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











Publication year range
1.
ASN Neuro ; 4(6): 409-21, 2012 Dec 03.
Article in English | MEDLINE | ID: mdl-23127255

ABSTRACT

PMP22 (peripheral myelin protein 22), also known as GAS 3 (growth-arrest-specific protein 3), is a disease-linked tetraspan glycoprotein of peripheral nerve myelin and constituent of intercellular junctions in epithelia. To date, our knowledge of the post-translational modification of PMP22 is limited. Using the CSS-Palm 2.0 software we predicted that C85 (cysteine 85), a highly conserved amino acid located between the second and third transmembrane domains, is a potential site for palmitoylation. To test this, we mutated C85S (C85 to serine) and established stable cells lines expressing the WT (wild-type) or the C85S-PMP22. In Schwann and MDCK (Madin-Darby canine kidney) cells mutating C85 blocked the palmitoylation of PMP22, which we monitored using 17-ODYA (17-octadecynoic acid). While palmitoylation was not necessary for processing the newly synthesized PMP22 through the secretory pathway, overexpression of C85S-PMP22 led to pronounced cell spreading and uneven monolayer thinning. To further investigate the functional significance of palmitoylated PMP22, we evaluated MDCK cell migration in a wound-healing assay. While WT-PMP22 expressing cells were resistant to migration, C85S cells displayed lamellipodial protrusions and migrated at a similar rate to vector control. These findings indicate that palmitoylation of PMP22 at C85 is critical for the role of the protein in modulating epithelial cell shape and motility.


Subject(s)
Cell Movement/genetics , Cell Size , Epithelial Cells/cytology , Epithelial Cells/physiology , Lipoylation/physiology , Myelin Proteins/metabolism , Animals , Bacterial Proteins/genetics , Caveolins/metabolism , Cell Movement/drug effects , Cells, Cultured , Cicatrix/metabolism , Cicatrix/pathology , Contactin 1/metabolism , Cysteine/genetics , Cysteine/metabolism , Dogs , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Fatty Acids, Unsaturated/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Lectins/metabolism , Lipoylation/drug effects , Lipoylation/genetics , Luminescent Proteins/genetics , Madin Darby Canine Kidney Cells , Mutation/genetics , Myelin Proteins/genetics , Radioimmunoprecipitation Assay , Rats , Schwann Cells/cytology , Schwann Cells/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection , Wounds and Injuries/pathology , rac GTP-Binding Proteins/metabolism
2.
Int J Cell Biol ; 2012: 150918, 2012.
Article in English | MEDLINE | ID: mdl-22536245

ABSTRACT

Cytoskeleton-associated protein 4 (CKAP4) is a reversibly palmitoylated and phosphorylated transmembrane protein that functions as a high-affinity receptor for antiproliferative factor (APF)-a sialoglycopeptide secreted from bladder epithelial cells of patients with interstitial cystitis (IC). Palmitoylation of CKAP4 by the palmitoyl acyltransferase, DHHC2, is required for its cell surface localization and subsequent APF signal transduction; however, the mechanism for APF signal transduction by CKAP4 is unknown. In this paper, we demonstrate that APF treatment induces serine phosphorylation of residues S3, S17, and S19 of CKAP4 and nuclear translocation of CKAP4. Additionally, we demonstrate that CKAP4 binds gDNA in a phosphorylation-dependent manner in response to APF treatment, and that a phosphomimicking, constitutively nonpalmitoylated form of CKAP4 localizes to the nucleus, binds DNA, and mimics the inhibitory effects of APF on cellular proliferation. These results reveal a novel role for CKAP4 as a downstream effecter for APF signal transduction.

3.
Expert Opin Drug Discov ; 5(2): 155-64, 2010 Feb.
Article in English | MEDLINE | ID: mdl-22822915

ABSTRACT

IMPORTANCE OF THE FIELD: Palmitoylation is the post-translational addition of a 16-carbon fatty acid, palmitate, to specific cysteines of proteins via a labile thioester bond. The transfer of palmitate to a substrate is mediated by palmitoyl acyltransferases (PATs). Nearly a third of the 23 genes that encode PATs are linked to human diseases, in particular cancer, and as such represent important targets for drug development. AREA COVERED IN THIS REVIEW: In this review, we summarize recent technical advances in the field of palmitoylation, how they will affect our ability to understand palmitoylation-related signaling, and outline a general strategy for the discovery of selective and potent palmitoylation inhibitors. WHAT THE READER WILL GAIN: The goals of this review are to increase awareness of the importance of palmitoylation in disease as well as our general lack of understanding of the complexity of the fundamental mechanisms of PAT regulation and specificity, and finally to suggest general strategies for the development of PAT inhibitors. TAKE HOME MESSAGE: Any reasonable hope of developing therapeutically useful, pharmacological modulators of palmitoylation will require that they be developed within the context of PAT-related signaling systems that are more extensively characterized than any we currently know. The successful creation of potent, specific drugs in other similarly complex systems suggests that development of useful drugs targeting PATs is certain.

4.
Mol Membr Biol ; 26(1): 14-31, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19191172

ABSTRACT

Thio-palmitoylation is the post-translational addition of the 16-carbon fatty acid, palmitate, to the thiol side chain of cysteine residues by a labile thioester bond. Palmitoylation increases the lipophilicity of a protein resulting in dramatic changes in its subcellular distribution such as moving from the endoplasmic reticulum to the plasma membrane or in subtle changes like an increased affinity for cholesterol-rich lipid rafts in membranes. Palmitoylation is also dynamic, making it unique among post-translational protein lipid modifications. Discovering the molecular identity of palmitoyl acyltransferases (PATs) was a watershed event that dramatically accelerated the pace of discovery in the field. Likewise, there has been increased interest in palmitoylation partly because many of the genes encoding PATs have been linked to cancer and other diseases. Now, with a greater understanding of how palmitate is enzymatically attached to proteins, some of the most interesting questions include: What are the substrates of each PAT?; how does a PAT recognize and palmitoylate a substrate?; how are PATs regulated?; and, how is depalmitoylation regulated? The answers to these questions are beginning to unfold due to the recent development of novel assays as well as the expansion and refinement of existing assays. Our ability to understand palmitoylation and its importance to human health and disease is only as good as the methods we use to test our hypotheses. The continued development of methods with increased sensitivity and selectivity is critical to this venture.


Subject(s)
Acyltransferases/metabolism , Lipoylation , Drug Discovery , Humans , Protein Processing, Post-Translational , Substrate Specificity
5.
Mol Biol Cell ; 20(5): 1454-63, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19144824

ABSTRACT

Previously, we identified cytoskeleton-associated protein 4 (CKAP4) as a major substrate of the palmitoyl acyltransferase, DHHC2, using a novel proteomic method called palmitoyl-cysteine identification, capture and analysis (PICA). CKAP4 is a reversibly palmitoylated and phosphorylated protein that links the ER to the cytoskeleton. It is also a high-affinity receptor for antiproliferative factor (APF), a small sialoglycopeptide secreted from bladder epithelial cells of patients with interstitial cystitis (IC). The role of DHHC2-mediated palmitoylation of CKAP4 in the antiproliferative response of HeLa and normal bladder epithelial cells to APF was investigated. Our data show that siRNA-mediated knockdown of DHHC2 and consequent suppression of CKAP4 palmitoylation inhibited the ability of APF to regulate cellular proliferation and blocked APF-induced changes in the expression of E-cadherin, vimentin, and ZO-1 (genes known to play a role in cellular proliferation and tumorigenesis). Immunocytochemistry revealed that CKAP4 palmitoylation by DHHC2 is required for its trafficking from the ER to the plasma membrane and for its nuclear localization. These data suggest an important role for DHHC2-mediated palmitoylation of CKAP4 in IC and in opposing cancer-related cellular behaviors and support the idea that DHHC2 is a tumor suppressor.


Subject(s)
Acyltransferases/physiology , Glycoproteins/metabolism , Membrane Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/physiology , Acyltransferases/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Half-Life , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins , Lipoylation , Models, Biological , Protein Transport , RNA Interference , RNA, Messenger/metabolism , Tumor Suppressor Proteins/metabolism
6.
Mol Cell Proteomics ; 7(7): 1378-88, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18296695

ABSTRACT

Protein palmitoylation is the post-translational addition of the 16-carbon fatty acid palmitate to specific cysteine residues by a labile thioester linkage. Palmitoylation is mediated by a family of at least 23 palmitoyl acyltransferases (PATs) characterized by an Asp-His-His-Cys (DHHC) motif. Many palmitoylated proteins have been identified, but PAT-substrate relationships are mostly unknown. Here we present a method called palmitoyl-cysteine isolation capture and analysis (or PICA) to identify PAT-substrate relationships in a living vertebrate system and demonstrate its effectiveness by identifying CKAP4/p63 as a substrate of DHHC2, a putative tumor suppressor.


Subject(s)
Acyltransferases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/physiology , Proteomics/methods , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dogs , Efficiency , HeLa Cells , Humans , Membrane Proteins/isolation & purification , Models, Biological , Palmitoyl Coenzyme A/metabolism , Protein Processing, Post-Translational , Sensitivity and Specificity , Substrate Specificity
7.
Assay Drug Dev Technol ; 5(1): 29-48, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17355198

ABSTRACT

High throughput image cytometers analyze individual cells in digital photomicrographs by first assigning pixels within each image to plasma membrane, cytoplasm, nucleus, or other regions. In this study, we report on a novel algorithm that: 1) identifies plasma membrane regions to measure changes in plasma membrane-associated proteins (protein kinase C [PKC] alpha, N-cadherin, E-cadherin, vascular endothelium [VE]-cadherin, and pan-cadherin) that regulate cell division, migration, and adhesion and 2) delineates the cell for generalized three-compartment image cytometry. Validation assays were performed for these proteins on cells cultured in 96-well plates and also for tissue sections obtained from transgenic and chemical carcinogenic models of skin cancer. The algorithm successfully quantified phorbol 12-myristate 13-acetate (PMA)-induced plasma membrane localization of PKCalpha in HeLa cells (Z' of 0.88). Additionally, PMA activated translocation to the plasma membrane at P < .01 of N-cadherin (in HeLa cells), E-cadherin (in A431 cells), and VE-cadherin (in human dermal microvascular endothelial cells), suggesting a relationship between PKCalpha activity and cadherin localization. For VE-cadherin, a Z' of 0.52 was obtained between serum-free medium, which increased VE-cadherin, and EGTA, which diminished VE-cadherin at the plasma membrane. For sections obtained from the transgenic skin cancer model, analysis of images with the plasma membrane algorithm revealed that tumor cells exhibited cadherin expression that was just 34% of that expressed by surrounding normal tissue; furthermore, tumor cells expressed elevated DNA content, consistent with development of aneuploidy. In contrast, increased DNA content did not occur for tumor cells produced by chemical carcinogenesis. The results demonstrate that this new algorithm for plasma membrane image cytometry enables statistically significant analyses in a variety of applications in both cultured cells and tissue sections.


Subject(s)
Biological Assay/methods , Cell Membrane/metabolism , Cell Separation/methods , Flow Cytometry/methods , Gene Expression Profiling/methods , Membrane Proteins/metabolism , Microscopy, Fluorescence, Multiphoton/methods , HeLa Cells , Humans
9.
Nat Biotechnol ; 21(12): 1505-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625562

ABSTRACT

Studies of protein function would be facilitated by a general method to inactivate selected proteins in living cells noninvasively with high spatial and temporal precision. Chromophore-assisted light inactivation (CALI) uses photochemically generated, reactive oxygen species to inactivate proteins acutely, but its use has been limited by the need to microinject dye-labeled nonfunction-blocking antibodies. We now demonstrate CALI of connexin43 (Cx43) and alpha1C L-type calcium channels, each tagged with one or two small tetracysteine (TC) motifs that specifically bind the membrane-permeant, red biarsenical dye, ReAsH. ReAsH-based CALI is genetically targeted, requires no antibodies or microinjection, and inactivates each protein by approximately 90% in <30 s of widefield illumination. Similar light doses applied to Cx43 or alpha1C tagged with green fluorescent protein (GFP) had negligible to slight effects with or without ReAsH exposure, showing the expected molecular specificity. ReAsH-mediated CALI acts largely via singlet oxygen because quenchers or enhancers of singlet oxygen respectively inhibit or enhance CALI.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium Channels, L-Type/radiation effects , Fluorescent Dyes/metabolism , Fluorescent Dyes/radiation effects , Gene Targeting/methods , Photochemistry/methods , Protein Engineering/methods , Calcium Channels, L-Type/genetics , Dose-Response Relationship, Radiation , Ion Channel Gating/physiology , Ion Channel Gating/radiation effects , Light , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/radiation effects
10.
Proc Natl Acad Sci U S A ; 99(12): 7877-82, 2002 Jun 11.
Article in English | MEDLINE | ID: mdl-12060735

ABSTRACT

All coelenterate fluorescent proteins cloned to date display some form of quaternary structure, including the weak tendency of Aequorea green fluorescent protein (GFP) to dimerize, the obligate dimerization of Renilla GFP, and the obligate tetramerization of the red fluorescent protein from Discosoma (DsRed). Although the weak dimerization of Aequorea GFP has not impeded its acceptance as an indispensable tool of cell biology, the obligate tetramerization of DsRed has greatly hindered its use as a genetically encoded fusion tag. We present here the stepwise evolution of DsRed to a dimer and then either to a genetic fusion of two copies of the protein, i.e., a tandem dimer, or to a true monomer designated mRFP1 (monomeric red fluorescent protein). Each subunit interface was disrupted by insertion of arginines, which initially crippled the resulting protein, but red fluorescence could be rescued by random and directed mutagenesis totaling 17 substitutions in the dimer and 33 in mRFP1. Fusions of the gap junction protein connexin43 to mRFP1 formed fully functional junctions, whereas analogous fusions to the tetramer and dimer failed. Although mRFP1 has somewhat lower extinction coefficient, quantum yield, and photostability than DsRed, mRFP1 matures >10 times faster, so that it shows similar brightness in living cells. In addition, the excitation and emission peaks of mRFP1, 584 and 607 nm, are approximately 25 nm red-shifted from DsRed, which should confer greater tissue penetration and spectral separation from autofluorescence and other fluorescent proteins.


Subject(s)
Luminescent Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cnidaria/metabolism , Dimerization , Kinetics , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Mammals , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection , Red Fluorescent Protein
11.
Science ; 296(5569): 913-6, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11988576

ABSTRACT

Many proteins associated with the plasma membrane are known to partition into submicroscopic sphingolipid- and cholesterol-rich domains called lipid rafts, but the determinants dictating this segregation of proteins in the membrane are poorly understood. We suppressed the tendency of Aequorea fluorescent proteins to dimerize and targeted these variants to the plasma membrane using several different types of lipid anchors. Fluorescence resonance energy transfer measurements in living cells revealed that acyl but not prenyl modifications promote clustering in lipid rafts. Thus the nature of the lipid anchor on a protein is sufficient to determine submicroscopic localization within the plasma membrane.


Subject(s)
Bacterial Proteins/metabolism , Luminescent Proteins/metabolism , Membrane Microdomains/metabolism , Oligopeptides/metabolism , Acylation , Animals , Bacterial Proteins/chemistry , Caveolin 1 , Caveolins/metabolism , Cell Line , Detergents , Dimerization , Dogs , Energy Transfer , Fluorescence , Green Fluorescent Proteins , Luminescent Proteins/chemistry , Myristic Acid/metabolism , Oligopeptides/chemistry , Palmitic Acid/metabolism , Protein Prenylation , Recombinant Fusion Proteins/metabolism , Solubility , Spectrometry, Fluorescence , Transfection
12.
Sci STKE ; 2002(131): pe23, 2002 May 07.
Article in English | MEDLINE | ID: mdl-11997581

ABSTRACT

Fluorescent proteins from sea creatures have revolutionized the study of cell biology and signal transduction in many ways. Zacharias discusses some of the technical caveats to working with these proteins when they are fused to cellular proteins to track protein localization and interactions. Special attention is paid to problems arising from oligomerization of these fluorescent proteins and how that impacts protein interactions detected by fluorescence resonance energy transfer (FRET).


Subject(s)
Cell Physiological Phenomena , Luminescent Proteins/metabolism , Oligopeptides/metabolism , Animals , Cell Physiological Phenomena/drug effects , Dimerization , Luminescent Proteins/adverse effects , Oligopeptides/adverse effects , Polymers/adverse effects , Polymers/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL