Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
ACS Synth Biol ; 2(4): 173-9, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23656475

ABSTRACT

By the delivery of specific natural or engineered proteins, mammalian cells can be programmed to perform increasingly sophisticated and useful functions. Here, we introduce a set of proteins that has potential value in cell-based therapies by programming a cell to target tumor cells. First, the delivery of VSV-G (vesicular stomatitis virus glycoprotein) allowed the cell to undergo membrane fusion with adjacent cells to form syncytia (i.e., a multinucleated cell) in conditions of low pH typically occurring at a tumor site. The formation of syncytia caused the clustering of nuclei along with an integration of the microtubule network and ER. Interestingly, the formation of syncytia between cells that are dynamically blebbing, a mode of migration preferred during tumor metastasis, resulted in the loss of these morphology changes. Lastly, the codelivery of VSV-G with L57R (an engineered photoactivated caspase-7) allowed cells to undergo low pH-dependent membrane fusion followed by blue light-dependent apoptosis. In cell-based therapies, the clearance of syncytia between tumor cells might further trigger an immune response against the tumor.


Subject(s)
Apoptosis/physiology , Membrane Fusion/physiology , Animals , Apoptosis/genetics , COS Cells , Caspase 7/genetics , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Nucleus/genetics , Cell Nucleus/physiology , Chlorocebus aethiops , Endoplasmic Reticulum/genetics , Giant Cells/physiology , HEK293 Cells , HeLa Cells , Humans , Membrane Fusion/genetics , Membrane Glycoproteins/genetics , Transcellular Cell Migration/genetics , Transcellular Cell Migration/physiology , Viral Envelope Proteins/genetics
2.
Chem Biol ; 20(4): 458-60, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601633

ABSTRACT

In this issue of Chemistry & Biology, Renicke et al. report a photosensitive degron (psd) consisting of the LOV2 domain fused to a protein degradation sequence. This design enabled light-dependent protein degradation in yeast. When psd was fused to cell-cycle-dependent proteins, it controlled cell cycle by light with spatiotemporal precision.

3.
Cell Calcium ; 53(3): 231-40, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23352230

ABSTRACT

Several recent reports have demonstrated how engineered proteins can control cell motility, an important functional module for ultimately programming cells as therapeutics. We have reported two engineered proteins that regulate the blebbing cell morphology using chimeras of RhoA, a protein that regulates cytoskeletal tension. Here, we show that engineered switching of blebbing can be used to regulate cell motility. First, the analysis of morphology and motility characteristics showed that blebbing cells wobbled, or shifted, faster and less linearly than cells with a wild type morphology. Second, activating engineered protein switches that regulate cell morphology led to predictable changes in motility characteristics. Last, exogenous stimuli such as blue light, acetylcholine and VEGF-A were used to show that groups of proteins could cooperatively increase cell motility in vitro. This work demonstrates that control of RhoA can program the motility patterns of living cells and has implications in studying the relationship between cell morphology and motility.


Subject(s)
Calcium/pharmacology , Calmodulin/metabolism , Cell Movement/drug effects , rhoA GTP-Binding Protein/metabolism , Cells, Cultured , HEK293 Cells , Humans , Protein Engineering , Vascular Endothelial Growth Factor A/metabolism
4.
Protein Eng Des Sel ; 26(3): 207-13, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23223808

ABSTRACT

Inteins are protein elements that covalently reassemble proteins from two precursor fragments in a process known as protein splicing. They are commonly used to reassemble a single target protein by protein splicing, but a second target protein can potentially reassemble by intein dimerization. Here, we use the naturally occurring split DnaE intein from Nostoc punctiforme (NpuDnaE) to demonstrate the simultaneous assembly of two target proteins in several examples studied with live cell imaging: yellow fluorescent protein (YFP) with monomeric red fluorescent protein (mRFP), dominant positive mutant of RhoA GTPase with YFP and GCaMP2 Ca(2+) indicator with mRFP. These examples showed the versatility of the strategy along with some interesting attributes: first, the two target proteins are in equal stoichiometry; second, the extent of protein splicing can be reported by a fluorescent protein. In particular, the split GCaMP2 with mRFP could find applications in tissue-specific Ca(2+) imaging in transgenic organisms, where mRFP could control for motion-related intensity changes.


Subject(s)
Bacterial Proteins/metabolism , Fluorescent Dyes/metabolism , Inteins , Luminescent Proteins/metabolism , Nostoc/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Bacterial Proteins/analysis , Bacterial Proteins/genetics , Calcium/metabolism , Cell Line , Cloning, Molecular/methods , Dimerization , Fluorescence , Fluorescent Dyes/analysis , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Models, Molecular , Nostoc/genetics , Protein Engineering/methods , rhoA GTP-Binding Protein/analysis , rhoA GTP-Binding Protein/genetics , Red Fluorescent Protein
5.
Science ; 338(6113): 1424-5, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23239720
6.
Cell Calcium ; 51(1): 57-64, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22133610

ABSTRACT

While genetically encoded Ca(2+) indicators (GECIs) allow Ca(2+) imaging in model organisms, the gene expression is often under the control of a single promoter that may drive expression beyond, the cell types of interest. To enable more cell-type specific targeting, GECIs can be brought under the, control of the intersecting expression from two promoters. Here, we present the splitting and, reassembly of two representative GECIs (TN-XL and GCaMP2) mediated by the split intein from Nostoc, punctiforme (NpuDnaE). While the split TN-XL biosensor offered ratiometric Ca(2+) imaging, it had a, diminished Ca(2+) response relative to the native TN-XL biosensor. In contrast, the split GCaMP2, biosensor retained similar Ca(2+) response to the native GCaMP2. The split GCaMP2 biosensor was, further targeted to the pharyngeal muscles of Caenorhabditis elegans where Ca(2+) signals from feeding C. elegans, were imaged. Thus, we envision that increased cell-type targetability of GECIs is feasible with two, complementary promoters.


Subject(s)
Calcium/metabolism , Inteins , Molecular Probes/metabolism , Protein Splicing , Animals , Caenorhabditis elegans/metabolism , Calcium Signaling , Cell Line , DNA Polymerase III/metabolism , Humans , Indicators and Reagents , Mammals , Nostoc/metabolism , Pharynx/metabolism
7.
ACS Synth Biol ; 1(3): 75-82, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-23651071

ABSTRACT

Apoptosis is a cell death program involved in the development of multicellular organisms, immunity, and pathologies ranging from cancer to HIV/AIDS. We present an engineered protein that causes rapid apoptosis of targeted cells in monolayer culture after stimulation with blue light. Cells transfected with the protein switch L57V, a tandem fusion of the light-sensing LOV2 domain and the apoptosis-executing domain from caspase-7, rapidly undergo apoptosis within 60 min after light stimulation. Constant illumination of under 5 min or oscillating with 1 min exposure had no effect, suggesting that cells have natural tolerance to a short duration of caspase-7 activity. Furthermore, the overexpression of Bcl-2 prevented L57V-mediated apoptosis, suggesting that although caspase-7 activation is sufficient to start apoptosis, it requires mitochondrial contribution to fully commit.


Subject(s)
Apoptosis/physiology , Caspase 7/genetics , Caspase 7/metabolism , Animals , Apoptosis/radiation effects , COS Cells , Caspase 7/chemistry , Chlorocebus aethiops , HeLa Cells , Humans , Models, Biological , Photochemical Processes , Protein Engineering , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synthetic Biology , Transfection
8.
ACS Synth Biol ; 1(6): 211-20, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-23651204

ABSTRACT

Mammalian cells reprogrammed with engineered transgenes have the potential to be useful therapeutic platforms because they can support large genetic networks, can be taken from a host or patient, and perform useful functions such as migration and secretion. Successful engineering of mammalian cells will require the development of modules that can perform well-defined, reliable functions, such as directed cell migration toward a chemical or physical signal. One inherently modular cellular pathway is the Ca(2+) signaling pathway: protein modules that mobilize and respond to Ca(2+) are combined across cell types to create complexity. We have designed a chimera of Rac1, a GTPase that controls cell morphology and migration, and calmodulin (CaM), a Ca(2+)-responsive protein, to control cell migration. The Rac1-CaM chimera (named RACer) controlled lamellipodia growth in response to Ca(2+). RACer was combined with LOVS1K (a previously engineered light-sensitive Ca(2+)-mobilizing module) and cytokine receptors to create protein networks where blue light and growth factors regulated cell morphology and, thereby, cell migration. To show the generalizability of our design, we created a Cdc42-CaM chimera that controls filopodia growth in response to Ca(2+). The insights that have been gained into Ca(2+) signaling and cell migration will allow future work to combine engineered protein systems to enable reprogrammed cell sensing of relevant therapeutic targets in vivo.


Subject(s)
Cell Movement/physiology , Protein Engineering/methods , Calcium Signaling , Calmodulin/genetics , Calmodulin/physiology , HEK293 Cells , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synthetic Biology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/physiology
9.
Chem Biol ; 18(12): 1611-9, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22195563

ABSTRACT

Synthetic biosystems have been engineered that enable control of metazoan cell morphology, migration, and death. These systems possess signal specificity, but lack flexibility of input signal. To exploit the potential of Ca(2+) signaling, we designed RhoA chimeras for reversible, Ca(2+)-dependent control over RhoA morphology and migration. First, we inserted a calmodulin-binding peptide into a RhoA loop that activates or deactivates RhoA in response to Ca(2+) signals depending on the chosen peptide. Second, we localized the Ca(2+)-activated RhoA chimera to the plasma membrane, where it responded specifically to local Ca(2+) signals. Third, input control of RhoA morphology was rewired by coexpressing the Ca(2+)-activated RhoA chimera with Ca(2+)-transport proteins using acetylcholine, store-operated Ca(2+) entry, and blue light. Engineering synthetic biological systems with input versatility and tunable spatiotemporal responses motivates further application of Ca(2+) signaling in this field.


Subject(s)
Calcium Signaling , Calcium/metabolism , rhoA GTP-Binding Protein/metabolism , Acetylcholine/pharmacology , Animals , COS Cells , Calcium Signaling/drug effects , Calmodulin/metabolism , Cell Line , Cell Membrane/metabolism , Cell Movement , Chlorocebus aethiops , Humans , Light , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thapsigargin/pharmacology , rhoA GTP-Binding Protein/genetics
10.
Chem Biol ; 18(7): 880-90, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21802009

ABSTRACT

Ca(2+) signals regulate diverse physiological processes through tightly regulated fluxes varying in location, time, frequency, and amplitude. Here, we developed LOVS1K, a genetically encoded and photoactivated synthetic protein to generate local or global Ca(2+) signals. With 300 ms blue light exposure, LOVS1K translocated to Orai1, a plasma membrane Ca(2+) channel, within seconds, generating a local Ca(2+) signal on the plasma membrane, and returning to the cytoplasm after tens of seconds. With repeated photoactivation, global Ca(2+) signals in the cytoplasm were generated to modulate engineered Ca(2+)-inducible proteins. Although Orai1 is typically associated with global store-operated Ca(2+) entry, we demonstrate that Orai1 can also generate local Ca(2+) influx on the plasma membrane. Our photoactivation system can be used to generate spatially and temporally precise Ca(2+) signals and to engineer synthetic proteins that respond to specific Ca(2+) signals.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , Humans , Light , Photochemical Processes , Plasmids/genetics , Protein Transport
11.
Cell Calcium ; 48(4): 195-201, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20888042

ABSTRACT

The Rho proteins are important regulators of cell morphology, and the prototypical protein RhoA is known to regulate contraction, blebbing and bleb retraction. We have identified and experimentally confirmed that RhoA has a binding site for calmodulin, a ubiquitous transducer of the Ca(2+) second messenger. Using structural modeling, a fusion protein was designed wherein RhoA activity was controlled by Ca(2+) via calmodulin. Living cells transfected with this synthetic protein underwent Ca(2+) sensitive and calmodulin-dependent bleb retraction within minutes. Further, the modularity of Ca(2+) signaling was exploited to induce bleb retraction in response to blue light (using channelrhodopsin-2) or exogenous chemicals (with acetylcholine receptor), showing input signal versatility. The widespread use of Ca(2+) signaling in nature suggests that fully exploring its signaling potential may allow powerful applications to other synthetic biological systems.


Subject(s)
Calcium/metabolism , rhoA GTP-Binding Protein/chemistry , Animals , Binding Sites , CHO Cells , Calcium Signaling , Calmodulin/chemistry , Calmodulin/metabolism , Cell Line , Cell Shape , Cricetinae , Cricetulus , Humans , Protein Structure, Tertiary , rhoA GTP-Binding Protein/metabolism
12.
Cell Calcium ; 47(4): 369-77, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20167369

ABSTRACT

Protein translocation is used by cells to regulate protein activity in time and space. Synthetic systems have studied the effect of second messengers and exogenous chemicals on translocation, and have used translocation-based sensors to monitor unrelated pathways such as caspase activity. We have created a synthetic Ca2+-inducible protein using calmodulin binding peptides that selectively reveal nuclear localization and export signals in low Ca2+ (0 microM) and high Ca2+ (10 microM) environments, respectively. Experiments in live cells showed that our construct translocates between the nucleolus and plasma membrane with time constants of approximately 2 h. Further, a single amino acid mutation (Cys20Ala) in our construct prevented translocation to the plasma membrane and instead targeted it the mitochondria as predicted by bioinformatic analysis. Lastly, we studied the effect of cell line, Ca2+ concentration, chemical inhibitors, and cell morphology on translocation and found these conditions affected the rate, extent and direction of translocation. Our work demonstrates the feasibility of engineering Ca2+/calmodulin-mediated modulation of protein translocation and suggests that more natural analogs may exist.


Subject(s)
Calcium Signaling , Cell Nucleus/metabolism , Peptide Fragments/metabolism , Recombinant Proteins/metabolism , Active Transport, Cell Nucleus , Animals , COS Cells , Calmodulin/metabolism , Cell Membrane/metabolism , Chlorocebus aethiops , Computational Biology , Mitochondria/metabolism , Mutagenesis, Site-Directed , Mutation/genetics , Nuclear Localization Signals/genetics , Nucleolus Organizer Region/metabolism , Peptide Fragments/genetics , Protein Binding , Protein Engineering , Recombinant Proteins/genetics
13.
IEEE Trans Nanobioscience ; 9(1): 24-30, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19887331

ABSTRACT

Deciphering and designing complex biomolecular networks in the cell are the goals of systems and synthetic biology, respectively. The effects of localization, spatial heterogeneity, and molecular fluctuations in biomolecular networks are not well understood. We present a theoretical approach based on physical principles to accurately simulate biomolecular networks using the Monte Carlo method. Incorporating this theory, a computational tool named Monte Carlo biomolecular simulator (MBS) was developed, enabling studies of biomolecular kinetics with both spatial and temporal resolutions. The accuracy of MBS was verified by comparison against the classical deterministic approaches. Furthermore, the effects of localization, spatial heterogeneity, and molecular fluctuations were studied in three simulated model systems, showing their impact on the overall reaction kinetics. This work demonstrates the unique insights that can be discovered by considering the subtle effects that can be created by the spatial and temporal kinetics of biomolecular reaction networks.


Subject(s)
Computational Biology/methods , Metabolic Networks and Pathways , Models, Biological , Monte Carlo Method , Signal Transduction , Calcium Signaling , Cells/chemistry , Cells/metabolism , DNA/chemistry , DNA/metabolism , Diffusion , Proteins/chemistry , Proteins/metabolism , Stochastic Processes
14.
BMC Syst Biol ; 3: 72, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19615050

ABSTRACT

BACKGROUND: While biological systems have often been compared with digital systems, they differ by the strong effect of crosstalk between signals due to diffusivity in the medium, reaction kinetics and geometry. Memory elements have allowed the creation of autonomous digital systems and although biological systems have similar properties of autonomy, equivalent memory mechanisms remain elusive. Any such equivalent memory system, however, must silence the effect of crosstalk to maintain memory fidelity. RESULTS: Here, we present a system of enzymatic reactions that behaves like an RS latch (a simple memory element in digital systems). Using both a stochastic molecular simulator and ordinary differential equation simulator, we showed that crosstalk between two latches operating in the same spatial localization disrupts the memory fidelity of both latches. Crosstalk was reduced or silenced when simple reaction loops were replaced with multiple step or cascading reactions, showing that cascading signaling pathways are less susceptible to crosstalk. CONCLUSION: Thus, the common biological theme of cascading signaling pathways is advantageous for maintaining the fidelity of a memory latch in the presence of crosstalk. The experimental implementation of such a latch system will lead to novel approaches to cell control using synthetic proteins and will contribute to our understanding of why cells behave differently even when given the same stimulus.


Subject(s)
Computer Storage Devices , Models, Biological , Signal Transduction , Enzymes/metabolism , Proteins/metabolism , Stochastic Processes
15.
Protein Sci ; 18(6): 1252-60, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19472343

ABSTRACT

Protein localization is an important regulatory mechanism in many cell signaling pathways such as cytoskeletal organization and genetic regulation. The specific mechanism of protein localization determines the kinetics and morphological constraints of protein translocation, and thus affects the rate and extent of localization. To investigate the affect of localization kinetics and morphology on protein localization, we designed a protein localization system based on Ca(2+)-calmodulin and Src homology 3 domain binding peptides that can translocate between specific localizations in response to a Ca(2+) signal. We used a stochastic biomolecular simulator to predict that such a protein localization system will exhibit slower and less complete translocations when the association kinetics of a binding domain and peptide are reduced. As well, we predicted that increasing the diffusion resistance by manipulating the morphology of the system would similarly impair translocation speed and completeness. We then constructed a network of synthetic fusion proteins and showed that these predictions could be qualitatively confirmed in vitro. This work provides a basis for explaining the different characteristics (rate and extent) of protein transport and localization in cells as a consequence of the kinetics and morphology of the transport mechanism.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Artificial Intelligence , Calcium/metabolism , Calmodulin/chemistry , Calmodulin/metabolism , Kinetics , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology
17.
Mol Oncol ; 2(1): 81-93, 2008 Jun.
Article in English | MEDLINE | ID: mdl-19383330

ABSTRACT

GPNMB is a melanoma-associated glycoprotein that is targeted by the CR011-vcMMAE antibody-drug conjugate (ADC). Previous studies have shown that CR011-vcMMAE induces the apoptosis of GPNMB-expressing tumor cells in vitro and tumor regression in xenograft models. This ADC is currently in clinical trials for melanoma. In the present investigation, a variety of compounds were examined for their ability to increase the expression of GPNMB by cancer cells. These experiments lead to the identification of three distinct groups of compounds that increased GPNMB, some of which were shown to enhance the sensitivity of melanoma cells to CR011-vcMMAE. These data indicate that it may be possible to increase the anticancer activity of CR011-vcMMAE through pharmacological enhancement of GPNMB expression for potential therapeutic benefit.


Subject(s)
Antibodies, Monoclonal/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Gene Expression Regulation/drug effects , Glioma/drug therapy , Immunoconjugates/pharmacology , Melanoma/drug therapy , Membrane Glycoproteins/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Glioma/pathology , Humans , Immunoconjugates/therapeutic use , Melanoma/pathology
18.
Int J Cancer ; 122(6): 1400-10, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18027850

ABSTRACT

Histone deacetylase inhibitors (HDACi) represent a promising new class of anticancer agents. In the current investigation, we examined the activity of the HDACi belinostat in preclinical models of prostate cancer. In vitro proliferation assays demonstrated that belinostat potently inhibited the growth of prostate cancer cell lines (IC(50) < 1.0 microM) and was cytotoxic to these cells. Washout experiments indicated that exposure to belinostat for relatively short periods of time (<12 hr) induced suboptimal growth-inhibition and that cells exposed to 1.0 microM belinostat for 48 hr retained the capacity for regrowth following drug withdrawal, while cells exposed to 4.0 microM belinostat were irreversibly growth-inhibited. Cell cycle analyses demonstrated that belinostat induced G2/M arrest and increased the percentage of cells with subG1 DNA content, thus confirming the growth-inhibitory and cytotoxic effects of this compound. Normal prostate epithelial cells were generally less susceptible to the effects of belinostat than were prostate cancer cells. In an orthotopic prostate cancer tumor model, belinostat inhibited tumor growth by up to 43%. Moreover, metastatic lung lesions were present in 47% of vehicle-treated animals but in none of the animals administered belinostat. Consistent with its observed antimetastatic activity, belinostat inhibited the migration of prostate tumor cells and increased the production of tissue inhibitor of metalloproteinase-1 (TIMP-1) by these cells, the latter effect being replicated by siRNA knockdown of HDAC3. Belinostat also increased the expression of p21 and decreased the expression of potentially oncogenic proteins (mutant p53 and ERG). These results support the clinical evaluation of belinostat for the treatment of prostate cancer.


Subject(s)
Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors , Hydroxamic Acids/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Cell Division/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Male , Mice , Mice, Nude , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Sulfonamides
19.
Biochem J ; 409(2): 581-9, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17868033

ABSTRACT

The human HDAC (histone deacetylase) family, a well-validated anticancer target, plays a key role in the control of gene expression through regulation of transcription. While HDACs can be subdivided into three main classes, the class I, class II and class III HDACs (sirtuins), it is presently unclear whether inhibiting multiple HDACs using pan-HDAC inhibitors, or targeting specific isoforms that show aberrant levels in tumours, will prove more effective as an anticancer strategy in the clinic. To address the above issues, we have tested a number of clinically relevant HDACis (HDAC inhibitors) against a panel of rhHDAC (recombinant human HDAC) isoforms. Eight rhHDACs were expressed using a baculoviral system, and a Fluor de Lystrade mark (Biomol International) HDAC assay was optimized for each purified isoform. The potency and selectivity of ten HDACs on class I isoforms (rhHDAC1, rhHDAC2, rhHDAC3 and rhHDAC8) and class II HDAC isoforms (rhHDAC4, rhHDAC6, rhHDAC7 and rhHDAC9) was determined. MS-275 was HDAC1-selective, MGCD0103 was HDAC1- and HDAC2-selective, apicidin was HDAC2- and HDAC3-selective and valproic acid was a specific inhibitor of class I HDACs. The hydroxamic acid-derived compounds (trichostatin A, NVP-LAQ824, panobinostat, ITF2357, vorinostat and belinostat) were potent pan-HDAC inhibitors. The growth-inhibitory effect of the HDACis on HeLa cells showed that both pan-HDAC and class-I-specific inhibitors inhibited cell growth. The results also showed that both pan-HDAC and class-I-specific inhibitor treatment resulted in increased acetylation of histones, but only pan-HDAC inhibitor treatment resulted in increased tubulin acetylation, which is in agreement with their activity towards the HDAC6 isoform.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Acetylation , Cell Proliferation , Cloning, Molecular , Enzyme Inhibitors/metabolism , HeLa Cells , Histone Deacetylases/classification , Histone Deacetylases/metabolism , Humans , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Biol Psychiatry ; 61(9): 1049-61, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17081505

ABSTRACT

BACKGROUND: Formation of long-term memories is critically dependent on extracellular-regulated kinase (ERK) signaling. Activation of the ERK pathway by the sequential recruitment of mitogen-activated protein kinases is well understood. In contrast, the proteins that inactivate this pathway are not as well characterized. METHODS: Here we tested the hypothesis that the brain-specific striatal-enriched protein tyrosine phosphatase (STEP) plays a key role in neuroplasticity and fear memory formation by its ability to regulate ERK1/2 activation. RESULTS: STEP co-localizes with the ERKs within neurons of the lateral amygdala. A substrate-trapping STEP protein binds to the ERKs and prevents their nuclear translocation after glutamate stimulation in primary cell cultures. Administration of TAT-STEP into the lateral amygdala (LA) disrupts long-term potentiation (LTP) and selectively disrupts fear memory consolidation. Fear conditioning induces a biphasic activation of ERK1/2 in the LA with an initial activation within 5 minutes of training, a return to baseline levels by 15 minutes, and an increase again at 1 hour. In addition, fear conditioning results in the de novo translation of STEP. Inhibitors of ERK1/2 activation or of protein translation block the synthesis of STEP within the LA after fear conditioning. CONCLUSIONS: Together, these data imply a role for STEP in experience-dependent plasticity and suggest that STEP modulates the activation of ERK1/2 during amygdala-dependent memory formation. The regulation of emotional memory by modulating STEP activity may represent a target for the treatment of psychiatric disorders such as posttraumatic stress disorder (PTSD), panic, and anxiety disorders.


Subject(s)
Amygdala/physiology , Fear/physiology , Long-Term Potentiation/physiology , Memory/physiology , Neostriatum/physiology , Protein Tyrosine Phosphatases/physiology , Acoustic Stimulation , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Behavior, Animal/drug effects , Cells, Cultured , Conditioning, Classical/physiology , Cycloheximide/pharmacology , Electric Stimulation , Enzyme Inhibitors/pharmacology , Female , Immunohistochemistry , In Vitro Techniques , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Neostriatum/metabolism , Patch-Clamp Techniques , Point Mutation/genetics , Point Mutation/physiology , Pregnancy , Protein Synthesis Inhibitors/pharmacology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Rats , Rats, Sprague-Dawley , Translocation, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...