Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Dev Cell ; 57(3): 387-397.e4, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35134345

ABSTRACT

Lipid droplets (LDs) are organelles of cellular lipid storage with fundamental roles in energy metabolism and cell membrane homeostasis. There has been an explosion of research into the biology of LDs, in part due to their relevance in diseases of lipid storage, such as atherosclerosis, obesity, type 2 diabetes, and hepatic steatosis. Consequently, there is an increasing need for a resource that combines datasets from systematic analyses of LD biology. Here, we integrate high-confidence, systematically generated human, mouse, and fly data from studies on LDs in the framework of an online platform named the "Lipid Droplet Knowledge Portal" (https://lipiddroplet.org/). This scalable and interactive portal includes comprehensive datasets, across a variety of cell types, for LD biology, including transcriptional profiles of induced lipid storage, organellar proteomics, genome-wide screen phenotypes, and ties to human genetics. This resource is a powerful platform that can be utilized to identify determinants of lipid storage.


Subject(s)
Databases as Topic , Lipid Droplets/metabolism , Animals , Cholesterol Esters/metabolism , Data Mining , Genome , Humans , Inflammation/pathology , Lipid Metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Phenotype , Phosphorylation , RNA Interference
2.
Cell Rep ; 34(13): 108928, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33789117

ABSTRACT

Flux through the RAF-MEK-ERK protein kinase cascade is shaped by phosphatases acting on the core components of the pathway. Despite being an established drug target and a hub for crosstalk regulation, little is known about dephosphorylation of MEK, the central kinase within the cascade. Here, we identify PPP6C, a phosphatase frequently mutated or downregulated in melanoma, as a major MEK phosphatase in cells exhibiting oncogenic ERK pathway activation. Recruitment of MEK to PPP6C occurs through an interaction with its associated regulatory subunits. Loss of PPP6C causes hyperphosphorylation of MEK at activating and crosstalk phosphorylation sites, promoting signaling through the ERK pathway and decreasing sensitivity to MEK inhibitors. Recurrent melanoma-associated PPP6C mutations cause MEK hyperphosphorylation, suggesting that they promote disease at least in part by activating the core oncogenic pathway driving melanoma. Collectively, our studies identify a key negative regulator of ERK signaling that may influence susceptibility to targeted cancer therapies.


Subject(s)
Carcinogenesis/pathology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphoprotein Phosphatases/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Phosphorylation , RNA, Small Interfering/metabolism , Substrate Specificity
3.
Proc Natl Acad Sci U S A ; 117(50): 31914-31922, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33257571

ABSTRACT

Inhibiting membrane association of RAS has long been considered a rational approach to anticancer therapy, which led to the development of farnesyltransferase inhibitors (FTIs). However, FTIs proved ineffective against KRAS-driven tumors. To reveal alternative therapeutic strategies, we carried out a genome-wide CRISPR-Cas9 screen designed to identify genes required for KRAS4B membrane association. We identified five enzymes in the prenylation pathway and SAFB, a nuclear protein with both DNA and RNA binding domains. Silencing SAFB led to marked mislocalization of all RAS isoforms as well as RAP1A but not RAB7A, a pattern that phenocopied silencing FNTA, the prenyltransferase α subunit shared by farnesyltransferase and geranylgeranyltransferase type I. We found that SAFB promoted RAS membrane association by controlling FNTA expression. SAFB knockdown decreased GTP loading of RAS, abrogated alternative prenylation, and sensitized RAS-mutant cells to growth inhibition by FTI. Our work establishes the prenylation pathway as paramount in KRAS membrane association, reveals a regulator of prenyltransferase expression, and suggests that reduction in FNTA expression may enhance the efficacy of FTIs.


Subject(s)
Cell Membrane/metabolism , Dimethylallyltranstransferase/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Neoplasms/pathology , Nuclear Matrix-Associated Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Estrogen/metabolism , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , CRISPR-Cas Systems/genetics , Computational Biology , Datasets as Topic , Gene Knockdown Techniques , Humans , Matrix Attachment Region Binding Proteins/genetics , Neoplasms/genetics , Nuclear Matrix-Associated Proteins/genetics , Protein Prenylation , Protein Subunits/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Estrogen/genetics
4.
Mol Cell ; 77(6): 1251-1264.e9, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32023484

