1
|
Biochemical Methods to Analyze the Subcellular Localization of NF-κB Proteins Using Cell Fractionation. Methods Mol Biol 2021; 2366:19-25. [PMID: 34236630 DOI: 10.1007/978-1-0716-1669-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cell fractionation is a method used to study different cellular events like protein translocation and sequestration by disrupting cells and fractionating their contents, thus allowing an enrichment of the protein of interest. Using different concentrations of sucrose or detergent buffer formulations in combination with centrifugations, the cell fractions are separated based on their density and size.
Collapse
|
2
|
Abstract
The ERK1 and ERK2 (ERK1/2) cascade is a central signaling pathway activated by a wide variety of extracellular agents that transmit the messages of G Protein Coupled Receptors (GPCRs) and Receptor Tyrosine Kinases (RTKs). Being such a central pathway, the activity of the cascade is well regulated, including by dynamic changes of the subcellular localization of components of the ERK1/2 cascade. In resting cells, ERK1/2 are localized in the cytosol due to their interactions with different anchoring proteins. After stimulation, ERK1/2 are phosphorylated by MEK1/2 on their regulatory TEY motif, which permits their detachment from the anchoring proteins. This detachment exposes ERK1/2 to additional phosphorylation on two serine residues (SPS motif) within the nuclear translocation signal (NTS) of the kinases. This additional phosphorylation allows ERK1/2 to interact with importin7, which consequently promotes their translocation to the nucleus. More studies are still required in order to better understand the mechanism and consequence of the nuclear translocation of ERK1/2. In this chapter, we describe some of the techniques used to study nuclear translocation of ERK1/2 in mammalian cells. We briefly mention methods such as digitonin permeabilization and cellular fractionation, as well as overexpression of reporter constructs. More thoroughly, we describe immunofluorescence, immunoprecipitation, and proximity ligation assay (PLA) approaches that are routinely used in our laboratory. Hopefully, the increase of knowledge based on these methods will open more opportunities for the identification of new therapeutic targets for diseases where the ERK1/2 cascade is dysregulated, such as cancer, neurodegenerative diseases, and diabetes.
Collapse
Affiliation(s)
- Denise A Berti
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Rony Seger
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
3
|
Luo Y, Hara T, Ishido Y, Yoshihara A, Oda K, Makino M, Ishii N, Hiroi N, Suzuki K. Rapid preparation of high-purity nuclear proteins from a small number of cultured cells for use in electrophoretic mobility shift assays. BMC Immunol 2014; 15:586. [PMID: 25527077 PMCID: PMC4339431 DOI: 10.1186/s12865-014-0062-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 12/12/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Highly purified nuclear protein is required when using an electrophoretic mobility shift assay (EMSA) to study transcription factors, e.g. nuclear factor-κB (NF-κB), a major transcription factor that regulates both innate and adaptive immune responses following infection. Although many protocols have been developed for nuclear protein extraction, they are not necessarily optimized for use in EMSA, often require a large number of cells and long processing times, and do not always result in complete separation of the nuclear and cytoplasmic fractions. RESULTS We have developed a simple, rapid and cost-effective method to prepare highly purified nuclear proteins from a small number of both suspended and adherent cultured cells that yields nuclear proteins comparable to those prepared by a standard large-scale method. The efficiency of the method was demonstrated by using EMSA to show the successful detection, in multilple concurrent samples, of NF-κB activation upon tetradecanoyl phorbol acetate (TPA) stimulation. CONCLUSIONS This method requires only a small number of cells and no specialized equipment. The steps have been simplified, resulting in a short processing time, which allows researchers to process multiple samples simultaneously and quickly. This method is especially optimized for use in EMSA, and may be useful for other applications that include proteomic analysis.
Collapse
Affiliation(s)
- Yuqian Luo
- Laboratory of Molecular Diagnostics, Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama-shi, 189-0002, Tokyo, Japan.
| | - Takeshi Hara
- Laboratory of Molecular Diagnostics, Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama-shi, 189-0002, Tokyo, Japan.
| | - Yuko Ishido
- Laboratory of Molecular Diagnostics, Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama-shi, 189-0002, Tokyo, Japan.
| | - Aya Yoshihara
- Laboratory of Molecular Diagnostics, Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama-shi, 189-0002, Tokyo, Japan.
| | - Kenzaburo Oda
- Laboratory of Molecular Diagnostics, Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama-shi, 189-0002, Tokyo, Japan.
| | - Masahiko Makino
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama-shi, 189-0002, Tokyo, Japan.
| | - Norihisa Ishii
- Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama-shi, 189-0002, Tokyo, Japan.
| | - Naoki Hiroi
- Department of Education Planning and Development, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan.
| | - Koichi Suzuki
- Laboratory of Molecular Diagnostics, Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama-shi, 189-0002, Tokyo, Japan.
| |
Collapse
|
4
|
Odeh AM, Craik JD, Ezzeddine R, Tovmasyan A, Batinic-Haberle I, Benov LT. Targeting mitochondria by Zn(II)N-alkylpyridylporphyrins: the impact of compound sub-mitochondrial partition on cell respiration and overall photodynamic efficacy. PLoS One 2014; 9:e108238. [PMID: 25250732 PMCID: PMC4177117 DOI: 10.1371/journal.pone.0108238] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/19/2014] [Indexed: 01/12/2023] Open
Abstract
Mitochondria play a key role in aerobic ATP production and redox control. They harness crucial metabolic pathways and control cell death mechanisms, properties that make these organelles essential for survival of most eukaryotic cells. Cancer cells have altered cell death pathways and typically show a shift towards anaerobic glycolysis for energy production, factors which point to mitochondria as potential culprits in cancer development. Targeting mitochondria is an attractive approach to tumor control, but design of pharmaceutical agents based on rational approaches is still not well established. The aim of this study was to investigate which structural features of specially designed Zn(II)N-alkylpyridylporphyrins would direct them to mitochondria and to particular mitochondrial targets. Since Zn(II)N-alkylpyridylporphyrins can act as highly efficient photosensitizers, their localization can be confirmed by photodamage to particular mitochondrial components. Using cultured LS174T adenocarcinoma cells, we found that subcellular distribution of Zn-porphyrins is directed by the nature of the substituents attached to the meso pyridyl nitrogens at the porphyrin ring. Increasing the length of the aliphatic chain from one carbon (methyl) to six carbons (hexyl) increased mitochondrial uptake of the compounds. Such modifications also affected sub-mitochondrial distribution of the Zn-porphyrins. The amphiphilic hexyl derivative (ZnTnHex-2-PyP) localized in the vicinity of cytochrome c oxidase complex, causing its inactivation during illumination. Photoinactivation of critical cellular targets explains the superior efficiency of the hexyl derivative in causing mitochondrial photodamage, and suppressing cellular respiration and survival. Design of potent photosensitizers and redox-active scavengers of free radicals should take into consideration not only selective organelle uptake and localization, but also selective targeting of critical macromolecular structures.
Collapse
Affiliation(s)
- Ahmad M. Odeh
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - James D. Craik
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Rima Ezzeddine
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Ludmil T. Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
- * E-mail:
| |
Collapse
|
5
|
Buşu C, Li W, Caldito G, Aw TY. Inhibition of glutathione synthesis in brain endothelial cells lengthens S-phase transit time in the cell cycle: Implications for proliferation in recovery from oxidative stress and endothelial cell damage. Redox Biol 2013; 1:131-139. [PMID: 23682351 PMCID: PMC3652486 DOI: 10.1016/j.redox.2013.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress-induced decrease in tissue or systemic glutathione (GSH) and damage to the vascular endothelium of the blood-brain barrier such as occurs in diabetes or stroke will have important implications for brain homeostasis. Endothelial proliferation or repair is crucial to preserving barrier function. Cell proliferation has been associated with increased intracellular GSH, but the kinetic and distribution of GSH during cell cycle is poorly understood. Here, we determined the influence of cellular GSH status on the early dynamics of nuclear-to-cytosol (N-to-C) GSH distribution (6-h interval) during proliferation in a human brain microvascular endothelial cell line (IHEC). Control IHECs exhibited two peak S-phases of the cell cycle at 48 and 60 h post seeding that temporally corresponded to peak nuclear GSH levels and expression of cdk1, the S-to-G2-to-M checkpoint controller, suggesting a link between cell cycle progression and nuclear GSH. Sustained inhibition of GSH synthesis delayed S-to-G2/M cell transition; cell arrest in the S-phase was correlated with decreased total nuclear GSH and increased nuclear expressions of chk2/phospho-chk2 and GADPH. The temporal correspondence of nuclear chk2 activation and GAPDH expression with S-phase prolongation is consistent with enhanced DNA damage response and extended time for DNA repair. Strikingly, when GSH synthesis was restored, cell transit time through S-phase remained delayed. Significantly, total nuclear GSH remained depressed, indicating a time lag between restored cellular GSH synthetic capacity and recovery of the nuclear GSH status. Interestingly, despite a delay in cell cycle recovery, nuclear expressions of chk2/phospho-chk2 and GAPDH resembled those of control cells. This means that restoration of nuclear DNA integrity preceded normalization of the cell cycle. The current results provide important insights into GSH control of endothelial proliferation with implications for cell repair or wound healing in recovery post-oxidative damage.
Collapse
Affiliation(s)
- Carmina Buşu
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center- Shreveport, 1501 Kings Highway, Shreveport, Louisiana, 71130, USA
| | | | | | | |
Collapse
|
6
|
A simple protocol for the subcellular fractionation of skeletal muscle cells and tissue. BMC Res Notes 2012; 5:513. [PMID: 22994964 PMCID: PMC3508861 DOI: 10.1186/1756-0500-5-513] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 08/31/2012] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND We describe a method for subcellular fractionation of mouse skeletal muscle, myoblast and myotubes to obtain relatively pure fractions of nuclear, cytosolic and mitochondrial compartments. Fractionation allows the analysis of a protein of interest (or other cellular component) based on its subcellular compartmental distribution and can also generate molecular information about the state of a cell and/or tissue and how the distribution of a protein may differ between different cellular compartments, tissues or cell types, in response to treatments or ageing. FINDINGS The described method was specifically developed for skeletal muscle and proliferating/differentiated muscle cells. The purity of the different fractions, representing the cytoplasmic, mitochondrial and nuclear subcellular compartments was validated by western blot analysis of "house-keeper" marker proteins specific for each cellular compartment. CONCLUSION This low cost method allowed the mitochondrial, cytoplasmic and nuclear subcellular compartments from the same starting muscle samples to be rapidly and simultaneously isolated with good purity and without the use of an ultracentrifuge. This method permits samples to be frozen at -80°C for future analysis and/or additional processing at a later date.
Collapse
|
7
|
Wild P, de Oliveira AP, Sonda S, Schraner EM, Ackermann M, Tobler K. The herpes simplex virus 1 U(S)3 regulates phospholipid synthesis. Virology 2012; 432:353-60. [PMID: 22789738 DOI: 10.1016/j.virol.2012.06.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 05/07/2012] [Accepted: 06/12/2012] [Indexed: 10/28/2022]
Abstract
Herpes simplex virus type 1 capsids bud at nuclear and Golgi membranes for envelopment by phospholipid bilayers. In the absence of U(S)3, nuclear membranes form multiple folds harboring virions that suggests disturbance in membrane turnover. Therefore, we investigated phospholipid metabolism in cells infected with the U(S)3 deletion mutant R7041(ΔU(S)3), and quantified membranes involved in viral envelopment. We report that (i) [(3)H]-choline incorporation into nuclear membranes and cytoplasmic membranes was enhanced peaking at 12 or 20 h post inoculation with wild type HSV-1 and R7041(ΔU(S)3), respectively, (ii) the surface area of nuclear membranes increased until 24 h of R7041(ΔU(S)3) infection forming folds that equaled ~45% of the nuclear surface, (iii) the surface area of viral envelopes between nuclear membranes equaled ~2400 R7041(ΔU(S)3) virions per cell, and (iv) during R7041(ΔU(S)3) infection, the Golgi complex expanded dramatically. The data indicate that U(S)3 plays a significant role in regulation of membrane biosynthesis.
Collapse
Affiliation(s)
- Peter Wild
- Institute of Veterinary Anatomy, University of Zürich, Switzerland.
| | | | | | | | | | | |
Collapse
|
8
|
Zhai D, Gomez-Mejiba SE, Gimenez MS, Deter ding LJ, Tomer KB, Mason RP, Ashby MT, Ramirez DC. Free radical-operated proteotoxic stress in macrophages primed with lipopolysaccharide. Free Radic Biol Med 2012; 53:172-81. [PMID: 22580125 PMCID: PMC4078023 DOI: 10.1016/j.freeradbiomed.2012.04.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 04/17/2012] [Accepted: 04/21/2012] [Indexed: 01/13/2023]
Abstract
The free-radical-operated mechanism of death of activated macrophages at sites of inflammation is unclear, but it is important to define it in order to find targets to prevent further tissue dysfunction. A well-defined model of macrophage activation at sites of inflammation is the treatment of RAW 264.7 cells with lipopolysaccharide (LPS), with the resulting production of reactive oxygen species (ROS). ROS and other free radicals can be trapped with the nitrone spin trap 5,5-dimethyl-1-pyrroline N-oxide (DMPO), a cell-permeable probe with antioxidant properties, which thus interferes with free-radical-operated oxidation processes. Here we have used immuno-spin trapping to investigate the role of free-radical-operated protein oxidation in LPS-induced cytotoxicity in macrophages. Treatment of RAW 264.7 cells with LPS resulted in increased ROS production, oxidation of proteins, cell morphological changes and cytotoxicity. DMPO was found to trap protein radicals to form protein-DMPO nitrone adducts, to reduce protein carbonyls, and to block LPS-induced cell death. N-Acetylcysteine (a source of reduced glutathione), diphenyleneiodonium (an inhibitor of NADPH oxidase), and 2,2'-dipyridyl (a chelator of Fe(2+)) prevented LPS-induced oxidative stress and cell death and reduced DMPO-nitrone adduct formation, suggesting a critical role of ROS, metals, and protein-radical formation in LPS-induced cell cytotoxicity. We also determined the subcellular localization of protein-DMPO nitrone adducts and identified some candidate proteins for DMPO attachment by LC-MS/MS. The LC-MS/MS data are consistent with glyceraldehyde-3-phosphate dehydrogenase, one of the most abundant, sensitive, and ubiquitous proteins in the cell, becoming labeled with DMPO when the cell is primed with LPS. This information will help find strategies to treat inflammation-associated tissue dysfunction by focusing on preventing free radical-operated proteotoxic stress and death of macrophages.
Collapse
Affiliation(s)
- Dili Zhai
- Department of Medicine, Gastroenterology Section, University of Chicago, Chicago, IL 60637, USA
| | - Sandra E. Gomez-Mejiba
- Laboratory of Experimental and Therapeutic Medicine, Instituto Multidisciplinario de Investigaciones Biológicas-San Luis (IMIBIO-SL)-Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), San Luis, San Luis 5700, Argentina
| | - Maria S. Gimenez
- Laboratory of Experimental and Therapeutic Medicine, Instituto Multidisciplinario de Investigaciones Biológicas-San Luis (IMIBIO-SL)-Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), San Luis, San Luis 5700, Argentina
| | - Leesa J. Deter ding
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Kenneth B. Tomer
- Laboratory of Structural Biology, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Ronald P. Mason
- Laboratory of Pharmacology and Chemistry, NIEHS, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Michael T. Ashby
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA
| | - Dario C. Ramirez
- Laboratory of Experimental and Therapeutic Medicine, Instituto Multidisciplinario de Investigaciones Biológicas-San Luis (IMIBIO-SL)-Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), San Luis, San Luis 5700, Argentina
- Department of Biochemistry and Biological Sciences, School of Chemistry, Biochemistry and Pharmacy, National University of San Luis, San Luis, San Luis 5700, Argentina
| |
Collapse
|
9
|
Sutter E, de Oliveira AP, Tobler K, Schraner EM, Sonda S, Kaech A, Lucas MS, Ackermann M, Wild P. Herpes simplex virus 1 induces de novo phospholipid synthesis. Virology 2012; 429:124-35. [PMID: 22560864 DOI: 10.1016/j.virol.2012.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 03/02/2012] [Accepted: 04/10/2012] [Indexed: 11/25/2022]
Abstract
Herpes simplex virus type 1 capsids bud at nuclear membranes and Golgi membranes acquiring an envelope composed of phospholipids. Hence, we measured incorporation of phospholipid precursors into these membranes, and quantified changes in size of cellular compartments by morphometric analysis. Incorporation of [³H]-choline into both nuclear and cytoplasmic membranes was significantly enhanced upon infection. [³H]-choline was also part of isolated virions even grown in the presence of brefeldin A. Nuclei expanded early in infection. The Golgi complex and vacuoles increased substantially whereas the endoplasmic reticulum enlarged only temporarily. The data suggest that HSV-1 stimulates phospholipid synthesis, and that de novo synthesized phospholipids are inserted into nuclear and cytoplasmic membranes to i) maintain membrane integrity in the course of nuclear and cellular expansion, ii) to supply membrane constituents for envelopment of capsids by budding at nuclear membranes and Golgi membranes, and iii) to provide membranes for formation of transport vacuoles.
Collapse
Affiliation(s)
- Esther Sutter
- Electron Microscopy, Institute of Veterinary Anatomy, University of Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wang Y, Sun DQ, Liu DG. Tumor suppression by RNA from C/EBPβ 3'UTR through the inhibition of protein kinase Cε activity. PLoS One 2011; 6:e16543. [PMID: 21283634 PMCID: PMC3026020 DOI: 10.1371/journal.pone.0016543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 01/03/2011] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Since the end of last century, RNAs from the 3'untranslated region (3'UTR) of several eukaryotic mRNAs have been found to exert tumor suppression activity when introduced into malignant cells independent of their whole mRNAs. In this study, we sought to determine the molecular mechanism of the tumor suppression activity of a short RNA from 3'UTR of C/EBPβ mRΝΑ (C/EBPβ 3'UTR RNA) in human hepatocarcinoma cells SMMC-7721. METHODOLOGY/PRINCIPAL FINDINGS By using Western blotting, immunocytochemistry, molecular beacon, confocal microscopy, protein kinase inhibitors and in vitro kinase assays, we found that, in the C/EBPβ 3'UTR-transfectant cells of SMMC-7721, the overexpressed C/EBPβ 3'UTR RNA induced reorganization of keratin 18 by binding to this keratin; that the C/EBPβ 3'UTR RNA also reduced phosphorylation and expression of keratin 18; and that the enzyme responsible for phosphorylating keratin 18 is protein kinase Cε. We then found that the C/EBPβ 3'UTR RNA directly inhibited the phosphorylating activity of protein kinase Cε; and that C/EBPβ 3'UTR RNA specifically bound with the protein kinase Cε-keratin 18 conjugate. CONCLUSION/SIGNIFICANCE Together, these facts suggest that the tumor suppression in SMMC-7721 by C/EBPβ 3'UTR RNA is due to the inhibition of protein kinase Cε activity through direct physical interaction between C/EBPβ 3'UTR RNA and protein kinase Cε. These facts indicate that the 3'UTR of some eukaryotic mRNAs may function as regulators for genes other than their own.
Collapse
Affiliation(s)
- Ying Wang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Da-Quan Sun
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ding-Gan Liu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
11
|
Suzuki K, Bose P, Leong-Quong RYY, Fujita DJ, Riabowol K. REAP: A two minute cell fractionation method. BMC Res Notes 2010; 3:294. [PMID: 21067583 PMCID: PMC2993727 DOI: 10.1186/1756-0500-3-294] [Citation(s) in RCA: 350] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 11/10/2010] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The translocation or shuttling of proteins between the nucleus and cytoplasm (nucleocytoplasmic transport [NCPT]) is often a rapid event following stimulation with growth factors or in response to stress or other experimental manipulations. Commonly used methods to separate nuclei from cytoplasm employ lengthy steps such as density gradient centrifugation which exposes cells to non-physiological hyperosmotic conditions for extended time periods resulting in varying degrees of leakage between the nucleus and cytoplasm. To help maintain and quantify nuclear:cytoplasmic ratios of proteins, agents such as leptomycin B have been employed to be able to better analyze NCPT by inhibiting nuclear export. To track NCPT in the absence of these experimental manipulations that could introduce unknown artefacts, we have developed a rapid method that appears to produce pure nuclear and cytoplasmic fractions, suitable for obtaining accurate estimates of the nuclear:cytoplasmic ratios of proteins known to undergo NCPT. FINDINGS We have developed a Rapid, Efficient And Practical (REAP) method for subcellular fractionation of primary and transformed human cells in culture. The REAP method is a two minute non-ionic detergent-based purification technique requiring only a table top centrifuge, micro-pipette and micro-centrifuge tubes. This inexpensive method has proven to efficiently separate nuclear from cytoplasmic proteins as estimated by no detectible cross-contamination of the nucleoporin and lamin A nuclear markers or the pyruvate kinase and tubulin cytoplasmic markers. REAP fractions also mirrored TNFα induced NF-κB NCPT observed in parallel by indirect immunofluorescence. CONCLUSIONS This method drastically reduces the time needed for subcellular fractionation, eliminates detectable protein degradation and maintains protein interactions. The simplicity, brevity and efficiency of this procedure allows for tracking ephemeral changes in subcellular relocalization of proteins while maintaining protein integrity and protein complex interactions.
Collapse
Affiliation(s)
- Keiko Suzuki
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, Canada
| | - Pinaki Bose
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, Canada
| | - Rebecca YY Leong-Quong
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, Canada
| | - Donald J Fujita
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, Canada
| | - Karl Riabowol
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, Canada
- Department of Oncology, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, Canada
| |
Collapse
|
12
|
Zhao J, Meng F, Zhu X, Han K, Liu S, Li G. Electrochemistry of Mitochondria: A New Way to Understand Their Structure and Function. ELECTROANAL 2008. [DOI: 10.1002/elan.200704205] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
13
|
Braun JS, Hoffmann O, Schickhaus M, Freyer D, Dagand E, Bermpohl D, Mitchell TJ, Bechmann I, Weber JR. Pneumolysin causes neuronal cell death through mitochondrial damage. Infect Immun 2007; 75:4245-54. [PMID: 17562768 PMCID: PMC1951198 DOI: 10.1128/iai.00031-07] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial toxins such as pneumolysin are key mediators of cytotoxicity in infections. Pneumolysin is a pore-forming toxin released by Streptococcus pneumoniae, the major cause of bacterial meningitis. We found that pneumolysin is the pneumococcal factor that accounts for the cell death pathways induced by live bacteria in primary neurons. The pore-forming activity of pneumolysin is essential for the induction of mitochondrial damage and apoptosis. Pneumolysin colocalized with mitochondrial membranes, altered the mitochondrial membrane potential, and caused the release of apoptosis-inducing factor and cell death. Pneumolysin induced neuronal apoptosis without activating caspase-1, -3, or -8. Wild-type pneumococci also induced apoptosis without activation of caspase-3, whereas pneumolysin-negative pneumococci activated caspase-3 through the release of bacterial hydrogen peroxide. Pneumolysin caused upregulation of X-chromosome-linked inhibitor of apoptosis protein and inhibited staurosporine-induced caspase activation, suggesting the presence of actively suppressive mechanisms on caspases. In conclusion, our results indicate additional functions of pneumolysin as a mitochondrial toxin and as a determinant of caspase-independent apoptosis. Considering this, blocking of pneumolysin may be a promising cytoprotective strategy in pneumococcal meningitis and other infections.
Collapse
Affiliation(s)
- Johann S Braun
- Department of Neurology, Charité Universitaetsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Eder AR, Arriaga EA. Capillary electrophoresis monitors enhancement in subcellular reactive oxygen species production upon treatment with doxorubicin. Chem Res Toxicol 2006; 19:1151-9. [PMID: 16978019 PMCID: PMC2626132 DOI: 10.1021/tx060083i] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study investigated the role of doxorubicin (DOX) accumulation in reactive oxygen species (ROS) production detected in individually electrophoresed organelles, including mitochondria, acidic organelles, and peroxisomes. While bulk measurements of ROS production in cells and organelles are not capable of discriminating between the effects of preparative procedures on measured ROS production, capillary electrophoresis with dual laser-induced detection of individual organelles demonstrated a difference in the measured ROS production as a result of various preparative procedures. Using this technique, the three different types of detected organelles (i) produce ROS and do not have detectable levels of DOX, (ii) contain detectable DOX but do not produce ROS, or (iii) produce ROS and accumulate DOX. The third type displays two subpopulations of organelles, one of which demonstrated a direct relationship between DOX uptake and subsequent ROS production, corresponding most likely to mitochondria, and a second one with low DOX uptake but large variation in ROS production, corresponding most likely to acidic organelles.
Collapse
Affiliation(s)
| | - Edgar A Arriaga
- *Author to whom correspondence should be addressed. Tel.: +1 612 624 8024; fax +1, 612 626 7541. E-mail address:
| |
Collapse
|
15
|
Ramirez DC, Mejiba SEG, Mason RP. Immuno–spin trapping of DNA radicals. Nat Methods 2006; 3:123-7. [PMID: 16432522 DOI: 10.1038/nmeth852] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Accepted: 12/23/2005] [Indexed: 11/09/2022]
Abstract
The detection of DNA radicals by immuno-spin trapping (IST) is based on the trapping of radicals with 5,5-dimethyl-1-pyrroline N-oxide (DMPO), forming stable nitrone adducts that are then detected using an anti-DMPO serum. DNA radicals are very reactive species, and because they are paramagnetic they have previously been detected only by electron spin resonance (ESR) with or without spin trapping, which is not available in most bioresearch laboratories. IST combines the simplicity, reliability, specificity and sensitivity of spin trapping with heterogeneous immunoassays for the detection of DNA radicals, and complements existing methods for the measurement of oxidatively generated DNA damage. Here we have used IST to demonstrate that DMPO traps Cu(II)-H(2)O(2)-induced DNA radicals in situ and in real time, forming DMPO-DNA nitrone adducts, but preventing both 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG) formation and DNA fragmentation. We also applied IST to detect DNA radicals in rat hepatocytes exposed to Cu(II) and H(2)O(2) under nonlethal conditions.
Collapse
Affiliation(s)
- Dario C Ramirez
- Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, North Carolina 27709, USA.
| | | | | |
Collapse
|
16
|
Liu DG, Sun L. Direct isolation of specific RNA-interacting proteins using a novel affinity medium. Nucleic Acids Res 2005; 33:e132. [PMID: 16126844 PMCID: PMC1192835 DOI: 10.1093/nar/gni133] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Isolation of proteins that specifically interact with a given RNA or RNA regulation element is essential for studies on the molecular mechanisms of gene expression. Here, a novel method for direct isolation of such interacting proteins is described. It uses an affinity medium that consists of an interacting RNA with an artificially added ‘tail’, which is annealed to one end of a DNA ‘arm’, the other end of which is fixed covalently on the surface of aminosilanized glass powder. Thus the RNA itself is fully suspending, facilitating its interactions with proteins in its natural conformation. The proteins bound on the interacting RNA are eluted and subjected to SDS–PAGE, and the Coomassie-stained protein bands are cut and subjected to mass spectrometry (MS) analysis. Using this method, three proteins specifically interacting with the C/EBPβ 3′-untranslated region (3′-UTR) RNA were isolated and identified. This method is simple and convenient, and the DNA-glass powder medium can be used repeatedly.
Collapse
Affiliation(s)
- Ding-Gan Liu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | | |
Collapse
|
17
|
McLaughlin LM, Demple B. Nitric Oxide–Induced Apoptosis in Lymphoblastoid and Fibroblast Cells Dependent on the Phosphorylation and Activation of p53. Cancer Res 2005; 65:6097-104. [PMID: 16024610 DOI: 10.1158/0008-5472.can-04-4254] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
When nitric oxide (NO) is produced at micromolar concentrations, as during inflammation, exposure to surrounding cells is potentially cytotoxic. The NO-dependent signaling pathways that initiate cell death are thought to involve the tumor suppressor protein p53, but the degree to which this factor contributes to NO-induced cell death is less clear. Various reports either confirm or negate a role for p53 depending on the cell type and NO donor used. In this study, we have used several pairs of cell lines whose only differences are the presence or absence of p53, and we have treated these cell lines with the same NO donor, spermineNONOate (SPER/NO). Treatment with SPER/NO induced such apoptotic markers as DNA fragmentation, nuclear condensation, poly(ADP-ribose) polymerase cleavage, cytochrome c release, and Annexin V staining. p53 was required for at least 50% of SPER/NO-induced apoptotic cell death in human lymphoblastoid cells and for almost all in primary and E1A-tranformed mouse embryonic fibroblasts, which highlights the possible importance of DNA damage for apoptotic signaling in fibroblasts. In contrast, p53 did not play a significant role in NO-induced necrosis. NO treatment also induced the phosphorylation of p53 at Ser15; pretreatment with phosphoinositide-3 kinase (PI3K) family inhibitors, wortmannin, LY294002, and caffeine, blocked such phosphorylation, but the p38 mitogen-activated protein kinase inhibitor, SB203580, did not. Pretreatment with the PI3K family inhibitors also led to a switch from NO-induced apoptosis to necrosis, which implicates a PI3K-related kinase such as ataxia telangiectasia mutated (ATM) or ATR (ATM and Rad3 related) in p53-dependent NO-induced apoptosis.
Collapse
Affiliation(s)
- Laura M McLaughlin
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts 02115-6021, USA
| | | |
Collapse
|
18
|
Caldas H, Jiang Y, Holloway MP, Fangusaro J, Mahotka C, Conway EM, Altura RA. Survivin splice variants regulate the balance between proliferation and cell death. Oncogene 2005; 24:1994-2007. [PMID: 15688031 DOI: 10.1038/sj.onc.1208350] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Survivin is an inhibitor of apoptosis protein that also plays critical roles in regulating the cell cycle and mitosis. Its prominent expression in essentially all human malignancies, and low or absent expression in most normal tissues, suggests that it would be an ideal target for cancer-directed therapy. Impeding development of safe and effective survivin antagonists for clinical use is a lack of understanding of the molecular mechanisms by which survivin differentially affects apoptosis and cell division, in normal and malignant cells. We show that the diverse functional roles of survivin can be explained, in part, by its heterodimerization with survivin splice variants in tumor cells. Survivin and survivin-DeltaEx3 interact within the mitochondria where they may inhibit mitochondrial-dependent apoptosis. If the expression of all survivin forms is eliminated by siRNA transfections, cells undergo both apoptosis and defective cell division. Overall, we provide new insights suggesting that targeting specific survivin isoforms, rather than survivin alone, may selectively and effectively destroy tumor cells. These findings are likely to have a significant impact in the design of biologic agents for clinical therapy.
Collapse
Affiliation(s)
- Hugo Caldas
- Center for Childhood Cancer, Columbus Children's Research Institute, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Srinivas KS, Chandrasekar G, Srivastava R, Puvanakrishnan R. A novel protocol for the subcellular fractionation of C3A hepatoma cells using sucrose density gradient centrifugation. ACTA ACUST UNITED AC 2005; 60:23-7. [PMID: 15236907 DOI: 10.1016/j.jbbm.2004.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2003] [Revised: 04/07/2004] [Accepted: 04/17/2004] [Indexed: 11/19/2022]
Abstract
In this paper, we describe a method to obtain a relatively pure mitochondrial and microsomal fractions by subcellular fractionation of human hepatoma cell line C3A using sucrose as the hypoosmotic medium. The cells were subjected to osmotic stress with sucrose and homogenized. Osmolarity was then restored to the cells and the organelles were separated by density gradient centrifugation. The protein profiles were examined by SDS-PAGE and the purity was analysed by marker enzymes and Western blotting. Our results indicate a good separation of mitochondrial and microsomal fractions from human hepatoma C3A cells.
Collapse
Affiliation(s)
- Kitambi Satish Srinivas
- Department of Biotechnology, Central Leather Research Institute, Adyar, Chennai-600 020, India
| | | | | | | |
Collapse
|
20
|
Gunasekera N, Olson KJ, Musier-Forsyth K, Arriaga EA. Capillary Electrophoretic Separation of Nuclei Released from Single Cells. Anal Chem 2004; 76:655-62. [PMID: 14750860 DOI: 10.1021/ac034916a] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report here the first capillary electrophoresis analysis of intact nuclei released on-column from single cells. Expression of the nuclear-targeted protein nuDsRed2 and the plasma membrane-bound farnesylated enhanced green fluorescent protein in cultured human DeltaH2-1 cells allowed fluorescent monitoring of the fate of these subcellular compartments upon injection of a single cell into the separation capillary. On-column treatment with digitonin allowed for the separation of the plasma membrane from the nucleus as indicated by their selective laser-induced fluorescence detection in two separate spectral regions. The data suggest that less than 0.1% of the plasma membrane remains bound to individually detected nuclei. In digitonin-treated cells, the electropherograms consisted of a prominent fluorescent peak attributed to nuDsRed2 localized to the nucleus and a collection of weakly fluorescent events (barely distinguishable from scattering) that seem to indicate additional localization of this protein to other subcellular regions. Taken together, this report points to the feasibility of studying intact organelles released from a single mammalian cell by capillary electrophoresis, which is a prerequisite to understanding the relevance of subcellular heterogeneity in biological systems.
Collapse
Affiliation(s)
- Nilhan Gunasekera
- Department of Chemistry, University of Minnesota, 207 Pleasant Street S.E., Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
21
|
Lefebvre T, Ferreira S, Dupont-Wallois L, Bussière T, Dupire MJ, Delacourte A, Michalski JC, Caillet-Boudin ML. Evidence of a balance between phosphorylation and O-GlcNAc glycosylation of Tau proteins--a role in nuclear localization. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1619:167-76. [PMID: 12527113 DOI: 10.1016/s0304-4165(02)00477-4] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Both phosphorylation and O-GlcNAc glycosylation posttranslationally modify microtubule-associated Tau proteins. Whereas the hyperphosphorylation of these proteins that occurs in Alzheimer's disease is well characterized, little is known about the O-GlcNAc glycosylation. The present study demonstrates that a balance exists between phosphorylation and O-GlcNAc glycosylation of Tau proteins, and furthermore that a dysfunction of this balance correlates with reduced nuclear localization. The affinity of Tau proteins for WGA lectin, together with evidence from [3H]-galactose transfer and analysis of beta-eliminated products, demonstrated the presence of O-GlcNAc residues on both cytosolic and nuclear Tau proteins. In addition, our data indicated the existence of a balance between phosphorylation and O-GlcNAc glycosylation events. Indeed, as demonstrated by 2D-electrophoresis and Western blotting, O-GlcNAc residues were mainly located on the less phosphorylated Tau 441 variants, whereas the more phosphorylated forms were devoid of O-GlcNAc residues. Furthermore, the Tau protein hyperphosphorylation induced by cellular okadaic acid treatment was correlated with reduced incorporation of O-GlcNAc residues into Tau proteins and with diminished Tau transfer into the nucleus. Hence, this paper establishes a direct relationship between O-GlcNAc glycosylation, phosphorylation and cellular localization of Tau proteins.
Collapse
Affiliation(s)
- Tony Lefebvre
- Laboratoire de Chimie Biologique, Unité Mixte de Recherches 8576 du CNRS, Université des Sciences et Technologies de Lille I, F-59655 Villeneuve d'Ascq, France
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Anderson AB, Ciriacks CM, Fuller KM, Arriaga EA. Distribution of zeptomole-abundant doxorubicin metabolites in subcellular fractions by capillary electrophoresis with laser-induced fluorescence detection. Anal Chem 2003; 75:8-15. [PMID: 12530812 DOI: 10.1021/ac020426r] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Doxorubicin (DOX) treatment of NS-1 mouse hybridoma cells results in the formation of zeptomole amounts of metabolites per cell that are difficult to determine by confocal microscopy or HPLC. The native fluorescence of DOX and its metabolites together with laser-induced fluorescence detection (HF) has previously been used to detect a maximum of four components. In this study, we use capillary electrophoresis with postcolumn LIF (CE-LIF) to separate and detect 12 components attributed to DOX metabolism, resulting from treatment of NS-1 cells with 25 microM DOX for 8 h. The so-called metabolites 8 and 10 have been identified as doxorubicinone (DOXone) and 7-deoxydoxorubicinone (7-deoxyDOXone), respectively, by comigration with the corresponding synthetic standard. Due to comigration of DOX with doxorubicinol (DOXone), the presence of DOXone had to be determined separately by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. The rest of the metabolites remain unidentified and are referred to by their number assignment. In comparison with the whole cell lysate, fractionation by differential centrifugation results in a better separation resolution of metabolites due to reduced amounts of metabolites in each fraction. This approach was chosen to compare the distribution of 13 metabolites in three subcellular fractions that form a pellet at < 1,400 g, 1,400-14,000 g, and > 14, 000 g and that generically are enriched in nuclei, organelles (mitochondria and lysosomes), and cytosolic components, respectively. The most abundant metabolite, DOXone, was estimated to be 90 +/- 15, 18 +/- 2, and 60 +/- 12 amol/cell (n = 5) in the nuclear-enriched, organelle-enriched, and cytosole-enriched fractions, respectively. In contrast, the total amount of other metabolites in a given fraction varied from 0 to 1,300 zmol. 7-DeoxyDOXone is the only metabolite that was present at similar levels in the three fractions. Other salient observations are metabolites 3, 7, and 11 are not detectable in the nuclear-enriched, organelle-enriched, and cytosole-enriched fractions, respectively; metabolite 9 and DOXone are more abundant in the nuclear-enriched fraction than in the other two fractions. The observations presented here suggest that subcellular fractionation followed by CE-LIF could be a powerful diagnostic for monitoring drug distribution, which is highly relevant to DOX cytoxicity studies.
Collapse
Affiliation(s)
- Adrian B Anderson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
23
|
Khoo SH, Hoggard PG, Williams I, Meaden ER, Newton P, Wilkins EG, Smith A, Tjia JF, Lloyd J, Jones K, Beeching N, Carey P, Peters B, Back DJ. Intracellular accumulation of human immunodeficiency virus protease inhibitors. Antimicrob Agents Chemother 2002; 46:3228-35. [PMID: 12234849 PMCID: PMC128776 DOI: 10.1128/aac.46.10.3228-3235.2002] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Intracellular accumulation of the protease inhibitors (PIs) saquinavir (SQV), ritonavir (RTV), and indinavir (IDV) was determined in 50 human immunodeficiency virus-positive patients. Following extraction, PIs were quantified by mass spectrometry. Paired plasma and intracellular samples were collected over a full dosing interval from patients (13 on SQV, 6 on RTV, 8 on IDV, 16 on SQV plus RTV, 7 on IDV plus RTV) with a plasma viral load of <400 copies/ml. Data were expressed as intracellular/plasma drug concentration ratios. A hierarchy of intracellular accumulation was demonstrated by the following medians: 9.45 for SQV > 1.00 for RTV > 0.51 for IDV. Coadministration of RTV did not boost ratios of SQV or IDV within the cell or in plasma, although absolute plasma and intracellular SQV concentrations were increased by RTV. Seven individuals receiving SQV in hard-gel capsule form (median, 32 months) had higher intracellular/plasma drug ratios than all other patients receiving SQV (median, 17.62 versus 4.83; P = 0.04), despite consistently low plasma SQV concentrations. How this occurs may provide insight into the mechanisms that limit adequate drug penetration into sanctuary sites.
Collapse
Affiliation(s)
- Saye H. Khoo
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Patrick G. Hoggard
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
- Corresponding author. Mailing address: Department of Pharmacology & Therapeutics, University of Liverpool, Block H, First Floor, 70 Pembroke Pl., Liverpool L69 3GF, United Kingdom. Phone: 44 151 794 5919. Fax: 44 151 794 5656. E-mail:
| | - Ian Williams
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - E. Rhiannon Meaden
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Philippa Newton
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Edmund G. Wilkins
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Alan Smith
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - John F. Tjia
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Judy Lloyd
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Kevin Jones
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Nick Beeching
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Peter Carey
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - Barry Peters
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| | - David J. Back
- Department of Pharmacology & Therapeutics, University of Liverpool, Department of Infectious Diseases, University Hospital Aintree, Department of Genitourinary Medicine, Royal Liverpool University Hospital, Liverpool, Department of Sexually Transmitted Diseases, Royal Free and University College Medical School, University College, Department of Genitourinary Medicine, St. Thomas' Hospital, London, Department of Infectious Diseases, North Manchester General Hospital, Manchester, United Kingdom
| |
Collapse
|
24
|
Boehning D, Joseph SK. Direct association of ligand-binding and pore domains in homo- and heterotetrameric inositol 1,4,5-trisphosphate receptors. EMBO J 2000; 19:5450-9. [PMID: 11032812 PMCID: PMC313997 DOI: 10.1093/emboj/19.20.5450] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP(3)Rs) are a family of intracellular Ca(2+) channels that exist as homo- or heterotetramers. In order to determine whether the N-terminal ligand-binding domain is in close physical proximity to the C-terminal pore domain, we prepared microsomal membranes from COS-7 cells expressing recombinant type I and type III IP(3)R isoforms. Trypsin digestion followed by cross-linking and co-immunoprecipitation of peptide fragments suggested an inter-subunit N- and C-terminal interaction in both homo- and heterotetramers. This observation was further supported by the ability of in vitro translated C-terminal peptides to interact specifically with an N-terminal fusion protein. Using a (45)Ca(2+) flux assay, we provide functional evidence that the ligand-binding domain of one subunit can gate the pore domain of an adjacent subunit. We conclude that common structural motifs are shared between the type I and type III IP(3)Rs and propose that the gating mechanism of IP(3)R Ca(2+) channels involves the association of the N-terminus of one subunit with the C-terminus of an adjacent subunit in both homo- and heterotetrameric complexes.
Collapse
MESH Headings
- Animals
- COS Cells
- Calcium/metabolism
- Calcium Channels/chemistry
- Calcium Channels/classification
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling
- Cross-Linking Reagents/metabolism
- Inositol 1,4,5-Trisphosphate/pharmacology
- Inositol 1,4,5-Trisphosphate Receptors
- Ion Channel Gating/drug effects
- Ligands
- Membrane Proteins/chemistry
- Membrane Proteins/metabolism
- Microsomes/metabolism
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Precipitin Tests
- Protein Binding
- Protein Isoforms
- Protein Structure, Quaternary/drug effects
- Protein Structure, Tertiary/drug effects
- Quaternary Ammonium Compounds/pharmacology
- Receptors, Cytoplasmic and Nuclear/chemistry
- Receptors, Cytoplasmic and Nuclear/classification
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/metabolism
- Succinimides/metabolism
- Trypsin/metabolism
Collapse
Affiliation(s)
- D Boehning
- Department of Pathology and Cell Biology, Thomas Jefferson University School of Medicine, Philadelphia, PA 19107, USA
| | | |
Collapse
|
25
|
Compromised glutamate transport in human glioma cells: reduction-mislocalization of sodium-dependent glutamate transporters and enhanced activity of cystine-glutamate exchange. J Neurosci 2000. [PMID: 10594060 DOI: 10.1523/jneurosci.19-24-10767.1999] [Citation(s) in RCA: 250] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Elevated levels of extracellular glutamate ([Glu](o)) can induce seizures and cause excitotoxic neuronal cell death. This is normally prevented by astrocytic glutamate uptake. Neoplastic transformation of human astrocytes causes malignant gliomas, which are often associated with seizures and neuronal necrosis. Here, we show that Na(+)-dependent glutamate uptake in glioma cell lines derived from human tumors (STTG-1, D-54MG, D-65MG, U-373MG, U-251MG, U-138MG, and CH-235MG) is up to 100-fold lower than in astrocytes. Immunohistochemistry and subcellular fractionation show very low expression levels of the astrocytic glutamate transporter GLT-1 but normal expression levels of another glial glutamate transporter, GLAST. However, in glioma cells, essentially all GLAST protein was found in cell nuclei rather than the plasma membrane. Similarly, brain tissues from glioblastoma patients also display reduction of GLT-1 and mislocalization of GLAST. In glioma cell lines, over 50% of glutamate transport was Na(+)-independent and mediated by a cystine-glutamate exchanger (system x(c)(-)). Extracellular L-cystine dose-dependently induced glutamate release from glioma cells. Glutamate release was enhanced by extracellular glutamine and inhibited by (S)-4-carboxyphenylglycine, which blocked cystine-glutamate exchange. These data suggest that the unusual release of glutamate from glioma cells is caused by reduction-mislocalization of Na(+)-dependent glutamate transporters in conjunction with upregulation of cystine-glutamate exchange. The resulting glutamate release from glioma cells may contribute to tumor-associated necrosis and possibly to seizures in peritumoral brain tissue.
Collapse
|
26
|
Lefebvre T, Alonso C, Mahboub S, Dupire MJ, Zanetta JP, Caillet-Boudin ML, Michalski JC. Effect of okadaic acid on O-linked N-acetylglucosamine levels in a neuroblastoma cell line. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1472:71-81. [PMID: 10572927 DOI: 10.1016/s0304-4165(99)00105-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
O-Linked N-Acetylglucosamine (O-GlcNAc) is a major form of post-translational modification found in nuclear and cytoplasmic proteins. Several authors have advanced the hypothesis according to which phosphorylation and O-GlcNAc glycosylation are reciprocally related to one another [1,2]. In order to test this hypothesis we have investigated the effect of a broad spectrum phosphatase inhibitor, okadaic acid (OA), generally used to induce protein hyperphosphorylation, on the GlcNAc content of cellular glycoproteins. We demonstrate that in neuronal cells lines OA decreases the level of O-GlcNAc in both nuclear and cytoplasmic proteins with a greater effect in the nuclear fraction. This phenomenon was demonstrated by the use of three different procedures for the detection of O-GlcNAc in conjunction with a systematic treatment with PNGase F. O-Linked GlcNAc was characterized using respectively lectin staining with WGA, galactosyltransferase labeling and metabolic labeling of cultured cells with [3H]glucosamine. Although the effects on individual proteins varied, a less pronounced effect was observed on HeLa or COS cell total homogenates. When Kelly cells were treated with OA, the major observation was a decrease in O-GlcNAc content of nuclear proteins. The measurement of the UDP-GlcNAc level clearly demonstrates that the decrease on the O-GlcNAc level in the neuroblastoma cell line after treatment with okadaic acid is not a consequence of the modification of the UDP-GlcNAc pool.
Collapse
Affiliation(s)
- T Lefebvre
- Unité Mixte de Recherches 8576 du CNRS, Laboratoire de Chimie Biologique, Villeneuve d'Ascq, France
| | | | | | | | | | | | | |
Collapse
|