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










Publication year range
1.
ACS Bio Med Chem Au ; 3(5): 461-470, 2023 Oct 18.
Article in English | MEDLINE | ID: mdl-37876499

ABSTRACT

Enzymes in glucose metabolism have been subjected to numerous studies, revealing the importance of their biological roles during the cell cycle. However, due to the lack of viable experimental strategies for measuring enzymatic activities particularly in living human cells, it has been challenging to address whether their enzymatic activities and thus anticipated glucose flux are directly associated with cell cycle progression. It has remained largely elusive how human cells regulate glucose metabolism at a subcellular level to meet the metabolic demands during the cell cycle. Meanwhile, we have characterized that rate-determining enzymes in glucose metabolism are spatially organized into three different sizes of multienzyme metabolic assemblies, termed glucosomes, to regulate the glucose flux between energy metabolism and building block biosynthesis. In this work, we first determined using cell synchronization and flow cytometric techniques that enhanced green fluorescent protein-tagged phosphofructokinase is adequate as an intracellular biomarker to evaluate the state of glucose metabolism during the cell cycle. We then applied fluorescence single-cell imaging strategies and discovered that the percentage of Hs578T cells showing small-sized glucosomes is drastically changed during the cell cycle, whereas the percentage of cells with medium-sized glucosomes is significantly elevated only in the G1 phase, but the percentage of cells showing large-sized glucosomes is barely or minimally altered along the cell cycle. Should we consider our previous localization-function studies that showed assembly size-dependent metabolic roles of glucosomes, this work strongly suggests that glucosome sizes are modulated during the cell cycle to regulate glucose flux between glycolysis and building block biosynthesis. Therefore, we propose the size-specific modulation of glucosomes as a behind-the-scenes mechanism that may explain functional association of glucose metabolism with the cell cycle and, thereby, their metabolic significance in human cell biology.

2.
PLoS One ; 18(8): e0289707, 2023.
Article in English | MEDLINE | ID: mdl-37540718

ABSTRACT

We have previously demonstrated that human liver-type phosphofructokinase 1 (PFK1) recruits other rate-determining enzymes in glucose metabolism to organize multienzyme metabolic assemblies, termed glucosomes, in human cells. However, it has remained largely elusive how glucosomes are reversibly assembled and disassembled to functionally regulate glucose metabolism and thus contribute to human cell biology. We developed a high-content quantitative high-throughput screening (qHTS) assay to identify regulatory mechanisms that control PFK1-mediated glucosome assemblies from stably transfected HeLa Tet-On cells. Initial qHTS with a library of pharmacologically active compounds directed following efforts to kinase-inhibitor enriched collections. Consequently, three compounds that were known to inhibit cyclin-dependent kinase 2, ribosomal protein S6 kinase and Aurora kinase A, respectively, were identified and further validated under high-resolution fluorescence single-cell microscopy. Subsequent knockdown studies using small-hairpin RNAs further confirmed an active role of Aurora kinase A on the formation of PFK1 assemblies in HeLa cells. Importantly, all the identified protein kinases here have been investigated as key signaling nodes of one specific cascade that controls cell cycle progression in human cells. Collectively, our qHTS approaches unravel a cell cycle-associated signaling network that regulates the formation of PFK1-mediated glucosome assembly in human cells.


Subject(s)
Aurora Kinase A , High-Throughput Screening Assays , Humans , HeLa Cells , Cell Cycle , Glucose/metabolism
3.
Methods Mol Biol ; 2487: 15-26, 2022.
Article in English | MEDLINE | ID: mdl-35687227

ABSTRACT

Fluorescence live-cell imaging that has contributed to our understanding of cell biology is now at the frontline of studying quantitative biochemistry in a cell. Particularly, technological advancements of fluorescence live-cell imaging and associated strategies in recent years have allowed us to discover various subcellular macromolecular assemblies in living human cells. Here we describe how real-time dynamics of a multienzyme metabolic assembly, the "glucosome," that is responsible for regulating glucose flux at subcellular levels, has been investigated in both 2- and 3-dimensional space of single human cells. We envision that such multi-dimensional fluorescence live-cell imaging will continue to revolutionize our understanding of how intracellular metabolic pathways and their network are functionally orchestrated at single-cell levels.


Subject(s)
Glucose , Imaging, Three-Dimensional , Glucose/metabolism , Humans , Microscopy, Fluorescence/methods
4.
J Biol Chem ; 298(5): 101845, 2022 05.
Article in English | MEDLINE | ID: mdl-35307352

ABSTRACT

Enzymes within the de novo purine biosynthetic pathway spatially organize into dynamic intracellular assemblies called purinosomes. The formation of purinosomes has been correlated with growth conditions resulting in high purine demand, and therefore, the cellular advantage of complexation has been hypothesized to enhance metabolite flux through the pathway. However, the properties of this cellular structure are unclear. Here, we define the purinosome in a transient expression system as a biomolecular condensate using fluorescence microscopy. We show that purinosomes, as denoted by formylglycinamidine ribonucleotide synthase granules in purine-depleted HeLa cells, are spherical and appear to coalesce when two come into contact, all liquid-like characteristics that are consistent with previously reported condensates. We further explored the biophysical and biochemical means that drive the liquid-liquid phase separation of these structures. We found that the process of enzyme condensation into purinosomes is likely driven by the oligomeric state of the pathway enzymes and not a result of intrinsic disorder, the presence of low-complexity domains, the assistance of RNA scaffolds, or changes in intracellular pH. Finally, we demonstrate that the heat shock protein 90 KDa helps to regulate the physical properties of the condensate and maintain their liquid-like state inside HeLa cells. We show that disruption of heat shock protein 90 KDa activity induced the transformation of formylglycinamidine ribonucleotide synthase clusters into more irregularly shaped condensates, suggesting that its chaperone activity is essential for purinosomes to retain their liquid-like properties. This refined view of the purinosome offers new insight into how metabolic enzymes spatially organize into dynamic condensates within human cells.


Subject(s)
HSP90 Heat-Shock Proteins , Purines , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Molecular Chaperones/genetics , Purines/metabolism , Ribonucleotides
5.
J Biol Chem ; 298(3): 101675, 2022 03.
Article in English | MEDLINE | ID: mdl-35122791

ABSTRACT

A multienzyme metabolic assembly for human glucose metabolism, namely the glucosome, has been previously demonstrated to partition glucose flux between glycolysis and building block biosynthesis in an assembly size-dependent manner. Among three different sizes of glucosome assemblies, we have shown that large-sized glucosomes are functionally associated with the promotion of serine biosynthesis in the presence of epidermal growth factor (EGF). However, due to multifunctional roles of EGF in signaling pathways, it is unclear which EGF-mediated signaling pathways promote these large glucosome assemblies in cancer cells. In this study, we used Luminex multiplexing assays and high-content single-cell imaging to demonstrate that EGF triggers temporal activation of extracellular signal-regulated kinases 1/2 (ERK1/2) in Hs578T cells. Subsequently, we found that treatments with a pharmacological inhibitor of ERK1/2, SCH772984, or short-hairpin RNAs targeting ERK1/2 promote the dissociation of large-sized assemblies to medium-sized assemblies in Hs578T cells. In addition, our Western blot analyses revealed that EGF treatment does not increase the expression levels of enzymes that are involved in both glucose metabolism and serine biosynthesis. The observed spatial transition of glucosome assemblies between large and medium sizes appears to be mediated by the degree of dynamic partitioning of glucosome enzymes without changing their expression levels. Collectively, our study demonstrates that EGF-ERK1/2 signaling pathways play an important role in the upregulation of large-sized glucosomes in cancer cells, thus functionally governing the promotion of glycolysis-derived serine biosynthesis.


Subject(s)
Epidermal Growth Factor , Glucose , MAP Kinase Signaling System , Multienzyme Complexes , Epidermal Growth Factor/metabolism , Glucose/metabolism , Humans , Multienzyme Complexes/metabolism , Phosphorylation , Serine/metabolism , Subcellular Fractions/metabolism
6.
Methods Enzymol ; 628: 1-17, 2019.
Article in English | MEDLINE | ID: mdl-31668224

ABSTRACT

Sequential metabolic enzymes have long been hypothesized to form multienzyme metabolic complexes to regulate metabolic flux in cells. Although in vitro biochemistry has not been fruitful to support the hypothesis, advanced biophysical technologies have successfully resurrected the hypothesis with compelling experimental evidence. As biochemistry has always evolved along with technological advancement over the century (e.g., recombinant protein expression, site-directed mutagenesis, advanced spectroscopy and structural biology techniques, etc.), there has been growing interest in advanced imaging-based biophysical methods to explore enzymes inside living cells. In this work, we describe how we visualize two phase-separated biomolecular condensates of multienzyme metabolic complexes that are associated with de novo purine biosynthesis and glucose metabolism in living human cells and how imaging-based data are quantitatively analyzed to advance our knowledge of enzymes and their assemblies in living cells. Therefore, we envision that the framework we describe here would be the starting point to investigate other metabolic enzymes and their assemblies in various cell types with an unprecedented potential to comprehend enzymes and their network in native habitats.


Subject(s)
Glucose/metabolism , Metabolomics/methods , Multienzyme Complexes/metabolism , Purines/metabolism , Biosynthetic Pathways , Cell Line , Humans , Metabolome , Microscopy, Fluorescence/methods , Optical Imaging/methods , Phase Transition , Single-Cell Analysis/methods
7.
J Biol Chem ; 292(22): 9191-9203, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28424264

ABSTRACT

Sequential metabolic enzymes in glucose metabolism have long been hypothesized to form multienzyme complexes that regulate glucose flux in living cells. However, it has been challenging to directly observe these complexes and their functional roles in living systems. In this work, we have used wide-field and confocal fluorescence microscopy to investigate the spatial organization of metabolic enzymes participating in glucose metabolism in human cells. We provide compelling evidence that human liver-type phosphofructokinase 1 (PFKL), which catalyzes a bottleneck step of glycolysis, forms various sizes of cytoplasmic clusters in human cancer cells, independent of protein expression levels and of the choice of fluorescent tags. We also report that these PFKL clusters colocalize with other rate-limiting enzymes in both glycolysis and gluconeogenesis, supporting the formation of multienzyme complexes. Subsequent biophysical characterizations with fluorescence recovery after photobleaching and FRET corroborate the formation of multienzyme metabolic complexes in living cells, which appears to be controlled by post-translational acetylation on PFKL. Importantly, quantitative high-content imaging assays indicated that the direction of glucose flux between glycolysis, the pentose phosphate pathway, and serine biosynthesis seems to be spatially regulated by the multienzyme complexes in a cluster-size-dependent manner. Collectively, our results reveal a functionally relevant, multienzyme metabolic complex for glucose metabolism in living human cells.


Subject(s)
Glucose/metabolism , Glycolysis/physiology , Multienzyme Complexes/metabolism , Pentose Phosphate Pathway/physiology , Phosphofructokinase-1, Liver Type/metabolism , Fluorescence Recovery After Photobleaching , Fluorescence Resonance Energy Transfer , Glucose/genetics , HeLa Cells , Humans , Multienzyme Complexes/genetics , Phosphofructokinase-1, Liver Type/genetics
9.
Biochemistry ; 54(3): 870-80, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25540829

ABSTRACT

Enzymes in human de novo purine biosynthesis have been demonstrated to form a reversible, transient multienzyme complex, the purinosome, upon purine starvation. However, characterization of purinosomes has been limited to HeLa cells and has heavily relied on qualitative examination of their subcellular localization and reversibility under wide-field fluorescence microscopy. Quantitative approaches, which are particularly compatible with human disease-relevant cell lines, are necessary to explicitly understand the purinosome in live cells. In this work, human breast carcinoma Hs578T cells have been utilized to demonstrate the preferential utilization of the purinosome under purine-depleted conditions. In addition, we have employed a confocal microscopy-based biophysical technique, fluorescence recovery after photobleaching, to characterize kinetic properties of the purinosome in live Hs578T cells. Quantitative characterization of the diffusion coefficients of all de novo purine biosynthetic enzymes reveals the significant reduction of their mobile kinetics upon purinosome formation, the dynamic partitioning of each enzyme into the purinosome, and the existence of three intermediate species in purinosome assembly under purine starvation. We also demonstrate that the diffusion coefficient of the purine salvage enzyme, hypoxanthine phosphoribosyltransferase 1, is not sensitive to purine starvation, indicating exclusion of the salvage pathway from the purinosome. Furthermore, our biophysical characterization of nonmetabolic enzymes clarifies that purinosomes are spatiotemporally different cellular bodies from stress granules and cytoplasmic protein aggregates in both Hs578T and HeLa cells. Collectively, quantitative analyses of the purinosome in Hs578T cells led us to provide novel insights for the dynamic architecture of the purinosome assembly.


Subject(s)
Multienzyme Complexes/metabolism , Purines/biosynthesis , Biophysical Phenomena , Biosynthetic Pathways , Cell Line, Tumor , Cell Survival , Cytoplasmic Granules/metabolism , Cytosol/metabolism , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/metabolism , Humans
10.
Elife ; 2: e00592, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23638301

ABSTRACT

α-Synuclein is a presynaptic protein that is implicated in Parkinson's and other neurodegenerative diseases. Physiologically, native α-synuclein promotes presynaptic SNARE-complex assembly, but its molecular mechanism of action remains unknown. Here, we found that native α-synuclein promotes clustering of synaptic-vesicle mimics, using a single-vesicle optical microscopy system. This vesicle-clustering activity was observed for both recombinant and native α-synuclein purified from mouse brain. Clustering was dependent on specific interactions of native α-synuclein with both synaptobrevin-2/VAMP2 and anionic lipids. Out of the three familial Parkinson's disease-related point mutants of α-synuclein, only the lipid-binding deficient mutation A30P disrupted clustering, hinting at a possible loss of function phenotype for this mutant. α-Synuclein had little effect on Ca(2+)-triggered fusion in our reconstituted single-vesicle system, consistent with in vivo data. α-Synuclein may therefore lead to accumulation of synaptic vesicles at the active zone, providing a 'buffer' of synaptic vesicles, without affecting neurotransmitter release itself. DOI:http://dx.doi.org/10.7554/eLife.00592.001.


Subject(s)
Phospholipids/metabolism , Synaptic Vesicles/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , alpha-Synuclein/physiology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/isolation & purification , Animals , Brain/metabolism , Mice , Molecular Mimicry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Synaptotagmin I/genetics , Synaptotagmin I/isolation & purification
11.
Bioessays ; 35(7): 658-65, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23625805

ABSTRACT

In vitro reconstitution assays are commonly used to study biological membrane fusion. However, to date, most ensemble and single-vesicle experiments involving SNARE proteins have been performed only with lipid-mixing, but not content-mixing indicators. Through simultaneous detection of lipid and small content-mixing indicators, we found that lipid mixing often occurs seconds prior to content mixing, or without any content mixing at all, during a 50-seconds observation period, for Ca(2+) -triggered fusion with SNAREs, full-length synaptotagmin-1, and complexin. Our results illustrate the caveats of commonly used bulk lipid-mixing fusion experiments. We recommend that proteoliposome fusion experiments should always employ content-mixing indicators in addition to, or in place of, lipid-mixing indicators.


Subject(s)
Membrane Fusion/physiology , Proteolipids/metabolism , SNARE Proteins/metabolism , Calcium/metabolism , Cryoelectron Microscopy , Evaluation Studies as Topic , Lipids/physiology , Models, Molecular , Proteolipids/genetics , SNARE Proteins/genetics , Software
12.
Nat Protoc ; 8(1): 1-16, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23222454

ABSTRACT

This protocol describes a single vesicle-vesicle microscopy system to study Ca(2+)-triggered vesicle fusion. Donor vesicles contain reconstituted synaptobrevin and synaptotagmin-1. Acceptor vesicles contain reconstituted syntaxin and synaptosomal-associated protein 25 (SNAP-25), and they are tethered to a PEG-coated glass surface. Donor vesicles are mixed with the tethered acceptor vesicles and incubated for several minutes at a zero-Ca(2+) concentration, resulting in a collection of single interacting vesicle pairs. The donor vesicles also contain two spectrally distinct fluorophores that allow simultaneous monitoring of temporal changes of the content and membrane. Upon Ca(2+) injection into the sample chamber, our system therefore differentiates between hemifusion and complete fusion of interacting vesicle pairs and determines the temporal sequence of these events on a sub-100-millisecond time scale. Other factors such as complexin can be easily added. Our system is unique in that it monitors both content and lipid mixing and starts from a metastable state of interacting vesicle pairs before Ca(2+) injection.


Subject(s)
Calcium/metabolism , Membrane Fusion , Microscopy, Fluorescence/methods , Synaptic Vesicles/metabolism , Image Processing, Computer-Assisted , Lipid Metabolism , R-SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmin I/metabolism , Synaptotagmins/metabolism
13.
Elife ; 1: e00109, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23240085

ABSTRACT

The molecular underpinnings of synaptic vesicle fusion for fast neurotransmitter release are still unclear. Here, we used a single vesicle-vesicle system with reconstituted SNARE and synaptotagmin-1 proteoliposomes to decipher the temporal sequence of membrane states upon Ca(2+)-injection at 250-500 µM on a 100-ms timescale. Furthermore, detailed membrane morphologies were imaged with cryo-electron microscopy before and after Ca(2+)-injection. We discovered a heterogeneous network of immediate and delayed fusion pathways. Remarkably, all instances of Ca(2+)-triggered immediate fusion started from a membrane-membrane point-contact and proceeded to complete fusion without discernible hemifusion intermediates. In contrast, pathways that involved a stable hemifusion diaphragm only resulted in fusion after many seconds, if at all. When complexin was included, the Ca(2+)-triggered fusion network shifted towards the immediate pathway, effectively synchronizing fusion, especially at lower Ca(2+)-concentration. Synaptic proteins may have evolved to select this immediate pathway out of a heterogeneous network of possible membrane fusion pathways.DOI:http://dx.doi.org/10.7554/eLife.00109.001.


Subject(s)
Calcium/metabolism , Membrane Fusion , Proteolipids/metabolism , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmin I/metabolism , Action Potentials , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Biological Transport , Calcium/pharmacology , Gene Expression , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proteolipids/ultrastructure , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synaptic Transmission , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Synaptosomal-Associated Protein 25/genetics , Synaptotagmin I/genetics , Syntaxin 1/genetics , Syntaxin 1/metabolism , Time Factors , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/metabolism
14.
Nat Chem Biol ; 7(12): 909-15, 2011 Oct 23.
Article in English | MEDLINE | ID: mdl-22020552

ABSTRACT

G protein-coupled receptors (GPCRs) transmit exogenous signals to the nucleus, promoting a myriad of biological responses via multiple signaling pathways in both healthy and cancerous cells. However, little is known about the response of cytosolic metabolic pathways to GPCR-mediated signaling. Here we applied fluorescent live-cell imaging and label-free dynamic mass redistribution assays to study whether purine metabolism is associated with GPCR signaling. Through a library screen of GPCR ligands in conjunction with live-cell imaging of a metabolic multienzyme complex for de novo purine biosynthesis, the purinosome, we demonstrated that the activation of endogenous Gα(i)-coupled receptors correlates with purinosome assembly and disassembly in native HeLa cells. Given the implications of GPCRs in mitogenic signaling and of the purinosome in controlling metabolic flux via de novo purine biosynthesis, we hypothesize that regulation of purinosome assembly and disassembly may be one of the downstream events of mitogenic GPCR signaling in human cancer cells.


Subject(s)
Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Protein Multimerization , Purines/biosynthesis , Receptors, G-Protein-Coupled/metabolism , HeLa Cells , Humans , Ligands , Multienzyme Complexes/biosynthesis , Purines/metabolism , Receptors, G-Protein-Coupled/agonists , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 108(29): E304-13, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21705659

ABSTRACT

Understanding the molecular principles of synaptic vesicle fusion is a long-sought goal. It requires the development of a synthetic system that allows manipulations and observations not possible in vivo. Here, we report an in vitro system with reconstituted synaptic proteins that meets the long-sought goal to produce fast content release in the millisecond time regime upon Ca(2+) triggering. Our system simultaneously monitors both content and lipid exchange, and it starts from stable interacting pairs of donor and acceptor vesicles, mimicking the readily releasable pool of synaptic vesicles prior to an action potential. It differentiates between single-vesicle interaction, hemifusion, and complete fusion, the latter mimicking quantized neurotransmitter release upon exocytosis of synaptic vesicles. Prior to Ca(2+) injection, the system is in a state in which spontaneous fusion events between donor and acceptor vesicles are rare. Upon Ca(2+) injection, a rapid burst of complete fusion events emerges, followed by a biphasic decay. The present study focuses on neuronal SNAREs, the Ca(2+) sensor synaptotagmin 1, and the modulator complexin. However, other synaptic proteins could be added and their function examined. Ca(2+) triggering is cooperative, requiring the presence of synaptotagmin, whereas SNAREs alone do not produce a fast fusion burst. Manipulations of the system mimic effects observed in vivo. These results also show that neuronal SNAREs alone do not efficiently produce complete fusion, that the combination of SNAREs with synaptotagmin lowers the activation barriers to full fusion, and that complexin enhances this kinetic control.


Subject(s)
Exocytosis/physiology , Models, Biological , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , SNARE Proteins/metabolism , Synaptic Vesicles/metabolism , Synaptotagmin I/metabolism , Animals , Calcium/metabolism , Cell Line , Escherichia coli , Fluorescence , Image Processing, Computer-Assisted , In Vitro Techniques , Lipids , Nerve Tissue Proteins/isolation & purification , Rats , SNARE Proteins/isolation & purification , Spodoptera , Synaptic Vesicles/physiology , Synaptotagmin I/isolation & purification
16.
Proc Natl Acad Sci U S A ; 107(29): 12872-6, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20615962

ABSTRACT

Evidence has been presented for a metabolic multienzyme complex, the purinosome, that participates in de novo purine biosynthesis to form clusters in the cytoplasm of living cells under purine-depleted conditions. Here we identified, using fluorescent live cell imaging, that a microtubule network appears to physically control the spatial distribution of purinosomes in the cytoplasm. Application of a cell-based assay measuring the rate of de novo purine biosynthesis confirmed that the metabolic activity of purinosomes was significantly suppressed in the absence of microtubules. Collectively, we propose a microtubule-assisted mechanism for functional purinosome formation in HeLa cells.


Subject(s)
Macromolecular Substances/metabolism , Microtubules/metabolism , Purines/metabolism , Actin Cytoskeleton/metabolism , Biological Transport/drug effects , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microtubules/drug effects , Multienzyme Complexes/metabolism , Nocodazole/pharmacology , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
17.
J Biol Chem ; 285(15): 11093-9, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20157113

ABSTRACT

The reversible association and dissociation of a metabolic multi-enzyme complex participating in de novo purine biosynthesis, the purinosome, was demonstrated in live cells to respond to the levels of purine nucleotides in the culture media. We also took advantage of in vitro proteomic scale studies of cellular substrates of human protein kinases (e.g. casein kinase II (CK2) and Akt), that implicated several de novo purine biosynthetic enzymes as kinase substrates. Here, we successfully identified that purinosome formation in vivo was significantly promoted in HeLa cells by the addition of small-molecule CK2-specific inhibitors (i.e. 4,5,6,7-tetrabromo-1H-benzimidazole, 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole, tetrabromocinammic acid, 4,4',5,5',6,6'-hexahydroxydiphenic acid 2,2',6,6'-dilactone (ellagic acid) as well as by silencing the endogenous human CK2alpha catalytic subunit with small interfering RNA. However, 4,5,6,7-tetrabromobenzotriazole, another CK2-specific inhibitor, triggered the dissociation of purinosome clusters in HeLa cells. Although the mechanism by which 4,5,6,7-tetrabromobenzotriazole affects purinosome clustering is not clear, we were capable of chemically reversing purinosome formation in cells by the sequential addition of two CK2 inhibitors. Collectively, we provide compelling cellular evidence that CK2-mediated pathways reversibly regulate purinosome assembly, and thus the purinosome may be one of the ultimate targets of kinase inhibitors.


Subject(s)
Casein Kinase II/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Casein Kinase II/metabolism , Catalysis , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Gene Silencing , HeLa Cells , Humans , Microscopy, Fluorescence/methods , Models, Biological , Models, Chemical , Purines/chemistry , RNA Interference , RNA, Small Interfering/metabolism
18.
Biophys J ; 95(12): 5789-97, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18931261

ABSTRACT

The protein machinery controlling membrane fusion (or fission) has been well studied; however, the role of vesicle diffusion near membranes in these critical processes remains unclear. We experimentally and theoretically investigated the dynamics of small vesicles (approximately 50 nm in diameter) that are diffusing near supported planar bilayers acting as "target" membranes. Using total internal reflection-fluorescence correlation spectroscopy, we examined the validity of theoretical analyses of vesicle-membrane interactions. Vesicles were hindered by hydrodynamic drag as a function of their proximity to the planar bilayer. The population distributions and diffusion kinetics of the vesicles were further affected by changing the ionic strength and pH of the buffer, as well as the lipid composition of the planar membrane. Effective surface charges on neutral bilayers were also analyzed by comparing experimental and theoretical data, and we show the possibility that vesicle dynamics can be modified by surface charge redistribution of the planar bilayer. Based on these results, we hypothesize that the dynamics of small vesicles, diffusing close to biomembranes, may be spatially restricted by altering local physiological conditions (e.g., salt concentration, lipid composition, and pH), which may represent an additional mechanism for controlling fusion (or fission) dynamics.


Subject(s)
Cell Membrane/metabolism , Diffusion , Unilamellar Liposomes/metabolism , Fluorescent Dyes/metabolism , Hydrogen-Ion Concentration , Lipid Bilayers/metabolism , Osmolar Concentration , Spectrometry, Fluorescence
19.
Biochim Biophys Acta ; 1778(11): 2461-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18703013

ABSTRACT

Biomembranes are complex, heterogeneous, dynamic systems playing essential roles in numerous processes such as cell signaling and membrane trafficking. Model membranes provide simpler platforms for studying biomembrane dynamics under well-controlled environments. Here we present a modified polymer lift-off approach to introduce chemical complexity into biomimetic membranes by constructing domains of one lipid composition (here, didodecylphosphatidylcholine) that are surrounded by a different lipid composition (e.g., dipentadecylphosphatidylcholine), which we refer to as patterned backfilled samples. Fluorescence microscopy and correlation spectroscopy were used to characterize this patterning approach. We observe two types of domain populations: one with diffuse boundaries and a minor fraction with sharp edges. Lipids within the diffuse domains in patterned backfilled samples undergo anomalous diffusion, which results from nonideally mixed clusters of gel phase lipid within the fluid domains. No lateral diffusion was observed within the minor population of domains with well-defined borders. These results suggest that, while membrane patterning by a variety of approaches is useful for biophysical and biosensor applications, a thorough and systematic characterization of the resulting biomimetic membrane, and its unpatterned counterpart, is essential.


Subject(s)
Membranes, Artificial , Boron Compounds , Diffusion , Fluorescence , Lipids/chemistry , Microscopy, Atomic Force , Phosphatidylcholines/chemistry , Polymers/chemistry , Unilamellar Liposomes/chemistry
20.
Biochim Biophys Acta ; 1598(1-2): 74-9, 2002 Jul 29.
Article in English | MEDLINE | ID: mdl-12147346

ABSTRACT

The active site of an apoptotic enzyme caspase-3 was characterized by measuring the intrinsic fluorescence of two tryptophan residues. Temperature dependence of the intrinsic fluorescence, the energy homotransfer between the tryptophan residues, and the fluorescence quenching by tetrapeptide inhibitors were investigated by the fluorescence lifetime measurements. It has been observed that the fluorescence lifetimes of caspase-3 in complex with inhibitors were significantly shortened by the electron transfer process.


Subject(s)
Caspases/chemistry , Caspase 3 , Caspases/isolation & purification , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Kinetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spectrometry, Fluorescence/methods , Tryptophan/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...