ABSTRACT

Lipid droplets (LDs) store lipids for energy and are central to cellular lipid homeostasis. The mechanisms coordinating lipid storage in LDs with cellular metabolism are unclear but relevant to obesity-related diseases. Here we utilized genome-wide screening to identify genes that modulate lipid storage in macrophages, a cell type involved in metabolic diseases. Among ∼550 identified screen hits is MLX, a basic helix-loop-helix leucine-zipper transcription factor that regulates metabolic processes. We show that MLX and glucose-sensing family members MLXIP/MondoA and MLXIPL/ChREBP bind LDs via C-terminal amphipathic helices. When LDs accumulate in cells, these transcription factors bind to LDs, reducing their availability for transcriptional activity and attenuating the response to glucose. Conversely, the absence of LDs results in hyperactivation of MLX target genes. Our findings uncover a paradigm for a lipid storage response in which binding of MLX transcription factors to LD surfaces adjusts the expression of metabolic genes to lipid storage levels.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Glucose/metabolism , Lipid Droplets/metabolism , Proteome/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cells, Cultured , Genetic Testing , Humans , Macrophages/cytology , Macrophages/metabolism , Protein Binding , Proteome/analysis , RNA, Small Interfering , Transcription, Genetic
5.
Nat Commun ; 7: 13516, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27869117

ABSTRACT

In humans and animals lacking functional LDL receptor (LDLR), LDL from plasma still readily traverses the endothelium. To identify the pathways of LDL uptake, a genome-wide RNAi screen was performed in endothelial cells and cross-referenced with GWAS-data sets. Here we show that the activin-like kinase 1 (ALK1) mediates LDL uptake into endothelial cells. ALK1 binds LDL with lower affinity than LDLR and saturates only at hypercholesterolemic concentrations. ALK1 mediates uptake of LDL into endothelial cells via an unusual endocytic pathway that diverts the ligand from lysosomal degradation and promotes LDL transcytosis. The endothelium-specific genetic ablation of Alk1 in Ldlr-KO animals leads to less LDL uptake into the aortic endothelium, showing its physiological role in endothelial lipoprotein metabolism. In summary, identification of pathways mediating LDLR-independent uptake of LDL may provide unique opportunities to block the initiation of LDL accumulation in the vessel wall or augment hepatic LDLR-dependent clearance of LDL.


Subject(s)
Activin Receptors, Type II/metabolism , Cholesterol, LDL/metabolism , Endothelial Cells/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type II/genetics , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Biological Transport , Cells, Cultured , Cholesterol, LDL/genetics , Cloning, Molecular , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Male , Mice , RNA Interference
6.
Proc Natl Acad Sci U S A ; 113(31): E4558-66, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27432991

ABSTRACT

Bromodomain and extraterminal domain protein inhibitors (BETi) hold great promise as a novel class of cancer therapeutics. Because acquired resistance typically limits durable responses to targeted therapies, it is important to understand mechanisms by which tumor cells adapt to BETi. Here, through pooled shRNA screening of colorectal cancer cells, we identified tripartite motif-containing protein 33 (TRIM33) as a factor promoting sensitivity to BETi. We demonstrate that loss of TRIM33 reprograms cancer cells to a more resistant state through at least two mechanisms. TRIM33 silencing attenuates down-regulation of MYC in response to BETi. Moreover, loss of TRIM33 enhances TGF-ß receptor expression and signaling, and blocking TGF-ß receptor activity potentiates the antiproliferative effect of BETi. These results describe a mechanism for BETi resistance and suggest that combining inhibition of TGF-ß signaling with BET bromodomain inhibition may offer new therapeutic benefits.


Subject(s)
Azepines/pharmacology , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Triazoles/pharmacology , Azepines/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance/genetics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HEK293 Cells , Humans , Molecular Structure , Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Triazoles/chemistry
7.
Proc Natl Acad Sci U S A ; 110(18): 7452-7, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23569269

ABSTRACT

Despite major advances in our understanding of many aspects of human papillomavirus (HPV) biology, HPV entry is poorly understood. To identify cellular genes required for HPV entry, we conducted a genome-wide screen for siRNAs that inhibited infection of HeLa cells by HPV16 pseudovirus. Many retrograde transport factors were required for efficient infection, including multiple subunits of the retromer, which initiates retrograde transport from the endosome to the trans-Golgi network (TGN). The retromer has not been previously implicated in virus entry. Furthermore, HPV16 capsid proteins arrive in the TGN/Golgi in a retromer-dependent fashion during entry, and incoming HPV proteins form a stable complex with retromer subunits. We propose that HPV16 directly engages the retromer at the early or late endosome and traffics to the TGN/Golgi via the retrograde pathway during cell entry. These results provide important insights into HPV entry, identify numerous potential antiviral targets, and suggest that the role of the retromer in infection by other viruses should be assessed.


Subject(s)
Genome, Human/genetics , Papillomaviridae/physiology , RNA Interference , RNA, Small Interfering/metabolism , Vesicular Transport Proteins/metabolism , Virus Internalization , Golgi Apparatus/virology , HeLa Cells , Human papillomavirus 16/physiology , Humans , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Protein Binding , Protein Transport , Reproducibility of Results , Viral Proteins/metabolism
8.
J Biomed Sci ; 16: 21, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19272191

ABSTRACT

BACKGROUND: The endocardial endothelium that lines the inner cavity of the heart is distinct from the microvascular endothelial cells and modulates cardiac muscle performance in a manner similar to the vascular endothelial modulation of vascular structure and vasomotor tone. Although the modulatory effects of endocardial endothelium (EE) on cardiomyocytes are firmly established, the regulatory effects of endocardial endothelium on the cardiac interstitium and its cellular components remain ill defined. METHODS AND RESULTS: We investigated whether the stimulatory effect of EE on cardiac fibroblasts would be altered when EECs are activated by the cytokine tumor necrosis factor-alpha (TNF-alpha) or the endotoxin bacterial lipopolysaccharide (LPS). Both TNF-alpha and LPS were found to independently attenuate the stimulatory effect of EE on cardiac fibroblasts. These agents lowered the synthesis or release of ET-1 and increased the secretion of TGF-beta and NO. CONCLUSION: The findings of this study using endocardial endothelial cells (EECs) and neonatal cardiac fibroblasts demonstrate that pro-inflammatory cytokines cause altered secretion of paracrine factors by EECs and inhibit proliferation and lower collagen synthesis in fibroblasts. These changes may influence fibroblast response and extra cellular matrix remodeling in pathological conditions of the heart.


Subject(s)
Endocardium/cytology , Endothelial Cells/metabolism , Fibroblasts/metabolism , Lipopolysaccharides/metabolism , Myocardium/cytology , Tumor Necrosis Factor-alpha/metabolism , Animals , Animals, Newborn , Cells, Cultured , Culture Media, Conditioned/chemistry , Endocardium/metabolism , Endothelial Cells/cytology , Fibroblasts/cytology , Myocardium/metabolism , Nitrites/metabolism , Paracrine Communication , Rats , Rats, Wistar , Swine
9.
Cell Biochem Biophys ; 47(1): 65-72, 2007.
Article in English | MEDLINE | ID: mdl-17406060

ABSTRACT

Given that vascular endothelial cells play an important role in the modulation of vascular structure and function, we hypothesized that endocardial endothelial cells (EECs) may have a modulator role in regulating the cardiac interstitial cells. Endocardial endothelial cells were isolated from freshly collected pig hearts and cardiac fibroblasts were isolated from 3- to 4-d-old Wistar rats. Fibroblasts were cultured in the presence or absence of conditioned medium from EECs. Proliferation of cardiac fibroblasts was measured by the incorporation of [3H]- Thymidine and collagen synthesis was assayed by the incorporation of [3H]-Proline. To determine the involvement of signaling mediators, in separate experiments, cardiac fibroblasts were incubated with BQ123 (selective ETA receptor antagonist), PD142893 (nonselective ETA/ETB receptor antagonist), Bis-indolylmaleimide (PKC inhibitor), PD 098059 (MEK inhibitor), or neutralizing anti-transforming growth factor (TGF)-beta-antibody. Endocardial endothelium-derived factors endothelin (ET)-1, TGF-beta, and Angiotensin (Ang)-II in the conditioned medium were assayed by enzyme-linked immunosorbent assay using commercially available kits. We report here evidence that suggest that endocardial endothelial cells stimulate both proliferation and collagen synthesis of cardiac fibroblasts. The response seems to be mediated by endothelin through its ETA receptor. Our results also indicate that protein kinase C (PKC) and mitogen-activated protein kinase (MAPK) pathways are essential for the EEC-induced proliferation of cardiac fibroblasts.


Subject(s)
Collagen/metabolism , Endocardium/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/metabolism , Myocardium/metabolism , Animals , Cell Line , Cell Proliferation , Culture Media, Conditioned/metabolism , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Protein Kinase C/metabolism , Rats , Rats, Wistar , Transforming Growth Factor beta/metabolism
10.
Endothelium ; 14(1): 35-43, 2007.
Article in English | MEDLINE | ID: mdl-17364895

ABSTRACT

Endocardial endothelial cells (EECs), which form the inner lining of the cavities of the heart, are a distinct cell population whose dysfunction can be critical in pathological conditions of heart. Insights into the role and organization of these cells in pathological states of the heart are limited mainly due to a dearth of experimental models. To date no endocardial endothelial cell line is available. The authors attempted to immortalize porcine ventricular EECs by transfecting the cells with human telomerase reverse transcriptase (hTERT). EECs immortalized by ectopic expression of hTERT exhibit phenotypic and functional characteristics similar to primary EECs. The EE cell line could be useful for the study of mechanisms involved in the interaction of EECs with the underlying myocardium and cardiac interstitium and as useful tools in understanding their role in diseased states of heart.


Subject(s)
Endocardium/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Heart Ventricles/cytology , Telomerase/metabolism , Animals , Blotting, Western , Cell Division/physiology , Cell Line, Transformed , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Direct , Fluorescent Dyes , Humans , Immunohistochemistry , Kinetics , Nitrites/analysis , Reverse Transcriptase Polymerase Chain Reaction , Swine , Telomerase/analysis , Telomerase/genetics , Transfection
11.
Biol Trace Elem Res ; 114(1-3): 85-92, 2006.
Article in English | MEDLINE | ID: mdl-17205990

ABSTRACT

Cerium has been implicated in the pathogenesis of cardiac disorders such as acute myocardial infarction and endomyocardial fibrosis (EMF). A geochemical hypothesis for the causation of EMF linked the cardiac lesions to magnesium deficiency consequent to malnutrition and increased cardiac levels of cerium derived from monazite soils in the coastal regions of the tropics. We tested the hypothesis that the stimulus for fibroblast proliferation and enhanced collagen synthesis in EMF is derived from cardiac endothelial cells activated or injured by cerium. We explored whether endocardial endothelial cells exposed to cerium secrete factors responsible for the increased proliferation and collagen synthesis in cardiac fibroblasts. Our results suggest that the growth response of cardiac fibroblasts to cerium is not mediated through growth factors secreted by endocardial endothelium and that the cardiac lesions in EMF result from direct stimulation of subendocardial fibroblasts by cerium.


Subject(s)
Cell Proliferation , Cerium/toxicity , Endothelium, Vascular/drug effects , Heart/drug effects , Myocardium/cytology , Animals , Cells, Cultured , Collagen/biosynthesis , Culture Media, Conditioned , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Myocardium/metabolism , Swine
12.
Mol Cell Biochem ; 253(1-2): 113-23, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14619961

ABSTRACT

Endothelium is now recognized as a massive, regionally specific, multifunctional organ. Given its strategic anatomic location between the circulating blood components and the vascular smooth muscle or the cardiac muscle, it is a biologically significant interface whose dysfunction can be a critical factor in various pathological conditions. Two types of endothelial cells are recognized in the heart, the endocardial endothelial (EE) cells and the microvascular endothelial cells (MVE). Both produce common autacoids and share similar roles in signal transduction induced by neurotransmitters, hormones or mechanical stimuli. They are however two distinct cell populations with dissimilar embryological origin, cytoskeletal organization, receptor mediated functions and electrophysiological properties. Both the MVE and EE are modulators of cardiac performance. Myocardial contraction may be modulated by cardioactive agents such as nitric oxide, prostanoids, endothelin, natriuretic peptides, angiotensin II, kinins, reactive oxygen species and adenyl purines released from the cardiac endothelium. Two mechanisms have been proposed for the signal transduction from EE to the underlying myocytes: stimulus-secretion-contraction coupling and blood-heart barrier. Nitric oxide, bradykinin and myofilament desensitizing agent are probably important in short-term regulation of myocardial functions. Endothelin and Angiotensin II are probably involved in long-term regulation. Besides its sensory function and paracrine modulation of myocardial performance, EE as a blood-heart barrier could be of significance for the ionic homeostasis of the cardiac interstitium. In cardiac diseases, the damage to EE or MVE leading to failure of the endothelial cells to perform its regulatory and modulator functions may have serious consequences. A better understanding of the endothelial signaling pathways in cardiac physiology and pathophysiology may lead to the development of novel therapeutic strategies.


Subject(s)
Endothelium, Vascular/metabolism , Myocardial Contraction/physiology , Myocardium/metabolism , Animals , Cell Communication/physiology , Humans , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL