Skip to main content

Rapid preparation of nuclei-depleted detergent-resistant membrane fractions suitable for proteomics analysis

Abstract

Background

Cholesterol-rich membrane microdomains known as lipid rafts have been implicated in diverse physiologic processes including lipid transport and signal transduction. Lipid rafts were originally defined as detergent-resistant membranes (DRMs) due to their relative insolubility in cold non-ionic detergents. Recent findings suggest that, although DRMs are not equivalent to lipid rafts, the presence of a given protein within DRMs strongly suggests its potential for raft association in vivo. Therefore, isolation of DRMs represents a useful starting point for biochemical analysis of lipid rafts. The physicochemical properties of DRMs present unique challenges to analysis of their protein composition. Existing methods of isolating DRM-enriched fractions involve flotation of cell extracts in a sucrose density gradient, which, although successful, can be labor intensive, time consuming and results in dilute sucrose-containing fractions with limited utility for direct proteomic analysis. In addition, several studies describing the proteomic characterization of DRMs using this and other approaches have reported the presence of nuclear proteins in such fractions. It is unclear whether these results reflect trafficking of nuclear proteins to DRMs or whether they arise from nuclear contamination during isolation. To address these issues, we have modified a published differential detergent extraction method to enable rapid DRM isolation that minimizes nuclear contamination and yields fractions compatible with mass spectrometry.

Results

DRM-enriched fractions isolated using the conventional or modified extraction methods displayed comparable profiles of known DRM-associated proteins, including flotillins, GPI-anchored proteins and heterotrimeric G-protein subunits. Thus, the modified procedure yielded fractions consistent with those isolated by existing methods. However, we observed a marked reduction in the percentage of nuclear proteins identified in DRM fractions isolated with the modified method (15%) compared to DRMs isolated by conventional means (36%). Furthermore, of the 21 nuclear proteins identified exclusively in modified DRM fractions, 16 have been reported to exist in other subcellular sites, with evidence to suggest shuttling of these species between the nucleus and other organelles.

Conclusion

We describe a modified DRM isolation procedure that generates DRMs that are largely free of nuclear contamination and that is compatible with downstream proteomic analyses with minimal additional processing. Our findings also imply that identification of nuclear proteins in DRMs is likely to reflect legitimate movement of proteins between compartments, and is not a result of contamination during extraction.

Background

Lipid rafts and caveolae, membrane microdomains that are enriched in cholesterol and sphingolipids, have been implicated in diverse physiologic mechanisms, such as signal transduction, trafficking and lipid transport [1, 2]. The invaginated and vesicular structures known as caveolae are a type of microdomain that harbor caveolin family proteins; however, non-caveolar 'flat' lipid rafts have also been shown to exist in cells that do not express caveolins [3]. Lipid rafts were originally defined as detergent-resistant membranes (DRMs) because of their relative insolubility in cold non-ionic detergents [4]. However, equating DRMs with lipid rafts is now believed to be inaccurate due to controversy over the biophysical and biochemical nature of rafts, and whether they even exist in vivo [3, 5–7]. In addressing these concerns, several recent papers have proposed a revision of the raft hypothesis. These studies suggest that, although DRMs do not correspond to lipid rafts as they exist in vivo, the presence of a given protein within a DRM, and its loss from that fraction when DRMs are perturbed by cholesterol depletion, for example, strongly suggests the potential for raft association of that protein in vivo [8–10]. In addition, it has been noted in a recent publication that, despite its limitations, isolation of DRMs is the only biochemical approach for assessing protein interactions with rafts [10].

DRMs are thought to be diverse in protein content and in functional roles. However, these membrane domains are still poorly understood and are currently being characterized using proteomics tools [11]. The cell signaling function of DRMs is thought to arise from the ability of these microdomains to selectively retain or exclude specific proteins, resulting in the formation of multiprotein complexes that process biochemical information across specific signaling axes. Because cholesterol-rich microdomains may be functionally altered by pathophysiologic alterations in lipid metabolism [12], systematic analysis of proteins present in DRMs is likely to provide novel insights into multiple signaling mechanisms that operate in the normal state and in disease.

The biophysical and biochemical properties of DRMs present unique challenges to studies of their protein composition. Membrane proteins possess hydrophobic regions that render them poorly soluble during extraction [13, 14]. In addition, DRMs represent a minor and transient component of the total membrane surface. The classical method of isolating DRM-enriched fractions is by flotation of cell extracts, prepared in cold detergents such as Triton X-100 or under detergent-free conditions, in a sucrose density gradient [15]. This approach exploits the detergent insolubility of DRMs at low temperatures as well as their light buoyant density. However, gradient centrifugation procedures are time-consuming and labor-intensive, requiring processing times on the order of 24 h. Moreover, the resultant dilute, sucrose-containing fractions have limited utility for proteomic analysis without additional processing steps [16].

We have employed a DRM isolation technique originally described by Solomon et al. [17, 18] that exploits the differential solubility of detergent-resistant microdomains in cold, non-ionic detergents [19–24]. This approach is rapid and yields fractions that are comparable to those isolated by flotation on density gradients. We have validated our observations from the detergent extraction procedure using alternative approaches, including sucrose density ultracentrifugation and immunofluorescence imaging [21, 23, 24]. Both unpublished and published data from us and others revealed the presence of nuclear proteins in DRM fractions isolated either by flotation in density gradients or by differential detergent solubility [16, 25–30](N.K.M. and R.M.A, unpublished observations). However, whether this unanticipated finding arose from intracellular trafficking of proteins between nuclei and DRMs, or, alternatively, was a reflection of nuclear contamination during extraction has not been determined. In this study, we describe a modification of the existing differential detergent extraction procedure that minimizes nuclear contamination of DRMs. Proteomic analysis of the resulting fractions suggests that the presence of 'nuclear' proteins in DRMs is likely to result from shuttling between the nucleus and other subcellular sites.

Results and discussion

To investigate whether the presence of nuclear proteins in DRMs was an artifact or instead reflected physiologic trafficking events, we modified the extraction procedure described by Solomon et al. by pelleting out nuclei and intact cells from detergent-free homogenates prior to detergent solubilization. We reasoned that this would reduce potential contamination from nuclear material. As a model system, we have used caveolin-negative LNCaP prostate cancer cells stably expressing myristoylated Akt1 (MyrAkt1)[24]. MyrAkt1 is a robust marker of DRMs in these cells based on (i) its presence in light buoyant density fractions following sucrose gradient centrifugation; (ii) its enrichment in DRMs following differential detergent extraction; and (iii) its accumulation at the plasma membrane based on localization to membranes that stain with CTxB, a marker of the DRM/lipid raft-restricted ganglioside GM1 (Figure 1A)[31].

Figure 1
figure 1

Modification of the detergent extraction procedure eliminates 'nuclear' proteins from detergent-resistant membranes. (A) Cholesterol- and sphingolipid-enriched membranes in LNCaP/MyrAkt1 cells were stained with 0.5 μg/mL Alexa 594-CTxB for 10 min prior to staining with anti-S473-P Akt (1:100) and FITC-conjugated secondary Ab (1:100). Nuclei were counterstained with DAPI prior to imaging. Original magnification, 63×. LNCaP cells stably expressing LacZ or MyrAkt1 were extracted using the conventional (1) or modified (2) detergent extraction procedure. Equal amounts (30 μg for detection of SAFB; 10 μg for other target proteins) of Triton-soluble (TS), Triton-insoluble, octylglucoside-soluble (TI) or nuclear (N) fractions were resolved by SDS-PAGE, transferred to nitrocellulose membranes and (B) stained with Ponceau S or (C) blotted with antibodies to the HA epitope tag, phospho-S473 Akt, PCNA, SAFB, hnRNP K and Giα2.

To determine the effect of pelleting nuclei and/or intact cells on the overall protein profiles of isolated fractions, we initially compared the conventional extraction method with the modified procedure. As starting material for the extraction, we generated membrane preparations from LNCaP/MyrHA-Akt1 or LNCaP/LacZ cells in log-phase growth by mechanical disruption of cells under detergent-free conditions, as described in Methods. In the conventional method, homogenized samples were centrifuged at 16,000 × g in a Beckman microcentrifuge for 10 min at 4°C. In the modified approach, samples were centrifuged at 500 × g for 5 min at 4°C to pellet intact cells and nuclei; importantly, no brake was applied during the deceleration of the centrifuge. The supernatant was decanted carefully and centrifuged at 16,000 × g in a Beckman microcentrifuge for 10 min at 4°C to pellet membranes. The pellet remaining from the low-speed spin, comprising predominantly nuclei, was lysed in 50 mM Tris-HCl pH 8.0, 150 mM NaCl, 1% Igepal CA-630, 0.5% sodium deoxycholate and 0.1% SDS plus protease and phosphatase inhibitors. In each case (conventional or modified), pellets from the high-speed centrifugation steps were then subjected to successive detergent extraction in Triton X-100 and octylglucoside to yield Triton-soluble (TS) and Triton-insoluble/octylglucoside-soluble (TI) fractions, as described in Methods.

Following determination of the protein concentration of fractions by MicroBCA assay, equal amounts of protein from each fraction were resolved by SDS-PAGE and transferred to nitrocellulose membranes. To verify the presence of protein in each fraction, membranes were stained with 0.1% (w/v) Ponceau S in 5% acetic acid (v/v) prior to blotting with the indicated antibodies. The staining pattern revealed both quantitative and qualitative differences in the overall protein profiles of TS and TI fractions generated by the two methods (Figure 1B). We confirmed the distribution of MyrAkt1 in TS, TI and nuclear (N) fractions by HA immunoblot, and observed robust enrichment of MyrAkt1 in TI fractions in both cases (Figure 1C). A small amount of MyrAkt1 was detected in the nuclear pellet from the low speed centrifugation, consistent with previous reports [32]. To determine the impact of the modified extraction procedure on protein distribution, we probed the membranes for selected proteins with defined subcellular localizations, including proliferating cell nuclear antigen (PCNA), a nuclear protein [33] and the small G-protein, Giα2, that resides in lipid rafts [34, 35]. As shown in Figure 1C, PCNA, although present in the TI fraction following the conventional extraction method, was undetectable in TI fractions prepared with the modified approach. As anticipated, the nuclear fraction showed robust signal corresponding to PCNA. In contrast, Giα2 was present in TI fractions generated with either extraction method, consistent with its localization to DRMs. These data were reproducible across three independent biological replicates and similar findings were obtained with fractions isolated from LNCaP cells stably expressing the irrelevant gene (LacZ). We also probed for two additional proteins that were identified as putative raft-resident binding partners of MyrAkt1 transiently expressed in HEK293 cells (N.K.M. and R.M.A, unpublished observations): scaffold attachment factor B (SAFB), a protein implicated in transcriptional regulation and chromatin organization [36]; and heterogeneous ribonucleoprotein K (hnRNP K), a pleiotropic nucleic acid binding protein that regulates gene and protein expression [37, 38]. Although SAFB was present in TI fractions generated by conventional means, it was absent from this material following elimination of nuclei, strongly suggesting that its presence in the TI fraction arose from nuclear contamination. In contrast, hnRNP K was detected in TS and TI fractions with both extraction methods, as well as in nuclei. These observations are consistent with localization of hnRNP K to multiple distinct subcellular sites [37].

To extend our analysis beyond characterization of specific proteins and to determine the potential utility of our modified fractionation procedure for proteomic applications, we used LC/MS as a read-out for the proteome compositions of TI fractions generated using either the conventional or modified methods. The TI fractions from the two extraction methods were subjected to in-solution tryptic digestion and mass spectrometry analysis, as described in Methods. According to the criteria outlined in Methods, 388 and 371 proteins were identified from the TI fractions isolated using the conventional and modified methods, respectively [see Additional file 1]. This proteomic analysis was not meant to be exhaustive. Instead we used the most confident protein identifications to a) ensure that the lipid raft isolation was successful and b) estimate the fraction of nuclear proteins. Common proteins identified in the TI fractions from both extraction methods included a number of proteins previously reported to be associated with DRMs, lipid rafts and/or caveolae (Table 1), such as flotillins [16, 25, 29, 39, 40], heterotrimeric G-protein subunits [25], components of the vacuolar ATP synthase protein family [16, 25, 29, 41] and others [29, 42–44]. Thus, the modified fractionation procedure did not alter the predicted composition of DRM fractions significantly and yielded fractions consistent with those isolated by density gradient-based methods.

Table 1 Proteins identified by tandem mass spectrometry.

To assess the impact of the modified extraction procedure on the protein profiles obtained from each fraction, we classified proteins according to subcellular localization. In particular, we focused on whether proteins were nuclear or non-nuclear in nature. Datasets were annotated using Gene Ontology Cellular Component (GO-CC) terms with the cross-reference converter [45]. As shown in Figure 2A, we observed a striking difference in the number of nuclear proteins differentially identified with the two approaches. Specifically, the modified extraction procedure led to identification of many fewer nuclear proteins, with only 54 out of 371 proteins (15%) classified as nuclear. This was in marked contrast to the conventional method in which 141 out of 388 proteins (36%) were categorized as nuclear. At the same time, the number of proteins annotated as non-nuclear increased from 195 (50%) to 258 (70%). The Venn diagrams in Figure 2B clearly show that the decrease of nuclear proteins in the modified protocol is concomitant to the increase in non-nuclear proteins. These findings are in agreement with our findings by immunoblot analysis that the nuclear proteins PCNA and SAFB are no longer detectable in TI fractions generated using the modified fractionation technique.

Figure 2
figure 2

Quantitative analysis of proteins in detergent-resistant membrane fractions analyzed by mass spectrometry. (A) Proteins in DRM (TI) fractions generated using the conventional or modified methods were analyzed by tandem mass spectrometry and classified as either nuclear, non-nuclear or unannotated based on Gene Ontology cellular component terms. The pie-charts demonstrate the marked reduction in proteins identified as 'nuclear' with the modified extraction procedure. (B) Venn diagrams illustrating the distinct and overlapping protein compositions of TI fractions isolated by each method.

Interestingly, of the 21 proteins annotated as nuclear in localization, that were found exclusively in the modified TI fraction, evidence from the literature and protein databases suggests that 16 of these are also present in additional subcellular locations, and may shuttle between the nucleus and other sites in the cell [46–58]. Thus, only 5 proteins (shaded in Table 1B), out of a total of 156 specifically identified in the modified TI fraction in this experiment, were present exclusively in nuclei. In addition, at least 60% of the 33 nuclear proteins identified with both approaches are also present in other subcellular locations. In contrast, only about 30% of the 108 nuclear proteins specifically identified in the conventional TI fraction are annotated to be present in additional locations.

