus that regulates endocytosis, thus adding an unexpected layer of complexity in the regulation of intracellular trafficking.


Subject(s)
Dynamins/metabolism , Endocytosis/physiology , MicroRNAs/metabolism , Animals , COS Cells , Caveolin 1/genetics , Caveolin 1/metabolism , Chlorocebus aethiops , Clathrin Heavy Chains/genetics , Clathrin Heavy Chains/metabolism , Dynamins/genetics , Gene Expression Regulation/physiology , HeLa Cells , Humans , MicroRNAs/genetics , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
5.
Mol Nutr Food Res ; 59(3): 552-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25522185

ABSTRACT

SCOPE: The increased prevalence of cardiovascular diseases (CVDs) has been hypothesized to be the result of an increased exposure to a host of atherogenic environmental factors, paramount among them being unhealthy dietary habits. Long-chain n-3 polyunsaturated fatty acids have been shown to have cardio protective effects, partially due to their ability to regulate gene expression. In this regard, increasing attention has been devoted to the role of miRNAs as regulators of multiple metabolic pathways whose deregulation has been associated with CVD risk. METHODS AND RESULTS: In this work, we investigated whether miRNA expression was regulated by docosahexanoic acid, conjugated linoleic acid, and cholesterol in Caco-2 cells. The modulated miRNAs, miR-107 was differentially expressed by all treatments and this modulation was independent of its hosting gene, PANK1, possibly through its own promoter, which contains binding sites for metabolically relevant transcription factors. Among the putative target genes of miR-107, we found some genes with key roles in circadian rhythm. Specifically, we demonstrated that binding of miR-107 to the CLOCK gene results in the deregulation of the circadian rhythm of the cells. CONCLUSION: Since chronodisruption has been linked to metabolic disorders such as type 2 diabetes, atherosclerosis, obesity, and CVD, our findings suggests that miR-107 could represent a new approach for pharmacological treatment of these diseases.


Subject(s)
Cholesterol/pharmacology , Circadian Clocks/genetics , Linoleic Acids, Conjugated/pharmacology , MicroRNAs/genetics , Animals , Atorvastatin/pharmacology , CLOCK Proteins/genetics , Caco-2 Cells/drug effects , Circadian Clocks/drug effects , Diet, High-Fat , Docosahexaenoic Acids/pharmacology , Fenofibrate/pharmacology , Gene Expression Regulation/drug effects , Humans , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/genetics , Promoter Regions, Genetic
6.
FASEB J ; 29(2): 597-610, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25392271

ABSTRACT

Atherosclerosis is the major cause of death and disability in diabetic and obese subjects with insulin resistance. Akt2, a phosphoinositide-dependent serine-threonine protein kinase, is highly express in insulin-responsive tissues; however, its role during the progression of atherosclerosis remains unknown. Thus, we aimed to investigate the contribution of Akt2 during the progression of atherosclerosis. We found that germ-line Akt2-deficient mice develop similar atherosclerotic plaques as wild-type mice despite higher plasma lipids and glucose levels. It is noteworthy that transplantation of bone marrow cells isolated from Akt2(-/-) mice to Ldlr(-/-) mice results in marked reduction of the progression of atherosclerosis compared with Ldlr(-/-) mice transplanted with wild-type bone marrow cells. In vitro studies indicate that Akt2 is required for macrophage migration in response to proatherogenic cytokines (monocyte chemotactic protein-1 and macrophage colony-stimulating factor). Moreover, Akt2(-/-) macrophages accumulate less cholesterol and have an alternative activated or M2-type phenotype when stimulated with proinflammatory cytokines. Together, these results provide evidence that macrophage Akt2 regulates migration, the inflammatory response and cholesterol metabolism and suggest that targeting Akt2 in macrophages might be beneficial for treating atherosclerosis.


Subject(s)
Atherosclerosis/physiopathology , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Animals , Blood Glucose/metabolism , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Cell Movement , Cholesterol/metabolism , Cytokines/metabolism , Disease Progression , Inflammation , Insulin/chemistry , Leukocytes/cytology , Lipids/blood , Lipoproteins, LDL/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Plaque, Atherosclerotic , Receptors, LDL/genetics
7.
Curr Opin Lipidol ; 25(4): 282-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24978143

ABSTRACT

PURPOSE OF REVIEW: The objective of this review article is to summarize the recent findings about the importance of microRNAs (miRNAs) in regulating lipoprotein metabolism. We highlight the recent findings that uncover the importance of miRNAs in controlling plasma LDL-cholesterol (LDL-C) levels. RECENT FINDINGS: In 2013, several studies reported a number of miRNAs that regulate plasma LDL-C levels, including miR-30c. In this review article, we discuss those miRNAs that modulate LDL-C levels and lipoprotein secretion. We also discuss the numerous studies that demonstrate the critical role of miRNAs in governing the many facets of HDL metabolism, such as the ATP transporters, ABCA1, and ABCG1, and the scavenger receptor, SRB1. SUMMARY: The understanding of how these miRNAs modulate lipoprotein metabolism promises to reveal new therapeutic targets to treat dyslipidemias and related cardiovascular disorders.


Subject(s)
Gene Expression Regulation , Lipoproteins/metabolism , MicroRNAs/genetics , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , Humans , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism
8.
Thromb Haemost ; 112(4): 796-802, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25008143

ABSTRACT

The miR-143/145 cluster regulates VSMC specific gene expression, thus controlling differentiation, plasticity and contractile function, and promoting the VSMC phenotypic switch from a contractile/non-proliferative to a migrating/proliferative state. More recently increased miR-145 expression was observed in human carotid atherosclerotic plaques from symptomatic patients. The goal of this study was to investigate the contribution of miR-143/145 during atherogenesis by generating mice lacking miR-143/145 on an Ldlr-deficient background. Ldlr-/- and Ldlr-/--miR-143/145-/- (DKO) were fed a Western diet (WD) for 16 weeks. At the end of the treatment, the lipid profile and the atherosclerotic lesions were assessed in both groups of mice. Absence of miR-143/145 significantly reduced atherosclerotic plaque size and macrophage infiltration. Plasma total cholesterol levels were lower in DKO and FLPC analysis showed decreased cholesterol content in VLDL and LDL fractions. Interestingly miR-143/145 deficiency per se resulted in increased hepatic and vascular ABCA1 expression. We further confirmed the direct regulation of miR-145 on ABCA1 expression by qRT-PCR, Western blotting and 3'UTR-luciferase reporter assays. In summary, miR-143/145 deficiency significantly reduces atherosclerosis in mice. Therapeutic inhibition of miR-145 might be useful for treating atherosclerotic vascular disease.


Subject(s)
Atherosclerosis/genetics , MicroRNAs/genetics , Receptors, LDL/genetics , 3' Untranslated Regions , Animals , Atherosclerosis/metabolism , Base Sequence , Carotid Arteries , Cell Differentiation , Cell Movement , Disease Progression , Female , Lipids/chemistry , Lipoproteins, LDL/metabolism , Macrophages/cytology , Male , Mice , Mice, Knockout , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Phenotype , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism
9.
Stem Cells ; 32(7): 1917-28, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24504902

ABSTRACT

Hair follicle stem cells (HF-SCs) alternate between periods of quiescence and proliferation, to finally differentiate into all the cell types that constitute the hair follicle. Also, they have been recently identified as cells of origin in skin cancer. HF-SCs localize in a precise region of the hair follicle, the bulge, and molecular markers for this population have been established. Thus, HF-SCs are good model to study the potential role of oncogenic activations on SC physiology. Expression of a permanently active form of Akt (myrAkt) in basal cells leads to Akt hyperactivation specifically in the CD34(+)Itga6(H) population. This activation causes bulge stem cells to exit from quiescence increasing their response to proliferative stimuli and affecting some functions such as cell migration. HF-SC identity upon Akt activation is preserved; in this sense, increased proliferation does not result in stem cell exhaustion with age suggesting that Akt activation does not affect self-renewal an important aspect for normal tissue maintenance and cancer development. Genome-wide transcriptome analysis of HF-SC isolated from myrAkt and wild-type epidermis underscores changes in metabolic pathways characteristic of cancer cells. These differences manifest during a two-step carcinogenesis protocol in which Akt activation in HF-SCs results in increased tumor development and malignant transformation.


Subject(s)
Carcinogenesis/metabolism , Epidermis/enzymology , Skin Neoplasms/enzymology , Stem Cells/enzymology , Animals , Cell Proliferation , Cells, Cultured , Enzyme Activation , Epidermis/pathology , Keratinocytes/enzymology , Mice, Transgenic , Proto-Oncogene Proteins c-akt , Re-Epithelialization , Signal Transduction
10.
Free Radic Biol Med ; 64: 31-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23871755

ABSTRACT

The regulation of the metabolism of cholesterol has been one of the most studied biological processes since its first isolation from gallstones in 1784. High levels of plasma low-density lipoprotein (LDL) cholesterol and reduced levels of plasma high-density lipoprotein (HDL) cholesterol are widely recognized as major risk factors of cardiovascular disease. An imbalance in the production of reactive oxygen species can oxidize LDL particles, increasing the levels of the highly proatherogenic oxidized LDL. Furthermore, under pathological scenarios, numerous molecules can function as pro-oxidants, such as iron or (high levels of) glucose. In addition to the classical mechanisms regulating lipid homeostasis, recent studies have demonstrated the important role of microRNAs (miRNAs) as regulators of lipoprotein metabolism, oxidative derivatives of lipoprotein, and redox balance. Here, we summarize recent findings in the field, highlighting the contributions of some miRNAs to lipid- and oxidative-associated pathologies. We also discuss how therapeutic intervention of miRNAs may be a promising strategy to decrease LDL, increase HDL, and ameliorate lipid- and oxidative-related disorders, including atherosclerosis, nonalcoholic fatty liver disease, and metabolic syndrome.


Subject(s)
Atherosclerosis/metabolism , Fatty Liver/metabolism , Lipid Metabolism/genetics , Metabolic Syndrome/metabolism , MicroRNAs/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression Regulation , Humans , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease , Oxidation-Reduction , Oxidative Stress , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
11.
Curr Atheroscler Rep ; 15(5): 322, 2013 May.
Article in English | MEDLINE | ID: mdl-23512606

ABSTRACT

MicroRNAs (miRNAs) are small, ~22 nucleotide (nt) sequences of RNA that regulate gene expression at posttranscriptional level. These endogenous gene expression inhibitors were primarily described in cancer but recent exciting findings have also demonstrated a key role in cardiovascular diseases (CVDs), including atherosclerosis. MiRNAs control endothelial cell (EC), vascular smooth muscle cell (VSMC), and macrophage functions, and thereby regulate the progression of atherosclerosis. MiRNA expression is modulated by different stimuli involved in every stage of atherosclerosis, and conversely miRNAs modulates several pathways implicated in plaque development such as cholesterol metabolism. In the present review, we focus on the importance of miRNAs in atherosclerosis, and we further discuss their potential use as biomarkers and therapeutic targets in CVDs.


Subject(s)
Atherosclerosis/genetics , Gene Expression Regulation/genetics , MicroRNAs/genetics , Endothelial Cells/metabolism , Humans , Lipid Metabolism/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism
12.
Circ Res ; 112(12): 1592-601, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23519695

ABSTRACT

RATIONALE: Foam cell formation because of excessive accumulation of cholesterol by macrophages is a pathological hallmark of atherosclerosis, the major cause of morbidity and mortality in Western societies. Liver X nuclear receptors (LXRs) regulate the expression of the adenosine triphosphate-binding cassette (ABC) transporters, including adenosine triphosphate-binding cassette transporter A1 (ABCA1) and adenosine triphosphate-binding cassette transporter G1 (ABCG1). ABCA1 and ABCG1 facilitate the efflux of cholesterol from macrophages and regulate high-density lipoprotein (HDL) biogenesis. Increasing evidence supports the role of microRNA (miRNAs) in regulating cholesterol metabolism through ABC transporters. OBJECTIVE: We aimed to identify novel miRNAs that regulate cholesterol metabolism in macrophages stimulated with LXR agonists. METHODS AND RESULTS: To map the miRNA expression signature of macrophages stimulated with LXR agonists, we performed an miRNA profiling microarray analysis in primary mouse peritoneal macrophages stimulated with LXR ligands. We report that LXR ligands increase miR-144 expression in macrophages and mouse livers. Overexpression of miR-144 reduces ABCA1 expression and attenuates cholesterol efflux to apolipoproteinA1 in macrophages. Delivery of miR-144 oligonucleotides to mice attenuates ABCA1 expression in the liver, reducing HDL levels. Conversely, silencing of miR-144 in mice increases the expression of ABCA1 and plasma HDL levels. Thus, miR-144 seems to regulate both macrophage cholesterol efflux and HDL biogenesis in the liver. CONCLUSIONS: miR-144 regulates cholesterol metabolism via suppressing ABCA1 expression and modulation of miRNAs may represent a potential therapeutical intervention for treating dyslipidemia and atherosclerotic vascular disease.


Subject(s)
Cholesterol, HDL/blood , Hepatocytes/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Anticholesteremic Agents/pharmacology , Apolipoprotein A-I/metabolism , COS Cells , Chlorocebus aethiops , Diet, High-Fat , Gene Expression Profiling/methods , Hep G2 Cells , Hepatocytes/drug effects , Homeostasis , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Oligonucleotides/metabolism , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Sulfonamides/pharmacology
13.
Mol Biol Cell ; 24(4): 483-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23264465

ABSTRACT

The endothelium maintains a barrier between blood and tissue that becomes more permeable during inflammation. Membrane rafts are ordered assemblies of cholesterol, glycolipids, and proteins that modulate proinflammatory cell signaling and barrier function. In epithelial cells, the MAL family members MAL, MAL2, and myeloid-associated differentiation marker (MYADM) regulate the function and dynamics of ordered membrane domains. We analyzed the expression of these three proteins in human endothelial cells and found that only MYADM is expressed. MYADM was confined in ordered domains at the plasma membrane, where it partially colocalized with filamentous actin and cell-cell junctions. Small interfering RNA (siRNA)-mediated MYADM knockdown increased permeability, ICAM-1 expression, and leukocyte adhesion, all of which are features of an inflammatory response. Barrier function decrease in MYADM-silenced cells was dependent on ICAM-1 expression. Membrane domains and the underlying actin cytoskeleton can regulate each other and are connected by ezrin, radixin, and moesin (ERM) proteins. In endothelial cells, MYADM knockdown induced ERM activation. Triple-ERM knockdown partially inhibited ICAM-1 increase induced by MYADM siRNA. Importantly, ERM knockdown also reduced ICAM-1 expression in response to the proinflammatory cytokine tumor necrosis factor-α. MYADM therefore regulates the connection between the plasma membrane and the cortical cytoskeleton and so can control the endothelial inflammatory response.


Subject(s)
Cytoskeletal Proteins/genetics , Human Umbilical Vein Endothelial Cells/drug effects , Intercellular Adhesion Molecule-1/genetics , Membrane Proteins/genetics , Microfilament Proteins/genetics , Myelin and Lymphocyte-Associated Proteolipid Proteins/genetics , Animals , Biological Transport/drug effects , Cell Adhesion/drug effects , Cytoskeletal Proteins/metabolism , Dogs , Gene Expression Regulation/drug effects , HeLa Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Madin Darby Canine Kidney Cells , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins/antagonists & inhibitors , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
14.
PLoS One ; 7(8): e42494, 2012.
Article in English | MEDLINE | ID: mdl-22880004

ABSTRACT

Mutations in the TP53 gene are very common in human cancers, and are associated with poor clinical outcome. Transgenic mouse models lacking the Trp53 gene or that express mutant Trp53 transgenes produce tumours with malignant features in many organs. We previously showed the transcriptome of a p53-deficient mouse skin carcinoma model to be similar to those of human cancers with TP53 mutations and associated with poor clinical outcomes. This report shows that much of the 682-gene signature of this murine skin carcinoma transcriptome is also present in breast and lung cancer mouse models in which p53 is inhibited. Further, we report validated gene-expression-based tests for predicting the clinical outcome of human breast and lung adenocarcinoma. It was found that human patients with cancer could be stratified based on the similarity of their transcriptome with the mouse skin carcinoma 682-gene signature. The results also provide new targets for the treatment of p53-defective tumours.


Subject(s)
Breast Neoplasms/genetics , Genome, Human/genetics , Genomics , Lung Neoplasms/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/classification , Adenocarcinoma/genetics , Animals , Breast Neoplasms/classification , Disease Models, Animal , Female , Gene Expression Profiling , Genes, Neoplasm/genetics , Genetic Engineering , Humans , Lung Neoplasms/classification , Mice , Mice, Transgenic , Multivariate Analysis , Mutation/genetics , Proportional Hazards Models , Reproducibility of Results , Skin/metabolism , Skin/pathology , Survival Analysis , Treatment Outcome , Tumor Suppressor Protein p53/antagonists & inhibitors
15.
Mol Biol Cell ; 22(8): 1252-62, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21325632

ABSTRACT

Membrane organization into condensed domains or rafts provides molecular platforms for selective recruitment of proteins. Cell migration is a general process that requires spatiotemporal targeting of Rac1 to membrane rafts. The protein machinery responsible for making rafts competent to recruit Rac1 remains elusive. Some members of the MAL family of proteins are involved in specialized processes dependent on this type of membrane. Because condensed membrane domains are a general feature of the plasma membrane of all mammalian cells, we hypothesized that MAL family members with ubiquitous expression and plasma membrane distribution could be involved in the organization of membranes for cell migration. We show that myeloid-associated differentiation marker (MYADM), a protein with unique features within the MAL family, colocalizes with Rac1 in membrane protrusions at the cell surface and distributes in condensed membranes. MYADM knockdown (KD) cells had altered membrane condensation and showed deficient incorporation of Rac1 to membrane raft fractions and, similar to Rac1 KD cells, exhibited reduced cell spreading and migration. Results of rescue-of-function experiments by expression of MYADM or active Rac1L61 in cells knocked down for Rac1 or MYADM, respectively, are consistent with the idea that MYADM and Rac1 act on parallel pathways that lead to similar functional outcomes.


Subject(s)
Antigens, Differentiation/metabolism , Cell Movement , Membrane Microdomains/metabolism , Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Antigens, Differentiation/genetics , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Dogs , Electroporation , Female , Gene Expression , Gene Silencing , Haplorhini , Humans , Male , Myelin and Lymphocyte-Associated Proteolipid Proteins , Phylogeny , Proteins/genetics , RNA, Small Interfering/metabolism , Transfection , rac1 GTP-Binding Protein/genetics
16.
Dev Cell ; 18(5): 814-27, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20493814

ABSTRACT

Transcytosis is a widespread pathway for apical targeting in epithelial cells. MAL2, an essential protein of the machinery for apical transcytosis, functions by shuttling in vesicular carriers between the apical zone and the cell periphery. We have identified INF2, an atypical formin with actin polymerization and depolymerization activities, which is a binding partner of MAL2. MAL2-positive vesicular carriers associate with short actin filaments during transcytosis in a process requiring INF2. INF2 binds Cdc42 in a GTP-loaded-dependent manner. Cdc42 and INF2 regulate MAL2 dynamics and are necessary for apical transcytosis and the formation of lateral lumens in hepatoma HepG2 cells. INF2 and MAL2 are also essential for the formation of the central lumen in organotypic cultures of epithelial MDCK cells. Our results reveal a functional mechanism whereby Cdc42, INF2, and MAL2 are sequentially ordered in a pathway dedicated to the regulation of transcytosis and lumen formation.


Subject(s)
Epithelial Cells/physiology , Hepatocytes/cytology , Hepatocytes/physiology , Microfilament Proteins/metabolism , Proteolipids/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/physiology , cdc42 GTP-Binding Protein/metabolism , Actins/genetics , Actins/metabolism , Bile/cytology , Bile/physiology , Cell Polarity , Epithelial Cells/cytology , Formins , Genes, Reporter , Golgi Matrix Proteins , Hep G2 Cells/cytology , Hep G2 Cells/physiology , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Myelin and Lymphocyte-Associated Proteolipid Proteins , RNA, Small Interfering/genetics
17.
Am J Physiol Cell Physiol ; 295(3): C661-78, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18596208

ABSTRACT

In the renal collecting duct, vasopressin controls transport of water and solutes via regulation of membrane transporters such as aquaporin-2 (AQP2) and the epithelial urea transporter UT-A. To discover proteins potentially involved in vasopressin action in rat kidney collecting ducts, we enriched membrane "raft" proteins by harvesting detergent-resistant membranes (DRMs) of the inner medullary collecting duct (IMCD) cells. Proteins were identified and quantified with LC-MS/MS. A total of 814 proteins were identified in the DRM fractions. Of these, 186, including several characteristic raft proteins, were enriched in the DRMs. Immunoblotting confirmed DRM enrichment of representative proteins. Immunofluorescence confocal microscopy of rat IMCDs with antibodies to DRM proteins demonstrated heterogeneity of raft subdomains: MAL2 (apical region), RalA (predominant basolateral labeling), caveolin-2 (punctate labeling distributed throughout the cells), and flotillin-1 (discrete labeling of large intracellular structures). The DRM proteome included GPI-anchored, doubly acylated, singly acylated, cholesterol-binding, and integral membrane proteins (IMPs). The IMPs were, on average, much smaller and more hydrophobic than IMPs identified in non-DRM-enriched IMCD. The content of serine 256-phosphorylated AQP2 was greater in DRM than in non-DRM fractions. Vasopressin did not change the DRM-to-non-DRM ratio of most proteins, whether quantified by tandem mass spectrometry (LC-MS/MS, n=22) or immunoblotting (n=6). However, Rab7 and annexin-2 showed small increases in the DRM fraction in response to vasopressin. In accord with the long-term goal of creating a systems-level analysis of transport regulation, this study has identified a large number of membrane-associated proteins expressed in the IMCD that have potential roles in vasopressin action.


Subject(s)
Chromatography, Liquid , Deamino Arginine Vasopressin/pharmacology , Detergents/chemistry , Kidney Tubules, Collecting/drug effects , Membrane Microdomains/drug effects , Membrane Proteins/metabolism , Tandem Mass Spectrometry , Animals , Aquaporin 2/metabolism , Deamino Arginine Vasopressin/analogs & derivatives , Hydrophobic and Hydrophilic Interactions , Immunoblotting , Kidney Tubules, Collecting/metabolism , Male , Membrane Microdomains/metabolism , Microscopy, Confocal , Molecular Weight , Phosphorylation , Rats , Rats, Sprague-Dawley , Serine
18.
Mol Pharmacol ; 70(5): 1654-63, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16928956

ABSTRACT

Plitidepsin (aplidin) is a marine cyclic depsipeptide in phase II clinical development against several neoplasias. Plitidepsin is a potent inducer of apoptosis through the sustained activation of Jun N-terminal kinase (JNK). We have reported that this activation depends on the early induction of oxidative stress, activation of Rac1 small GTPase, and the later down-regulation of MKP-1 phosphatase. Using Scatchard and saturation binding analyses, we have found that (14)C-labeled plitidepsin binds to a moderately high-affinity receptor (K(d) of 44.8 +/- 3.1 and 35.5 +/- 4.8 nM, respectively) in MDA-MB-231 breast cancer cells. Two minutes after addition to cells, half of the drug was membrane-bound and was subsequently found in the cytosolic fraction. At 4 degrees C, plitidepsin cellular binding was around 10-fold lower than at 37 degrees C but sufficed to induce cell death, suggesting that this process is triggered from the membrane. Depletion of plasma membrane cholesterol by short treatment with methyl-beta-cyclodextrin diminished plitidepsin binding and Rac1 and JNK activation. Rac1 is targeted to the plasma membrane by plitidepsin as shown by subcellular fractioning and immunofluorescence analysis followed by confocal microscopy. Methyl-beta-cyclodextrin blocked this effect. A subline of HeLa cells (HeLa-R), partially resistant to plitidepsin, showed similar affinity (K(d) of 79.5 +/- 2.5 versus 37.7 +/- 8.2 nM) but 7.5-fold lower binding capacity than wild-type HeLa cells. Moreover, HeLa-R cells had lower total (71%) and membrane (67%) cholesterol content and membrane-bound Rac1, and showed no Rac1 activation upon plitidepsin treatment. In conclusion, cellular plitidepsin uptake and induction of apoptosis via activation of the Rac1-JNK pathway is membrane-cholesterol dependent.


Subject(s)
Cell Membrane/drug effects , Cholesterol/metabolism , Depsipeptides/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Cholesterol/deficiency , Depsipeptides/pharmacokinetics , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , HeLa Cells , Humans , Peptides, Cyclic , Protein Transport/drug effects , Tumor Cells, Cultured
19.
Endocrinology ; 145(2): 1011-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14576188

ABSTRACT

Polarized transport of newly synthesized proteins to the apical surface of epithelial cells takes place by a direct pathway from the Golgi or by an indirect route involving the delivery of the protein to the basolateral surface, followed by its endocytosis and transport across the cell. The indirect pathway, named transcytosis, is also used to translocate external material across the cell. MAL, a raft-associated integral membrane protein required for the direct apical route, is known to be expressed in the thyroid epithelium. MAL2, a member of the MAL protein family, has been recently identified as an essential component of the machinery for the transcytotic route in human hepatoma cells. Herein, we have investigated the expression and distribution of MAL2 in the human thyroid. MAL2 mRNA species were detected in the thyroid. Immunohistochemical analysis of thyroid follicles indicated that, in contrast to MAL, which predominantly distributed to the Golgi region, MAL2 distributed to the apical membrane. Biochemical analysis in primary thyrocyte cultures indicated that MAL2 exclusively resides in raft membranes. Confocal immunofluorescence analysis of thyrocyte cultures revealed that MAL2 predominantly localized in a subapical endosome compartment that was positive for Rab11a. Alterations in MAL2 expression, distribution, and appearance were found in specific types of follicular cell-derived carcinomas. Although the role of MAL2 has not been directly addressed in this study, the simultaneous expression of MAL and MAL2 suggests that traffic to the apical membrane in thyrocytes may rely on MAL for the direct route and on MAL2 for the transcytotic pathway.


Subject(s)
Carrier Proteins/analysis , Carrier Proteins/genetics , Gene Expression , Proteolipids/analysis , Proteolipids/genetics , Thyroid Gland/chemistry , Vesicular Transport Proteins , Biological Transport , Carrier Proteins/physiology , Cell Membrane/chemistry , Endocytosis , Endosomes/chemistry , Epithelial Cells/chemistry , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Microscopy, Confocal , Myelin and Lymphocyte-Associated Proteolipid Proteins , Proteolipids/physiology , RNA, Messenger/analysis , Thyroid Neoplasms/chemistry , rab GTP-Binding Proteins/analysis
20.
J Cell Biol ; 163(1): 155-64, 2003 Oct 13.
Article in English | MEDLINE | ID: mdl-14530381

ABSTRACT

MAL is an integral protein component of the machinery for apical transport in epithelial Madin-Darby canine kidney (MDCK) cells. To maintain its distribution, MAL cycles continuously between the plasma membrane and the Golgi complex. The clathrin-mediated route for apical internalization is known to differ from that at the basolateral surface. Herein, we report that MAL depends on the clathrin pathway for apical internalization. Apically internalized polymeric Ig receptor (pIgR), which uses clathrin for endocytosis, colocalized with internalized MAL in the same apical vesicles. Time-lapse confocal microscopic analysis revealed cotransport of pIgR and MAL in the same endocytic structures. Immunoelectron microscopic analysis evidenced colabeling of MAL with apically labeled pIgR in pits and clathrin-coated vesicles. Apical internalization of pIgR was abrogated in cells with reduced levels of MAL, whereas this did not occur either with its basolateral entry or the apical internalization of glycosylphosphatidylinositol-anchored proteins, which does not involve clathrin. Therefore, MAL is critical for efficient clathrin-mediated endocytosis at the apical surface in MDCK cells.


Subject(s)
Clathrin/metabolism , Endocytosis/physiology , Membrane Proteins/metabolism , Animals , Dogs , Glycosylphosphatidylinositols/metabolism , Kidney/metabolism , Mice , Oligonucleotides, Antisense/metabolism , Receptors, Polymeric Immunoglobulin/metabolism , Receptors, Transferrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL