Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Dev Cell ; 59(2): 244-261.e6, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38154460

ABSTRACT

WNT morphogens trigger signaling pathways fundamental for embryogenesis, regeneration, and cancer. WNTs are modified with palmitoleate, which is critical for binding Frizzled (FZD) receptors and activating signaling. However, it is unknown how WNTs are released and spread from cells, given their strong lipid-dependent membrane attachment. We demonstrate that secreted FZD-related proteins and WNT inhibitory factor 1 are WNT carriers, potently releasing lipidated WNTs and forming active soluble complexes. WNT release occurs by direct handoff from the membrane protein WNTLESS to the carriers. In turn, carriers donate WNTs to glypicans and FZDs involved in WNT reception and to the NOTUM hydrolase, which antagonizes WNTs by lipid moiety removal. WNT transfer from carriers to FZDs is greatly facilitated by glypicans that serve as essential co-receptors in Wnt signaling. Thus, an extracellular network of carriers dynamically controls secretion, posttranslational regulation, and delivery of WNT morphogens, with important practical implications for regenerative medicine.


Subject(s)
Glypicans , Wnt Proteins , Wnt Proteins/metabolism , Glypicans/metabolism , Wnt Signaling Pathway , Embryonic Development , Lipids , Frizzled Receptors/chemistry , Frizzled Receptors/metabolism
2.
ACS Chem Biol ; 18(3): 583-594, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36795767

ABSTRACT

Biomolecular condensates formed by liquid-liquid phase separation have been implicated in multiple diseases. Modulation of condensate dynamics by small molecules has therapeutic potential, but so far, few condensate modulators have been disclosed. The SARS-CoV-2 nucleocapsid (N) protein forms phase-separated condensates that are hypothesized to play critical roles in viral replication, transcription, and packaging, suggesting that N condensation modulators might have anti-coronavirus activity across multiple strains and species. Here, we show that N proteins from all seven human coronaviruses (HCoVs) vary in their tendency to undergo phase separation when expressed in human lung epithelial cells. We developed a cell-based high-content screening platform and identified small molecules that both promote and inhibit condensation of SARS-CoV-2 N. Interestingly, these host-targeted small molecules exhibited condensate-modulatory effects across all HCoV Ns. Some have also been reported to exhibit antiviral activity against SARS-CoV-2, HCoV-OC43, and HCoV-229E viral infections in cell culture. Our work reveals that the assembly dynamics of N condensates can be regulated by small molecules with therapeutic potential. Our approach allows for screening based on viral genome sequences alone and might enable rapid paths to drug discovery with value for confronting future pandemics.


Subject(s)
COVID-19 , Coronavirus 229E, Human , Coronavirus OC43, Human , Humans , SARS-CoV-2 , Nucleocapsid Proteins
3.
Neuro Oncol ; 25(1): 185-198, 2023 01 05.
Article in English | MEDLINE | ID: mdl-35640920

ABSTRACT

BACKGROUND: Supratentorial RELA fusion (ST-RELA) ependymomas (EPNs) are resistant tumors without an approved chemotherapeutic treatment. Unfortunately, the molecular mechanisms that lead to chemoresistance traits of ST-RELA remain elusive. The aim of this study was to assess RELA fusion-dependent signaling modules, specifically the role of the Hedgehog (Hh) pathway as a novel targetable vulnerability in ST-RELA. METHODS: Gene expression was analyzed in EPN from patient cohorts, by microarray, RNA-seq, qRT-PCR, and scRNA-seq. Inhibitors against Smoothened (SMO) (Sonidegib) and Aurora kinase A (AURKA) (Alisertib) were evaluated. Protein expression, primary cilia formation, and drug effects were assessed by immunoblot, immunofluorescence, and immunohistochemistry. RESULTS: Hh components were selectively overexpressed in EPNs induced by the RELA fusion. Single-cell analysis showed that the Hh signature was primarily confined to undifferentiated, stem-like cell subpopulations. Sonidegib exhibited potent growth-inhibitory effects on ST-RELA cells, suggesting a key role in active Hh signaling; importantly, the effect of Sonidegib was reversed by primary cilia loss. We, thus, tested the effect of AURKA inhibition by Alisertib, to induce cilia stabilization/reassembly. Strikingly, Alisertib rescued ciliogenesis and synergized with Sonidegib in killing ST-RELA cells. Using a xenograft model, we show that cilia loss is a mechanism for acquiring resistance to the inhibitory effect of Sonidegib. However, Alisertib fails to rescue cilia and highlights the need for other strategies to promote cilia reassembly, for treating ST-RELA tumors. CONCLUSION: Our study reveals a crucial role for the Hh pathway in ST-RELA tumor growth, and suggests that rescue of primary cilia represents a vulnerability of the ST-RELA EPNs.


Subject(s)
Ependymoma , Supratentorial Neoplasms , Humans , Hedgehog Proteins , Cilia/metabolism , Cilia/pathology , Aurora Kinase A/genetics , Ependymoma/pathology , Supratentorial Neoplasms/pathology , Transcription Factor RelA
4.
Dev Cell ; 57(5): 670-685.e8, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35231446

ABSTRACT

The dually lipidated Sonic hedgehog (SHH) morphogen signals through the tumor suppressor membrane protein Patched1 (PTCH1) to activate the Hedgehog pathway, which is fundamental in development and cancer. SHH engagement with PTCH1 requires the GAS1 coreceptor, but the mechanism is unknown. We demonstrate a unique role for GAS1, catalyzing SHH-PTCH1 complex assembly in vertebrate cells by direct SHH transfer from the extracellular SCUBE2 carrier to PTCH1. Structure of the GAS1-SHH-PTCH1 transition state identifies how GAS1 recognizes the SHH palmitate and cholesterol modifications in modular fashion and how it facilitates lipid-dependent SHH handoff to PTCH1. Structure-guided experiments elucidate SHH movement from SCUBE2 to PTCH1, explain disease mutations, and demonstrate that SHH-induced PTCH1 dimerization causes its internalization from the cell surface. These results define how the signaling-competent SHH-PTCH1 complex assembles, the key step triggering the Hedgehog pathway, and provide a paradigm for understanding morphogen reception and its regulation.


Subject(s)
Hedgehog Proteins , Patched-1 Receptor , Signal Transduction , Catalysis , Cholesterol/metabolism , Hedgehog Proteins/metabolism , Patched-1 Receptor/genetics , Patched-1 Receptor/metabolism , Structure-Activity Relationship
5.
J Cell Sci ; 135(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35142364

ABSTRACT

Glypicans are a family of cell surface heparan sulfate proteoglycans that play critical roles in multiple cell signaling pathways. Glypicans consist of a globular core, an unstructured stalk modified with sulfated glycosaminoglycan chains, and a glycosylphosphatidylinositol anchor. Though these structural features are conserved, their individual contribution to glypican function remains obscure. Here, we investigate how glypican 3 (GPC3), which is mutated in Simpson-Golabi-Behmel tissue overgrowth syndrome, regulates Hedgehog signaling. We find that GPC3 is necessary for the Hedgehog response, surprisingly controlling a downstream signal transduction step. Purified GPC3 ectodomain rescues signaling when artificially recruited to the surface of GPC3-deficient cells but has dominant-negative activity when unattached. Strikingly, the purified stalk, modified with heparan sulfate but not chondroitin sulfate, is necessary and sufficient for activity. Our results demonstrate a novel function for GPC3-associated heparan sulfate and provide a framework for the functional dissection of glycosaminoglycans by in vivo biochemical complementation. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Abnormalities, Multiple , Glypicans , Hedgehog Proteins , Heparitin Sulfate , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Arrhythmias, Cardiac , Genetic Diseases, X-Linked , Gigantism , Glypicans/genetics , Glypicans/metabolism , Heart Defects, Congenital , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Heparan Sulfate Proteoglycans , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Signal Transduction
6.
Nat Commun ; 12(1): 6966, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845226

ABSTRACT

The membrane protein Dispatched (Disp), which belongs to the RND family of small molecule transporters, is essential for Hedgehog (Hh) signaling, by catalyzing the extracellular release of palmitate- and cholesterol-modified Hh ligands from producing cells. Disp function requires Furin-mediated proteolytic cleavage of its extracellular domain, but how this activates Disp remains obscure. Here, we employ cryo-electron microscopy to determine atomic structures of human Disp1 (hDisp1), before and after cleavage, and in complex with lipid-modified Sonic hedgehog (Shh) ligand. These structures, together with biochemical data, reveal that proteolytic cleavage opens the extracellular domain of hDisp1, removing steric hindrance to Shh binding. Structure-guided functional experiments demonstrate the role of hDisp1-Shh interactions in ligand release. Our results clarify the mechanisms of hDisp1 activation and Shh morphogen release, and highlight how a unique proteolytic cleavage event enabled acquisition of a protein substrate by a member of a family of small molecule transporters.


Subject(s)
Hedgehog Proteins/chemistry , Membrane Transport Proteins/chemistry , Amino Acid Sequence , Binding Sites , Cryoelectron Microscopy , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Ligands , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
7.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33526656

ABSTRACT

Hedgehog signaling is fundamental in animal embryogenesis, and its dysregulation causes cancer and birth defects. The pathway is triggered when the Hedgehog ligand inhibits the Patched1 membrane receptor, relieving repression that Patched1 exerts on the GPCR-like protein Smoothened. While it is clear how loss-of-function Patched1 mutations cause hyperactive Hedgehog signaling and cancer, how other Patched1 mutations inhibit signaling remains unknown. Here, we develop quantitative single-cell functional assays for Patched1, which, together with mathematical modeling, indicate that Patched1 inhibits Smoothened enzymatically, operating in an ultrasensitive regime. Based on this analysis, we propose that Patched1 functions in cilia, catalyzing Smoothened deactivation by removing cholesterol bound to its extracellular, cysteine-rich domain. Patched1 mutants associated with holoprosencephaly dampen signaling by three mechanisms: reduced affinity for Hedgehog ligand, elevated catalytic activity, or elevated affinity for the Smoothened substrate. Our results clarify the enigmatic mechanism of Patched1 and explain how Patched1 mutations lead to birth defects.


Subject(s)
Hedgehog Proteins/metabolism , Mutation/genetics , Patched-1 Receptor/genetics , Signal Transduction , Allosteric Regulation , Animals , Biocatalysis , Cholesterol/metabolism , Cilia/metabolism , Holoprosencephaly/genetics , Ligands , Mice , Models, Biological , Patched-1 Receptor/metabolism , Phenotype , Protein Domains , Smoothened Receptor/chemistry , Smoothened Receptor/metabolism
8.
Dev Cell ; 55(4): 450-467.e8, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33038332

ABSTRACT

Hedgehog signaling governs critical processes in embryogenesis, adult stem cell maintenance, and tumorigenesis. The activating ligand, Sonic hedgehog (SHH), is highly hydrophobic because of dual palmitate and cholesterol modification, and thus, its release from cells requires the secreted SCUBE proteins. We demonstrate that the soluble SCUBE-SHH complex, although highly potent in cellular assays, cannot directly signal through the SHH receptor, Patched1 (PTCH1). Rather, signaling by SCUBE-SHH requires a molecular relay mediated by the coreceptors CDON/BOC and GAS1, which relieves SHH inhibition by SCUBE. CDON/BOC bind both SCUBE and SHH, recruiting the complex to the cell surface. SHH is then handed off, in a dual lipid-dependent manner, to GAS1, and from GAS1 to PTCH1, initiating signaling. These results define an essential step in Hedgehog signaling, whereby coreceptors activate SHH by chaperoning it from a latent extracellular complex to its cell-surface receptor, and point to a broader paradigm of coreceptor function.


Subject(s)
Hedgehog Proteins/metabolism , Lipids/chemistry , Receptors, Cell Surface/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/metabolism , Catalysis , Cell Cycle Proteins/metabolism , Cholesterol/metabolism , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Ligands , Mice , Models, Biological , NIH 3T3 Cells , Palmitic Acid/pharmacology , Patched-1 Receptor/metabolism
9.
Dev Cell ; 55(3): 314-327.e7, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32860743

ABSTRACT

Cholesterol plays two critical roles in Hedgehog signaling, a fundamental pathway in animal development and cancer: it covalently modifies the Sonic hedgehog (SHH) ligand, restricting its release from producing cells, and directly activates Smoothened in responding cells. In both contexts, a membrane protein related to bacterial RND transporters regulates cholesterol: Dispatched1 controls release of cholesterylated SHH, and Patched1 antagonizes Smoothened activation by cholesterol. The mechanism and driving force for eukaryotic RND proteins, including Dispatched1 and Patched1, are unknown. Here, we show that Dispatched1 acts enzymatically to catalyze SHH release. Dispatched1 uses the energy of the plasma membrane Na+ gradient, thus functioning as an SHH/Na+ antiporter. In contrast, Patched1 repression of Smoothened requires the opposing K+ gradient. Our results clarify the transporter activity of essential eukaryotic RND proteins and demonstrate that the two main cation gradients of animal cells differentially power cholesterol transport at two crucial steps in the Hedgehog pathway.


Subject(s)
Cholesterol/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Biocatalysis , Biological Transport , Cations , Cell Membrane/metabolism , Conserved Sequence , Humans , Membrane Proteins , Mice , Models, Biological , NIH 3T3 Cells , Patched-1 Receptor/metabolism , Sodium/metabolism
11.
Cell ; 174(2): 312-324.e16, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29804838

ABSTRACT

The seven-transmembrane-spanning protein Smoothened is the central transducer in Hedgehog signaling, a pathway fundamental in development and in cancer. Smoothened is activated by cholesterol binding to its extracellular cysteine-rich domain (CRD). How this interaction leads to changes in the transmembrane domain and Smoothened activation is unknown. Here, we report crystal structures of sterol-activated Smoothened. The CRD undergoes a dramatic reorientation, allosterically causing the transmembrane domain to adopt a conformation similar to active G-protein-coupled receptors. We show that Smoothened contains a unique inhibitory π-cation lock, which is broken on activation and is disrupted in constitutively active oncogenic mutants. Smoothened activation opens a hydrophobic tunnel, suggesting a pathway for cholesterol movement from the inner membrane leaflet to the CRD. All Smoothened antagonists bind the transmembrane domain and block tunnel opening, but cyclopamine also binds the CRD, inducing the active transmembrane conformation. Together, these results define the mechanisms of Smoothened activation and inhibition.


Subject(s)
Hedgehog Proteins/metabolism , Smoothened Receptor/chemistry , Xenopus Proteins/chemistry , Allosteric Regulation , Animals , Binding Sites , Cell Line , Cholesterol/chemistry , Cholesterol/metabolism , Crystallography, X-Ray , Flow Cytometry , Hedgehog Proteins/genetics , Humans , Mice , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Protein Structure, Tertiary , Signal Transduction , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism , Veratrum Alkaloids/chemistry , Veratrum Alkaloids/metabolism , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
12.
Annu Rev Cell Dev Biol ; 33: 145-168, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28693388

ABSTRACT

Communication between cells pervades the development and physiology of metazoans. In animals, this process is carried out by a relatively small number of signaling pathways, each consisting of a chain of biochemical events through which extracellular stimuli control the behavior of target cells. One such signaling system is the Hedgehog pathway, which is crucial in embryogenesis and is implicated in many birth defects and cancers. Although Hedgehog pathway components were identified by genetic analysis more than a decade ago, our understanding of the molecular mechanisms of signaling is far from complete. In this review, we focus on the biochemistry and cell biology of the Hedgehog pathway. We examine the unique biosynthesis of the Hedgehog ligand, its specialized release from cells into extracellular space, and the poorly understood mechanisms involved in ligand reception and pathway activation at the surface of target cells. We highlight several critical questions that remain open.


Subject(s)
Hedgehog Proteins/metabolism , Signal Transduction , Animals , Feedback, Physiological , Humans , Ligands , Models, Biological
13.
Proc Natl Acad Sci U S A ; 113(40): E5866-E5875, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647915

ABSTRACT

The Hedgehog cell-cell signaling pathway is crucial for animal development, and its misregulation is implicated in numerous birth defects and cancers. In unstimulated cells, pathway activity is inhibited by the tumor suppressor membrane protein, Patched. Hedgehog signaling is triggered by the secreted Hedgehog ligand, which binds and inhibits Patched, thus setting in motion the downstream events in signal transduction. Despite its critical importance, the mechanism by which Hedgehog antagonizes Patched has remained unknown. Here, we show that vertebrate Patched1 inhibition is caused by direct, palmitate-dependent interaction with the Sonic Hedgehog ligand. We find that a short palmitoylated N-terminal fragment of Sonic Hedgehog binds Patched1 and, strikingly, is sufficient to inhibit it and to activate signaling. The rest of Sonic Hedgehog confers high-affinity Patched1 binding and internalization through a distinct binding site, but, surprisingly, it is not absolutely required for signaling. The palmitate-dependent interaction with Patched1 is specifically impaired in a Sonic Hedgehog mutant causing human holoprosencephaly, the most frequent congenital brain malformation, explaining its drastically reduced potency. The palmitate-dependent interaction is also abolished in constitutively inhibited Patched1 point mutants causing the Gorlin cancer syndrome, suggesting that they might adopt a conformation distinct from the wild type. Our data demonstrate that Sonic Hedgehog signals via the palmitate-dependent arm of a two-pronged contact with Patched1. Furthermore, our results suggest that, during Hedgehog signaling, ligand binding inhibits Patched by trapping it in an inactive conformation, a mechanism that explains the dramatically reduced activity of oncogenic Patched1 mutants.


Subject(s)
Hedgehog Proteins/metabolism , Patched Receptors/metabolism , Signal Transduction , 3T3 Cells , Animals , Endocytosis/drug effects , Holoprosencephaly/metabolism , Holoprosencephaly/pathology , Humans , Lipoylation , Mice , Models, Molecular , Mutation/genetics , Oncogenes , Palmitic Acid/metabolism , Peptides/pharmacology , Protein Binding/drug effects , Signal Transduction/drug effects
14.
Cell ; 166(5): 1176-1187.e14, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27545348

ABSTRACT

In vertebrates, sterols are necessary for Hedgehog signaling, a pathway critical in embryogenesis and cancer. Sterols activate the membrane protein Smoothened by binding its extracellular, cysteine-rich domain (CRD). Major unanswered questions concern the nature of the endogenous, activating sterol and the mechanism by which it regulates Smoothened. We report crystal structures of CRD complexed with sterols and alone, revealing that sterols induce a dramatic conformational change of the binding site, which is sufficient for Smoothened activation and is unique among CRD-containing receptors. We demonstrate that Hedgehog signaling requires sterol binding to Smoothened and define key residues for sterol recognition and activity. We also show that cholesterol itself binds and activates Smoothened. Furthermore, the effect of oxysterols is abolished in Smoothened mutants that retain activation by cholesterol and Hedgehog. We propose that the endogenous Smoothened activator is cholesterol, not oxysterols, and that vertebrate Hedgehog signaling controls Smoothened by regulating its access to cholesterol.


Subject(s)
Cholesterol/metabolism , Hedgehog Proteins/metabolism , Smoothened Receptor/agonists , Animals , Cholesterol/chemistry , Crystallography, X-Ray , Mice , NIH 3T3 Cells , Oxysterols/chemistry , Oxysterols/metabolism , Protein Binding , Protein Conformation , Signal Transduction , Smoothened Receptor/chemistry , Smoothened Receptor/metabolism , Xenopus laevis
15.
Cell Rep ; 15(9): 1920-9, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27210761

ABSTRACT

Upon Notch pathway activation, the receptor is cleaved to release the Notch intracellular domain (NICD), which translocates to the nucleus to activate gene transcription. Using Xenopus egg extracts, we have identified a Notch1-specific destruction signal (N1-Box). We show that mutations in the N1-Box inhibit NICD1 degradation and that the N1-Box is transferable for the promotion of degradation of heterologous proteins in Xenopus egg extracts and in cultured human cells. Mutation of the N1-Box enhances Notch1 activity in cultured human cells and zebrafish embryos. Human cancer mutations within the N1-Box enhance Notch1 signaling in transgenic zebrafish, highlighting the physiological relevance of this destruction signal. We find that binding of the Notch nuclear factor, CSL, to the N1-Box blocks NICD1 turnover. Our studies reveal a mechanism by which degradation of NICD1 is regulated by the N1-Box to minimize stochastic flux and to establish a threshold for Notch1 pathway activation.


Subject(s)
Proteolysis , Receptor, Notch1/chemistry , Receptor, Notch1/metabolism , Sequence Homology, Amino Acid , Amino Acid Sequence , Animals , Cell Extracts , Embryo, Nonmammalian/metabolism , F-Box Proteins/metabolism , HEK293 Cells , Humans , Muscle Proteins/metabolism , Mutation/genetics , Protein Binding , Protein Domains , Protein Stability , Regulatory Sequences, Nucleic Acid/genetics , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism , Xenopus , Zebrafish/embryology
16.
Chem Commun (Camb) ; 51(99): 17529-31, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26469153

ABSTRACT

Dynamic constitutional frameworks, based on squalene, PEG and PEI components, reversibly connected to core centers, allow the efficient identification of adaptive vectors for good DNA transfection efficiency and are well tolerated by mammalian cells.


Subject(s)
DNA/chemistry , Genetic Vectors , Transfection , DNA/genetics , HeLa Cells , Humans , Microscopy, Electron, Transmission
17.
Org Biomol Chem ; 13(34): 9005-11, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26171592

ABSTRACT

We introduce Dynamic Constitutional Frameworks (DCFs), macromolecular structures that efficiently bind and transfect double stranded DNA. DCFs are easily synthesizable adaptive 3D networks consisting of core connection centres reversibly linked via labile imine bonds both to linear polyethyleneglycol (PEG, ∼1500 Da) and to branched polyethyleneimine (bPEI, ∼800 Da). DCFs bind linear and plasmid DNA, forming particulate polyplexes of 40-200 nm in diameter. The polyplexes are stable during gel electrophoresis, well tolerated by cells in culture, and exhibit significant transfection activity. We show that an optimal balance of PEG and bPEI components is important for building DCFs that are non-toxic and exhibit good cellular transfection activity. Our study demonstrates the versatility and effectiveness of DCFs as promising new vectors for DNA delivery.


Subject(s)
DNA/chemistry , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Cell Survival , HeLa Cells , Humans , Microscopy, Electron, Transmission , Molecular Weight , Photoelectron Spectroscopy , Plasmids/administration & dosage
18.
Chembiochem ; 16(4): 611-7, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25663046

ABSTRACT

Cholesterol is a fundamental lipid component of eukaryotic membranes and a precursor of potent signaling molecules, such as oxysterols and steroid hormones. Cholesterol and oxysterols are also essential for Hedgehog signaling, a pathway critical in embryogenesis and cancer. Despite their importance, the use of imaging sterols in cells is currently very limited. We introduce a robust and versatile method for sterol microscopy based on C19 alkyne cholesterol and oxysterol analogues. These sterol analogues are fully functional; they rescue growth of cholesterol auxotrophic cells and faithfully recapitulate the multiple roles that sterols play in Hedgehog signal transduction. Alkyne sterol analogues incorporate efficiently into cellular membranes and can be imaged with high resolution after copper(I)-catalyzed azide-alkyne cycloaddition reaction with fluorescent azides. We demonstrate the use of alkyne sterol probes for visualizing the subcellular distribution of cholesterol and for two-color imaging of sterols and choline phospholipids. Our imaging strategy should be broadly applicable to studying the role of sterols in normal physiology and disease.


Subject(s)
Hedgehog Proteins/metabolism , Optical Imaging , Signal Transduction , Sterols/analysis , Alkynes/chemistry , Animals , Azides/chemistry , Cholesterol/analogs & derivatives , Click Chemistry , Copper/chemistry , Cycloaddition Reaction , Humans , Mice , Microscopy/methods , Molecular Probes/chemistry , NIH 3T3 Cells , Optical Imaging/methods , Sterols/metabolism
19.
Chembiochem ; 16(3): 472-6, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25586136

ABSTRACT

Phospholipids with a choline head group are abundant components of all biological membranes, performing critical functions in cellular structure, metabolism, and signaling. In spite of their importance, our ability to visualize choline phospholipids in vivo remains very limited. We present a simple and robust chemical strategy to image choline phospholipids, based on the metabolic incorporation of azidocholine analogues, that accurately reflects the normal biosynthetic incorporation of choline into cellular phospholipids. Azidocholine-labeled phospholipids can be imaged in cells with high sensitivity and resolution, following derivatization with fluorophores, by bio-orthogonal chemical reactions compatible with live-cell imaging. We used this method to visualize the subcellular localization of choline phospholipids. We also demonstrate that double metabolic labeling with azidocholine and propargylcholine allows sensitive two-color imaging of choline phospholipids. Our method represents a powerful approach to directly image phospholipids, and to study their dynamics in cells and tissues.


Subject(s)
Click Chemistry , Molecular Imaging/methods , Phospholipids/analysis , Phospholipids/chemistry , Animals , Azides/chemistry , Choline/analogs & derivatives , Choline/chemistry , Fluorescent Dyes/chemistry , Mice , Molecular Structure , NIH 3T3 Cells/metabolism , Phosphatidylcholines/analysis , Phosphatidylcholines/chemistry , Phospholipids/metabolism , Sensitivity and Specificity
20.
ACS Chem Biol ; 9(10): 2217-22, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25122546

ABSTRACT

Choline-containing phospholipids (Cho-PLs) are major components of all cellular membranes. We developed an electron microscopic technique to investigate the poorly understood problem of how Cho-PLs are distributed between membrane leaflets. Our method relies on generating freeze-fracture replicas of cells metabolically labeled with the choline analog, propargylcholine, followed by "click" reaction to conjugate biotin to propargylcholine head groups, and immunodetection of biotin with colloidal gold. Using this method in budding yeast, we found that, surprisingly, the Golgi and plasma membrane display a cytoplasmic leaflet-dominant asymmetry in Cho-PL distribution; in contrast, Cho-PLs are evenly distributed between the exoplasmic and cytoplasmic leaflets of other organelle membranes. In mammalian culture cells, the plasma membrane shows symmetrical Cho-PL distribution between leaflets, suggesting a fundamental difference between yeast and mammals. Our method should be expandable to other classes of lipids and will be useful for deciphering the mechanism responsible for generating lipid asymmetry in biological membranes.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Choline/metabolism , Click Chemistry/methods , Freeze Fracturing/methods , Liver Neoplasms/metabolism , Microscopy, Electron/methods , Phospholipids/metabolism , Saccharomyces cerevisiae/metabolism , Cell Membrane/metabolism , Chromatography, Thin Layer , Cytoplasm/metabolism , Golgi Apparatus/metabolism , Humans , Saccharomyces cerevisiae/growth & development , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...