These findings strongly suggest that the modified DRM extraction method generates fractions that are largely free of nuclear contamination. They also imply that, given the high frequency with which so-called 'nuclear proteins' are found in other organelles, due to shuttling between compartments, nuclear proteins are likely to be detected by most extraction procedures, even when performed under the most stringent conditions. Canonical nuclear proteins, such as nuclear hormone receptors, can shuttle between the cytoplasm and nuclear compartments as part of their hormone-dependent functions. Recently, the androgen receptor was found to process androgenic signals non-genomically by forming a complex with Akt1 in lipid raft microdomains [59]. The identification of such proteins in DRMs may thus reflect a legitimate physiologic process and not an experimental artifact.

Conclusion

In summary, we describe a rapid, robust method for isolation of detergent-resistant membrane fractions that are compatible with downstream proteomic analyses with minimal additional processing. The present approach allows for a significant reduction of nuclear protein contamination of DRM samples. We believe the results presented here indicate that the DRM isolation procedure we describe can be applied with confidence to settings where a robust characterization of DRM constituents by mass spectrometry is desired, and also to more traditional biochemical methods involving antibody-based analyses.

Methods

Membrane preparations and isolation of detergent resistant membrane fractions

Populations of LNCaP/MyrHA-Akt1 or LNCaP/LacZ cells in log-phase growth were rinsed twice with phosphate-buffered saline (PBS), resuspended in Buffer M [50 mM HEPES pH 7.4, 10 mM NaCl, 5 mM MgCl2, 0.1 mM EDTA plus protease inhibitor cocktail (Complete Mini, Roche Applied Science), 1 mM Na3VO4, 1 mM NaF and 1 mM Na4P2O7.10 dH2O] and homogenized at 4°C using a Potter-Elvehjem tissue grinder (12 strokes, 1800 rpm). 107-108 cells were used for each extraction method. High-speed (16,000 × g) pellets from either the conventional or modified approach were subjected to successive detergent extraction essentially as described [19, 21, 24]. Briefly, pellets were resuspended in Buffer A [25 mM 2-(N-morpholino)-ethanesulfonic acid, 150 mM NaCl, pH 6.5] and samples combined with an equal volume of Buffer A containing 2% Triton X-100, and phosphatase and protease inhibitors. Samples were incubated on ice for 60 min, centrifuged at 16,000 × g for 20 min at 4°C and supernatants collected as Triton-soluble (TS) material. Pellets were rinsed briefly with Buffer A and resuspended in Buffer B [10 mM Tris-Cl, pH 7.6, 150 mM NaCl, 60 mM β-octylglucoside and phosphatase and protease inhibitors]. Samples were incubated on ice for 30 min, centrifuged at 16,000 × g for 20 min at 4°C and supernatants collected as Triton-insoluble (TI) material.

Tryptic Digestion and Mass Spectrometry Analysis

Four micrograms of each TI fraction was subjected to in-solution tryptic digestion. Three MS experiments were performed: TI fraction from the conventional extraction procedure; TI fraction from the modified extraction procedure; and extraction buffer control with a single replicate for each. Approximately 2 μL protein extracts or extraction buffer as control, were mixed with 8 μL 0.2% RapiGest™ SF (Waters Corporation, Milford, MA) in 50 mM ammonium bicarbonate, boiled for 5 min and cooled to room temperature. Dithiothreitol was added to 5 mM final concentration, samples heated at 60°C for 30 min and cooled to room temperature. Iodoacetamide was added to 15 mM final concentration and samples were incubated protected from light for 30 min prior to addition of 100 ng MS-grade trypsin and further incubation at 37°C for 2 h. To hydrolyze the RapiGest™ surfactant, trifluoroacetic acid was added to a final concentration of 0.5% and samples were incubated at 37°C for 45 min. Following centrifugation at 14,000 × g for 10 min, the supernatants were transferred to screw-top vials. Acetonitrile and acetic acid were added to a final concentration of 7.5% and 1.5%, respectively. Samples were analyzed by online C18 nanoflow reversed-phase HPLC (Eksigent nanoLC·2D™) hyphenated to a Thermo Scientific LTQ Orbitrap mass spectrometer. The digest was loaded onto a 100 μm i.d. × 15 cm C18 column and the peptides were separated at 200 nL/min with 80 min gradients from 5 to 31.5% acetonitrile in 0.4% formic acid. Survey spectra were acquired in the orbitrap with the resolution set to a value of 30,000. Up to five of the most intense ions per cycle were fragmented and analyzed in the linear trap.

Database searching was performed using the Mascot search engine (Matrix Science, v.2.1). All MS datasets were searched against the International Protein Index (IPI) human protein database of the International Protein Index (v3.10; 57479 sequences). Protein modifications were selected as carbamidomethyl (C) (fixed) and oxidation (M) (variable). Up to one missed cleavage was allowed. The mass tolerance was set as ± 20 ppm for MS spectra and ± 0.5 Da for MS/MS spectra. The following criteria were used to generate a high-confidence data set: at least two peptides must be identified for each protein, with a score not less than 33, corresponding to a significance level of p < 0.01 as determined by reverse database searching. Known contaminants (e.g. keratins and trypsin) were removed from the protein lists. Applying these parameters, no false positives were identified through searching the reversed IPI_Human database.

References

  1. Simons K, Toomre D: Lipid rafts and signal transduction. Nat Rev Mol Cell Biol. 2000, 1 (1): 31-39. 10.1038/35036052.

    Article  CAS  PubMed  Google Scholar 

  2. Rajendran L, Simons K: Lipid rafts and membrane dynamics. J Cell Sci. 2005, 118 (Pt 6): 1099-1102. 10.1242/jcs.01681.

    Article  CAS  PubMed  Google Scholar 

  3. Hancock JF: Lipid rafts: contentious only from simplistic standpoints. Nat Rev Mol Cell Biol. 2006, 7 (6): 456-462. 10.1038/nrm1925.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Brown DA, Rose JK: Sorting of GPI-anchored proteins to glycolipid-enriched membrane subdomains during transport to the apical cell surface. Cell. 1992, 68 (3): 533-544. 10.1016/0092-8674(92)90189-J.

    Article  CAS  PubMed  Google Scholar 

  5. Munro S: Lipid rafts: elusive or illusive?. Cell. 2003, 115 (4): 377-388. 10.1016/S0092-8674(03)00882-1.

    Article  CAS  PubMed  Google Scholar 

  6. Lai EC: Lipid rafts make for slippery platforms. J Cell Biol. 2003, 162 (3): 365-370. 10.1083/jcb.200307087.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  7. Edidin M: The state of lipid rafts: from model membranes to cells. Annu Rev Biophys Biomol Struct. 2003, 32: 257-283. 10.1146/annurev.biophys.32.110601.142439.

    Article  CAS  PubMed  Google Scholar 

  8. Brown DA: Lipid rafts, detergent-resistant membranes, and raft targeting signals. Physiology (Bethesda). 2006, 21: 430-439.

    Article  CAS  Google Scholar 

  9. Brown DA: Analysis of raft affinity of membrane proteins by detergent-insolubility. Methods Mol Biol. 2007, 398: 9-20.

    Article  CAS  PubMed  Google Scholar 

  10. Lingwood D, Simons K: Detergent resistance as a tool in membrane research. Nat Protoc. 2007, 2 (9): 2159-2165. 10.1038/nprot.2007.294.

    Article  CAS  PubMed  Google Scholar 

  11. Raimondo F, Ceppi P, Guidi K, Masserini M, Foletti C, Pitto M: Proteomics of plasma membrane microdomains. Expert Rev Proteomics. 2005, 2 (5): 793-807. 10.1586/14789450.2.5.793.

    Article  CAS  PubMed  Google Scholar 

  12. Li YC, Park MJ, Ye SK, Kim CW, Kim YN: Elevated levels of cholesterol-rich lipid rafts in cancer cells are correlated with apoptosis sensitivity induced by cholesterol-depleting agents. Am J Pathol. 2006, 168 (4): 1107-18; quiz 1404-5. 10.2353/ajpath.2006.050959.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Magee AI, Parmryd I: Detergent-resistant membranes and the protein composition of lipid rafts. Genome Biol. 2003, 4 (11): 234-10.1186/gb-2003-4-11-234.

    Article  PubMed Central  PubMed  Google Scholar 

  14. Wu CC, Yates JR: The application of mass spectrometry to membrane proteomics. Nat Biotechnol. 2003, 21 (3): 262-267. 10.1038/nbt0303-262.

    Article  CAS  PubMed  Google Scholar 

  15. Parkin ET, Turner AJ, Hooper NM: Amyloid precursor protein, although partially detergent-insoluble in mouse cerebral cortex, behaves as an atypical lipid raft protein. Biochem J. 1999, 344 Pt 1: 23-30. 10.1042/0264-6021:3440023.

    Article  CAS  PubMed  Google Scholar 

  16. Blonder J, Hale ML, Lucas DA, Schaefer CF, Yu LR, Conrads TP, Issaq HJ, Stiles BG, Veenstra TD: Proteomic analysis of detergent-resistant membrane rafts. Electrophoresis. 2004, 25 (9): 1307-1318. 10.1002/elps.200405891.

    Article  CAS  PubMed  Google Scholar 

  17. Solomon KR, Mallory MA, Finberg RW: Determination of the non-ionic detergent insolubility and phosphoprotein associations of glycosylphosphatidylinositol-anchored proteins expressed on T cells. Biochem J. 1998, 334 ( Pt 2): 325-333.

    Article  CAS  Google Scholar 

  18. Solomon KR, Mallory MA, Hanify KA, Finberg RW: The nature of membrane anchorage determines kinase association and detergent solubility of CD4. Biochem Biophys Res Commun. 1998, 242 (2): 423-428. 10.1006/bbrc.1997.7983.

    Article  CAS  PubMed  Google Scholar 

  19. Zhuang L, Lin J, Lu ML, Solomon KR, Freeman MR: Cholesterol-rich lipid rafts mediate akt-regulated survival in prostate cancer cells. Cancer Res. 2002, 62 (8): 2227-2231.

    CAS  PubMed  Google Scholar 

  20. Stehr M, Adam RM, Khoury J, Zhuang L, Solomon KR, Peters CA, Freeman MR: Platelet derived growth factor-BB is a potent mitogen for rat ureteral and human bladder smooth muscle cells: dependence on lipid rafts for cell signaling. J Urol. 2003, 169 (3): 1165-1170. 10.1097/01.ju.0000041501.01323.b9.

    Article  CAS  PubMed  Google Scholar 

  21. Kim J, Adam RM, Solomon KR, Freeman MR: Involvement of cholesterol-rich lipid rafts in interleukin-6-induced neuroendocrine differentiation of LNCaP prostate cancer cells. Endocrinology. 2004, 145 (2): 613-619. 10.1210/en.2003-0772.

    Article  CAS  PubMed  Google Scholar 

  22. Stehr M, Estrada CR, Khoury J, Danciu TE, Sullivan MP, Peters CA, Solomon KR, Freeman MR, Adam RM: Caveolae are negative regulators of transforming growth factor-beta1 signaling in ureteral smooth muscle cells. J Urol. 2004, 172 (6 Pt 1): 2451-2455. 10.1097/01.ju.0000138084.53577.ca.

    Article  CAS  PubMed  Google Scholar 

  23. Zhuang L, Kim J, Adam RM, Solomon KR, Freeman MR: Cholesterol targeting alters lipid raft composition and cell survival in prostate cancer cells and xenografts. J Clin Invest. 2005, 115 (4): 959-968.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Adam RM, Mukhopadhyay NK, Kim J, Di Vizio D, Cinar B, Boucher K, Solomon KR, Freeman MR: Cholesterol sensitivity of endogenous and myristoylated Akt. Cancer Res. 2007, 67 (13): 6238-6246. 10.1158/0008-5472.CAN-07-0288.

    Article  CAS  PubMed  Google Scholar 

  25. Foster LJ, De Hoog CL, Mann M: Unbiased quantitative proteomics of lipid rafts reveals high specificity for signaling factors. Proc Natl Acad Sci U S A. 2003, 100 (10): 5813-5818. 10.1073/pnas.0631608100.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. MacLellan DL, Steen H, Adam RM, Garlick M, Zurakowski D, Gygi SP, Freeman MR, Solomon KR: A quantitative proteomic analysis of growth factor-induced compositional changes in lipid rafts of human smooth muscle cells. Proteomics. 2005, 5 (18): 4733-4742. 10.1002/pmic.200500044.

    Article  CAS  PubMed  Google Scholar 

  27. Karsan A, Blonder J, Law J, Yaquian E, Lucas DA, Conrads TP, Veenstra T: Proteomic analysis of lipid microdomains from lipopolysaccharide-activated human endothelial cells. J Proteome Res. 2005, 4 (2): 349-357. 10.1021/pr049824w.

    Article  CAS  PubMed  Google Scholar 

  28. Guo Y, Singleton PA, Rowshan A, Gucek M, Cole RN, Graham DR, Van Eyk JE, Garcia JG: Quantitative proteomics analysis of human endothelial cell membrane rafts: evidence of MARCKS and MRP regulation in the sphingosine 1-phosphate-induced barrier enhancement. Mol Cell Proteomics. 2007, 6 (4): 689-696. 10.1074/mcp.M600398-MCP200.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  29. Kowalski MP, Dubouix-Bourandy A, Bajmoczi M, Golan DE, Zaidi T, Coutinho-Sledge YS, Gygi MP, Gygi SP, Wiemer EA, Pier GB: Host resistance to lung infection mediated by major vault protein in epithelial cells. Science. 2007, 317 (5834): 130-132. 10.1126/science.1142311.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  30. Yanagida M, Nakayama H, Yoshizaki F, Fujimura T, Takamori K, Ogawa H, Iwabuchi K: Proteomic analysis of plasma membrane lipid rafts of HL-60 cells. Proteomics. 2007, 7 (14): 2398-2409. 10.1002/pmic.200700056.

    Article  CAS  PubMed  Google Scholar 

  31. Blank N, Schiller M, Krienke S, Wabnitz G, Ho AD, Lorenz HM: Cholera toxin binds to lipid rafts but has a limited specificity for ganglioside GM1. Immunol Cell Biol. 2007, 85 (5): 378-382. 10.1038/sj.icb.7100045.

    Article  CAS  PubMed  Google Scholar 

  32. Burgering BM, Coffer PJ: Protein kinase B (c-Akt) in phosphatidylinositol-3-OH kinase signal transduction. Nature. 1995, 376 (6541): 599-602. 10.1038/376599a0.

    Article  CAS  PubMed  Google Scholar 

  33. Kelman Z: PCNA: structure, functions and interactions. Oncogene. 1997, 14 (6): 629-640. 10.1038/sj.onc.1200886.

    Article  CAS  PubMed  Google Scholar 

  34. Wedegaertner PB, Wilson PT, Bourne HR: Lipid modifications of trimeric G proteins. J Biol Chem. 1995, 270 (2): 503-506. 10.1074/jbc.270.2.503.

    Article  CAS  PubMed  Google Scholar 

  35. Moffett S, Brown DA, Linder ME: Lipid-dependent targeting of G proteins into rafts. J Biol Chem. 2000, 275 (3): 2191-2198. 10.1074/jbc.275.3.2191.

    Article  CAS  PubMed  Google Scholar 

  36. Oesterreich S: Scaffold attachment factors SAFB1 and SAFB2: Innocent bystanders or critical players in breast tumorigenesis?. J Cell Biochem. 2003, 90 (4): 653-661. 10.1002/jcb.10685.

    Article  CAS  PubMed  Google Scholar 

  37. Bomsztyk K, Denisenko O, Ostrowski J: hnRNP K: one protein multiple processes. Bioessays. 2004, 26 (6): 629-638. 10.1002/bies.20048.

    Article  CAS  PubMed  Google Scholar 

  38. Mikula M, Dzwonek A, Karczmarski J, Rubel T, Dadlez M, Wyrwicz LS, Bomsztyk K, Ostrowski J: Landscape of the hnRNP K protein-protein interactome. Proteomics. 2006, 6 (8): 2395-2406. 10.1002/pmic.200500632.

    Article  CAS  PubMed  Google Scholar 

  39. Salzer U, Prohaska R: Stomatin, flotillin-1, and flotillin-2 are major integral proteins of erythrocyte lipid rafts. Blood. 2001, 97 (4): 1141-1143. 10.1182/blood.V97.4.1141.

    Article  CAS  PubMed  Google Scholar 

  40. Li N, Shaw AR, Zhang N, Mak A, Li L: Lipid raft proteomics: analysis of in-solution digest of sodium dodecyl sulfate-solubilized lipid raft proteins by liquid chromatography-matrix-assisted laser desorption/ionization tandem mass spectrometry. Proteomics. 2004, 4 (10): 3156-3166. 10.1002/pmic.200400832.

    Article  CAS  PubMed  Google Scholar 

  41. Bae TJ, Kim MS, Kim JW, Kim BW, Choo HJ, Lee JW, Kim KB, Lee CS, Kim JH, Chang SY, Kang CY, Lee SW, Ko YG: Lipid raft proteome reveals ATP synthase complex in the cell surface. Proteomics. 2004, 4 (11): 3536-3548. 10.1002/pmic.200400952.

    Article  CAS  PubMed  Google Scholar 

  42. McIntosh DP, Schnitzer JE: Caveolae require intact VAMP for targeted transport in vascular endothelium. Am J Physiol. 1999, 277 (6 Pt 2): H2222-32.

    CAS  PubMed  Google Scholar 

  43. Hooper NM: Detergent-insoluble glycosphingolipid/cholesterol-rich membrane domains, lipid rafts and caveolae (review). Mol Membr Biol. 1999, 16 (2): 145-156. 10.1080/096876899294607.

    Article  CAS  PubMed  Google Scholar 

  44. Bini L, Pacini S, Liberatori S, Valensin S, Pellegrini M, Raggiaschi R, Pallini V, Baldari CT: Extensive temporally regulated reorganization of the lipid raft proteome following T-cell antigen receptor triggering. Biochem J. 2003, 369 (Pt 2): 301-309. 10.1042/BJ20020503.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  45. Reference Database of Immune Cells. [http://refdic.rcai.riken.jp/tools/xrefconv.cgi]

  46. Chen M, Zsengeller Z, Xiao CY, Szabo C: Mitochondrial-to-nuclear translocation of apoptosis-inducing factor in cardiac myocytes during oxidant stress: potential role of poly(ADP-ribose) polymerase-1. Cardiovasc Res. 2004, 63 (4): 682-688. 10.1016/j.cardiores.2004.04.018.

    Article  CAS  PubMed  Google Scholar 

  47. Yoon JH, Singh P, Lee DH, Qiu J, Cai S, O'Connor TR, Chen Y, Shen B, Pfeifer GP: Characterization of the 3' --> 5' exonuclease activity found in human nucleoside diphosphate kinase 1 (NDK1) and several of its homologues. Biochemistry. 2005, 44 (48): 15774-15786. 10.1021/bi0515974.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  48. Tourriere H, Gallouzi IE, Chebli K, Capony JP, Mouaikel J, van der Geer P, Tazi J: RasGAP-associated endoribonuclease G3Bp: selective RNA degradation and phosphorylation-dependent localization. Mol Cell Biol. 2001, 21 (22): 7747-7760. 10.1128/MCB.21.22.7747-7760.2001.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Tamanini F, Bontekoe C, Bakker CE, van Unen L, Anar B, Willemsen R, Yoshida M, Galjaard H, Oostra BA, Hoogeveen AT: Different targets for the fragile X-related proteins revealed by their distinct nuclear localizations. Hum Mol Genet. 1999, 8 (5): 863-869. 10.1093/hmg/8.5.863.

    Article  CAS  PubMed  Google Scholar 

  50. Schmidt C, Lipsius E, Kruppa J: Nuclear and nucleolar targeting of human ribosomal protein S6. Mol Biol Cell. 1995, 6 (12): 1875-1885.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  51. Blackstone C, Roberts RG, Seeburg DP, Sheng M: Interaction of the deafness-dystonia protein DDP/TIMM8a with the signal transduction adaptor molecule STAM1. Biochem Biophys Res Commun. 2003, 305 (2): 345-352. 10.1016/S0006-291X(03)00767-8.

    Article  CAS  PubMed  Google Scholar 

  52. Xie B, Li H, Wang Q, Xie S, Rahmeh A, Dai W, Lee MY: Further characterization of human DNA polymerase delta interacting protein 38. J Biol Chem. 2005, 280 (23): 22375-22384. 10.1074/jbc.M414597200.

    Article  CAS  PubMed  Google Scholar 

  53. Yedavalli VS, Neuveut C, Chi YH, Kleiman L, Jeang KT: Requirement of DDX3 DEAD box RNA helicase for HIV-1 Rev-RRE export function. Cell. 2004, 119 (3): 381-392. 10.1016/j.cell.2004.09.029.

    Article  CAS  PubMed  Google Scholar 

  54. Soulet F, Al Saati T, Roga S, Amalric F, Bouche G: Fibroblast growth factor-2 interacts with free ribosomal protein S19. Biochem Biophys Res Commun. 2001, 289 (2): 591-596. 10.1006/bbrc.2001.5960.

    Article  CAS  PubMed  Google Scholar 

  55. Shim J, Lim H, J RY, Karin M: Nuclear export of NF90 is required for interleukin-2 mRNA stabilization. Mol Cell. 2002, 10 (6): 1331-1344. 10.1016/S1097-2765(02)00730-X.

    Article  CAS  PubMed  Google Scholar 

  56. Sutherland HG, Lam YW, Briers S, Lamond AI, Bickmore WA: 3D3/lyric: a novel transmembrane protein of the endoplasmic reticulum and nuclear envelope, which is also present in the nucleolus. Exp Cell Res. 2004, 294 (1): 94-105. 10.1016/j.yexcr.2003.11.020.

    Article  CAS  PubMed  Google Scholar 

  57. Lemos TA, Passos DO, Nery FC, Kobarg J: Characterization of a new family of proteins that interact with the C-terminal region of the chromatin-remodeling factor CHD-3. FEBS Lett. 2003, 533 (1-3): 14-20. 10.1016/S0014-5793(02)03737-7.

    Article  CAS  PubMed  Google Scholar 

  58. Weighardt F, Biamonti G, Riva S: Nucleo-cytoplasmic distribution of human hnRNP proteins: a search for the targeting domains in hnRNP A1. J Cell Sci. 1995, 108 ( Pt 2): 545-555.

    CAS  Google Scholar 

  59. Cinar B, Mukhopadhyay NK, Meng G, Freeman MR: Phosphoinositide 3-kinase-independent non-genomic signals transit from the androgen receptor to Akt1 in membrane raft microdomains. J Biol Chem. 2007, 282 (40): 29584-29593. 10.1074/jbc.M703310200.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Dr. Michael Freeman, Dr. Heidi Rayala and Dr. Keith Solomon for helpful discussions. This work was funded in part by NIH grant R21 DK66412 (to R.M.A.), the Children's Urological Foundation (to R.M.A.) and the American Italian Cancer Foundation (to D.D.V.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rosalyn M Adam.

Additional information

Authors' contributions

RMA conceived of the study, performed the fractionation and immunoblot analyses and drafted the manuscript. WY performed mass spectrometry and data analysis on DRM fractions. DDV performed immunofluorescence staining. NKM provided data on localization of hnRNP K and SAFB to detergent-resistant membrane and nuclear fractions. HS assisted with mass spectrometry analysis and data mining and helped to draft the manuscript. All authors have read and approved the final manuscript.

Electronic supplementary material

12860_2008_308_MOESM1_ESM.xls

Additional file 1: MS Data Summary. The table summarizes MS data obtained from DRM fractions isolated using either the conventional or modified extraction procedures. (XLS 552 KB)

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Adam, R.M., Yang, W., Di Vizio, D. et al. Rapid preparation of nuclei-depleted detergent-resistant membrane fractions suitable for proteomics analysis. BMC Cell Biol 9, 30 (2008). https://doi.org/10.1186/1471-2121-9-30

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2121-9-30

Keywords